Next Article in Journal
Effect of Insect Live Larvae as Environmental Enrichment on Poultry Gut Health: Gut Mucin Composition, Microbiota and Local Immune Response Evaluation
Next Article in Special Issue
Survival of Embryos and Fry of Sea Trout (Salmo trutta m. trutta) Growing from Eggs Exposed to Different Concentrations of Selenium during Egg Swelling
Previous Article in Journal
Elephant Endotheliotropic Herpesvirus Impact in the European Asian Elephant (Elephas maximus) Population: Are Hereditability and Zoo-Associated Factors Linked with Mortality?
Previous Article in Special Issue
Reproductive Potential of Stone Moroko (Pseudorasbora parva, Temminck et Schlegel, 1846) (Teleostei: Cypriniformes: Gobionidae) Inhabiting Central Europe
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review on Environmental Contaminants-Related Fertility Threat in Male Fishes: Effects and Possible Mechanisms of Action Learned from Wildlife and Laboratory Studies

by
Sayyed Mohammad Hadi Alavi
1,*,
Sepideh Barzegar-Fallah
1,
Parastoo Rahdar
1,
Mohammad Mahdi Ahmadi
1,
Mina Yavari
1,
Azadeh Hatef
2,
Mahdi Golshan
3 and
Otomar Linhart
4
1
School of Biology, College of Science, University of Tehran, Tehran P.O. Box 14155-6655, Iran
2
Toxicology Centre, University of Saskatchewan, Saskatoon, SK S7N 5B3, Canada
3
Iranian Fisheries Science Research Institute, Agricultural Research, Education and Extension Organization, Tehran P.O. Box 15745-133, Iran
4
South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Faculty of Fisheries and Protection of Waters, University of South Bohemia in České Budějovice, 389 25 Vodňany, Czech Republic
*
Author to whom correspondence should be addressed.
Animals 2021, 11(10), 2817; https://doi.org/10.3390/ani11102817
Submission received: 27 July 2021 / Revised: 14 September 2021 / Accepted: 19 September 2021 / Published: 27 September 2021

Abstract

:

Simple Summary

Public concern regarding environmental contaminants (ECs)-related reproductive disorders has increased due to increasing global rates of infertility. All kinds of ECs are on rise rapidly in developing and industrializing low- and middle-income countries. The aquatic environments throughout the world are repositories for enormous amounts of ECs. As the biology of the reproductive system is highly conserved in vertebrates, wildlife or laboratory studies on fish provide significant information to establish a detailed risk assessment, and to identify novel or more sensitive endpoints for ECs-related reproductive disorders. The adverse effects of ECs on endocrine regulation of reproduction in male fishes have been extensively studied and reviewed; however, our knowledge on the effects and mechanisms of action of ECs on determinants of male fertility is limited. The present study is a state-of-the-art comprehensive review on the ECs-related fertility threat in male fishes with emphasis on the ECs effects on sperm production, morphology, genome, and motility kinetics. After a brief introduction to reproductive biology, fertility indicators, and determinants of fertility in male fishes, wildlife evidences for reproductive disorders were reviewed in fishes from the polluted aquatic environment. The laboratory studies show that ECs detected in aquatic environment are capable of causing fertility threat at environmentally relevant concentrations associated with a decrease in fertility determinant(s). This study suggests an urgent need to better elucidate mechanisms through which ECs affect sperm functions to cause fertility threat.

Abstract

Increasing global rates of diminished fertility in males has been suggested to be associated with exposure to environmental contaminants (ECs). The aquatic environments are the final repository of ECs. As the reproductive system is conserved in vertebrates, studies on the effects of ECs on fertility endpoints in fishes provide us with valuable information to establish biomarkers in risk assessment of ECs, and to understand the ECs-related fertility threat. The aim of the present review was to evaluate associations between ECs and fertility determinants to better understand ECs-related male fertility threat in male fishes. Wildlife studies show that the reproductive system has been affected in fishes sampled from the polluted aquatic environment. The laboratory studies show the potency of ECs including natural and synthetic hormones, alkylphenols, bisphenols, plasticizers, pesticides, pharmaceutical, alkylating, and organotin agents to affect fertility determinants, resulting in diminished fertility at environmentally relevant concentrations. Both wildlife and laboratory studies reveal that ECs adverse effects on male fertility are associated with a decrease in sperm production, damage to sperm morphology, alternations in sperm genome, and decrease in sperm motility kinetics. The efficiency of ECs to affect sperm quality and male fertility highly depends on the concentration of the contaminants and the duration of exposure. Our review highlights that the number of contaminants examined over fertility tests are much lower than the number of contaminants detected in our environment. The ECs effects on fertility are largely unknown when fishes are exposed to the contaminants at early developmental stages. The review suggests the urgent need to examine ECs effects on male fertility when a fish is exposed at different developmental stages in a single or combination protocol. The ECs effects on the sperm genome are largely unknown to understand ECs-related inheritance of reproductive disorders transmitted to the progeny. To elucidate modes of action of ECs on sperm motility, it is needed to study functional morphology of the motility apparatus and to investigate ECs-disrupted motility signaling.

1. Introduction

Global rates of environmental contaminants (ECs)-related reproductive disorders have been increasing over the past 50 years. In human beings, the incidences of testicular dysgenesis syndrome, including hypospadias (urethra opens on the underside of the penis instead of the tip), cryptorchidism (one or both testes not descended into the scrotum), testicular cancer, low semen quality, and infertile men, show global increases associated with ECs [1,2,3,4,5,6,7,8,9]. Landrigan et al. [10] reported that all kinds of ECs are all on the rise rapidly in developing and industrializing low-income and middle-income countries. The public concern regarding ECs-related reproductive disorders was originally linked to observations of reduced fertility, birth defects, and sexual developmental disorders in wildlife [11]. For over 30 years, the World Health Organization (WHO), National Institute of Health (NIH, USA), European Food Safety Authority, and the other organizations composed of working groups of experts in endocrinology, risk assessment, and toxicology, have conducted studies to examine the adverse effects of ECs on reproduction in humans and wildlife. These studies have shown that there are about 800 natural and man-made chemicals known or suspected to interfere with physiological and endocrinological regulation of reproduction [12]. However, our knowledge on the ECs-related hormonal dysfunctions that cause diminished fertility is limited to a small fraction of these chemicals. To reduce ECs-related fertility threat in males, it is critical to identify the contaminants that interfere with determinants of fertility, including sperm production, morphology, genome, and motility, and to characterize their modes of action on reproductive endocrine system. In this regard, interdisciplinary efforts that combine knowledge from wildlife, experimental animals, and human infertility clinics are needed to provide a more holistic approach for ECs-related reproductive disorders and fertility threat.
The aquatic environment is at greatest risk from pollutants since all chemicals will eventually find themselves in the rivers, lakes, and oceans as the final repository [13]. As biology of reproduction is highly conserved in vertebrates [14,15,16], studies on fishes as model organisms provide significant information to establish a detailed risk assessment and to establish novel or more sensitive endpoints for ECs-related fertility threat. Frequent clear evidences show reproductive disorders in fishes from polluted aquatic environments (see Section 4). The adverse effects of ECs on endocrine regulation of reproduction in male fishes have been extensively studied and reviewed in laboratory studies [13,17,18,19,20,21,22,23,24,25]. In contrast, our knowledge to understand whether ECs-disrupted hormonal functions result in diminished fertility is poor. To answer, it is critical to uncover the adverse effects of ECs on sperm production, morphology, genome, and motility kinetics as key determinants of fertility.
We have recently reviewed the toxicity of ECs on sperm morphology and motility in fishes, in vitro [26]. The review showed that ECs, in a dose-dependent manner, cause damage to sperm morphology and interfere with sperm energetics and motility kinetics, and thus affect male fertility. However, significant decreases or complete suppression of sperm motility and fertilizing ability occurred mostly at concentrations considerably higher than those reported in the aquatic environment or exceeding the WHO recommended limits for surface waters. Recently, Carnevali et al. [27] and Golshan and Alavi [25] suggested that ECs are capable of affecting sperm quality in fishes associated with alternations in hormonal functions of hypothalamus–pituitary–testis (HPT). These reviews have mostly focused on studies that show the adverse effects of ECs on sperm functions using in vitro approaches, and, moreover, the effects of a few ECs have been reviewed.
The present study is a state-of-the-art comprehensive review on the ECs-related fertility threat in male fishes with emphasis on the adverse effects of ECs on determinants of fertility, including sperm production, morphology, genome, and motility kinetics. After a brief introduction to reproductive biology, fertility indicators, and determinants of fertility in male fishes, wildlife evidences for reproductive disorders are reviewed in fishes from the polluted aquatic environment. To understand whether reproductive disorders in wildlife is associated with ECs, we review laboratory studies in which the adverse effects of particular ECs are studied on determinants of fertility, in vivo. Finally, we discuss urgent needs to better elucidate mechanisms through which ECs affect sperm functions to cause fertility threat. The present review provides us with valuable information to understand ecotoxicological impacts of ECs on fish fertility, which can be useful to establish biomarkers in ECs risk assessment.

2. An Introduction to Reproductive Biology in Male Fishes

It is essential to review reproductive biology, fertility indicators, and determinants of fertility in male fishes before delving into the ECs-related fertility threat. These provide the basic information to better understand multiplicity of sites through which ECs interfere with fertility. To clarify the terminology, “semen” refers to seminal plasma and sperm and “sperm” refers to sperm cells in the present review.

2.1. Anatomy of Reproductive Organ

In general, the male reproductive organ consists of a paired testes, the testicular duct, and the sperm duct in fishes [28,29] (Figure 1). In some primitive fishes (such as sturgeons), the testes release sperm into the testicular ducts, which pass the kidneys. At spawning, semen is released into the aquatic environment through the urinary ducts opened into the urogenital opening (Figure 1A,C). In most bony fishes (teleosts), neither testicular ducts nor sperm ducts attach to the kidneys. The sperm is released from the testes into sperm ducts where seminal plasma is secreted. At spawning, semen is released into the aquatic environment through the sperm ducts opened in the urogenital opening (Figure 1B, bookmark0D).
The testes are divided into the “tubular type” and the “lobular type” according to the distributions of spermatogonia in the seminiferous region [30,31,32,33] (Figure 1E, bookmark0F).
In the tubular type, as spermatogonia divide and enter in meiosis, the cysts migrate towards the region of the spermatic ducts located in the central region of the testis, where the cysts open to release sperm (Figure 1E). This type of testicular arrangement is found in zebrafish (Danio rerio) and guppy (Poecilia reticulate).
In the lobular type, the testis is composed of numerous lobules that are separated from each other by a thin layer of fibrous connective tissue, and spermatogonia are spread along the germinal compartment throughout the testis. The cysts do not migrate or become displaced during their development, and sperm is released into the lobular lumen (Figure 1F). This type of testicular arrangement is found in Japanese medaka (Oryzias latipes), common carp (Cyprinus carpio), goldfish (Carassius auratus), and rainbow trout (Oncorhynchus mykiss).
The testicular compartment contains Sertoli cells, Leydig cells, blood/lymphatic vessels, macrophages and mast cells, and neural and connective tissue cells (Figure 1G). The Leydig and Sertoli cells are involved in biosynthesis of steroid hormones to regulate sperm production and maturation.
The testicular ducts are located adjacent to the testes, which continue into the sperm ducts on the ventral sides. Testicular and sperm ducts possess very similar structural and enzyme-histochemical characteristics, and play key roles in nutrition of sperm, storage of sperm, synthesis of steroids, secretion of proteins and enzymes, and formation of the seminal plasma [34,35]. Maturation of sperm to acquire potential for motility and fertilizing ability occurs in the sperm ducts [36,37].

2.2. Spermatogenesis

Sperm is produced from spermatogonia following divisions [15,38,39]. During the process of spermatogenesis, diploid spermatogonia type A divides mitotically to produce diploid spermatogonia type B. The final mitotic division of spermatogonia type B produces diploid primary spermatocytes that undergo the first meiotic division to form haploid secondary spermatocytes. The second meiotic division produces haploid spermatids that transform into the flagellated sperm.

2.3. Sperm Morphology

Sperm is differentiated into a head, midpiece, and flagellum in fishes [40,41,42] (Figure 2). The head of sperm contains DNA for transferring a haploid set of the chromosomes into the oocyte upon fertilization. Mitochondria and proximal and distal centrioles are located in the midpiece. Mitochondria deliver energy that is required for the beating of the sperm motility apparatus with a “9 + 2” structure called the “axoneme” [43,44]. Both proximal and distal centrioles consist of nine peripheral triplets of microtubules. The distal centriole forms the basal body of the axoneme. Sperm is acrosomeless in teleostean fishes, while it possesses acrosome in primitive fishes, including hagfish and sturgeons [45,46].

2.4. Sperm Physiology

The seminal plasma is a product of Sertoli cells, testicular ducts, and sperm ducts, and its composition is different among fishes that may reflect species variations. The main role of seminal plasma is to create an optimal environment for the storage of sperm during maturation in the sperm ducts. Seminal plasma maintains sperm viability, motility, and fertilizing ability, and protects sperm against damage caused by proteolytic or oxidative attacks [50,51].

2.5. Sperm Motility

Sperm is generally immotile in the seminal plasma and the sperm ducts of fishes, and motility is triggered upon discharge into the aquatic environment (Figure 3A). In most freshwater and marine fishes, osmolality of the seminal plasma is the key factor to maintain sperm in the quiescent state in the sperm ducts [52]. In some freshwater fishes, including Salmonidae and Acipenseridae, high concentrations of potassium (K+) ions inhibits sperm motility in the seminal plasma [53,54,55]. At spawning, a hypo-osmotic and a hyper-osmotic signal is necessary for initiation of sperm motility in freshwater and marine fishes, respectively [42,56,57,58,59]. Changes of osmolality around sperm accompanied by K+ efflux in freshwater fishes and water efflux in marine fishes trigger sperm motility signaling. Activation of sperm motility is associated with an increase in intracellular pH and calcium (Ca2+) ions in both freshwater and marine fishes, while cyclic adenosine monophosphate (cAMP) remains unchanged. However, studies show that demembranated sperm of salmonid and sturgeon fishes require cAMP for the axonemal beating [60,61,62]. In some marine fishes, it has been shown that 17,20β,21-trihydroxy-4-pregnen-3-one (17,20β-P) is capable to induce sperm hypermotility by increasing cAMP and intracellular Ca2+ through a membrane progesterone receptor [63,64].
After initiation of sperm motility in the aquatic environment, duration of motility is very short in fishes from a few seconds to several minutes or hours depending on the species [28,29,51,59,65,66]. The inter-species differences probably depend on the capacity of the sperm to restore intracellular ATP and creatine phosphate concentrations [67,68]. Once sperm motility is initiated, the percentage of motile sperm and sperm velocity rapidly decrease, which are associated with a large, but not complete depletion of ATP [46,59,66] (Figure 3B). Fish sperm can regenerate ATP from phosphocreatine and ADP; however, this ATP regeneration system does not prevent the precipitous decline in ATP levels during motility [69,70,71].

3. Fertility Indicators and Assessments in Fishes

Fertilization and hatching rates are calculated to assess fertility in fishes (Figure 4A). The fertilization rate is the percentage of oocytes that become fertilized upon spawning or artificial insemination, and is calculated as number of fertilized eggs/initial number of oocytes × 100. Successful fertilization depends on onset of release of sperm from males and ova from females [28,72]. During the short period of motility, sperm must penetrate the oocyte through a funnel called the “micropyle” to fertilize it [32,73]. A fertilized egg can be easily identified by the presence of a multi-cellular blastodisc (cleavage), which occurs from several hours to a few days post fertilization and depends on fish species and environmental factors including temperature. The hatching rate is the percentage of hatched larvae, and calculated as number of hatched larvae/initial number of oocytes × 100. Once embryonic development is completed, larvae hatch [74,75].

4. Determinants of Fertility in Male Fishes

Analyses of sperm production, morphology, genome, and motility kinetics are basically important to assess fertility in male fishes (Figure 4B).

4.1. Sperm Production

Frequent studies have shown that fertilization rate positively correlates with sperm volume, sperm density, number of sperm per oocyte, and density of sperm in the water during fertilization [76,77,78,79,80,81,82,83]. One can weigh semen mass, measure semen volume, or count sperm density to evaluate sperm production.

4.2. Sperm Morphology

There is a species-specific relationship between the head size of sperm and diameter of micropyle in fishes [49,73,74]. This indicates that sperm of one species can penetrate only into the oocyte of similar species. A change in the size of sperm head is a mirror of the size of nucleus [84,85,86]. It has also reported that sperm with a smaller head can move faster than those with a larger head [86,87,88]. Additionally, both positive and negative correlations have been reported between the length of flagellum and the sperm velocity [86,87,88,89,90,91]. These suggest that alternations in the size of sperm head and length of flagellum can result in diminished fertility by affecting sperm penetration into the oocytes or sperm motility performance. Various microscopic techniques including scanning and electron microscopy are valuable methods to assess sperm morphology [43,45,92].

4.3. Sperm Genome

Upon fertilization, sperm with a haploid number of chromosomes transmit a parental genome to the next generation. Alternation of chromosome material, Y chromosome deletion, and ploidy level are among factors that affect fertility. The integrity of sperm DNA correlates with fertilization and embryonic development in fishes [93,94]. Fertility threat has been frequently reported in polyploid fish, which were associated with failure of testicular development [95], enlarged head size making penetration of sperm through a normal-sized micropyle difficult [96,97], reduced sperm production [84,98], and increased abnormal sperm with malformation of the head, mitochondria, and flagellum resulting in decreasing motility and velocity [84,99]. One can assess the integrity of DNA using a comet assay, sperm chromatin structure assay, or terminal deoxynucleotidyl transferase dUTP nick end labelling assay (TUNEL) [100,101]. Chromosome number and DNA content can be counted or assessed using a flow cytometry, respectively [84,86,95].

4.4. Sperm Motility Kinetics

Duration of sperm motility, percentage of motile sperm, and sperm velocity are key determinants for fertility in male fishes [28,79,102]. It has been shown that sperm with faster movement and a longer period of motility have more chance to approach an oocyte to fertilize it [78,80,103,104]. In addition, it has been suggested that sperm velocity and the duration of motility are positively correlated with ATP content of sperm [59,66]. A computer-assisted sperm analysis (CASA) provides a valuable tool to assess sperm motility kinetics [105,106,107]. Percentage of sperm motility is evaluated by counting the number of motile sperm and total number of sperm. The sperm velocity is the distance between the starting and ending points of the motility track divided by the time spent for this movement. Based on sperm head positions during the period of motility, various sperm velocity parameters are identified, including curvilinear velocity (VCL, the velocity along the sperm head trajectory), straight line velocity (VSL, the straight line distance between the start and end points of the sperm head trajectory), and the angular path velocity (VAP, the velocity along a derived smoothed path) (Figure 3C).

5. Wildlife Evidences for Environmental Contaminants (ECs)-Related Fertility Threat in Male Fishes

Wildlife studies have been frequently conducted to investigate reproductive disorders in male fishes inhabiting the aquatic environment that receive bleached kraft pulp mill effluent, municipal wastewater effluent, sewage treatment plant effluent, wastewater treatment plant effluent, and agricultural and industrial runoffs. Various types of hormonal mimic or genotoxic ECs have been detected including natural and synthetic hormones, pesticides, metals, industrial chemicals (nonylphenol, alkylphenol, bisphenols, phthalates, polychlorinated biphenyls, hexachlorocyclohexane and hexachlorobenzene), and phytosterols. These studies have shown various reproductive disorders in males from the polluted aquatic environments (Table 1).
Delay in sexual maturity, decrease in gonadosomatic index (gonad mass/body mass × 100) and secondary sexual characteristics, and increases in female-biased sex ratio and intersex (occurrence of oocytes in the testes) are predominant reproductive disorders reported in fishes from polluted aquatic environments [108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125]. The incidence of intersex has been shown to be mostly associated with increased circulating levels of vitellogenin (Vtg) or 17β-estradiol (E2) [111,113,115,117,118,123,124,126,127,128,129,130].
Compared to a number of studies that reported incidence of intersex and the size of testis, there are a few studies that investigated sperm quality. These studies demonstrate the adverse effects of ECs to decrease number of spermiating males, sperm production (volume and density), viability, motility, velocity, and fertilizing ability, and to increase number of abnormal sperm and sperm chromosome breakage [115,117,121,131,132,133,134,135]. Decreases in sperm production have been shown to be mostly associated with decreases in circulating androgen levels (testosterone (T) or 11-ketotestosterone (11-KT)), which lead to disorders in testicular development [115,121,122,129,130,134,135,136,137]. It has been also shown that decreases in fertilizing ability of fish from polluted aquatic environments were associated with decreases in sperm production, motility, or velocity [115,132,134].
Taken together, wildlife studies reveal that ECs are capable of impairing spermatogenesis and sperm development in fishes. Subsequently, morphological damage to sperm, abnormality of the sperm genome, decrease in sperm production, and reduction of sperm motility kinetics result in diminished fertility in male fishes. It is worth to note that wildlife studies also showed ECs-related fertility threat in male fishes which were associated with disruption of hormonal functions of the hypothalamus–pituitary–testis axis that regulate spermatogenesis and sperm development (Table 1).
Table 1. Wildlife evidences for environmental contaminants-related reproductive disorders in male fishes.
Table 1. Wildlife evidences for environmental contaminants-related reproductive disorders in male fishes.
RegionFish SpeciesContaminantsReproductive EndpointsDescriptionAuthors
Jackfish Bay, North Shore of Lake Superior, CanadaWhite suckerBKME that includes resin acidDelayed sexual maturation
Decreased testicular size
Reduced secondary sexual characters
Decreased LH, T, 11-KT, and 17,20β-P levels
Failed to increase 17,20β-P levels in response to stimulation of spermiation by sGnRH-A
Polluted site: Jackfish bay
Reference site: Mountain bay
Sampling time: May–Aug 1988 (Munkittrick et al. 1991); Aug 1988, Aug 1989 and Sep 1990 (Munkittrick et al. 1992); May 1990 and 1991 (Van Der Kraak et al. 1992)
[109,110,138]
Estuarine, UKFlounderSewage effluentsNo difference in GSI, increased Vtg levels at all polluted sites
Intersex at the Mersey estuary (17%)
Polluted sites: Tyne, Crouch, Thames and Mersey estuaries
Reference site: Alde River
Sampling time: Sep–Dec 1996
[111]
The Moselle and the Rhone Watershed, FranceChubMetals and organic chemicalsNo difference in GSI
Increased Vtg levels at Igney
Necrotic sperm cells observed at Igney
Polluted site: Igney (the Moselle river)
Reference site: Saillans (the Drome river)
Sampling time: June
[139]
Saint Louis, Missouri, USAShovelnose sturgeonChlordane, p,p’. DDE, PCBsIntersex (29%)Polluted site: Downstream of St. Louis, Prairie de Rocher, IL
Reference site: Upstream of St. Louis, Davenport, IA
Sampling time: Autumn
[112]
Tokyo Bay, JapanFlounderSewage effluents that include Alkylphenol polyethoxylates and NPDecreased GSI in males with ovo-testis
Increased Vtg levels
Intersex (15% of individuals)
Polluted site: Tokyo Bay, Japan
Reference site: Shiriuchi, Hokkaido, Japan
Concentrations (μg/L): Alkylphenol polyethoxylates (37.5), Nonylphenol: (0.2–1.1)
Sampling time: Jan 1997–May 1998
[113]
The Po River, ItalyBarbelNot determinedIntersex (50%)Polluted site: Downstream of the Po River
Reference site: Upstream of the Po River
Sampling time: April 1999
[114]
Nene and Aire Rivers, UKRoachSTWsDecreased GSI
Intersex at Nene and Aire (100%) (1–4% intersex at reference site)
Delayed spermatogenesis
Increased Vtg, T, and E2 levels in intersex males compared to normal and to intersex males of reference sitesNo difference in 11-KT levels between intersex males of polluted sites and reference sites
Decreased number of spermiating males
Decreased sperm volume and density
Decreased sperm motility and velocity
Decreased fertilizing ability of sperm
Polluted sites: Nene (Northamptonshire) and Aire (Yorkshire)
Reference cites: Royal Canal (Ireland), Grantham Canal (Leicestershire) and a spring-fed lake (Lake Wartnaby, Leicestershire)
Sampling time: October 1995 and 1996; May 1998, 1999, and 2000
[115,132]
The St. Johns RiverLargemouth bassDDT and its metabolites, PCBs, high and low molecular weight PAHs, cyclodiene and
chlorinated pesticides
At pre-spawning season (Sep 1996):
Increased E2 levels at polluted sites
Decreased 11-KT levels at Palatka and Julington Creek
At Spawning season (February 1997):
Decreased GSI at Palatka, increased E2 levels at Palatka and Green Cove, decreased E2 levels at Julington CreekDecreased 11-KT levels at all polluted sites
Polluted sites: Downstream of the St. Johns River: Palatka (North of Welaka), Green Cove (North of Palatka) and Julington Creek (North of Green Cove). The Palatka and Green Cove sites are representative of urban, industrial, and agricultural development. The Palatka site is near to a paper mill plant. The Julington Creek site receives discharges of wastewater and runoff from recreational boating marinas
Reference site: Upstream of the St. Johns River
Sampling time: September 1996 and February 1997
[116]
The Anoia River, SpainCarpSTW that includes estrogenic compoundsDecreased testicular size
Occurrence of testicular atrophy
Intersex (19%)
Decreased T levels, Increased Vtg levels
Polluted sites: Downstream of the Anoia River
Reference site: None
Sampling time: January and March 2000
[126]
The St. Lawrence River, Montreal, CanadaSpottail ShinerSTW that includes xenoestrogensIntersexes at Îles de la Paix, Île Dorval, Îlet Vert and Île Beauregard (2.6, 15, 31, and 27, respectively)
Increased Vtg mRNA levels at Îlet Beauregard, Îlet Vert, and Île Saint-Ours compared to the Îles de Boucherville, Île Dorval, Îles de la Paix, and Ottawa river
Delayed spermatogenesis at Îlet Vert and Île Beauregard
Decreased sperm density, motility, and velocity at Îlet Vert compared to the Îles de la Paix
Polluted site: The downstream sites of the St. Lawrence River (Îlet Vert, Île Beauregard and Île St. Ours)
Reference sites: The upstream sites of the St. Lawrence River (Îles de la Paix, Île Dorval, Îles de Boucherville) and the Ottawa River
Sampling time: June 1999–2002
[117]
Hamilton Harbour, Western Lake Ontario, CanadaWhite PerchTreated domestic sewageIntersex (22–83%)
Increased Vtg levels
Polluted site: The Cootes Paradise region
Reference site: Deal Lake and hatchery fish
Sampling time: August 1998 and 2000, October 1999, July and September 2002
[118]
Boulder Creek and the South Platte River, Colorado, USAWhite suckerWWTPIntersex
Increased percentage of female compared to males (83% vs. 45%)
Polluted site: Boulder Creek downstream of the Boulder WWTP
Reference site: A stream reach beginning 2 km upstream of the Boulder
Sampling time: March May, October and November 2002
[120]
Danish Streams in Aarhus County, Jutland, DenmarkRoachDomestic STWsIntersex (6.7–6.5% at polluted sites compared to 4.5–5% at reference sites)
The highest intersex 26.5% was observed in the stream Kristrup Landkanal.
Polluted sites: Aarhus Brook, Egaa, and Kristrup Landkanal streams receive sewage effluent discharges from STW
Reference sites: Lake Almind and Lake Ravn receiving no or small amounts of STW, respectively
Sampling time: June and September 1999
[119]
Boulder Creek, a Rributary of the South Platte River, Colorado, USA WWTP that includes E2, EE2, estrone, BPA, NPDecreased GSI
Intersex (18–22%)
Increased Vtg levels
Decreased sperm abundance
Polluted site: Downstream of the Boulder WWTP
Reference site: Upstream of the Boulder WWTP
The Boulder WWTP uses filter/activated sludge treatment process with nitrification/denitrification and chlorination/dechlorination. The mean annual concentrations of NH3-N, NO3-N, biological oxygen demand, and total suspended solids in the WWTP effluent were 7, 12, 15, and 6 mg/L, respectively. At downstream of the WWTP outfall, NH3-N and NO3-N were 1 and 4 mg/L, respectively
Concentrations (ng/L): E2 (1.6–2.1), EE2 (0.7–<2.0), estrone (36), BPA (2.5-35), NP (39–340) at polluted site, and E2 (<0.2–2.9), EE2 (<0.8–<2.0), estrone (<5), BPA (3.1–27), NP (<33–120) at reference site.
Sampling time: Autumn 2003 and Spring 2004
[127]
Oldman, Bow and Red Deer Rivers, Alberta, CanadaLongnose daceMunicipal wastewater and agricultural runoffIncreased female-biased sex ratio at Oldman river
Increased GSI at downstream of Oldman and Bow rivers
Increased Vtg levels at downstream Oldman, Bow, and Red Deer rivers
Polluted sites: Downstream of Oldman, Bow and Red deer rivers (Discharges of Lethbridge, Calgary and Red Deer, respectively)
Reference sites: Upstream of the above rivers
Concentrations (ng/L) of selected contaminants at Red Deer and Oldman rivers: α-estradiol (0.2), E2 (0.6), T (1.7), Equilin (1.8), Estrone (1.8), BPA (3.8), α-Zearalanol (5.2), Ergosterol (25.4), Stigmastanol (37.5), β-sitosterol (100.5), Stigmasterol (144.7), Cholesterol (189.2), Fucosterol (212.1)
Sampling time: April 2005 (Jeffries et al. 2008), Oct and No 2005 (Jeffries et al. 2010)
[123,124]
The Luvuvhu River, Limpopo Province, South AfricaTilapiaDDT and metabolitesDecreased GSI at XW
Intersex at AD, ND, and XW (55%, 60%, and 59%, respectively)
Increased sperm velocity at ND
Trends toward decrease in sperm motility at ND and XW
Polluted sites: Nandoni Dam (ND) and Xikundu Weir (XW)
Reference site: Albasini Dam (AD) (outside of the DDT-sprayed area)
Sampling surveys were carried out during high and low flow seasons
Concentrations (μg/L): DDT and metabolites: 0.1–1
Sampling time: March and October 2007, February 2008
[121,122]
African catfishA trend toward decrease in sperm motility at ND
Trends toward decrease in sperm velocity at ND and XW
Testicular abnormalities at ND and XW
The Elbe River, Czech RepublicChubHg, PCBs, DDT and its metabolites, HCHs, HCB, OCSIntersex at downstream of Usti nad Labem
Decreased GSI at downstream of Pardubice and Neratovice
Decreased 11-KT levels at upstream and downstream of Neratovice
Polluted sites: Downstream and upstream of Pardubice, Neratovice, and Usti nad Labem. Pardubice receives effluents of the chemical factory containing primarily active components for making medicines and pesticides. Neratovice receives effluents of the chemical plant producing caprolactam as a raw material for polyamide fiber and the making of plastics, polyvinylchloride, and several inorganic compounds. Usti nad Labem is affected by agricultural and industrial activity
Reference site: The upper reaches of the Vltava River (the tributary of the Elbe River).
Concentrations (mg/kg lipid): HCHs (0.021–0.486), DDT (1.14–6.48), HCB (0.109–0.521) at polluted sites and HCHs (0.018), DDT (0.92), HCB (0.033) at reference site
Sampling time: May 2004
[140]
The Luvuvhu River, Limpopo Province, South AfricaTilapiaDDT and its metabolitesIntersex at AD, ND, and XW (49%, 63%, and 58%, respectively)
No intersex in the aquaria-reared fish
Polluted sites: Nandoni Dam (ND) and Xikundu Weir (XW)
Reference site: Albasini Dam (AD) and reared fish
[125]
North West Province of South AfricaCatfishDBP, NP, SeTesticular abnormal growth in fish at HD during HF period.
Trends toward decrease in sperm motility at HD during LF period
Decreased sperm velocity at HD during both LF and HF periods
Polluted site: Hartbeespoort Dam (HD) characterized by eutrophication due to the high levels of phosphates and nitrates originating from STWs and agricultural effluent
Reference site: Marico Bosveld Dam (MD) receiving water from the Marico River, an unmodified natural ecological state, Concentrations (μg/L): DBP: 3.5, NP: 4, Se 24, water chemistry: pH: 7.5 (MD) vs. 9–10.4 (HD), O2 (mg/L): 7.9 (MD) vs. 9.5–10.6 (HD), sampling time: At a low flow (LF) in Oct and at a high flow (HF) in March (MD was sampled only in Oct)
[133]
Lake Erie, Ontario, CanadaGoldfishPCBNo difference in GSI
Decreased T and 11-KT levels
Decreased Vtg levels
Polluted site: Wheatley Harbour, Northwest shore of Lake Erie
Reference site: Hillman Marsh
[128]
The Grand River Watershed in Southern Ontario, CanadaRainbow darterMWWESampling time: September 2010
Decreased GSI at DS2 and DS3.5 compared to US1
Intersex at DS2 and DS3.5 compared to US1
Increased Vtg mRNA levels at DS2 compared to US1 and DS3.5
Sampling time: April and May 2011
Intersex (%): DS1 (27.3), INT1 (48.5), DS2 (84.6), DS2x (80), DS3.5 (53.3), DS Gp (6.7) vs. US1 (6.3), US2 (0), US Gp (25)
No difference in GSI of normal males among US1, US2, DS1, INT1, DS2, DS2x, and DS3.5
Higher GSI of normal males at DS Gp than UP Gp
No difference in GSI of intersex males
No difference in E2 levels
Decreased T levels at DS1, DS2, and DS3.5
Decreased 11-KT levels at DS1, INT1, DS2, and DS3.5
Decreased T and 11-KT levels in intersex males
Decreased proportion of spermatogonia at DS2
Decreased proportion of spermatocytes and increased spermatids at INT1, DS2x, and DS3.5, no difference in proportion of sperm
Polluted site: Downstream Waterloo treatment plant outfall (DS1), between Waterloo and Kitchener MWWTPs (INT1), downstream Kitchener treatment plant outfall (DS2), downstream Kitchener outfall out of the plum. (DS2x), far downstream Kitchener treatment plant outfall (DS3.5) and downstream Guelph treatment plant outfall (DS Gp)
Reference sites: Upstream urban areas (US1), upstream Waterloo outfall (US2) and upstream Guelph treatment plant outfall (US Gp)
Sampling time: September 2010, April and May 2011
[129,130]
The Alibori River, Benin, West AfricaAfrican catfisho,p′-p,p′-DDT, o,p′-p,p′-DDE, o,p′-p,p′-DDD, endosulfan, HeptachlorIntersex (25–50%)
Decreased 11-KT levels at S3-RS, S4-RS, S3-DS
Increased E2 levels at S2-RS, S3-RS and S4-RS, S4-DS
Decreased spermatocytes, spermatids, and sperm in the testes of fish from S2-S4 compared to S1
Polluted sites: The Alibori River in Sori, Gogounou (S2), Alibori K, Kandi (S3), and Batran, Banikoara (S4). Alibori River collects the drainage from agricultural areas in the cotton-producing basin
Reference site: The Pendjari River within the Pendjari National Park (S1). Agriculture is strictly forbidden
Concentrations (μg/L): p,p′-DDT (0.11 RS, 0.07 DS, S4), o,p′-DDE (0.17, RS-S2, 0.19 RS-S3, 0.09 RS-S4, 0.18 DS-S2, 0.14 DS-S3, 0.17 DS-S4), p,p′-DDE, (0.13 RS-S2, 0.10 RS-S3, 0.18 RS-S4, 0.07 DS-S2, 0.08 DS-S3, 0.14 DS-S4), o,p′-DDD (0.07 RS-S2, 0.09 RS-S3, 0.09 RS-S4), p,p′-DDD (0.09 RS-S2, 0.08 RS-S4, 0.08 DS-S4), endosulfan (8.2 RS-S2, 8.8 RS-S3, 13.8 RS-S4, 2.9 DS-S2, 3.4 DS-S3, 3.7 DS-S4), Heptachlor (0.5 RS-S2, 0.5 RS-S3, 0.6 RS-S4, 1.4 DS-S2, 1.5 DS-S3, 1.3 DS-S4)
Sampling time: Rainy season (RS), Sep 2010 and 2011, period of flooding with intense use of pesticides in cotton fields; dry season (DS), Feb 2011 and 2012, period of no agriculture but limited use of pesticides
[136]
Guinean tilapiaIntersex (12.4-40%)
Decreased 11-KT levels at S2-RS, S4-RS, and S4DS
Increased E2 levels at S2, S3, and S4 both RS and DS
Decreased spermatids and sperm in the testes of fish from S2–S4 compared to S1
Grand River Watershed, Southern Ontario,
Canada
Rainbow darterMWWEDecreased GSI at DSK and DSK2
Higher intersex at DSK, DSK2, and USK
Decreased 11-KT levels at DSK, DSK2, DSW, USK, and USWDecreased T levels at DSK
Increased sperm volume at DSK and DSK2
Decreased fertilization rate at DSK and USK
Decreased hatching rate at DSK, USK, DSW, and USW
Polluted sites: Sites 1 km downstream of the Kitchener MWWTP (DSK) and Waterloo MWWTP (DSW), and 3 km downstream of Kitchener MWWTP (DSK2)
Reference site: Sites 1 km upstream of the Kitchener MWWTP (USK) and Waterloo MWWTP (USW), and a site upstream of the city-limits (R)
Sampling time: Apr and May 2012, May 2013
[134]
Lake Mead National Recreation Area, USACarpDDT
PCB
PCP
PBDEs
Decreased GSI at BB, GB, LVB and WB
Decreased 11-KT levels at BB, GB, LVB, and WB
Decreased E2 levels at BB, GB, and WB
Decreased sperm motility at LVB and WB
Decreased sperm viability at LVB
Decreased sperm mitochondrial functions at LVB
Increased sperm chromosome breakage at LVB
Polluted sites: Boulder Basin (BB), Gregg Basin (GB), Las Vegas Bay (LVB) and Willow beach (WB), Reference site: Overton Arm (OA)
DDT (ng/g wet weight): 43.4 (BB), 10.5 (GB), 107.5 (LVB), 49.7 (WB), 21.3 (OA)
PCBs (ng/g wet weight): 56 (BB), 14 (GB), 94 (LVB), 381 (WB), 16 (OA)
PCP (ng/g wet weight): 11 (BB), 0 (GB), 82 (LVB), 0 (WB), 0 (OA)
PBDEs (ng/g wet weight): 62.9 (BB), 6.4 (GB), 118.8 (LVB), 88.4 (WB), 21.1 (OA)
Sampling time: May 1998, May 1999–May 2000, March–April 2006
[137]
Fish species: African catfish (Clarias gariepinus), Barbel (Barbus plebejus), Black bass (Micropterus spp.), Channel catfish (Ictalurus punctatus), Chub (Leuciscus cephalus), Carp (Cyprinus carpio), Chub (Leuciscus cephalus), Flounder (Pleuronectes yokohamae and Platichthys flesus), Goldfish (Carassius auratus), Guinean tilapia (Tilapia guineensis), Largemouth bass (Micropterus salmoides floridanus), Longnose dace (Rhinichthys cataractae), Mosquitofish (Gambusia holbrooki), Rainbow darter (Etheostoma caeruleum), Roach (Rutilus rutilus), Shovelnose sturgeon (Scaphirhynchus platyorynchus), Spottail shiner (Notropis hudsonius), Tilapia (Oreochromis mossambicus), White perch (Morone americana), White sucker (Catostomus commersoni). Hormones: 11-ketotestosterone (11-KT), 17α-ethynilestradiol (EE2), 17α,20β-dihydroxy-4-pregnen-3-one (17,20β-P), 17β-estradiol (E2), salmon gonadotropin-releasing hormone analog (D-Arg6,Pro9N-Et salmon GnRH) (sGnRH-A), luteinizing hormone (LH), testosterone (T). Chemicals: 1,1,1-trichloro-2,2-bis(p-chlorophenyl)ethane (DDT), p,p’-dichlorodiphenyldi-chloroethylene (p,p′.DDE), α-, β-, and γ-isomers of hexachlorocyclohexane (HCHs), Bisphenol A (BPA), di-n-Butyl phthalate (DBP), hexachlorobenzene (HCB), hydrophobic fragrance galaxolide (HHCB), mercury (Hg), nonylphenol (NP), octachlorostyrene (OCS), octylphenol (OP), personal care products (PCPs), polybrominated diphenyl ethers (PBDEs), polychlorinated biphenyls (PCBs), selenium (Se). Source of contaminants: Bleached kraft pulp mill effluent (BKME), municipal wastewater effluent (MWWE), sewage treatment plant (STP), sewage treatment works (STW), wastewater treatment plant effluent (WWTP).

6. Laboratory Evidences for Environmental Contaminants (ECs)-Related Fertility Threat in Male Fishes

Laboratory studies help us to screen the ECs adverse effects on fertility determinants to understand ECs-related fertility threat in fishes. Basically, recording male fertility at the individual level in the laboratory studies provide us with ecologically important understanding of EC-related male reproductive disorders. Moreover, there are large numbers of contaminants in the aquatic environment with agonist/antagonist interactions [141,142], which may cause difficulties to understand which kind of contaminant cause male infertility. To elucidate how each of these ECs interferes with fertility in male fishes, it is suggested to examine their effects on determinants of fertility in a single exposure protocol. This will provide valuable information on how each of these EC affects the testicular functions, which may result in diminished sperm quality.
To best of our knowledge, we found a few contaminants that have been examined over fertility tests or their adverse effects have been studied on sperm production, morphology, genome, and motility kinetics in fishes. The examined ECs could be classified into: (a) Natural or synthetic hormones (E2; 17α-ethinylestradiol, EE2; 17α-methyltestosterone, 17α-MT; progesterone, P), (b) alkylphenols (4-nonylphenol, 4-NP), (c) bisphenols (bisphenol A, BPA; bisphenol AF, BPAF; bisphenol S, BPS), (d) plasticizers (di-2-ethylhexyl phthalate, DEHP; mono-(2-ethylhexyl)-phthalate, MEHP), (e) pesticides (DDT and its metabolites; vinclozolin, VZ; monocrotophos, MCP; methoxychlor, DMDT), (f) pharmaceutical compounds (alderin, clofibrate, diethylstilbestrol, DES; fluoxetine, FLX; flutamide, FLU; levonorgestrel, LNG), (g) alkylating agent (methyl methane sulfonate, MMS), and (h) organotin compound (tributyltin, TBT).
In this section, we first review the laboratory studies that have investigated the adverse effects of ECs on fertility in male fishes. Then, the effects of ECs on sperm production and motility kinetics are reviewed in the laboratory studies.

6.1. Effects of Environmental Contaminants (ECs) on Fertility

The adverse effects of examined ECs on fertility in male fishes are summarized in Table 2. Most of these studies have shown reduction of fertilization rate or hatching rate when exposed males were mated with unexposed or exposed females, in vivo. Among ECs, exposure of fathead minnow (Pimephales promelas) to 0.1 µg/L EE2 and to 50 µg/L 17α-MT for 21 d [143,144], of medaka to 0.38 µg/L 17α-MT for 21 d [145], and of rainbow trout to 0.75 µg/L 4-NP for 60 d [146] resulted in complete fertility loss (Table 2). Bisphenol A [147], VZ [148], EE2 [149,150], and MMS [151] as high as 3120, 450, 0.5, and 50 µg/L were without effects on fertility of adult zebrafish, fathead minnow, medaka, and salmon exposed for 21, 28, 21, 21 d, respectively.
Decreases in fertility have been seen when duration of exposure of males to ECs was increased [145,146,152,153]. Additionally, increasing concentrations of a particular contaminant resulted in lower fertilization rate or hatching rate [143,145,152,153,154,155,156,157,158,159,160,161,162,163,164,165], suggesting a correlation between EC concentration and fertility. The adverse effects of a particular contaminant on male fertility show differences among species that may suggest a species- specific sensitivity to the contaminant. For instance, following exposure to 17α-MT for 21 d, fertilization rate is significantly decreased at 0.05 µg/L in medaka [145] and at 5 µg/L in fathead minnow [143]. If similar developmental stage in exposure test (exposure of adults or embryos to ECs) is considered, it is likely that species-specificity effects on fertilization and hatching rates exist for EE2 [144,149,150,155,161], BPA [147,157], and 4-NP [146,152] (Table 2).
Any changes in fertilization rate following exposure to the contaminants are similar to that of hatching rate at the same concentration [145,150,151]. This suggests a positive correlation between fertilization rate and hatching rate. Thus, fertilization rate, hatching rate, or both can be recorded to assess ECs-related male fertility in fishes.
Table 2. Laboratory studies on the potential adverse effects of environmental contaminants (ECs) to impair fertility in male fishes.
Table 2. Laboratory studies on the potential adverse effects of environmental contaminants (ECs) to impair fertility in male fishes.
ECsFish
model
Dose (µg/L)ExposureFertility endpoints (%)DescriptionAuthors
Fertilization RateHatching Rate
E2Rainbow troutCtrlAdult, 50 d62 Sperm were prediluted in MIS1 and added into eggs at ratio 55,000:1 to 65,000:1. Fertilizing medium was water at 6 mL. The eyed eggs were recoded 30–35 d post fertilization.[156] A
0.000459
0.001121 *
0.00267 *
MedakaCtrlAdult, 25 d9976Pairs were exposed for 21 d for females and for 25 d for males. During the exposure period, the spawned eggs were collected and the fertilized eggs (fertility of the mating pairs) were counted. During the last 3 d of exposure, the fertilized eggs from each pair were collected and the hatching rate was recorded.[154] A
0.0299962
0.0569071
0.1169761
0.2279864
0.46378 *50
EE2Fighting fishCtrlAdult, 28 d78 Pairs were exposed. Nests were removed and the pairs were kept in tanks for 3 d to determine fertilization rate by counting fry numbers.[161] N
0.0176
0.164 *
ZebrafishCtrlAdult, 21 d64 Males were mated with untreated females. After 14 d, the hatching rate was calculated.[166] N
0.0256 *
MedakaCtrlAdult, 21 d9886Exposed spermiating male were kept in tanks with unexposed females for 7 d. All of the eggs were collected from each pair and fertilization and hatching rates were calculated.
[150] 3
0.039884
0.068661
0.128763
0.489481
Fathead minnowCtrlAdult, 21 d98 Pairs were exposed. After exposure, they were transferred to aquaria with clean water. The spawned eggs were collected and the fertilized eggs (fertility of the mating pairs) were counted 2 d after spawning.
[144] 4A
0.000189
0.00192
0.00384
0.0130 *
0.10 *
Rainbow troutCtrlAdult, 62 d60 a, 46 b, 25 c Sperm was prediluted in MIS1 and added into eggs at ratio 300,000:1 (a), 50,000:1 (b), and 10,000:1 (c). Fertilization medium was ovarian fluid, 60 mM NaHCO3, 50 mM Tris, pH 9. The eyed eggs were recoded 28 d post fertilization[155] A
0.01636 a,*, 23 b,*,19 c
0.13135 a,*, 22 b,*,17 c,*
MedakaCtrlAdult, 21 d92 Pairs were exposed and the spawned eggs were collected during the exposure period. The fertilized eggs (fertility of the mating pairs) were recorded.
[149] A
0.03395
0.06493
0.11692
0.26191
0.48883
17α-MTMedakaCtrlAdult, 21 d99 a, 98 b68 bPairs were exposed. During the exposure period, the spawned eggs were collected and the fertilized eggs (fertility of the mating pairs) and the hatching rate were recorded following 7 d (a) or 21 d (b) of exposure.
[145] A
0.02398 a, 97 b60 b
0.04786 a, 80 b,*39 b,*
0.08890 a, 33 b,*18 b,*
0.18896 a, 0 b,*0 b,*
0.38096 a, 0 b,*0 b,*
Fathead minnowCtrlAdult, 21 d98 After exposure, 3–4 pairs were transferred to aquaria with clean water. The spawned eggs were collected and the fertilized eggs (fertility of the mating pairs) were counted 2 d after spawning.[143] 5A
0.198
159
531 *
500 *
4-NPMedaka0Adult, 21 d9794Pairs were exposed. For 21 consecutive d, spawned eggs were collected, counted, and assessed for fertilization rate. The fertilized eggs were maintained at the same treatments, and number of hatched embryos was recorded to assess hatching rate. [167] A
1.279698
2.959796
9.819599
27.89599
89.49577 *
Rainbow troutCtrlAdult, 60 d 97 a, 65 bSperm was prediluted in MIS1 and added into eggs at ratios of 55,000:1 to 65,000:1. Fertilizing medium was water. The eyed eggs were recoded 35 d post fertilization. Fertilization was recorded following 30 d (a) and 60 d (b) of exposure.[146] N
0.13 96 a, 64 b
0.28 94 a, 68 b
0.75 95 a, 0 b,*
MedakaCtrlAdult, 21 d98a, 98b Pairs were exposed. During the exposure period, the spawned eggs were collected and the fertilized eggs (fertility of the mating pairs) were counted following 7 d (a) or 21 d (b) of exposure.
[152] A
24.898a, 96b
50.995a, 95b
10198 a, 89 b
18496 a, 78 b,*
BPARare minnowCtrlAdult, 21 d96 a,b,c Males were exposed to BPA for 7 d (a), 14 d (b), or 21 d (c). Fertilization was performed in vitro, and evaluated at 4 h post fertilization. [168] A
1192 a,*, 90 b,*, 89 c,*
20690 a,*, 89 b,*, 87 c,*
ZebrafishCtrlLarvae, 150 d9287BPA exposure was from larvae (6 d post fertilization) to 5 months post fertilization.
[164] A
0.2289061 *
2.288962 *
22.887 *86
Zebrafish0.032Embryos, 150 dn.s.47Four females and 4 males within the same group were maintained together to produce offspring for assessment of fertilization and hatching rates. Solvent contains 0.03 BPA.[163] A
0.372n.s.24 *
Brown troutCtrlAdult, 76 d66 Sperm of exposed males was prediluted in MIS1 and added into eggs at ratios of 45,000:1 to 60,000:1. Fertilizing medium was water. The eyed eggs were recoded 30–35 d post fertilizat[157] N
1.7573
2.476
5.028 *
MedakaCtrlAdult, 21 d99 Pairs were exposed. During the exposure period, the spawned eggs were collected and the fertilized eggs were counted.
[147] A
83798
172099
312099
BPSZebrafishCtrlEmbryos, 75 d 90Embryos were exposed to BPS. Males and females were assigned to new tanks and acclimatized for 3 d. Numbers of spawned eggs were recorded daily for the next 7 d.
[162] N
0.1 88
1 85
10 48 *
100 42 *
CtrlAdult, 21 d 98Fertilized eggs of exposed female and male were collected at 16 d post exposure, and were exposed to same BPS concentrations until 6 d post fertilization, and hatching rate was determined.[159] A
0.5 28 *
5 21 *
50 10 *
BPAFZebrafishCtrlEmbryos, 120 d7699Embryos were exposed to BPAF for 120 d. The number embryo/larvae was recorded at 7 d post fertilization.
[160] N
57091
257592
12549 *94
MEHPZebrafishCtrlPre-adult (2 month-old), 81 d 82 a, 82 bOne hundred fertilized eggs of male and female exposed to MEHP were collected and divided into group (a) receiving the same MEHP concentrations, and group (b) receiving no further MEHP. The hatching rate was determined during 6 d post fertilization. [165] A
0.47 82 a, 54 b,*
4.0 67 a, 57 b,*
37.5 65 a,*, 47 b,*
DEHPZebrafishCtrlAdult, 21 d 64Males were mated with untreated females. Over a period of 14 d, the hatching rate was calculated.[166] N
0.2 3 *
20 2 *
CtrlAdult, 10 d83 a, 81 b Males were injected into the intraperitoneal cavity (mg/kg). Each aquarium contains 2 females and 2 males. Following 1–5 d (a) and 6–10 d (b) of treatment, eggs were collected and fertilization rate was recorded.[153] A
0.578 a, 80 b
5088 a, 78 b
500075 a, 48 b,*
FLUMedakaCtrlAdult, 21 d95 a, 97 b, 95 c Pairs were exposed. All spawned eggs were collected from female fish and fertility of each pair were checked daily for 3 weeks: (a) 1 week, (b) 2 weeks, and (c) 3 weeks post exposure.
[169] A
10096 a, 96 b, 94 c
20099 a, 95 b, 99 c
40094 a, 100 b, 95 c
79096 a, 99 b, 95 c
156018 a,*, 50 b,*, 49 c,*
VZFathead minnow0, 60, 250. 450Adult, 28 dn.s.n.s.Pairs were exposed. The spawned eggs were collected and the fertilized eggs (fertility of the mating pairs) and hatching rate were counted during exposure.[148] A
MCPGuppyCtrlEmbryos, 90 d 34Pairs of guppies were exposed in a semi-static exposure system. Number of newly hatched offspring produced per female were counted.
[158] N
10 21 *
100 10 *
1000 9 *
MMSBrown troutCtrlAdult, 21 d9493Males were injected into the intraperitoneal cavity (mg/kg). Sperm (100 μL) were added into 40 g eggs. Fertilization medium was SAFD2. Fertilization rate was checked after 120 degree-days of development of eggs.[151] A
509594
Arctic charr0Adult, 21 d8475
508376
(*) Values with asterisk show statistically significant difference compared to the control. (A) Superscript of A in the Author’s column shows the actual concentrations of the contaminant. (N) Superscript of N in the Author’s column shows the nominal concentrations of the contaminant. (n.s.) No significant effects were observed. Fish species: Arctic charr (Salvelinus alpinus), brown trout (Salmo trutta f. fario), fathead minnow (Pimephales promelas), fighting fish (Betta splendens), guppy (Poecilia reticulate), medaka (Oryzias latipes), rainbow trout (Oncorhynchus mykiss), rare minnow (Gobiocypris rarus), zebrafish (Danio rerio). Environmental contaminants (ECs): 17α-methyltestosterone (17α-MT), 17α-ethinylestradiol (EE2), 17β-estradiol (E2), 4-nonylphenol (4-NP), bisphenol A (BPA); bisphenol AF (BPAF), bisphenol S (BPS), di-2-ethylhexyl phthalate (DEHP), flutamide (FLU), mono-(2-ethylhexyl)-phthalate (MEHP), monocrotophos (MCP), methyl methane sulfonate (MMS), vinclozolin (VZ). 1 Composition of the motility-inhibiting saline solution (MIS): 103 mM NaCl, 40 mM KCl, 1 mM CaCl2, 0.8 mM MgSO2, 20 mM Tris, pH 7.8. 2 Composition of the salmonid artificial fertilization diluent: NaCl solution, 250 mOsmol/kg, 0.05 M-glycine, 0.02 M-Tris-buffer, pH 9.0. 3 Actual concentrations of EE2: 31 and 245 ng/L (nominal 60 and 480 ng/L), respectively. 4 Actual concentrations of EE2: 0.7 and 0.8 ng/L (nominal 1 ng/L) and 8.1 and 7.8 ng/L (nominal 10ng/L) for male and female aquaria, respectively. 5 Actual concentrations of 17α-MT: 0.11 and 0.09 μg/L (nominal 0.1 μg/L) and 42.5 and 48.2 μg/L (nominal 50 μg/L) for male and female aquaria, respectively.
The number of sperm per an oocyte influences fertilization success, in vitro (see Section 4.1). Schultz et al. [155] examined various numbers of sperm per an oocyte to examine the effects of EE2 on male fertility in rainbow trout. They observed that sensitivity for detecting EE2 adverse effects on sperm fertilizing ability are lost at lower number of sperm per an oocyte, since the fertilization rate was low in the control group. Therefore, at first, it is essential to determine the minimum number of sperm per an oocyte to achieve an adequate fertilization rate if one uses an in vitro fertilizing assay to examine the effects of ECs on fertility.
Taken together, laboratory studies indicate that ECs are capable of reducing fertility in male fishes. However, the ECs-related fertility threat shows differences among species, and depends on concentration of the contaminant and duration of exposure. In the next sections, the biological targets through which ECs effect on male fertility are discussed.

6.2. Effects of Environmental Contaminants (ECs) on Sperm Production

There are a few studies that have clarified the percentage of males undergone spermiation following exposure to a contaminant (Table 3). Schoenfuss et al. [170] and Lahnsteiner et al. [157] reported significant decrease in the number of spermiating males of adult goldfish and brown trout (Salmo trutta f. fario) exposed to 0.05 µg/L E2 and 5 µg/L BPA for 70 and 76 d, respectively. McAllister and Kime [171] reported full suppression of spermiation in zebrafish males that have been exposed to 0.01 µg/L TBT from early developmental stages. These show that ECs are capable of inhibiting spermiation in male fishes.
Studies have shown reduction of sperm volume or density in male fishes exposed to E2 [156,170], EE2 [155,161], 4-NP [146], BPA [157,163,164,172,173,174], BPS [162], DEHP [175], MEHP [165], VZ [176,177,178], Flu [176,179], DDT [176], MCP [158], clofibrate acid [180], FLX [181], DES [179], and TBT [171,172] (Table 3). The adverse effects of ECs on sperm volume and density highly depend on concentrations of the contaminant. Generally, significant decreases in sperm volume or density have been observed when the concentration of contaminants or duration of exposure was increased. The effective concentration to affect sperm volume and density varies for a particular contaminant during similar duration of exposure. For examples, following 90 d of exposure, BPA significantly decreased sperm volume in goldfish at 0.2 µg/L, while sperm density was reduced at 20 µg/L [173,174]. In brown trout exposed to BPA for 38 d, sperm volume and density were decreased at 5 and 1.75 µg/L, respectively [157]. In zebrafish exposed to the metabolite of DEHP for 81 d, sperm volume and density were decreased at 37.5 and 4.9 µg/L MEHP, respectively [165]. However, a particular contaminant may affect one of the sperm production indices. For an example, in rainbow trout exposed to 0.13–0.75 µg/L 4-NP for 30 d, sperm volume was decreased at lowest dose (0.13 µg/L), while sperm density remained unchanged [146].
Laboratory studies also show that effectiveness of a contaminant on sperm production depends on duration of exposure [146,156,157,171,181]. For instances, exposure of rainbow trout to 0.001 µg/L E2 resulted in significant reduction of sperm volume and density following 35 and 50 d of exposure, respectively [156].
Similar to the effects of ECs on fertility, it is likely that there are species-specific effects of ECs on sperm production when similar developmental stage is considered in exposure test. For an example, when mature goldfish [174] and rainbow trout [157] were exposed to BPA, sperm volume was reduced at 0.2 and 5 µg/L, respectively. Exposure to 0.01 µg/L EE2 decreased sperm density in mature rainbow trout, while it was without effects on one-sided livebearer at 0.01–0.15 µg/L [155,182].
Taken together, laboratory studies indicate that ECs interfere with sperm production to affect male fertility. The degree of ECs effects depends on concentration and duration of exposure.
Table 3. Laboratory studies on the potential adverse effects of environmental contaminants (ECs) to impair spermiation and sperm production indices (volume and density) in male fishes.
Table 3. Laboratory studies on the potential adverse effects of environmental contaminants (ECs) to impair spermiation and sperm production indices (volume and density) in male fishes.
ECsModelDose
(µg/L)
ExposureSpermiation
(%)
Volume
(mL)
Density
(×109 sperm/mL)
Authors and Notes
E2GraylingCtrlAdult, 50 d 0.3 Authors: [156]
0.0011 0.1 *
Rainbow troutCtrlAdult, 50 d 4.1 a, 3.8 b6.1 a, 6.3 bAuthors: [156]; Analysis performed 35 d (a) and 50 d (b) of exposure; Sperm volume: Sperm mass (g)
0.0004 4.0 a, 2.8 b6.0 a, 6.5 b
0.0011 2.0 a,*, 1.5b,*5.6 a, 5.4 b,*
0.0026 2.2 a, 1.4 b,*5.7 a, 3.9 b,*
GoldfishCtrlAdult, 70 d91 a, 94 b Authors: [170]; This study was performed in winter (a) and summer (b).
0.0567 a,*, 40 b,*decrease (data not shown)decrease (data not shown)
EE2One-sided livebearerCtrlAdult, 28 d 2.4 Authors: [182]; Sperm density: ×106 sperm/mL.
0.005 2.2
0.064 2.2
0.127 2.0
Rainbow troutCtrlAdult, 62 d 7.9Authors: [155]
0.016 20 *
0.131 30 *
4-NPRainbow troutCtrlAdults, 60 d 5.9 a, 6.0 b7.2 a, 7.0 bAuthors: [146]; Analysis performed at 30 d (a) and 60 d (b) of exposure
0.13 3.8 a,*, 1.5 b,*7.4 a, 6.8 b
0.28 4.0 a,*, 1.0 b,*6.9 a, 6.6 b
0.75 2.7 a,*, 0.1 b,*7.3 a, n.d. b,*
BPARare minnowCtrlAdult, 21 d 2.0 a, 2.0 b, 1.9 cAuthors: [168]; Analysis performed at 7 d (a), 14 d (b), and 21 d (c) of exposure; Sperm density: ×1013 sperm/male
11 2.0 a, 2.1 b, 1.9 c
206 2.2 a, 1.9 b, 1.8 c
ZebrafishCtrlEmbryos, 150 d 1.24.0 Authors: [164]; Sperm volume: ×107 sperm/testis; Sperm density: ×109 sperm/g testis
0.228 1.0 *4.0
2.28 1.14.0
22.8 1.2 4.0
CtrlLarvae, 150 d 1.34.5
0.228 0.8 *3.5 *
2.28 1.24.5
22.8 1.34.5
CtrlAdult, 150 d 1.23.5
0.228 1.13.5
2.28 1.23.5
22.8 0.6 *1.8 *
ZebrafishSolventEmbryos, 150 d 2.8Authors: [163]; Solvent contains 0.03 BPA. Sperm density: ×109 sperm/g testis
0.37 2.2 *
GoldfishCtrlAdult, 90 d 207.112.9Authors: [173,174]; Sperm volume: μL
0.2 52.9 *11.1
2.0
20
73.3 *
58.8 *
10.8
9.2 *
Brown troutCtrlAdult, 76 d70 a, 80 b, 70c0.5 a, 0.5 b, 0.5 c29.1 a, 25.1 b, 23.3cAuthors: [157]; Analysis performed at day 38 (a), 56 (b), and 76 (c) of exposure; Sperm volume: Sperm mass (g)
1.75 80 a, 80 b, 60c0.6 a, 0.6 b, 0.5 c26.6 a,*, 26.0 b, 24.1 c
2.4 60 a, 80 b, 60c0.6 a, 0.5 b, 0.7 c26.8 a,*, 25.8 b, 24.2 c
5 10 a, b, c,و0.1 a,*, 0.02 b,*, 0.1 c26.2 a,*, 25.2 b, 25.4 c
GuppyCtrlAdult, 21 d 8.0 Authors: [172]; Sperm density: ×106 sperm/mL
274 5.1 *
549 2.0 *
BPSZebrafishCtrlEmbryos, 75 d 7.8Authors: [162]
0.1 7.1
1 6.9
10 4.5 *
100 2.9 *
FLXMosquitofishCtrlAdult, 30 d 13Authors: [183]; Sperm density: ×106 sperm/g testis
0.042 19 *
0.479 17 *
GoldfishCtrlAdult, 14 d 23 a, 0.9 b Authors: [181]; Analysis performed at 7 d (a) and 14 d (b) of exposure; Sperm volume: μL
0.375 10 a,*, 0.5 b
45 0.7 a,*,0.2 b,*
VZGoldfishCtrlAdult, 30 d 90 Authors: [178]; Sperm volume: μL
100 53 *
400 15 *
800 6 *
GuppyCtrlAdult, 30 d 3.0 Authors: [177]; VZ: μg per mg dry food; Sperm density: ×106 sperm/male
0.1 2.3
1 1.7 *
10 1.0 *
GuppyCtrlAdult, 30 d 4.9Authors: [176]; VZ: μg per mg dry food; Sperm density: ×106 sperm/male
1 3.6
10 3.1
MEHPZebrafishCtrlPre-adult (2 month-old), 81 d 8.39.8Authors: [165]; Sperm volume: ×107 sperm/d; Sperm density: ×1010 sperm/mL
0.46 7.49.1
4.0 7.18.6 *
37.5 6.8 *7.2
DEHPGoldfishCtrlAdult, 30 d 90.4 Authors: [175]; Sperm volume: μL
1 3.7 *
10 40.8 *
100 8.7 *
FLUZebrafishCtrlAdult, 30 d 1.5Authors: [179]; Sperm density: ×107 sperm/mL
0.279 0.7 *
Guppy CtrlAdult, 30 d 4.9Authors: [176]; Flu: μg Flu per mg dry food Sperm density: ×106 sperm/male
1 2.9 *
10 1.4 *
DESZebrafishCtrlAdult, 30 d 1.5Authors: [179]; Sperm density: ×107 sperm/mL
0.095 0.5 *
TBTGuppyCtrlAdult, 21 d 8.0Authors: 170; Sperm density: ×106 sperm/mL
0.011 3.0 *
0.022 2.7 *
ZebrafishCtrlEmbryos,
70 d
0.5 a, 0.7 b Authors: [171]; Analysis performed at day 30 (a) and 70 (b) of exposure. At 0.01 µg/L no male was spermiated.
0.00001 0.4 a, 1.1 b
0.001 0.6 a, 2.1 b,*
0.1 1.9 a,*, 3.0 b,*
p,p ′-DDEGuppyCtrlAdult, 30 d 4.9Authors: [176]; p,p′-DDE: μg per mg dry food; Sperm density: ×106 sperm/male
0.1 3.8 *
1 6.7 *
MCPGuppyCtrlEmbryos,
90 d
3.4Authors: [158]; Sperm density: ×106 sperm/mL
10 1.9 *
100 2.2 *
1000 2.0 *
ClofibrateFathead
minnow
CtrlAdult, 21 d 4.8 a, 1.2 b, 1.0 c Authors: [180]; a, 53–57 females: 8–12 males; b, 6 females: 6 males, and c, 2 females: 10 males; Sperm volume is measured in ×106 sperm/mg testis
1 0.8 c
10 0.3 b,*, 0.8 c
1000 1.7 a,*, 0.5 b,*, 1.1 c
MSTPsGoldfishCtrlAdult, 70 d91a, 94b Authors: [170]; This study was performed in winter (a) and summer (b)
100%100a, 93bn.s.n.s.
(*) Values with asterisk show statistically significant difference from the control. (n.d.) Not determined. (n.s.) No significant difference was observed compared to control (data are not shown in the article). Fish model: Arctic charr (Salvelinus alpinus), brown trout (Salmo trutta f. fario), fathead minnow (Pimephales promelas), goldfish (Carassius auratus), grayling (Thymallus thymallus), guppy (Poecilia reticulate), medaka (Oryzias latipes), mosquitofish (Gambusia holbrooki), one-sided livebearer (Jenynsia multidentata), rainbow trout (Oncorhynchus mykiss), rare minnow (Gobiocypris rarus), zebrafish (Danio rerio). Environmental contaminants (ECs): 17α-ethinylestradiol (EE2), 17β-estradiol (E2), 4-nonylphenol (4-NP), bisphenol A (BPA), bisphenol S (BPS), di-2-ethylhexyl phthalate (DEHP), diethylstilbestrol (DES), flutamide (Flu), mono-(2-ethylhexyl)-phthalate (MEHP), monocrotophos (MCP), fluoxetine (FLX); p,p’-1,1-dichloro-2,2-bis (p-chlorophenyl) ethylene (p,p’-DDE), tributyltin (TBT), vinclozolin (VZ). Source of contaminants: Municipal sewage treatment plants (MSTPs).

6.3. Effects of Environmental Contaminants (ECs) on Sperm Morphology

Abnormalities in sperm morphology has been observed in zebrafish exposed to 0.0001 and 0.01 µg/L TBT for 30-70 d [171], in African catfish (Clarias gariepinus) exposed to 100 µg/L 4-NP for 15 d [184], in goldfish exposed to 400–800 µg/L VZ for 30 d [178], and in rare minnow (Gobiocypris rarus) exposed to 15 and 225 µg/L BPA [168].
The nuclear vacuoles observed in the sperm of African catfish exposed to 4-NP [184] suggest that 4-NP possess potency to damage DNA contents of sperm as there is a positive relationship between sperm nuclear vacuoles and DNA damage [185]. Recently, Zhang et al. [168] reported that number of swelling sperm was increased in rare minnow exposed to 15 and 225 µg/L BPA for two or three weeks. It is unclear whether the swelling of sperm was due to the presence of vacuoles; however, if not artifact, some vacuole-like structures were observed. Abnormalities in the sperm plasma membrane, mitochondria and flagellum were observed in sperm of zebrafish, goldfish, and rare minnow exposed to TBT, VZ, and BPA, respectively [168,171,178]. Morphological abnormalities of sperm were accompanied by decrease in the duration of sperm motility in BPA-exposed rare minnow and decreases in sperm motility or velocity in TBT-exposed zebrafish and VZ-exposed goldfish, suggesting that ECs are capable of interfering with sperm motility, as plasma membrane, mitochondria, and flagellum are involved in the osmolality-induced ATP-dependent axonemal beating of sperm (see Section 2.5).
Taken together, ECs cause abnormalities in sperm that can negatively affect sperm motility.

6.4. Effects of Environmental Contaminants (ECs) on Sperm Genome

In reproduction, a haploid set of the chromosomes for normal development of embryos comes from the sperm [85,186]. Sayed et al. [184] reported a decrease in the size of sperm nucleus following exposure of African catfish to 100 µg/L 4-NP. This suggests that 4-NP affected the sperm genome as a correlation exists between the nucleus size and ploidy level of sperm [86].
It has been shown that changes in the sperm chromosome number and DNA content may lead to unsuccessful fertilization or to severe genetic disease in the offspring [84,187]. Brown et al. [188] reported increases in aneuploidy sperm formation in rainbow trout exposed to 0.01 µg/L EE2 for 50 d. In addition, embryos propagated from exposed males to EE2 have shown increases in aneuploidy levels, both hypoploid (haploid) and hyperploid (triploid). Therefore, it could be speculated that observed fertility loss in rainbow trout [155] and fathead minnow [144] exposed to EE2 might be also related to alternations in sperm genome.
In addition to ECs-related aneuploidy sperm formation, ECs may damage DNA or cause epigenetic modifications that can shape susceptibility to disease and result in diverse phenotypes. Damage to the DNA of sperm has been reported in males of brown trout and Arctic charr following 21 d of a single intraperitoneal injection of genotoxicant MMS [151]. In this study, the author also reported aneuploidy-induced mortality and morphological abnormalities in progeny, suggesting that MMS changes in sperm genome were inherited to offspring. Corradetti et al. [166] also reported an increase in the DNA fragmentation rate in the germ cells of zebrafish exposed to 0.2 and 20 µg/L DEHP, suggesting the potential of phthalates to induce oxidative stress in the testes with consequent damage to the sperm genome.
Regarding epigenetic modifications, it has been reported that BPA affect DNA methylation in the gonads of rare minnow [189,190]. These studies have shown hypermethylation of global DNA in the testes of rare minnow exposed to 15 and 225 µg/L BPA for 7 d. The BPA-induced hypermethylation was associated with increases in DNA methyltransferase proteins that function as methylation writers and maintainers at 225 µg/L, and with decrease in ten-eleven translocation proteins that function as methylation eraser at 15 µg/L [190,191].
Taken together, these studies indicate that ECs are capable of causing damage to DNA, changing ploidy level or causing epigenetic modifications. Thus, ECs-related fertility threat might be related to alternations in sperm genome.

6.5. Effects of Environmental Contaminants (ECs) on Sperm Motility Kinetic

Over the past 20 years, studying sperm motility kinetics received high considerations to elucidate ECs-related fertility threat in fishes (Table 4). Studies show that sperm motility has been decreased in fish exposed to E2 and EE2 [156,170,182], 4-NP [146], BPA [157,163,164,173,174], DEHP [175], VZ [178], LNG [192], and Cu [193]. The effects of P and inclofibrate acid, alderin, FLX, and DMDT were uncertain or non-significant trends toward decrease in sperm motility were observed [180,194,195,196]. The efficiency of the contaminants to interfere with sperm motility highly depends on concentrations of the contaminants and duration of exposure. Generally, a greater decrease in sperm motility is reported when concentrations of ECs are increased (Table 4). As examples, McAllister and Kime [171] reported TBT potential to decrease and totally suppress sperm motility in zebrafish at 0.001 and 0.1 µg/L following 30–70 d of exposure from the early developmental stage, respectively. Hatef et al. [178] and Golshan et al. [175] reported that sperm motility was decreased following 30 d exposure of adult goldfish exposed to 400 and 800 µg/L VZ, and to 100 µg/L DEHP, while 100 µg/L VZ and 1 or 10 µg/L DEHP were without effects. Prolongation of the duration of exposure also results in higher effects on sperm motility. As examples, sperm motility was decreased in goldfish exposed to 4.5 µg/L BPA for 20 or 30 d, while it was not affected following 10 d of exposure [173]. Additionally, the adverse effects of a particular EC on sperm motility seems to be species-specific, if sperm motility evaluated at a similar time post initiation of motility among species with exposure test performed at similar developmental stage. For instance, exposure of adult grayling to E2 was reported to decrease sperm motility at 0.001 µg/L, while E2 was without effect on sperm motility in rainbow trout exposed to >0.001 µg/L [156]. In addition, the adverse effects of a particular contaminant are comparable in a same fish species. For an example, VZ, DEHP, and BPA reduced sperm motility in goldfish following 30 d exposure to 800, 100, and 4.5 µg/L, respectively [173,175]. In rainbow trout, significant effects of 4-NP on sperm motility were observed at 0.75 µg/L following 60 d exposure [146], while EE2 up to 0.1 µg/L were without effects on sperm motility following a 62 d exposure [155].
Most studies have evaluated sperm motility at one time post sperm activation. However, analyses of sperm motility at various time post activation could result in better elucidation of dose-dependent effects of ECs [173,174,175,178]. For an example, sperm motility was decreased in goldfish exposed to BPA for 90 d at 20 µg/L and 0.2 µg/L when it was evaluated at 15 and 60 s post sperm activation, respectively [174]. In another study, when sperm motility in goldfish exposed to VZ for 30 d was evaluated at 15 and 60 s post sperm activation, decreases were observed at 800 and 400 µg/L, respectively [178].
The ECs effects on sperm velocity have been shown by analyses of VCL, VSL, and VAP in the laboratory studies (Table 4). The VCL was decreased in fishes exposed to P [194], LNG [192], BPA [164,173,174], DEHP [175], VZ [178], LNG [192], clofibrate acid [180], and TBT [171]. The VSL showed significant decreases following exposure of fishes to P [194], LNG [192], BPA [164], and clofibrate acid [180]. The VAP of motile sperm were decreased in fishes exposed to E2 [156], P [194], and BPA [157,164].
Changes in velocity parameters identify the wave form of the flagellum of sperm during motility period. There are three models identified from the effects of ECs on sperm velocity parameters in vitro [26]. In the first model, VCL does not change, while VSL or VAP decreases. These changes have been observed in fishes exposed to E2 or BPA (Table 4), suggesting that sperm motility trajectory was changed from a straight direction to a circular direction. In the second model, all three parameters including VCL, VSL, and VAP are decreased. This model has been reported in fishes exposed to P, EE2, and BPA (Table 4), which clearly indicating changes in sperm trajectory from straight to circular. Compared to the first model, the diameter of sperm trajectories is generally smaller in the second model. There is also another model that has not been seen. The VCL decreases but VAP and VSL remain unchanged. In this case, sperm move a shorter distance over a similar period of time, while the smoothly curved direction of sperm movement remains unchanged.
Similar to the effects of ECs on sperm motility, ECs effects on sperm velocity also depend on concentrations and duration of exposure, and might be species-specific (Table 4). As an example, in adults exposure, E2 decreased sperm velocity in grayling at 0.001 µg/L, while it was without effects on sperm velocity in rainbow trout, goldfish and one-sided livebearer at 0.0026, 0.05, and 0.25 µg/L, respectively [156,170,197].
Similar to sperm motility, time point post sperm activation is very important to assess the effects of ECs on sperm velocity. For an example, VCL at 15 s post sperm activation was not differed in goldfish exposed to 0.6–11 µg/L BPA for 10, 20, or 30 d, while VCL showed a significant decrease when it was evaluated at 60 s post sperm activation [174].
Taken together, these studies indicate ECs effects on sperm motility and velocity, suggesting that ECs-related diminished fertility in males might be due to decreases in sperm motility kinetics.
Table 4. Laboratory studies on the potential adverse effects of environmental contaminants (ECs) to impair sperm motility kinetics in male fishes. Motility shows percentage of motile sperm evaluated at time post activation (TPA) following activation of sperm motility in an activation medium (AM). Sperm velocity characters are the curvilinear velocity (VCL), the straight line velocity (VSL), and the angular path velocity (VAP). Some studies have used an immobilizing medium (IM) to prevent spontaneous initiation of sperm motility at stripping or to predilute sperm before analysis of the motility.
Table 4. Laboratory studies on the potential adverse effects of environmental contaminants (ECs) to impair sperm motility kinetics in male fishes. Motility shows percentage of motile sperm evaluated at time post activation (TPA) following activation of sperm motility in an activation medium (AM). Sperm velocity characters are the curvilinear velocity (VCL), the straight line velocity (VSL), and the angular path velocity (VAP). Some studies have used an immobilizing medium (IM) to prevent spontaneous initiation of sperm motility at stripping or to predilute sperm before analysis of the motility.
ECsModelDose (µg/L)ExposureTPA (s)Motility (%)Velocity (µm/s)Authors and Notes
VCLVSLVAP
E2One-sided livebearerCtrlAdult, 28 d240 (every 10 s)79113110 Authors: [197]; AM: HAMF-10 medium
0.0578115111
0.1070115111
0.2575118116
Rainbow troutCtrlAdult, 50 d1089 78Authors: [156]; IM (mM): 103 NaCl, 40 KCl, 1 CaCl2, 0.8 MgSO2, 20 Tris, pH 7.8; AM (mM): 60 NaHCO3, 20 Glycin, pH 9.0; Analysis done at 35 d post exposure.
0.000487 82
0.001185 84
0.002680 90
GraylingCtrlAdult, 50 d1074 105Authors: [156]; IM and AM: See above row
0.00146 * 78 *
GoldfishCtrlAdult, 70 dn.d.2500 a, 3000 b 100 a, 62 b Authors: [170]; IM: Ringer solution; AM: DW; Motility was absolute number of motile sperm per fish. Exposures were performed in winter (a) and summer (b).
0.05 <10 a,*, <10 b,* 112 a, 75 b
PFathead minnowCtrlAdult, 7 d80 (every 15 s)53947590Authors: [194]; IM: 0.8 mM NaCl solution. AM: DW
0.02563.5785273
0.3393960 *38 *53 *
EE2ZebrafishCtrlEmbryos,
240 d
44 (every 2 s)588956 Authors: [198]; IM: n.d.; AM: n.d.; Solvent: DMSO
Solvent 498047
0.0002538749
0.00024387949
0.001478852
one-sided livebearerCtrlAdult, 28 d240 (every 10 s)82103101 Authors: [182]; AM: HAMF-10 medium
0.00575109105
0.06476107103
0.12765 *106109
Fighting fishCtrlAdult, 28 d60 (every 6 s) Authors: [161]; IM (g/L): 5.5 NaCl, 2 KCl, 3.8 Glycine, 2.4 Tris, pH 7.5; AM: Water containing EE2
0.01n.s.n.s.n.s.n.s.
0.1n.s.n.s.decreasen.s.
MedakaCtrlAdult, 21 d30 7060 Authors: [150]; AM (g/L): 1 NaCl, 0.03 KCl, 0.04 CaCl2, 0.1 MgCl2, 0.2 NaHCO3, pH 7.3
0.06 8885
0.12 8583
0.24 9075
0.48 8381
Rainbow troutCtrlAdult, 62 d12 (every 15 s) Authors: [155]; IM (mM): 103 NaCl, 40 KCl, 1 CaCl2, 0.8 MgSO2, 20 Tris, pH 7.8; AM (mM): 60 NaHCO3, 50 Tris, pH 9
0.016n.s.n.s.n.s.
0.131n.s.n.s.n.s.
LNGFathead minnow0Adult, 14 d80 (every 15 s)85688152Authors: [192]
0.01274 *6062 *48
0.12745 *47*58 *46
BPARare minnowCtrlAdult, 21 dn.d.97 a, b, c Authors: [168]; IM: Hank’s balance; AM: 0.55% NaCl
1197 a, 97 b, 96 c
20697 a, 96 b, 95 c
ZebrafishCtrlEmbryos, 150 dn.d. (15 frames each 0.05 s)651287487Authors: [164]; IM: Hank’s balance; AM: n.d.
0.22853*120 *58 *73*
2.2861113 *50 *68*
22.8611166276
CtrlLarvae, 150 d681286686
0.22855 *1286684
2.2861117 *6276
22.86212058 *72 *
CtrlAdult, 150 d651307487
0.22860125 *6884
2.2859125 *62 *76 *
22.845 *125 *60 *76 *
ZebrafishSolvent Embryos, 150 dn.d. (15 frames each 0.05 s)75100 Authors: [163]; IM: Hank’s balance; AM: n.d. Solvent contains 0.03 BPA
0.3736 *68
GoldfishCtrlAdult, 30 d1597 a, 100 b, 99 c157 a, 146 b, 166 c Authors: [173];AM (mM): 50 NaCl, 20 Tris, pH 8.5, osmolality 110 mOsmol/kg; Analyses performed at day 10 (a), 20 (b), and 30 (c) of exposure
0.694 a, 94 b, 95 c148 a, 156 b, 159 c
4.596 a,80 b,*,88 c,*158 a, 136 b, 158 c
11.098 a, 85 b,*,91 c,*144 a,138 b, 154 c
Ctrl6090 a, 79 b, 55 a, 30 b, 38 c
0.678 a, 74 b, 70 c,*34 a,*, 25 b,*, 30 c,*
4.585a, 61b,*, 68c,*29 a,*, 34 b,*, 38 c,*
11.085 a, 65 b,*, 61 c,*30 a,*, 29 b,*, 37 c,*
GoldfishCtrlAdult, 90 d1599168 Authors: [174]; AM (mM): 50 NaCl, 5 KCl, 20 Tris, pH 8.5, osmolality 110 mOsmol/kg
0.294155
295150 *
2088*140 *
Ctrl606460
0.251 *59
246 *48
2041 *47 *
Brown troutCtrlAdult, 76 d1084 a, 88 b, 86 c 97 a, 107 b, 106 cAuthors: [157]; IM (mM): 103 NaCl, 40 KCl, 1 CaCl2, 0.8 MgSO2, 20 Tris, pH 7.8; AM (mM): 60 NaHCO3, 20 Glycin, pH 9.0; Analyses were performed at day 38 (a), 56 (b), and 76 (c) of exposure
1.7533 a,*, 73 b, 70 c 62 a,*, 75 b,*, 111 c
2.440 a,*, 36 b,*, 81 c 75 a,*, 75 b,*, 92 c
54 a,*, 1 b,*, 4 c 32 a,*, 49 b,*, 41 c,*
4-NPRainbow troutCtrlAdult, 60 d1068 a, 75 b, 69 c 82 a, 92 b, 89 cAuthors: [146]; IM (mM): 103 NaCl, 40 KCl, 1 CaCl2, 0.8 MgSO2, 20 Tris, pH 7.8; AM (mM): 60 NaHCO3, 20 Glycin, pH 9.0; Analyses were performed at day 0 (a), 30 (b), and 60 (c) of exposure
0.1369 a, 70 b, 65 c 77 a, 96 b, 84 c
0.2872a, 75b, 81c 81a, 101b, 78c
0.7566a, 70b, n.d.c,* 82a, 91b, n.d.c,*
FLXMosquitofishCtrlAdult, 35 d 851126280Authors: [196]; IM (mM): 207 NaCl, 5.4 KCl, 1.3 CaCl2, 0.49 MgCl2, 0.41 MgSO4, 10 Tris, pH 7.5; AM: 150 mM KCl with 2 mg/mL BSA
0.031831156584
0.375841086177
VZGoldfishCtrlAdult, 30 d1599165 Authors: [178]; AM (mM): 50 NaCl, 20 Tris, pH 8.5 Osmolality 110 mOsmol/kg
10097159
40080137
80019*130*
Ctrl606464
1005258
40044*65
80014*60
DEHPGoldfishCtrlAdult, 30 d1598161 Authors: [175]; AM (mM): 50 NaCl, 5 mM KCl. 20 Tris, pH 8.5
186142
1088128*
10074*107*
Ctrl606767
146*60
1047*63
10038*68
ClofibrateFathead minnowCtrlAdult, 21 d5623022 Authors: [180]; IM (mM): 94 NaCl, 27 KCl, 50 Glycine, 15 Tris pH. 7.6; AM: Water; Sperm velocity in nm/s.
1562616
10512418
10005021 *11 *
TBTZebrafishCtrlFry,
70 d
5 78 a, 60 b Authors: [171]; IM (g/L): 5.8 NaCl, 0.2 KCl, 0.2 CaCl2, 0.04 MgCl2, 2.1 NaHCO3, 0.04 NaH2PO4, 3.8 glycine, pH 8.6; AM: DW; Analyses were performed at day 30 (a) and 70 (b) of exposure
0.00001 62 a, 62b
0.001 44 a,*, 19 b,*
0.01 n.d. a,*, 20 b,*
0.1 36 a,*, 0 b,*
MSTPsGoldfishCtrlAdult, 70n.d.2500 a, 3000 b 100 a, 62 b Authors: [170]; IM: Ringer solution; AM: DW; Sperm motility shows absolute number of motile sperm per fish. Exposures were performed in winter (a) and summer (b)
100%2000 a, 2700 b 100 a, 95 b
CuKillifishCtrlFry, 345 d 83 Authors: [193]; Ctrl contains 1.8 Cu
7.1 26 *
10.9 17 *
AlderinCatfish0 7510989103Authors: [195]; IM (g): 5.49 NaCl, 2.01 KCl, 3.75 glycine, 1.82 Tris; AM: Tank water
0.14 711008091
DMDT0.23 68867181
(*) Values with asterisk show statistically significant difference from the control. (DW) distilled water. (n.d.) Not determined. (n.s.) No significant difference was observed compared to control (data are not shown in the article). Environmental contaminants (ECs): 17α-ethinylestradiol (EE2), 17β-estradiol (E2), 4-nonylphenol (4-NP); bisphenol A (BPA), copper (Cu), di-2-ethylhexyl phthalate (DEHP), fluoxetine (FLX), methoxychlor (DMDT), levonorgestrel (LNG); progesterone (P), tributyltin (TBT), vinclozolin (VZ). Municipal sewage treatment plants, STPs; fish models: African catfish (Clarias gariepinus), brown trout (Salmo trutta f. fario), fathead minnow (Pimephales promelas), fighting fish (Betta splendens), goldfish (Carassius auratus), grayling (Thymallus Thymallus), killifish (Poecilia vivipara), medaka (Oryzias latipes), mosquitofish (Gambusia holbrooki), one-sided livebearer (Jenynsia multidentata), rainbow trout (Oncorhynchus mykiss), rare minnow (Gobiocypris rarus), zebrafish (Danio rerio). Source of contaminants: Municipal sewage treatment plants (MSTPs). Chemicals: Bovine serum albumin (BSA).
Table 5. Environmental contaminants (ECs)-related male fertility endpoints at environmentally relevant concentrations. The lowest and highest tested concentrations are shown as LTC and HTC, respectively.
Table 5. Environmental contaminants (ECs)-related male fertility endpoints at environmentally relevant concentrations. The lowest and highest tested concentrations are shown as LTC and HTC, respectively.
ECsEnvironmental
Concentration (ng/L)
Fish SpeciesLTC (ng/L)HTC
(ng/L)
FertilitySpermAuthors
ProductionMotilityVelocity
E20.1–5.01, 0.2–2.92, 0.63, 0.5–5.24, 0.4–3.35, 0.3–55.06, 2.7–48.07, LOD-3.711, 7.4820, LOD-7.426, LOD-84.327, LOD-33.428, <1–17529Zebrafish-25+n.d.n.d.n.d.[166] N
One-sided livebearer50250n.d.n.d.--[197] A
Rainbow trout0.42.6++--[156] N
Medaka29463-n.d.n.d.n.d.[154] A
Grayling-1.0n.d.+++[156] N
Goldfish-50n.d.++-[170] A
P7.4–11.81, 5.4–6.18Fathead minnow25339n.d.n.d.--[194] A
EE20.7–<2.02, 0.1–8.94, 0.045, 0.2–7.59, LOD-42.010, LOD-0.811; LOD-35.628, <0.8–34.029Fighting fish10100-n.d.--[161] N
Zebrafish0.21n.d.n.d.--[198] A
Medaka30480-n.d.n.d.-[150] A
Fathead minnow0.1100+n.d.n.d.n.d.[144] 1
Rainbow trout16131++--[155] A
Medaka33488-n.d.n.d.n.d.[149] A
One-sided livebearer10150n.d.---[182] A
17α-MT1.3–1.88, <0.9–14.512, 1.3313Medaka23380-n.d.n.d.n.d.[145] A
Fathead minnow10050,000-n.d.n.d.n.d.[143] 2
4-NPLOD-37,00011, LOD-10,18614, 15–38615, 77–114216, 112.4–2065.726, <0.5–211.029Medaka1,27089,400-n.d.n.d.n.d.[167] A
Rainbow trout130750++++[146] N
Medaka248184,000-n.d.n.d.n.d.[152] A
BPALOD-12,20514, LOD-112516, LOD-800017, 0.5–70218, 6.6–74.919, 12.3–755.626, <1–14529Rare minnow11,000206,000+--n.d.[168] A
Zebrafish22822,800++++[163,164] A
Brown trout1,7505,000++ + + [157] N
Medaka837,0003,120,000-n.d.n.d.n.d.[147] A
Goldfish20020,000n.d.+++[173] A, [174] N
Guppy274,000549,000n.d.-n.d.n.d.[172] N
BPS0.3–19.019Zebrafish100100,000--n.d.n.d.[162] N
Zebrafish50050,000-+ n.d.n.d.[159] A
BPAFLOD-15,00043Zebrafish5000125,000-n.d.n.d.n.d.[160] N
DEHP330–182,00018, 230–73020, 364–2.6821, 61.6–4352.022, 150–12,10041Zebrafish20020,000+n.d.n.d.n.d.[166] N
Zebrafish5005,000,000-n.d.n.d.n.d.[153] A
Goldfish1000100,000n.d.+++[175] N
VZ0.5–2023, 524, <1025Fathead minnow60,000450,000-n.d.n.d.n.d.[148] A
Goldfish100,000800,000n.d.---[178] N
Guppy10010,000n.d.-n.d.n.d.[177] A
Guppy100010,000n.d.-n.d.n.d.[176] A
DESLOD-3.327, LOD-8.528Zebrafish-95n.d.-n.d.n.d.[179] A
FLX0.4–2.630, 2.0–19.531; 4.7–9.432; LOD-12833Mosquitofish42479n.d.+--[183] A, [197] A
Goldfish37545,000n.d.±n.d.n.d.[181] A
FLU0.55–1.134, 12–3044Zebrafish-279n.d.±n.d.n.d.[179] A
Medaka100,0001,560,000- [169] A
Guppy100010,000n.d.±n.d.n.d.[176] A
ClofibrateLOD-17.232; 0.2–0.735, 6–700044Fathead minnow10001,000,000n.d.±--[180] A
MCP8.336, 16537, LOD-400038Guppy10,0001,000,000±±n.d.n.d.[158] N
MMS0.139Brown trout-50,000--n.d.n.d.[151] N
Arctic charr-50,000--n.d.n.d.[151] N
TBT0.4240Guppy1122n.d.±n.d.n.d.[172] N
Zebrafish0.01100n.d.±n.d.±[171] N
DDE36042Guppy1001000n.d.+n.d.n.d.[176] A
Aldrin14042African catfish 140n.d.n.d.--[196] N
DMDT23042African catfish 230n.d.n.d.--[196] N
(+) A significant decrease was observed at environmental relevant concentration. (–) No significant decrease was observed at the maximum tested concentration of the contaminant. (+/–) The lowest examined dose was higher than environmental relevant concentration, however significant decrease was observed. (A) Superscript of A in the Author’s column shows the actual concentrations of the contaminant. (N) Superscript of N in the Author’s column shows the nominal concentrations of the contaminant. (n.d.) Not determined. (LOD) Lower of Detection. Environmental contaminants (ECs): 17α-ethinylestradiol (EE2), 17α-methyltestosterone (17α-MT), 17β-estradiol (E2), 4-nonylphenol (4-NP); bisphenol A (BPA), Bisphenol AF (BPAF), Bisphenol S (BPS), p,p′-1,1-dichloro-2,2-bis (p-chlorophenyl) ethylene (p,p′-DDE), Di-2-ethylhexyl phthalate (DEHP), Diethylstilbestrol (DES), Flutamide (FLU), Fluoxetine (FLX), Methoxychlor (DMDT), Monocrotophos (MCP), Progesterone (P), Tributyltin (TBT), Vinclozolin (VZ). References for environmental concentrations of the contaminants: 1. Velicu and Suri [199]; 2. Vajda et al. [127]; 3. Jeffries et al. [123]; 4. Kuch and Ballschmiter [200]; 5. Baronti et al. [201]; 6. Nasu et al. [202]; 7. Desbrow et al. [203]; 8. Liu et al. [204]; 9. Belfroid et al. [205]; 10. Ternes et al. [206]; 11. Snyder et al. [207]; 12.Yu et al. [208]; 13. Blankvoort et al. [209]; 14. Höhne and Püttmann [210]; 15. Cailleaud et al. [211]; 16. Mohapatra et al. [212]; 17. Kang et al. [213]; 18. Fromme et al. [214], 19. Yang et al. [215]; 20. Bókony et al. [216]; 21. Wen et al. [217]; 22. Zhang et al. [218]; 23. Reedman et al. [219]; 24. Kreuger et al. [220]; 25. Zheng et al. [221]; 26. Wang et al. [222]; 27. Chen et al. [223]; 28. Lei et al. [224]; 29. Pojana et al. [225]; 30. Wu et al. [226]; 31. Paíga et al. [227]; 32. Lindim et al. [228]; 33. Sanots et al. [229]; 34. Yan et al. [230]; 35. Nentwig et al. [231]; 36. Anjum and Malik [232]; 37. Kang and Zhang [233]; 38. Kumari et al. [234]; 39. Canty et al. [235], 40. Guo et al. [236]; 41. Li et al. [237]; 42. Nibamureke et al. [238]; 43. Song et al. [239]; 44. Corcoran et al. [240]. Fish species: African Catfish (Clarias gariepinus), Arctic charr (Salvelinus alpinus), Brown trout (Salmo trutta f. fario), Fathead minnow (Pimephales promelas), Fighthing fish (Betta splendens), Goldfish (Carassius auratus), Grayling (Thymallus Thymallus), Guppy (Poecilia reticulate), Medaka (Oryzias latipes), Mosquitofish (Gambusia holbrooki), One-sided livebearer (Jenynsia multidentata), Rainbow trout (Oncorhynchus mykiss), Rare minnow (Gobiocypris rarus), Zebrafish (Danio rerio). 1 Actual concentrations of EE2: 0.7 and 0.8 ng/L (nominal 1 ng/L) and 8.1 and 7.8 ng/L (nominal 10 ng/L) for male and female aquaria, respectively. 2 Actual concentrations of 17α-MT: 0.11 and 0.09 μg/L (nominal 0.1 μg/L) and 42.5 and 48.2 μg/L (nominal 50 μg/L) for male and female aquaria, respectively.

7. Conclusions

Both wildlife and laboratory studies show the adverse effects of ECs on fertility in male fishes. However, the sites of action of ECs to reduce male fertility are highly diverse. They may reduce sperm production, cause damage to sperm morphology, alter the sperm genome, or decrease sperm motility and velocity (Figure 5).
It has been shown that in vitro exposure of semen (or sperm) to ECs causes damage to sperm and decreases sperm motility and velocity to decline sperm fertilizing ability; however, the effective concentrations are far exceeding the WHO recommended limits [26]. The present review on laboratory studies highlights that adverse effects of ECs on determinants of fertility (including sperm production, morphology, genome, and motility kinetics) can appear at environmentally relevant concentrations when male fishes are exposed to ECs in vivo (Table 5). These support the hypothesis that diminished fertility in fishes from wildlife might be related to the contaminants in the aquatic environment.
The present review shows that: (a) Unlike the source of contaminants from industrial, municipal, treatment plant, or agricultural activities, abnormalities in sperm morphology [137,139], damage to the sperm genome [137], decrease in sperm production (volume or density) [115,117,127,132,136], decrease in sperm motility or velocity [115,117,121,122,132,133,137], and diminished male fertility [115,132,134] have been observed in fishes from wildlife that were associated with delay in sexual maturation, decrease in testicular size, histological defects in the testis, and increase in female bias sex ratio and intersex (Table 1). (b) The laboratory studies reveal that ECs reduce male fertility in concentration-dependent and exposure time-dependent manners. There is at least one study that shows E2, EE2, 17α-MT, 4-NP, BPA, BPS, and DEHP and its metabolite (MEHP) cause diminished fertility at environmentally relevant concentration (Table 2 and Table 5). (c) The ECs-related diminished fertility in males at environmentally relevant concentrations is associated with the adverse effects of ECs on at least one of the determinants of fertility. There is at least one study that shows the adverse effects of almost all examined ECs on sperm production, morphology, genome, or motility kinetics at environmentally relevant concentrations (Table 3, Table 4 and Table 5). It is worth to note that both wildlife and laboratory studies [13,17,18,19,20,21,22,23,24,25] suggest that the adverse effects of ECs on the determinants of fertility that cause diminished fertility were associated with alternations in hormonal functions of HPT and increased circulating Vtg levels (Table 1). However, our aim in the present study was not to review the effects of ECs on hormonal functions of HPT.

8. Future Research Directions

The present review highlights the following aspects that should be taken into account in further studies to better elucidate the ECs-related fertility threat in male fishes.
(a) One can easily distinguish that the number of contaminants that have been used in fertility tests are comparable to the number of contaminants in our environment. Future studies should examine the effects of other chemicals with major public concerns. Among them, there is an immediate need to examine the effects of pharmaceuticals (such as antidepressants, antibiotics, antidiabetics, and contraceptives) that are highly consumed in global medicine use, agriculture, and animal husbandry [24,241]. Fertilizers and biocides (such as antimicrobial compounds and pesticides), personal care products (such as antifungal agents and cosmetics), and industrial contaminants (such as heavy metals, plasticizers and polycyclic aromatic compounds) are other types of contaminants that need immediate consideration [242,243,244].
(b) Examination of the adverse effects of ECs at environmentally relevant concentrations that cover the lowest and highest detected concentrations is highly needed. Most studies have failed to include the lowest environmental concentration among treatments (Table 5).
(c) The ontogeny-dependent effects of the ECs on male fertility are largely unknown. Basically, developmental stages are the main parameters to be carefully defined based on the objective. For instance, if the study is aiming at investigating the adverse effect of ECs on sperm production, it is a prerequisite to expose the fish from the early developmental stage of the testis. However, the present review shows that a number of studies that exposed fishes to ECs at early life stages is considerably lower than those that examined the adverse effects of ECs in adult-exposed individuals (Table 2, Table 3 and Table 4). When embryos are exposed, the study reports the adverse effects of ECs throughout the life, which is close to the physiological condition in the environment. It is worth to note that it has been hypothesized that the effects of ECs is high at the early developmental stage due to the fact that alternations in normal developmental and physiological phenomena are irreversible [245]. One of the key advantages and significant reasons for studying the adverse effects of ECs at different developmental stages is to elucidate the condition in which a fish species displays migration within its life cycle. In this regard, we could find only one study that compared the adverse effects of ECs at different developmental stages [164]. The authors reported that the adverse effects of BPA on sperm production and number of spermiated males were highly differed if embryos, larvae, or adults were exposed (Table 3 and Table 4).
(d) Assessment various types of determinants of fertility to better elucidate site of action of an EC that cause diminished fertility. There are a few studies that have evaluated the ECs effects on sperm production and motility (Table 5).
(e) The present study shows that ECs may cause phenotypic changes in the reproductive system and affect determinants of fertility (Figure 6). It has been well known that ECs disrupts hormonal regulation of spermatogenesis to decrease sperm production [17,20,23,25,27]. However, our knowledge on mechanisms of action of ECs on sperm morphology, genome, and motility are largely unknown.
To better elucidate ECs effects on sperm morphology, future studies should investigate functional structure of sperm including nucleus, mitochondria, centrioles, and axoneme, with particular consideration to the organization of the microtubules and motor protein called “dynein” in the latter case [168,171,178,184,246,247]. Studying structure and components, integrity, and permeability of the sperm plasma membrane can help us to better understand association between ECs-induced abnormalities of sperm and its motility kinetics.
Regarding the ECs effects on sperm genome, it is needed to examine effects of ECs on incidence of aneuploidy [188], to study nuclear proteins including DNA binding proteins and DNA transcription and replication using omics tools [246], and to study sperm epigenetic functions (see Section 8).
Regarding sperm motility, there are a few studies that show that the ECs effects on sperm motility kinetics might be related to a decrease in the ATP source required for flagellar beating or to damage to sperm morphology [135,161,168,171,178,184,247]. However, the effects of ECs to interfere with intracellular messengers (such as pH, Ca2+, and cAMP) that regulate sperm motility signaling are fully unknown and should be considered in future studies [246]. Schiffer et al. [248] showed that ECs alters intracellular Ca2+ through activation of a Ca2+ channel to interfere sperm motility in human. The laboratory studies suggest that ECs may affect sperm swimming pattern as sperm velocity parameters are decreased (Table 4). Future researches may investigate organization and functions of axonemal proteins involved in regulation of sperm motility and velocity as well as generation of reactive oxygen species (ROS) that may disrupt sperm motility signaling and may affect sperm velocity.
(f) Sperm requires energy for motility and flagellar beating force [66,68,106]. Our knowledge on the adverse effects of ECs on ATP content of sperm is very limited. Montgomery et al. [161] reported a decrease in sperm ATP content when a fighting fish (Betta splendens) was exposed to 0.1 µg/L EE2, which was associated with decrease in VSL. This suggests that the adverse effects of EE2 on sperm velocity could be related to amount of ATP available to generate flagellar beating force. Further studies are needed to investigate sperm energetics along with sperm motility kinetics to better understand the mechanisms through which ECs cause fertility threat.
However, assessment of the adenylate energy charge (AEC = ATP + 0.5 ADP/ATP + ADP + AMP) is suggested to better understand the adverse effects of ECs on sperm energetics and metabolism of energy [26,66,249]. Moreover, ECs may affect phosphocreatine production, which is necessary for maintenance of motility [246].
(g) In an aquatic environment, there are mixtures of ECs that may exhibit pharmacodynamic interactions. These interactions can be antagonistic or synergistic, which may cause an increase in the toxicity of a particular chemical through the cocktail effect [141,142]. For instance, Schoenfuss et al. [170] reported no changes in sperm production, motility, and velocity when goldfish adults were treated with municipal sewage treatment plants. However, they observed significant decrease in sperm production and motility when the goldfish were exposed to E2. Therefore, another very interesting issue that requires an immediate consideration is to examine the interaction effects of various contaminants in combination exposure tests. The results will also provide significant information to better understand the mechanistic effects of ECs on fertility.
(h) Our current knowledge on the ECs-related male fertility threat is mainly from freshwater fishes inhabiting lakes or rivers (Table 1). Future wildlife and laboratory studies should sample from the primitive fishes as well as marine fishes due to their significance in ecological impacts in the aquatic environment (such as fishes of coral reefs), biological conservation program (such as sturgeons and eels), or in commercial fisheries (such as ornamental fishes).
(i) Recently, epigenetic inheritance of the adverse effects of ECs received high scientific and public considerations. As ECs have been shown to affect the sperm genome, further studies need to characterize transgenerational inheritance of the adverse effects of ECs to the offspring. There are studies that show the adverse effects of bisphenols and 4-NP that were inherited to the next generations [163,167]; however, it is largely unknown for the other ECs. Chen et al. [163] reported that male offspring of adult zebrafish that have been exposed to BPA from early developmental stages showed a decrease in sperm production, while no effects on sperm motility or fertility were seen. These were reported for the offspring grown in BPA-free water. However, growing of offspring of zebrafish in the same concentrations of BPA resulted in decreases in sperm production, motility, and fertility, similar to their parents that were exposed to BPA from early developmental stages.
(j) The threat of ECs to fish populations is still an open question. There are historical reports that show populations of fishes have declined [250,251,252]; however, the contributions of ECs were not known. Hamilton et al. [252] suggested linking exposure and fish health at the population level, examining combinations of ECs, and studying the implications of population-genetic methods to address the adverse effects of ECs on fish populations. On their comments, the establishment of a long-term study that uses a model species from wildlife, investigates chronic effects of ECs, considers genetic diversity, and elucidates fertility within individuals of the population is needed. Moreover, development of an ecological habitat (such as artificial river) where a small fish population is routinely exposed to ECs in situ would be a great help.

9. Suggestions to Normalize Future Studies

We also would like to offer the following suggestions to normalize the studies for better understanding of the ECs-related fertility threat in fishes.
(a) The large variations among results of studies that have examined a particular contaminant suggest the need to optimize a protocol for harmonization of studies, which should encompass both biological and technical issues. These may include a protocol for optimizing the design of exposure tests, methods for evaluating male fertility, and laboratory tools to assess determinants of fertility. For example, (a-i) CASA has become very popular for the analyses of sperm motility kinetics. Technical conditions for recording of sperm motility and setting of CASA affect the results [253], thus need to be standardized for laboratory models. (a-ii) Most laboratory studies have used distilled water and/or activation medium to assess sperm motility kinetics (Table 4). To approach real effects of ECs, future studies should use the water of the aquarium where the fish were exposed to EC. (a-iii) Analysis of sperm motility kinetics needs to be performed at earliest time post sperm activation. It would be also useful if studies considered similar time periods to better understand the risk assessment of ECs. In this regard, analysis of sperm motility kinetics within 10 to 30 s post activation is recommended, as (a) one can operate the microscope and CASA easily [106] and (b) sperm fertilizing ability highly decreases after 30 s post activation [104]. (a-iv) As another example, indices of sperm production (sperm volume and density) could be expressed as total or normalized to body weight [28,106].
(b) The present study suggests species-specific adverse effects of an EC on male fertility and its determinants. To better understand sensitivity of a species to an EC, the effects of ECs could be examined on small laboratory fishes including zebrafish, guppy and medaka, which serve as aquatic model organisms. Application of species with different biology of reproduction will provide us with valuable information to assess ecological risk assessment of ECs. Among species, zebrafish and Medaka, in contrast to guppy, are external fertilizers, and the testes are of the tubular type in zebrafish and guppy compared to those of medaka that are of the lobular type.
(c) Although all studies have considered this note, it is worth reminding that one should take into account renewal of exposure medium based on the half-life of the EC.

Author Contributions

Yiteng Wu and Hongtao Yu proposed mathematical description of combination method; Yiteng Wu proposed and proved the theoretical limit theorem; Yiteng Wu and Ruiyang Huang designed the experiments and analyzed the results. Yingle Li and Senjie Lin wrote part of code.

Funding

This study was supported by the Ministry of Education, Youth and Sports of the Czech Republic (CENAKVA project No. LM2018099 and Biodiversity project No. CZ.02.1.01./0.0/0.0/16–025/0007370) to O. Linhart, and by the University of Tehran-Department of Environment of Iran (project No. 192/S/1400) to S.M.H. Alavi.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data analyzed for the present study are included in this published article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Richiardi, L.; Bellocco, R.; Adami, H.O.; Torrang, A.; Barlow, L.; Hakulinen, T.; Rahu, M.; Stengrevics, A.; Storm, H.; Tretli, S.; et al. Testicular cancer incidence in eight Northern European countries: Secular and recent trends. Cancer Epidemiol. Prev. Biomark. 2004, 13, 2157–2166. [Google Scholar]
  2. Andersson, A.M.; Jørgensen, N.; Main, K.M.; Toppari, J.; Meyts, E.R.D.; Leffers, H.; Juul, A.; Jensen, T.K.; Skakkebæk, N.E. Adverse trends in male reproductive health: We may have reached a crucial ‘tipping point’. Int. J. Androl. 2008, 31, 74–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Phillips, K.P.; Tanphaichitr, N. Human exposure to endocrine disrupters and semen quality. J. Toxicol. Environ. Health B Crit. Rev. 2008, 11, 188–220. [Google Scholar] [CrossRef]
  4. Lund, L.; Engebjerg, M.C.; Pedersen, L.; Ehrenstein, V.; Nørgaard, M.; Sørensen, H.F. Prevalence of hypospadias in Danish boys: A longitudinal study, 1977–2005. Eur. Urol. 2009, 55, 1022–1026. [Google Scholar] [CrossRef]
  5. Toppari, J.; Virtanen, H.E.; Main, K.M.; Skakkebaek, N.E. Cryptorchidism and hypospadias as a sign of testicular dysgenesis syndrome (TDS): Environmental connection. Birth Defects Res. A Clin. Mol. Teratol. 2010, 88, 910–919. [Google Scholar] [CrossRef] [PubMed]
  6. Mascarenhas, M.N.; Flaxman, S.R.; Boerma, T.; Vanderpoel, S.; Stevens, G.A. National, regional, and global trends in infertility prevalence since 1990: A systematic analysis of 277 health surveys. PLoS Med. 2012, 9, e1001356. [Google Scholar] [CrossRef] [Green Version]
  7. Agarwal, A.; Mulgund, A.; Hamada, A.; Chyatte, M.R. A unique view on male infertility around the globe. Reprod. Biol. Endocrinol. 2015, 13, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Inhorn, M.C.; Patrizio, P. Infertility around the globe: New thinking on gender, reproductive technologies and global movements in the 21st century. Hum. Reprod. Update 2015, 21, 411–426. [Google Scholar] [CrossRef] [Green Version]
  9. Skakkebaek, N.E.; Rajpert-De Meyts, E.; Buck Louis, G.M.; Toppari, J.; Andersson, A.M.; Eisenberg, M.L.; Jensen, T.K.; Jørgensen, N.; Swan, S.H.; Sapra, K.J.; et al. Male reproductive disorders and fertility trends: Influences of environment and genetic susceptibility. Physiol. Rev. 2016, 96, 55–97. [Google Scholar] [CrossRef] [Green Version]
  10. Landrigan, P.J.; Fuller, R.; Acosta, N.J.R.; Adeyi, O.; Arnold, R.; Basu, N.N.; Baldé, A.B.; Bertollini, R.; Bose-O’Reilly, S.; Boufford, J.I.; et al. Lancet Commission on pollution and health. Lancet 2018, 391, 462–512. [Google Scholar] [CrossRef] [Green Version]
  11. EFSA Scientific Committee. Scientific Opinion on the Hazard Assessment of Endocrine Disruptors: Scientific Criteria for Identification of Endocrine Disruptors and Appropriateness of Existing Test Methods for Assessing Effects Mediated by These Substances on Human Health and the Environment. EFSA J. 2013, 11, 3132. Available online: www.efsa.europa.eu/efsajournal (accessed on 10 April 2021). [CrossRef]
  12. Bergman, Å.; Heindel, J.J.; Jobling, S.; Kidd, K.A.; Zoeller, R.T. The State-of-the-Science of Endocrine Disrupting Chemicals; World Health Organization and United Nations Environment Programme Report–2012; WHO: Geneva, Switzerland, 2013; Available online: www.who.int/ceh/publications/endocrine/en/index.html (accessed on 10 April 2021).
  13. Kime, D.E. Endocrine Disruptors in Fish; Kluwer Academic Publishers: Boston, MA, USA, 1998. [Google Scholar]
  14. Kah, O. Endocrine targets of the hypothalamus and pituitary. In Fish Physiology: Fish Neuroendocrinology; Bernier, N.J., Van Der Kraak, G., Farrell, A.P., Brauner, C.J., Eds.; Elsevier Inc.: Amsterdam, The Netherlands, 2009; Volume 28, pp. 75–112. [Google Scholar] [CrossRef]
  15. Schulz, R.W.; de França, L.R.; Lareyre, J.J.; Le Gac, F.; Chiarini-Garcia, H.; Nobrega, R.H.; Miura, T. Spermatogenesis in fish. Gen. Comp. Endocrinol. 2010, 165, 390–411. [Google Scholar] [CrossRef]
  16. Dufour, S.; Quérat, B.; Tostivint, H.; Pasqualini, C.; Vaudry, H.; Rousseau, K. Origin and evolution of the neuroendocrine control of reproduction in vertebrates, with special focus on genome and gene duplications. Physiol. Rev. 2020, 100, 869–943. [Google Scholar] [CrossRef]
  17. Gregory, M.; Aravindakshan, J.; Nadzialek, S.; Cyr, D.G. Effects of endocrine disrupting chemicals on testicular functions. In Fish Spermatology; Alavi, S.M.H., Cosson, J., Coward, K., Rafiee, G., Eds.; Alpha Science Ltd.: Oxford, UK, 2008; pp. 161–214. [Google Scholar]
  18. Ankley, G.T.; Bencic, D.C.; Breen, M.S.; Collette, T.W.; Conolly, R.B.; Denslow, N.D.; Edwards, S.W.; Ekman, D.R.; Garcia-Reyero, N.; Jensen, K.M.; et al. Endocrine disrupting chemicals in fish: Developing exposure indicators and predictive models of effects based on mechanism of action. Aquat. Toxicol. 2009, 92, 168–178. [Google Scholar] [CrossRef]
  19. Scholz, S.; Klüver, N. Effects of endocrine disrupters on sexual, gonadal development in fish. Sex. Dev. 2009, 3, 136–151. [Google Scholar] [CrossRef] [PubMed]
  20. Bosker, T.; Munkittrick, K.R.; MacLatchy, D.L. Challenges and opportunities with the use of biomarkers to predict reproductive impairment in fishes exposed to endocrine disrupting substances. Aquat. Toxicol. 2011, 100, 9–16. [Google Scholar] [CrossRef] [PubMed]
  21. Leet, J.K.; Gall, H.E.; Sepúlveda, M.S. A review of studies on androgen and estrogen exposure in fish early life stages: Effects on gene and hormonal control of sexual differentiation. J. Appl. Toxicol. 2011, 31, 379–398. [Google Scholar] [CrossRef]
  22. Mennigen, J.A.; Stroud, P.; Zamora, J.M.; Moon, T.W.; Trudeau, V.L. Pharmaceuticals as neuroendocrine disruptors: Lessons learned from fish on prozac. J. Toxicol. Environ. Health. B Crit. Rev. 2011, 14, 387–412. [Google Scholar] [CrossRef]
  23. Hano, T. Studies on the evaluation of the effects of endocrine disrupting chemicals using transgenic see-through medaka (Oryzias latipes), olvas-GFP/STII-YI strain. Bull. Fish. Res. Agency 2012, 36, 1–56. [Google Scholar]
  24. Abdel-Moneim, A.; Coulter, D.P.; Mahapatra, C.T.; Sepúlveda, M.S. Intersex in fishes and amphibians: Population implications, prevalence, mechanisms and molecular biomarkers. J. Appl. Toxicol. 2015, 35, 1228–1240. [Google Scholar] [CrossRef]
  25. Golshan, M.; Alavi, S.M.H. Androgen signaling in male fishes: Examples of anti-androgenic chemicals that cause reproductive disorders. Theriogenology 2019, 139, 58–71. [Google Scholar] [CrossRef]
  26. Hatef, A.; Alavi, S.M.H.; Golshan, M.; Linhart, O. Toxicity of environmental contaminants to fish spermatozoa functions in vitro—A review. Aquat. Toxicol. 2013, 140–141, 134–144. [Google Scholar] [CrossRef] [PubMed]
  27. Carnevali, O.; Santangeli, S.; Forner-Piquer, I.; Basili, D.; Maradonna, F. Endocrine disrupting chemicals in aquatic environment: What are the risks for fish gametes? Fish Physiol. Biochem. 2018, 44, 1561–1576. [Google Scholar] [CrossRef] [PubMed]
  28. Alavi, S.M.H.; Linhart, O.; Coward, K.; Rodina, M. Fish spermatology: Implication for aquaculture management. In Fish Spermatology; Alavi, S.M.H., Cosson, J., Coward, K., Rafiee, G., Eds.; Alpha Science Ltd.: Oxford, UK, 2008; pp. 397–460. [Google Scholar]
  29. Dzyuba, V.; Shelton, W.L.; Kholodnyy, V.; Boryshpolets, S.; Cosson, J.; Dzyuba, B. Fish sperm biology in relation to urogenital system structure. Theriogenology 2019, 132, 153–163. [Google Scholar] [CrossRef]
  30. Grier, H.J. Cellular organization of the testis and spermatogenesis in fishes. Am. Zool. 1981, 21, 345–357. [Google Scholar] [CrossRef] [Green Version]
  31. Billard, R. Spermatogenesis and spermatology of some teleost fish species. Reprod. Nutr. Dev. 1986, 2, 877–920. [Google Scholar] [CrossRef]
  32. Nagahama, Y. The functional morphology of teleosts gonads. In Fish Physiology, Reproduction Part A.; Hoar, W.S., Randall, D.J., Donaldson, E.M., Eds.; Academic Press Inc.: London, UK, 1983; Volume IX, pp. 223–275. [Google Scholar]
  33. Parenti, L.R.; Grier, H.J. Evolution and phylogeny of gonad morphology in bony fishes. Integr. Comp. Biol. 2004, 44, 333–348. [Google Scholar] [CrossRef] [Green Version]
  34. Lahnsteiner, F.; Patzner, R.A. The spermatic duct of blenniid fish (Teleostei, Blenniidae): Fine structure, histochemistry and function. Zoomorphology 1990, 110, 63–73. [Google Scholar] [CrossRef]
  35. Lahnsteiner, F.; Patzner, R.A.; Weismann, T. The spermatic ducts of salmonid fishes (Salmonidae, Teleostei). Morphology, histochemistry and composition of the secretion. J. Fish Biol. 1993, 42, 79–93. [Google Scholar] [CrossRef]
  36. Miura, T.; Yamauchi, K.; Takahashi, H.; Nagahama, Y. The role of hormone in the acquisition of sperm motility in salmonid fish. J. Exp. Zool. 1992, 261, 359–363. [Google Scholar] [CrossRef]
  37. Morisawa, S.; Ishida, K.; Okuno, M.; Morisawa, M. Roles of pH and cyclic adenosine monophosphate in the acquisition of potential for sperm motility during migration from the sea to the river in chum salmon. Mol. Reprod. Dev. 1993, 34, 420–426. [Google Scholar] [CrossRef] [PubMed]
  38. Vizziano, D.; Fostier, A.; Loir, M.; Le Gac, F. Testis development, its hormonal regulation and spermiation induction in teleost fish. In Fish Spermatology; Alavi, S.M.H., Cosson, J., Coward, K., Rafiee, G., Eds.; Alpha Science Ltd.: Oxford, UK, 2008; pp. 103–140. [Google Scholar]
  39. Watanabe, A.; Onitake, K. The regulation of spermatogenesis in fish. In Fish Spermatology; Alavi, S.M.H., Cosson, J., Coward, K., Rafiee, G., Eds.; Alpha Science Ltd.: Oxford, UK, 2008; pp. 141–160. [Google Scholar]
  40. Jamieson, B.G.M. Fish Evolution and Systematics: Evidence from Spermatozoa; Cambridge University Press: Cambridge, UK, 1991. [Google Scholar]
  41. Hara, M.; Okiyama, M. An ultrastructural review on the spermatozoa of Japanese fishes. Bull. Oce. Res. Inst. Univ. Tokyo 1998, 33, 1–138. [Google Scholar]
  42. Lahnsteiner, F.; Patzner, R.A. Sperm morphology and ultrastructure in fish. In Fish Spermatology; Alavi, S.M.H., Cosson, J., Coward, K., Rafiee, G., Eds.; Alpha Science Ltd.: Oxford, UK, 2008; pp. 1–61. [Google Scholar]
  43. Ingermann, R.L. Energy metabolism and respiration in fish spermatozoa. In Fish Spermatology; Alavi, S.M.H., Cosson, J., Coward, K., Rafiee, G., Eds.; Alpha Science Ltd.: Oxford, UK, 2008; pp. 241–266. [Google Scholar]
  44. Inaba, K.; Shiba, K. Microscopic analysis of sperm movement: Links to mechanisms and protein components. Microscopy 2018, 67, 144–155. [Google Scholar] [CrossRef]
  45. Morisawa, S.; Cherr, G.N. Acrosome reaction in spermatozoa from hagfish (Agnatha) Eptatretus burgeri and Eptatretus stouti: Acrosomal exocytosis and identification of filamentous actin. Dev. Growth Differ. 2002, 44, 337–344. [Google Scholar] [CrossRef] [Green Version]
  46. Alavi, S.M.H.; Hatef, A.; Pšenička, M.; Kašpar, V.; Boryshpolets, S.; Dzyuba, B.; Cosson, J.; Bondarenko, V.; Rodina, M.; Gela, D.; et al. Sperm biology and control of reproduction in sturgeon: (II) Sperm morphology, acrosome reaction, motility and cryopreservation. Rev. Fish Biol. Fish. 2012, 22, 861–886. [Google Scholar] [CrossRef]
  47. Hatef, A.; Alavi, S.M.H.; Rodina, M.; Linhart, O. Morphology and fine structure of the Russian sturgeon, Acipenser gueldenstaedtii (Acipenseridae, Chondrostei) spermatozoa. J. Appl. Ichthyol. 2012, 28, 978–983. [Google Scholar] [CrossRef]
  48. Alavi, S.M.H.; Butts, I.A.E.; Hatef, A.; Mommens, M.; Trippel, E.A.; Litvak, M.K.; Babiak, I. Sperm morphology, ATP content and analysis of motility in Atlantic halibut (Hippoglossus hippoglossus L.). Can. J. Zool. 2011, 89, 219–228. [Google Scholar] [CrossRef]
  49. Inaba, K. Molecular architecture of sperm flagella: Molecules for motility and signaling. Zool. Sci. 2003, 20, 1043–1056. [Google Scholar] [CrossRef]
  50. Ciereszko, A. Chemical composition of seminal plasma and its physiological relationship with sperm motility, fertilizing capacity and cryopreservation success in fish. In Fish Spermatology; Alavi, S.M.H., Cosson, J., Coward, K., Rafiee, G., Eds.; Alpha Science Ltd.: Oxford, UK, 2008; pp. 215–240. [Google Scholar]
  51. Kowalski, R.K.; Cejko, B.I. Sperm quality in fish: Determinants and affecting factors. Theriogenology 2019, 135, 94–108. [Google Scholar] [CrossRef]
  52. Morisawa, M.; Suzuki, K. Osmolality and potassium ions: Their roles in initiation of sperm motility in teleosts. Science 1980, 210, 1145–1147. [Google Scholar] [CrossRef]
  53. Morisawa, M.; Suzuki, K.; Morisawa, S. Effects of potassium and osmolality on spermatozoan motility of salmonid fishes. J. Exp. Biol. 1983, 107, 105–113. [Google Scholar] [CrossRef]
  54. Billard, R. Reproduction in rainbow trout: Sex differentiation, dynamics of gametogenesis, biology and preservation of gametes. Aquaculture 1992, 100, 263–298. [Google Scholar] [CrossRef]
  55. Alavi, S.M.H.; Cosson, J.; Karami, M.; Amiri, B.M.; Akhoundzadeh, M.A. Spermatozoa motility in the Persian sturgeon, Acipenser persicus: Effects of pH, dilution rate, ions and osmolality. Reproduction 2004, 128, 819–828. [Google Scholar] [CrossRef]
  56. Morisawa, M.; Oda, S.; Yoshida, M.; Takai, H. Transmembrane signal transduction for the regulation of sperm motility in fishes and ascidians. In The Male Gamete: From Basic to Clinical Applications; Gagnon, C., Ed.; Cache River Press: Vienna, IL, USA, 1999; pp. 149–160. [Google Scholar]
  57. Alavi, S.M.H.; Cosson, J. Sperm motility in fishes: (II) Effects of ions and osmotic pressure. Cell Biol. Int. 2006, 30, 1–14. [Google Scholar] [CrossRef] [PubMed]
  58. Morisawa, M. Adaptation and strategy for fertilization in the sperm of teleosts fish. J. Appl. Ichthyol. 2008, 24, 362–370. [Google Scholar] [CrossRef]
  59. Alavi, S.M.H.; Cosson, J.; Bondarenko, O.; Linhart, O. Sperm motility in fishes: (III) diversity of regulatory signals from membrane to the axoneme. Theriogenology 2019, 136, 143–165. [Google Scholar] [CrossRef]
  60. Morisawa, M.; Okuno, M. Cyclic AMP induces maturation of trout sperm axoneme to initiate motility. Nature 1982, 295, 703–704. [Google Scholar] [CrossRef]
  61. Cosson, M.P.; Cosson, J.; Andre, F.; Billard, R. cAMP/ATPrelationshipinthe activation of trout sperm motility: Their interaction in membrane-deprived models and in live spermatozoa. Cell Motil. Cytoskelet. 1995, 31, 159–176. [Google Scholar] [CrossRef] [PubMed]
  62. Linhart, O.; Cosson, J.; Mims, S.D.; Shelton, W.L.; Rodina, M. Effects of ions on the motility of fresh and demembranated paddlefish (Polyodon spathula) spermatozoa. Reproduction 2002, 124, 713–719. [Google Scholar] [CrossRef]
  63. Tubbs, C.; Thomas, P. Progestin signaling through an olfactory G protein and membrane progestin receptor-a in Atlantic croaker sperm: Potential role in induction of sperm hypermotility. Endocrinology 2009, 150, 473–484. [Google Scholar] [CrossRef] [Green Version]
  64. Tan, W.; Aizen, J.; Thomas, P. Membrane progestin receptor-alpha mediates progestin-induced sperm hypermotility and increased fertilization success in southern flounder (Paralichthys lethostigma). Gen. Comp. Endocrinol. 2014, 200, 18–26. [Google Scholar] [CrossRef] [PubMed]
  65. Alavi, S.M.H.; Cosson, J. Sperm motility in fishes: (I) Effects of pH and temperature. Cell Biol. Int. 2005, 29, 101–110. [Google Scholar] [CrossRef]
  66. Cosson, J. Frenetic activation of fish spermatozoa flagella entails short-term motility, portending their precocious decadence. J. Fish Biol. 2010, 76, 240–279. [Google Scholar] [CrossRef]
  67. Ziętara, M.S.; Biegniewska, A.; Rurangwa, E.; Swierczynski, J.; Ollevier, F.; Skorkowski, E.F. Bioenergetics of fish spermatozoa during semen storage. Fish Physiol. Biochem. 2009, 35, 607–614. [Google Scholar] [CrossRef]
  68. Dzyuba, B.; Bondarenko, O.; Fedorov, P.; Gazo, I.; Prokopchuk, G.; Cosson, J. Energetics of fish spermatozoa: The proven and the possible. Aquaculture 2017, 472, 60–72. [Google Scholar] [CrossRef]
  69. Lahnsteiner, F.; Patzner, R.A.; Weismann, T. Energy resources of spermatozoa of the rainbow trout Oncorhynchus mykiss (Pisces, Teleostei). Reprod. Nut. Dev. 1993, 33, 349–360. [Google Scholar] [CrossRef] [PubMed]
  70. Saudrais, C.; Fierville, F.; Loir, M.; Le Remeur, E.; Cibert, C.; Cosson, J. The use of phosphocreatine plus ADP as energy source for motility of membrane-derived trout spermatozoa. Cell Motil. Cytoskel. 1998, 41, 91–106. [Google Scholar] [CrossRef]
  71. Woolsey, J.; Ingermann, R.L. Acquisition of the potential for sperm motility in steelhead (Oncorhynchus mykiss): Effect of pH on dynein ATPase. Fish Physiol. Biochem. 2003, 29, 47–56. [Google Scholar] [CrossRef]
  72. Kinsey, W.H.; Sharma, D.; Kinsey, S.C. Fertilization and egg activation in fishes. In The Fish Oocyte: From Basic Studies to Biotechnological Applications; Babin, P.J., Cerdà, J., Lubzens, E., Eds.; Springer: Dordrecht, The Netherlands, 2007; pp. 397–409. [Google Scholar] [CrossRef]
  73. Kudo, S. Fertilization, cortical reaction, polyspermy preventing and anti-microbial mechanisms in fish eggs. Bull. Inst. Zool. Acad. Sinica 1991, 16, 313–340. [Google Scholar]
  74. Ginzburg, A.S. Fertilization in Fishes and Problem of Polyspermy; Israel Program for Scientific Translations; National Technical Information Service US Department of Commerce: Springfield, VA, USA, 1972.
  75. Iwamatsu, T. Stages of normal development in the medaka Oryzias latipes. Mech. Dev. 2004, 121, 605–618. [Google Scholar] [CrossRef]
  76. Tvedt, H.B.; Benfey, T.J.; Martin-Robichaud, D.J.; Power, J. The relationship between sperm density, spermatocrit, sperm motility and fertilization success in Atlantic halibut, Hippoglossus hippoglossus. Aquaculture 2001, 194, 191–200. [Google Scholar] [CrossRef]
  77. Rideout, R.M.; Trippel, E.A.; Litvak, M.K. Relationship between sperm density, spermatocrit, sperm motility and spawning date in wild and cultured haddock. J. Fish Biol. 2004, 65, 319–332. [Google Scholar] [CrossRef]
  78. Kaspar, V.; Kohlmann, K.; Vandeputte, M.; Rodina, M.; Gela, D.; Kocour, M.; Alavi, S.M.H.; Hulak, M.; Linhart, O. Equalizing sperm concentrations in a common carp (Cyprinus carpio) sperm pool does not affect variance in proportions of larvae sired in competition. Aquaculture 2007, 272 (Suppl. S1), S204–S209. [Google Scholar] [CrossRef]
  79. Hatef, A.; Niksirat, H.; Alavi, S.M.H. Composition of ovarian fluid in endangered Caspian brown trout, Salmo trutta caspius, and its effects on spermatozoa motility and fertilizing ability compared to freshwater and a saline medium. Fish Physiol. Biochem. 2009, 35, 695–700. [Google Scholar] [CrossRef]
  80. Butts, I.A.E.; Trippel, E.A.; Litvak, M.K. The effect of sperm to egg ratio and gamete contact time on fertilization success in Atlantic cod Gadus morhua. Aquaculture 2009, 286, 89–94. [Google Scholar] [CrossRef]
  81. Butts, I.A.E.; Roustaian, P.; Litvak, M.K. Fertilization strategies for winter flounder: Effects of spermatozoa density and the duration of gamete receptivity. Aquac. Biol. 2012, 16, 115–124. [Google Scholar] [CrossRef] [Green Version]
  82. Linhart, O.; Cheng, Y.; Xin, M.M.; Rodina, M.; Tučková, V.; Shelton, W.L.; Kašpar, V. Standardization of egg activation and fertilization in sterlet (Acipenser ruthenus). Aquac. Rep. 2020, 17, 100381. [Google Scholar] [CrossRef]
  83. Cheng, Y.; Franek, R.; Rodina, M.; Xin, M.; Cosson, J.; Zhang, S.; Linhart, O. Optimization of Sperm Management and Fertilization in Zebrafish (Danio rerio (Hamilton)). Animals 2021, 11, 1558. [Google Scholar] [CrossRef] [PubMed]
  84. Linhart, O.; Rodina, M.; Flajšhans, M.; Mavrodiev, N.; Nebesárová, J.; Gela, D.; Kocour, M. Studies on sperm of diploid and triploid tench, Tinca tinca (L.). Aquac. Int. 2006, 14, 9–25. [Google Scholar] [CrossRef]
  85. Piferrer, F.; Beaumont, A.; Falguière, J.C.; Flajšhans, M.; Haffray, P.; Colombo, L. Polyploid fish and shellfish: Production, biology and applications to aquaculture for performance improvement and genetic containment. Aquaculture 2009, 293, 125–156. [Google Scholar] [CrossRef] [Green Version]
  86. Alavi, S.M.H.; Drozd, B.; Hatef, A.; Flajšhans, M. Sperm morphology, motility and velocity in naturally polyploid European weatherfish (Misgurnus fossilis L.). Theriogenology 2013, 80, 153–160. [Google Scholar] [CrossRef]
  87. Pšenička, M.; Flajšhans, M.; Hulák, M.; Kašpar, V.; Rodina, M.; Borishpolets, S.; Linhart, O. The influence of ploidy level on ultrastructure and motility of tench Tinca tinca (L.) spermatozoa. Rev. Fish Biol. Fish. 2010, 20, 331–338. [Google Scholar] [CrossRef]
  88. Pšenička, M.; Kašpar, V.; Rodina, M.; Gela, D.; Hulák, M.; Flajšhans, M. Comparative study on ultrastructure and motility parameters of spermatozoa of tetraploid and hexaploid Siberian sturgeon Acipenser baerii. J. Appl. Ichthyol. 2011, 27, 683–686. [Google Scholar] [CrossRef]
  89. Gage, M.J.G.; MacFarlane, C.; Yeates, S.; Shackleton, R.; Parker, G.A. Relationships between sperm morphometry and sperm motility in the Atlantic salmon. J. Fish Biol. 2002, 60, 1528–1539. [Google Scholar] [CrossRef]
  90. Alavi, S.M.H.; Psenicka, M.; Rodina, M.; Policar, T.; Linhart, O. Changes of sperm morphology, volume, density and motility and seminal plasma composition in Barbus barbus (Cyprinidae: Teleostei) during the reproductive season. Aquat. Living Resour. 2008, 21, 75–80. [Google Scholar] [CrossRef]
  91. Alavi, S.M.H.; Rodina, M.; Viveiros, A.T.M.; Cosson, J.; Gela, D.; Boryshpolets, S.; Linhart, O. Effects of osmolality on sperm morphology, motility and flagellar wave parameters in Northern pike (Esox lucius L.). Theriogenology 2009, 72, 32–43. [Google Scholar] [CrossRef]
  92. Ishijima, S.; Hara, M.; Okiyama, M. Comparative studies on spermatozoan motility of Japanese fishes. Bull. Oce. Res. Inst. Univ. Tokyo 1998, 33, 139–152. [Google Scholar]
  93. Beirão, J.; Boulais, M.; Gallego, V.; O’Brien, J.K.; Peixoto, S.; Robeck, T.R.; Cabrita, E. Sperm handling in aquatic animals for artificial reproduction. Theriogenology 2019, 133, 161–178. [Google Scholar] [CrossRef] [PubMed]
  94. Figueroa, E.; Lee-Estévez, M.; Valdebenito, I.; Farías, J.G.; Romero, J. Potential biomarkers of DNA quality in cryopreserved fish sperm: Impact on gene expression and embryonic development. Rev. Aquac. 2020, 12, 382–391. [Google Scholar] [CrossRef]
  95. Kim, D.S.; Jo, J.Y.; Lee, T.Y. Induction of triploidy in mud loach (Misgurnus mizolepis) and its effect on gonad development and growth. Aquaculture 1994, 120, 263–270. [Google Scholar] [CrossRef]
  96. Chourrout, D.; Chevassus, B.; Krieg, F.; Happe, A.; Burger, G.; Renard, P. Production of second generation triploid and tetraploid rainbow trout by mating tetraploid males and diploid females-potential of tetraploid fish. Theor. Appl. Genet. 1986, 72, 193–206. [Google Scholar] [CrossRef]
  97. Oshima, K.; Morishima, K.; Yamaha, E.; Arai, K. Reproductive capacity of triploid loaches obtained from Hokkaido Island, Japan. Ichthyol. Res. 2005, 52, 1–8. [Google Scholar] [CrossRef]
  98. Fujimoto, T.; Yasui, G.S.; Yoshikawa, H.; Yamaha, E.; Arai, K. Genetic and reproductive potential of spermatozoa of diploid and triploid males obtained from interspecific hybridization of Misgurnus anguillicaudatus female with M. mizolepis male. J. Appl. Ichthyol. 2008, 24, 430–437. [Google Scholar] [CrossRef] [Green Version]
  99. Kawamura, K.; Ueda, T.; Aoki, K.; Hosoya, K. Spermatozoa in triploids of the rosy bitterling, Rhodeus ocellatus ocellatus. J. Fish Biol. 1999, 55, 420–432. [Google Scholar] [CrossRef]
  100. Cabrita, E.; Robles, V.; Rebordinos, L.; Sarasquete, C.; Herraez, M.P. Evaluation of DNA damage in rainbow trout (Oncorhynchus mykiss) and gilthead sea bream (Sparus aurata) cryopreserved sperm. Cryobiology 2005, 50, 144–153. [Google Scholar] [CrossRef] [PubMed]
  101. Shaliutina, A.; Hulak, M.; Gazo, I.; Linhartova, P.; Linhart, O. Effect of short-term storage on quality parameters, DNA integrity, and oxidative stress in Russian (Acipenser gueldenstaedtii) and Siberian (Acipenser baerii) sturgeon sperm. Anim. Reprod. Sci. 2013, 139, 127–135. [Google Scholar] [CrossRef] [PubMed]
  102. Linhart, O.; Rodina, M.; Gela, D.; Kocour, M.; Vandeputte, M. Spermatozoal competition in common carp (Cyprinus carpio): What is the primary determinant of competition success? Reproduction 2005, 130, 705–711. [Google Scholar] [CrossRef] [Green Version]
  103. Gage, M.J.G.; MacFarlane, C.P.; Yeates, S.; Ward, R.G.; Searle, J.B.; Parker, G.A. Spermatozoal traits and sperm competition in Atlantic salmon: Relative sperm velocity is the primary determinant of fertilization success. Curr. Biol. 2004, 14, 44–47. [Google Scholar] [CrossRef]
  104. Linhart, O.; Alavi, S.M.H.; Rodina, M.; Gela, D.; Cosson, J. Comparison of sperm velocity, motility and fertilizing ability between firstly and secondly activated spermatozoa of common carp (Cyprinus carpio). J. Appl. Ichthyol. 2008, 24, 386–392. [Google Scholar] [CrossRef]
  105. Wilson-Leedy, J.G.; Ingermann, R.L. Development of a novel CASA system based on open source software for characterization of zebrafish sperm motility parameters. Theriogenology 2007, 67, 661–672. [Google Scholar] [CrossRef]
  106. Cosson, J. Methods to analyse the movements of the spermatozoa and their flagella. In Fish Spermatology; Alavi, S.M.H., Cosson, J., Coward, K., Rafiee, G., Eds.; Alpha Science Ltd.: Oxford, UK, 2008; pp. 63–102. [Google Scholar]
  107. Amann, R.P.; Waberski, D. Computer-assisted sperm analysis (CASA): Capabilities and potential developments. Theriogenology 2014, 81, 5–17. [Google Scholar] [CrossRef]
  108. McMaster, M.E.; Van Der Kraak, G.J.; Portt, C.B.; Munkittrick, K.R.; Sibley, P.K.; Smith, I.R.; Dixon, D.G. Changes in hepatic mixed function oxygenase (MFO) activity, plasma steroid levels, and age at maturity of a white sucker (Catostomus commersoni) population exposed to bleached kraft pulp mill effluent. Aquat. Toxicol. 1991, 21, 199–217. [Google Scholar] [CrossRef]
  109. Munkittrick, K.R.; Portt, C.B.; Van Der Kraak, G.J.; Smith, I.R.; Rokosh, D. Impact of bleached kraft mill effluent on population characteristics, liver MFO activity and serum steroid levels of a Lake Superior white sucker (Catostomus commersoni) population. Can. J. Fish. Aquat. Sci. 1991, 48, 1371–1380. [Google Scholar] [CrossRef]
  110. Munkittrick, K.R.; McMaster, M.E.; Portt, C.B.; Van Der Kraak, G.J.; Smith, I.R.; Dixon, D.G. Changes in maturity, plasma sex steroid levels, hepatic mixed-function oxygenase activity and the presence of external lesions in lake whitefish (Coregonus clupeaformis) exposed to bleached kraft mill effluent. Can. J. Fish. Aquat. Sci. 1992, 49, 1560–1569. [Google Scholar] [CrossRef]
  111. Allen, Y.; Scott, A.P.; Matthiessen, P.; Haworth, S.; Thain, J.E.; Feist, S. Survey of estrogenic activity in United Kingdom estuarine and coastal waters and its effects on gonadal development of the flounder Platichthys flesus. Environ. Toxicol. Chem. 1999, 18, 1791–1800. [Google Scholar] [CrossRef]
  112. Harshbarger, J.C.; Coffey, M.J.; Young, M.Y. Inter-sexes in Mississippi River shovelnose sturgeon sampled below Saint Louis, Missouri, USA. Mar. Environ. Res. 2000, 50, 247–250. [Google Scholar] [CrossRef]
  113. Hashimoto, S.; Bessho, H.; Hara, A.; Nakamura, M.; Iguchi, T.; Fujita, K. Elevated serum vitellogenin levels and gonadal abnormalities in wild male flounder (Pleuronectes yokohamae) from Tokyo Bay, Japan. Mar. Environ. Res. 2000, 49, 37–53. [Google Scholar] [CrossRef]
  114. Viganò, L.; Arillo, A.; Bottero, S.; Massari, A.; Mandich, A. First observation of intersex cyprinids in the Po River (Italy). Sci. Total Environ. 2001, 269, 189–194. [Google Scholar] [CrossRef]
  115. Jobling, S.; Beresford, N.; Nolan, M.; Rodgers-Gray, T.; Brighty, G.C.; Sumpter, J.P.; Tyler, C.R. Altered sexual maturation and gamete production in wild roach (Rutilus rutilus) living in rivers that receive treated sewage effluents. Biol. Reprod. 2002, 66, 272–281. [Google Scholar] [CrossRef]
  116. Sepúlveda, M.S.; Johnson, W.E.; Higman, J.C.; Denslow, N.D.; Schoeb, T.R.; Gross, T.S. An evaluation of biomarkers of reproductive function and potential contaminant effects in Florida largemouth bass (Micropterus salmoides floridanus) sampled from the St. Johns River. Sci. Total Environ. 2002, 289, 133–144. [Google Scholar] [CrossRef]
  117. Aravindakshan, J.; Paquet, V.; Gregory, M.; Dufresne, J.; Fournier, M.; Marcogliese, D.J.; Cyr, D.J. Consequences of xenoestrogen exposure on male reproductive function in Spottail Shiners (Notropis hudsonius). Toxicol. Sci. 2004, 78, 156–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Kavanagh, R.J.; Balch, G.C.; Kiparissis, Y.; Niimi, A.J.; Sherry, J.; Tinson, C.; Metcalfe, C.D. Endocrine disruption and altered gonadal development in white perch (Morone americana) from the lower Great Lakes region. Environ. Health Perspect. 2004, 112, 898–902. [Google Scholar] [CrossRef] [Green Version]
  119. Bjerregaard, L.B.; Korsgaard, B.; Bjerregaard, P. Intersex in wild roach (Rutilus rutilus) from Danish sewage effluent-receiving streams. Ecotoxicol. Environ. Saf. 2006, 64, 321–328. [Google Scholar] [CrossRef]
  120. Woodling, J.D.; Lopez, E.M.; Maldonado, T.A.; Norris, D.O.; Vajda, A.M. Intersex and other reproductive disruption of fish in wastewater effluent dominated Colorado streams. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2006, 144, 10–15. [Google Scholar] [CrossRef]
  121. Marchand, M.J.; Pieterse, G.M.; Barnhoorn, I.E.J. Preliminary results on sperm motility and testicular histology of two feral fish species, Oreochromis mossambicus and Clarias gariepinus, from a currently DDT-sprayed area, South Africa. J. Appl. Ichthyol. 2008, 24, 423–429. [Google Scholar] [CrossRef]
  122. Marchand, M.J.; Pieterse, G.M.; Barnhoorn, I.E.J. Sperm motility and testicular histology as reproductive indicators of fish health of two feral fish species from a currently DDT sprayed area, South Africa. J. Appl. Ichthyol. 2010, 26, 707–714. [Google Scholar] [CrossRef]
  123. Jeffries, K.M.; Nelson, E.R.; Jackson, L.J.; Habibi, H.R. Basin-wide impacts of compounds with estrogen-like activity on longnose dace (Rhinichthys cataractae) in two Prairie Rivers of Alberta, Canada. Environ. Toxicol. Chem. 2008, 27, 2042–2052. [Google Scholar] [CrossRef]
  124. Jeffries, K.M.; Jackson, L.J.; Ikonomou, M.G.; Habibi, H.R. Presence of natural and anthropogenic organic contaminants and potential fish health impacts along two river gradients in Alberta, Canada. Environ. Toxicol. Chem. 2010, 29, 2379–2387. [Google Scholar] [CrossRef] [PubMed]
  125. Barnhoorn, I.E.J.; van Dyk, J.C.; Pieterse, G.M.; Bornman, M.S. Intersex in feral indigenous freshwater Oreochromis mossambicus, from various parts in the Luvuvhu River, Limpopo Province, South Africa. Ecotoxicol. Environ. Saf. 2010, 73, 1537–1542. [Google Scholar] [CrossRef] [PubMed]
  126. Solé, M.; Raldúa, D.; Piferrer, F.; Barceló, D.; Porte, C. Feminization of wild carp, Cyprinus carpio, in a polluted environment: Plasma steroid hormones, gonadal morphology and xenobiotic metabolizing system. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2003, 136, 145–156. [Google Scholar] [CrossRef]
  127. Vajda, A.M.; Barber, L.B.; Gray, J.L.; Lopez, E.M.; Woodling, J.D.; Norris, D.O. Reproductive disruption in fish downstream from an estrogenic wastewater effluent. Environ. Sci. Technol. 2008, 42, 3407–3414. [Google Scholar] [CrossRef]
  128. Gilroy, E.A.M.; McMaster, M.E.; Parrott, J.L.; Hewitt, L.M.; Park, B.J.; Brown, S.B.; Sherry, J.P. Assessment of the health status of wild fish from the Wheatley Harbour Area of Concern, Ontario, Canada. Environ. Toxicol. Chem. 2012, 31, 2798–2811. [Google Scholar] [CrossRef]
  129. Bahamonde, P.A.; Tetreault, G.R.; McMaster, M.E.; Servos, M.R.; Martyniuk, C.J.; Munkittrick, K.R. Molecular signatures in rainbow darter (Etheostoma caeruleum) inhabiting an urbanized river reach receiving wastewater effluents. Aquat. Toxicol. 2014, 148, 211–220. [Google Scholar] [CrossRef] [PubMed]
  130. Bahamonde, P.A.; Fuzzen, M.L.; Bennett, C.J.; Tetreault, G.R.; McMaster, M.E.; Servos, M.R.; Martyniuk, C.J.; Munkittrick, K.R. Whole organism responses and intersex severity in rainbow darter (Etheostoma caeruleum) following exposures to municipal wastewater in the Grand River basin, ON, Canada. Part A. Aquat. Toxicol. 2015, 159, 290–301. [Google Scholar] [CrossRef] [PubMed]
  131. McMaster, M.E.; Portt, C.B.; Munkittrick, K.R.; Dixon, D.G. Milt characteristics, reproductive performance, and larval survival and development of white sucker exposed to bleached kraft mill effluent. Ecotoxicol. Environ. Saf. 1992, 23, 103–117. [Google Scholar] [CrossRef]
  132. Jobling, S.; Coey, S.; Whitmore, J.G.; Kime, D.E.; Van Look, K.J.W.; McAllister, B.G.; Beresford, N.; Henshaw, A.C.; Brighty, G.; Tyler, C.R.; et al. Wild intersex roach (Rutilus rutilus) have reduced fertility. Biol. Reprod. 2002, 67, 515–524. [Google Scholar] [CrossRef]
  133. Wagenaar, G.; Botha, T.; Barnhoorn, I. Sperm motility and testicular histology as reproductive indicators in Clarias gariepinus from an eutrophic impoundment, South Africa. J. Appl. Ichthyol. 2012, 28, 990–997. [Google Scholar] [CrossRef]
  134. Fuzzen, M.L.M.; Bennett, C.J.; Tetreault, G.R.; McMaster, M.E.; Servos, M.R. Severe intersex is predictive of poor fertilization success in populations of rainbow darter (Etheostoma caeruleum). Aquat. Toxicol. 2015, 160, 106–116. [Google Scholar] [CrossRef]
  135. Jenkins, J.A.; Olivier, H.M.; Draugelis-Dale, R.O.; Eilts, B.E.; Torres, L.; Patiño, R.; Nilsen, E.; Goodbred, S.L. Assessing reproductive and endocrine parameters in male largescale suckers (Catostomus macrocheilus) along a contaminant gradient in the lower Columbia River, USA. Sci. Total Environ. 2014, 484, 365–378. [Google Scholar] [CrossRef] [Green Version]
  136. Agbohessi, P.T.; Toko, I.I.; Ouédraogo, A.; Jauniaux, T.; Mandiki, S.N.M.; Kestemont, P. Assessment of the health status of wild fish inhabiting a cotton basin heavily impacted by pesticides in Benin (West Africa). Sci. Total Environ. 2015, 506-507, 567–584. [Google Scholar] [CrossRef] [PubMed]
  137. Jenkins, J.A.; Rosen, M.R.; Draugelis-Dale, R.O.; Echols, K.R.; Torres, L.; Wieser, C.M.; Kersten, C.A.; Goodbred, S.L. Sperm quality biomarkers complement reproductive and endocrine parameters in investigating environmental contaminants in common carp (Cyprinus carpio) from the Lake Mead National Recreation Area. Environ. Res. 2018, 163, 149–164. [Google Scholar] [CrossRef] [PubMed]
  138. Van der Kraak, G.J.; Munkittrick, K.R.; McMaster, M.E.; Portt, C.B.; Chang, J.P. Exposure to bleached kraft pulp mill effluent disrupts the pituitary-gonadal axis of white sucker at multiple sites. Toxicol. Appl. Pharmacol. 1992, 115, 224–233. [Google Scholar] [CrossRef]
  139. Flammarion, P.; Brion, F.; Babut, M.; Garric, J.; Migeon, B.; Noury, P.; Thybaud, E.; Palazzi, X.; Tyler, C.R. Induction of fish vitellogenin and alterations in testicular structure: Preliminary results of estrogenic effects in Chub (Leuciscus cephalus). Ecotoxicology 2000, 9, 127–135. [Google Scholar] [CrossRef] [Green Version]
  140. Randák, T.; Žlábek, V.; Pulkrabová, J.; Kolářová, J.; Kroupová, H.; Široká, Z.; Velíšek, J.; Svobodová, Z.; Hajšlová, J. Effects of pollution on chub in the River Elbe, Czech Republic. Ecotoxicol. Environ. Saf. 2009, 72, 737–746. [Google Scholar] [CrossRef]
  141. Celander, M.C. Cocktail effects on biomarker responses in fish. Aquat. Toxicol. 2011, 105, 72–77. [Google Scholar] [CrossRef] [PubMed]
  142. Hamid, N.; Junaid, M.; Pei, D.S. Combined toxicity of endocrine-disrupting chemicals: A review. Ecotoxicol. Environ. Saf. 2021, 215, 112136. [Google Scholar] [CrossRef]
  143. Pawlowski, S.; Sauer, A.; Shears, J.A.; Tyler, C.R.; Braunbeck, T. Androgenic and estrogenic effects of the synthetic androgen 17α-methyltestosterone on sexual development and reproductive performance in the fathead minnow (Pimephales promelas) determined using the gonadal recrudescence assay. Aquat. Toxicol. 2004, 68, 277–291. [Google Scholar] [CrossRef]
  144. Pawlowski, S.; van Aerle, R.; Tyler, C.R.; Braunbeck, T. Effects of 17α-ethinylestradiol in a fathead minnow (Pimephales promelas) gonadal recrudescence assay. Ecotoxicol. Environ. Saf. 2004, 57, 330–345. [Google Scholar] [CrossRef]
  145. Kang, I.J.; Yokota, H.; Oshima, Y.; Tsuruda, Y.; Shimasaki, Y.; Honjo, T. The effects of methyltestosterone on the sexual development and reproduction of adult medaka (Oryzias latipes). Aquat. Toxicol. 2008, 87, 37–46. [Google Scholar] [CrossRef]
  146. Lahnsteiner, F.; Berger, B.; Grubinger, F.; Weismann, T. The effect of 4-nonylphenol on semen quality, viability of gametes, fertilization success, and embryo and larvae survival in rainbow trout (Oncorhynchus mykiss). Aquat. Toxicol. 2005, 71, 297–306. [Google Scholar] [CrossRef] [PubMed]
  147. Kang, I.J.; Yokota, H.; Oshima, Y.; Tsuruda, Y.; Oe, T.; Imada, N.; Tadokoro, H.; Honjo, T. Effects of bisphenol a on the reproduction of Japanese medaka (Oryzias latipes). Environ. Toxicol. Chem. 2002, 21, 2394–2400. [Google Scholar] [CrossRef]
  148. Martinović, D.; Blake, L.S.; Durhan, E.J.; Greene, K.J.; Kahl, M.D.; Jensen, K.M.; Makynen, E.A.; Villeneuve, D.L.; Ankley, G.T. Reproductive toxicity of vinclozolin in the fathead minnow: Confirming an anti-androgenic mode of action. Environ. Toxicol. Chem. 2008, 27, 478–488. [Google Scholar] [CrossRef] [PubMed]
  149. Seki, M.; Yokota, H.; Matsubara, H.; Tsuruda, Y.; Maeda, M.; Tadokoro, H.; Kobayashi, K. Effect of ethinylestradiol on the reproduction and induction of vitellogenin and testis-ova in medaka (Oryzias latipes). Environ. Toxicol. Chem. 2002, 21, 1692–1698. [Google Scholar] [CrossRef]
  150. Hashimoto, S.; Watanabe, E.; Ikeda, M.; Terao, Y.; Strüssmann, C.A.; Inoue, M.; Hara, A. Effects of ethinylestradiol on medaka (Oryzias latipes) as measured by sperm motility and fertilization success. Arch. Environ. Contam. Toxicol. 2009, 56, 253–259. [Google Scholar] [CrossRef] [PubMed]
  151. Devaux, A.; Fiat, L.; Gillet, C.; Bony, S. Reproduction impairment following paternal genotoxin exposure in brown trout (Salmo trutta) and Arctic charr (Salvelinus alpinus). Aquat. Toxicol. 2011, 101, 405–411. [Google Scholar] [CrossRef] [PubMed]
  152. Kang, I.J.; Yokota, H.; Oshima, Y.; Tsuruda, Y.; Hano, T.; Maeda, M.; Imada, N.; Tadokoro, H.; Honjo, T. Effects of 4-nonylphenol on reproduction of Japanese medaka, Oryzias latipes. Environ. Toxicol. Chem. 2003, 22, 2438–2445. [Google Scholar] [CrossRef] [PubMed]
  153. Uren-Webster, T.M.; Lewis, C.; Filby, A.L.; Paull, G.C.; Santos, E.M. Mechanisms of toxicity of di (2-ethylhexyl) phthalate on the reproductive health of male zebrafish. Aquat. Toxicol. 2010, 99, 360–369. [Google Scholar] [CrossRef]
  154. Kang, I.J.; Yokota, H.; Oshima, Y.; Tsuruda, Y.; Yamaguchi, T.; Maeda, M.; Imada, N.; Tadokoro, H.; Honjo, T. Effect of 17β-estradiol on the reproduction of Japanese medaka (Oryzias latipes). Chemosphere 2002, 47, 71–80. [Google Scholar] [CrossRef]
  155. Schultz, I.R.; Skillman, A.; Nicolas, J.M.; Cyr, D.G.; Nagler, J.J. Short-term exposure to 17α-ethynylestradiol decreases the fertility of sexually maturing male rainbow trout (Oncorhynchus mykiss). Environ. Toxicol. Chem. 2003, 22, 1272–1280. [Google Scholar] [CrossRef]
  156. Lahnsteiner, F.; Berger, B.; Kletzl, M.; Weismann, T. Effect of 17β-estradiol on gamete quality and maturation in two salmonid species. Aquat. Toxicol. 2006, 79, 124–131. [Google Scholar] [CrossRef]
  157. Lahnsteiner, F.; Berger, B.; Kletzl, M.; Weismann, T. Effect of bisphenol A on maturation and quality of semen and eggs in the brown trout, Salmo trutta f. fario. Aquat.Toxicol. 2005, 75, 213–224. [Google Scholar] [CrossRef] [PubMed]
  158. Tian, H.; Li, Y.; Wang, W.; Wu, P.; Ru, S. Exposure to monocrotophos pesticide during sexual development causes the feminization/demasculinization of the reproductive traits and a reduction in the reproductive success of male guppies (Poecilia reticulata). Toxicol. Appl. Pharmacol. 2012, 263, 163–170. [Google Scholar] [CrossRef]
  159. Ji, K.; Hong, S.; Kho, Y.; Choi, K. Effects of bisphenol S exposure on endocrine functions and reproduction of zebrafish. Environ. Sci. Technol. 2013, 47, 8793–8800. [Google Scholar] [CrossRef]
  160. Shi, J.; Jiao, Z.; Zheng, S.; Li, M.; Zhang, J.; Feng, Y.; Yin, J.; Shao, B. Long-term effects of bisphenol AF (BPAF) on hormonal balance and genes of hypothalamus-pituitary-gonad axis and liver of zebrafish (Danio rerio), and the impact on offspring. Chemosphere 2015, 128, 252–257. [Google Scholar] [CrossRef]
  161. Montgomery, T.M.; Brown, A.C.; Gendelman, H.K.; Ota, M.; Clotfelter, E.D. Exposure to 17α-ethinylestradiol decreases motility and ATP in sperm of male fighting fish Betta splendens. Environ. Toxicol. 2014, 29, 243–252. [Google Scholar] [CrossRef] [PubMed]
  162. Naderi, M.; Wong, M.Y.L.; Gholami, F. Developmental exposure of zebrafish (Danio rerio) to bisphenol-S impairs subsequent reproduction potential and hormonal balance in adults. Aquat. Toxicol. 2014, 148, 195–203. [Google Scholar] [CrossRef] [PubMed]
  163. Chen, J.; Xiao, Y.; Gai, Z.; Li, R.; Zhu, Z.; Bai, C.; Tanguay, R.L.; Xu, X.; Huang, C.; Dong, Q. Reproductive toxicity of low level bisphenol A exposures in a two-generation zebrafish assay: Evidence of male-specific effects. Aquat. Toxicol. 2015, 169, 204–214. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, J.; Saili, K.S.; Liu, Y.; Li, L.; Zhao, Y.; Jia, Y.; Bai, C.; Tanguay, R.L.; Dong, Q.; Huang, C. Developmental bisphenol A exposure impairs sperm function and reproduction in zebrafish. Chemosphere 2017, 169, 262–270. [Google Scholar] [CrossRef]
  165. Zhu, Y.; Hua, R.; Zhou, Y.; Li, H.; Quan, S.; Yu, Y. Chronic exposure to mono-(2-ethylhexyl)-phthalate causes endocrine disruption and reproductive dysfunction in zebrafish. Environ. Toxicol. Chem. 2016, 35, 2117–2124. [Google Scholar] [CrossRef]
  166. Corradetti, B.; Stronati, A.; Tosti, L.; Manicardi, G.; Carnevali, O.; Bizzaro, D. Bis-(2-ethylexhyl) phthalate impairs spermatogenesis in zebrafish (Danio rerio). Reprod. Biol. 2013, 13, 195–202. [Google Scholar] [CrossRef]
  167. Watanabe, H.; Horie, Y.; Takanobu, H.; Koshio, M.; Flynn, K.; Iguchi, T.; Tatarazako, N. Medaka extended one-generation reproduction test evaluating 4-nonylphenol. Environ. Toxicol. Chem. 2017, 36, 3254–3266. [Google Scholar] [CrossRef]
  168. Zhang, Y.; Guan, Y.; Zhang, T.; Yuan, C.; Liu, Y.; Wang, Z. Adult exposure to bisphenol A in rare minnow Gobiocypris rarus reduces sperm quality with disruption of testicular aquaporins. Chemosphere 2018, 193, 365–375. [Google Scholar] [CrossRef] [PubMed]
  169. Kang, I.J.; Hano, T.; Oshima, Y.; Yokota, H.; Tsuruda, Y.; Shimasaki, Y.; Honjo, T. Anti-androgen flutamide affects gonadal development and reproduction in medaka (Oryzias latipes). Mar. Environ. Res. 2006, 62, S253–S257. [Google Scholar] [CrossRef]
  170. Schoenfuss, H.L.; Levitt, J.T.; Van Der Kraak, G.; Sorensen, P.W. Ten-week exposure to treated sewage discharge has relatively minor, variable effects on reproductive behavior and sperm production in goldfish. Environ. Toxicol. Chem. 2002, 21, 2185–2190. [Google Scholar] [CrossRef] [PubMed]
  171. McAllister, B.G.; Kime, D.E. Early life exposure to environmental levels of the aromatase inhibitor tributyltin causes masculinisation and irreversible sperm damage in zebrafish (Danio rerio). Aquat. Toxicol. 2003, 65, 309–316. [Google Scholar] [CrossRef]
  172. Haubruge, E.; Petit, F.; Gage, M.J.G. Reduced sperm counts in guppies (Poecilia reticulata) following exposure to low levels of tributyltin and bisphenol A. Proc. Biol. Sci. 2000, 267, 2333–2337. [Google Scholar] [CrossRef] [Green Version]
  173. Hatef, A.; Alavi, S.M.H.; Abdulfatah, A.; Fontaine, P.; Rodina, M.; Linhart, O. Adverse effects of bisphenol A on reproductive physiology in male goldfish at environmentally relevant concentrations. Ecotoxicol. Environ. Saf. 2012, 76, 56–62. [Google Scholar] [CrossRef]
  174. Hatef, A.; Zare, A.; Alavi, S.M.H.; Habibi, H.R.; Linhart, O. Modulations of androgen and estrogen mediating genes and testicular response in male goldfish exposed to bisphenol A. Environ. Toxicol. Chem. 2012, 31, 2069–2077. [Google Scholar] [CrossRef]
  175. Golshan, M.; Hatef, A.; Socha, M.; Milla, S.; Butts, I.A.E.; Carnevali, O.; Rodina, M.; Sokołowska-Mikołajczyk, M.; Fontaine, P.; Linhart, O.; et al. Di-(2-ethylhexyl)-phthalate disrupts pituitary and testicular hormonal functions to reduce sperm quality in mature goldfish. Aquat. Toxicol. 2015, 163, 16–26. [Google Scholar] [CrossRef]
  176. Baatrup, E.; Junge, M. Antiandrogenic pesticides disrupt sexual characteristics in the adult male guppy (Poecilia reticulata). Environ. Health Perspect. 2001, 109, 1063–1070. [Google Scholar] [CrossRef] [PubMed]
  177. Bayley, M.; Foged Larsen, P.; Bækgaard, H.; Baatrup, E. The effects of vinclozolin, an anti-androgenic fungicide, on male guppy secondary sex characters and reproductive success. Biol. Reprod. 2003, 69, 1951–1956. [Google Scholar] [CrossRef] [Green Version]
  178. Hatef, A.; Alavi, S.M.H.; Milla, S.; Křišťan, J.; Golshan, M.; Fontaine, P.; Linhart, O. Anti-androgen vinclozolin impairs sperm quality and steroidogenesis in goldfish. Aquat. Toxicol. 2012, 122–123, 181–187. [Google Scholar] [CrossRef]
  179. Yin, P.; Li, Y.W.; Chen, Q.L.; Liu, Z.H. Diethylstilbestrol, flutamide and their combination impaired the spermatogenesis of male adult zebrafish through disrupting HPG axis, meiosis and apoptosis. Aquat. Toxicol. 2017, 185, 129–137. [Google Scholar] [CrossRef]
  180. Runnalls, T.; Hala, D.N.; Sumpter, J.P. Preliminary studies into the effects of the human pharamecutical Clofibtric acid on sperm parameters in adult fathead minnows. Aquat. Toxicol. 2007, 84, 111–118. [Google Scholar] [CrossRef] [PubMed]
  181. Mennigen, J.A.; Lado, W.E.; Zamora, J.M.; Duarte-Guterman, P.; Langlois, V.S.; Metcalfe, C.D.; Chang, J.P.; Moon, T.W.; Trudeau, V.L. Waterborne fluoxetine disrupts the reproductive axis in sexually mature male goldfish, Carassius auratus. Aquat. Toxicol. 2010, 100, 354–364. [Google Scholar] [CrossRef]
  182. Roggio, M.A.; Guyon, N.F.; Hued, A.C.; Ame, M.V.; Valdes, M.E.; Giojalas, L.C.; Wunderlin, D.A.; Bistoni, M.A. Effects of the synthetic estrogen 17α-ethinylestradiol on aromatase expression, reproductive behavior and sperm quality in the fish Jenynsia multidentata. Bull. Environ. Contam. Toxicol. 2014, 92, 579–584. [Google Scholar] [CrossRef] [PubMed]
  183. Bertram, M.G.; Ecker, T.E.; Wong, B.B.M.; O’Bryan, M.K.; Baumgartner, J.B.; Martin, J.M.; Saaristo, M. The antidepressant fluoxetine alters mechanisms of pre- and post-copulatory sexual selection in the eastern mosquitofish (Gambusia holbrooki). Environ. Pollut. 2018, 238, 238–247. [Google Scholar] [CrossRef]
  184. Sayed, A.E.D.H.; Mahmoud, U.M.; Mekkawy, I.A. Reproductive biomarkers to identify endocrine disruption in Clarias gariepinus exposed to 4-nonylphenol. Ecotoxicol. Environ. Saf. 2012, 78, 310–319. [Google Scholar] [CrossRef] [PubMed]
  185. Franco, J.G., Jr.; Baruffi, R.L.; Mauri, A.L.; Petersen, C.G.; Oliveira, J.B.; Vagnini, L. Significance of large nuclear vacuoles in human spermatozoa: Implications for ICSI. Reprod. Biomed. Online 2008, 17, 42–45. [Google Scholar] [CrossRef]
  186. Yoshikawa, H.; Morishima, K.; Fujimoto, T.; Arias-Rodriguez, L.; Yamaha, E.; Arai, K. Ploidy manipulation using diploid sperm in the loach, Misgurnus anguillicaudatus: A review. J. Appl. Ichthyol. 2008, 24, 410–414. [Google Scholar] [CrossRef] [Green Version]
  187. Na-Nakorn, U.; Rangsin, W.; Boonngam, J. Allotriploidy increases sterility in the hybrid between Clarias macrocephalus and Clarias gariepinus. Aquaculture 2004, 237, 73–88. [Google Scholar] [CrossRef]
  188. Brown, K.H.; Schultz, I.R.; Cloud, J.G.; Nagler, J.J. Aneuploid sperm formation in rainbow trout exposed to the environmental estrogen 17α-ethynylestradiol. Proc. Natl. Acad. Sci. USA 2008, 105, 19786–19791. [Google Scholar] [CrossRef] [Green Version]
  189. Liu, Y.; Yuan, C.; Chen, S.; Zheng, Y.; Zhang, Y.; Gao, J.; Wang, Z. Global and cyp19a1a gene specific DNA methylation in gonads of adult rare minnow Gobiocypris rarus under Bisphenol A exposure. Aquat. Toxicol. 2014, 156, 10–16. [Google Scholar] [CrossRef] [PubMed]
  190. Yuan, C.; Zhang, Y.; Liu, Y.; Zhang, T.; Wang, Z. Enhanced GSH synthesis by Bisphenol A exposure promoted DNA methylation process in the testes of adult rare minnow Gobiocypris rarus. Aquat. Toxicol. 2016, 178, 99–105. [Google Scholar] [CrossRef]
  191. Horii, T.; Hatada, I. Regulation of CpG methylation by Dnmt and Tet in pluripotent stem cells. J. Reprod. Dev. 2016, 62, 331–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Frankel, T.; Yonkos, L.; Ampy, F.; Frankel, J. Exposure to levonorgestrel increases nest acquisition success and decreases sperm motility in the male fathead minnow (Pimephales promelas). Environ. Toxicol. Chem. 2018, 37, 1131–1137. [Google Scholar] [CrossRef]
  193. Zebral, Y.D.; Anni, I.S.A.; Junior, A.S.V.; Corcini, C.D.; da Silva, J.C.; Caldas, J.S.; Acosta, I.B.; Afonso, S.B.; Bianchini, A. Life-time exposure to waterborne copper IV: Sperm quality parameters are negatively affected in the killifish Poecilia vivipara. Chemosphere 2019, 236, 124332. [Google Scholar] [CrossRef]
  194. Murack, P.J.; Parrish, J.; Barry, T.P. Effects of progesterone on sperm motility in fathead minnow (Pimephales promelas). Aquat. Toxicol. 2011, 104, 121–125. [Google Scholar] [CrossRef]
  195. Das Neves, J.; Barnhoorn, I.E.J.; Wagenaar, G.M. The effects of environmentally relevant concentrations of aldrin and methoxychlor on the testes and sperm of male Clarias gariepinus (Burchell, 1822) after short-term exposure. Fish Physiol. Biochem. 2018, 44, 1421–1434. [Google Scholar] [CrossRef]
  196. Martin, J.M.; Bertram, M.G.; Saaristo, M.; Ecker, T.E.; Hannington, S.L.; Tanner, J.L.; Michelangeli, M.; O’Bryan, M.K.; Wong, B.B.M. Impact of the widespread pharmaceutical pollutant fluoxetine on behaviour and sperm traits in a freshwater fish. Sci. Total Environ. 2019, 650, 1771–1778. [Google Scholar] [CrossRef]
  197. Guyón, N.F.; Roggio, M.A.; Amé, M.V.; Hued, A.C.; Valdés, M.E.; Giojalas, L.C.; Wunderlin, D.A.; Bistoni, M.A. Impairments in aromatase expression, reproductive behavior, and sperm quality of male fish exposed to 17β-estradiol. Environ. Toxicol. Chem. 2012, 31, 935–940. [Google Scholar] [CrossRef]
  198. Soares, J.; Coimbra, A.M.; Reis-Henriques, M.A.; Monteiro, N.M.; Vieira, M.N.; Oliveira, J.M.A.; Guedes-Dias, P.; Fontaínhas-Fernandes, A.; Silva Parra, S.; Carvalho, A.P.; et al. Disruption of zebrafish (Danio rerio) embryonic development after full life-cycle parental exposure to low levels of ethinylestradiol. Aquat. Toxicol. 2009, 95, 330–338. [Google Scholar] [CrossRef]
  199. Velicu, M.; Suri, R. Presence of steroid hormones and antibiotics in surface water of agricultural, suburban and mixed-use areas. Environ. Monit. Assess. 2009, 154, 349–359. [Google Scholar] [CrossRef] [PubMed]
  200. Kuch, H.M.; Ballschmitter, K. Determination of endocrine-disrupting phenolic compounds and estrogens in surface and drinking water by HRGC-(NCI)-MS in the picogram per liter range. Environ. Sci. Technol. 2001, 35, 3201–3206. [Google Scholar] [CrossRef]
  201. Baronti, C.; Curini, R.; D’Ascenzo, G.; Di Corcia, A.; Gentili, A.; Samperi, R. Monitoring natural and synthetic estrogens at activated treatment plants and in receiving river water. Environ. Sci. Technol. 2000, 34, 5059–5066. [Google Scholar] [CrossRef]
  202. Nasu, M.; Goto, M.; Kato, H.; Oshima, Y.; Tanaka, H. Study on endocrine disrupting chemicals in wastewater treatment plants. Water Sci. Technol. 2000, 43, 101–108. [Google Scholar] [CrossRef]
  203. Desbrow, C.; Routledge, E.J.; Brighty, G.C.; Sumpter, J.P.; Waldock, M. Identification of estrogenic chemicals in STW effluent: 1. Chemical fractionation and in vitro biological screening. Environ. Sci. Technol. 1998, 32, 1549–1558. [Google Scholar] [CrossRef]
  204. Liu, S.; Ying, G.G.; Zhao, J.L.; Chen, F.; Yang, B.; Zhou, L.J.; Lai, H.J. Trace analysis of 28 steroids in surface water, wastewater and sludge samples by rapid resolution liquid chromatography-electrospray ionization tandem mass spectrometry. J. Chromatogr. A 2011, 1218, 1367–1378. [Google Scholar] [CrossRef] [PubMed]
  205. Belfroid, A.C.; Van der Horst, A.; Vethaak, A.D.; Schafer, A.J.; Ris, G.B.J.; Wegener, J.; Cofino, W.P. Analysis and occurrence of estrogenic hormones and their glucuronides in surface water and waste water in the Netherlands. Sci. Total Environ. 1999, 225, 101–108. [Google Scholar] [CrossRef]
  206. Ternes, T.A.; Stumpf, M.; Mueller, J.; Haberer, K.; Wilken, R.D.; Servos, M. Behaviour and occurrence of estrogens in municipal sewage treatment plants—I. Investigations in Germany, Canada and Brazil. Sci. Total Environ. 1999, 225, 81–90. [Google Scholar] [CrossRef]
  207. Snyder, S.; Giesy, J.; Kannan, K.; Gross, T.; Snyder, E.; Verbrugge, D.; Keith, T. Analytical methods for detection of selected estrogenic compounds in aqueous mixtures. Environ. Sci. Technol. 1999, 33, 2814–2820. [Google Scholar] [CrossRef]
  208. Yu, Q.; Geng, J.; Ren, H. Occurrence and fate of androgens in municipal wastewater treatment plants in China. Chemosphere 2019, 237, 124371. [Google Scholar] [CrossRef] [PubMed]
  209. Blankvoort, B.M.G.; Rodenburg, R.J.T.; Murk, A.J.; Koeman, J.H.; Schilt, R.; Aarts, J.M.M.J.G. Androgenic activity in surface water samples detected using the AR-LUX assay: Indication for mixture effects. Environ. Toxicol. Pharmacol. 2005, 19, 263–272. [Google Scholar] [CrossRef] [PubMed]
  210. Höhne, C.; Püttmann, W. Occurrence and temporal variations of the xenoestrogens bisphenol A, 4-tert-octylphenol, and tech. 4-nonylphenol in two German wastewater treatment plants. Environ. Sci. Pollut. Res. Int. 2008, 15, 405–416. [Google Scholar] [CrossRef]
  211. Cailleaud, K.; Forget-Leray, J.; Souissi, S.; Lardy, S.; Augagneur, S.; Budzinski, H. Seasonal variation of hydrophobic organic contaminant concentrations in the water-column of the Seine Estuary and their transfer to a planktonic species Eurytemora affinis (Calanoid, copepod). Part 2: Alkylphenol-polyethoxylates. Chemosphere 2007, 70, 281–287. [Google Scholar] [CrossRef] [PubMed]
  212. Mohapatra, S.; Huang, C.H.; Mukherji, S.; Padhye, L.P. Occurrence and fate of pharmaceuticals in WWTPs in India and comparison with a similar study in the United States. Chemosphere 2016, 159, 526–535. [Google Scholar] [CrossRef]
  213. Kang, J.H.; Aasi, D.; Katayama, Y. Bisphenol A in the aquatic environment and its endocrine-disruptive effects on aquatic organisms. Crit. Rev. Toxicol. 2007, 37, 607–625. [Google Scholar] [CrossRef]
  214. Fromme, H.; Küchler, T.; Otto, T.; Pilz, K.; Müller, J.; Wenzel, A. Occurrence of phthalates and bisphenol A and F in the environment. Water Res. 2002, 36, 1429–1438. [Google Scholar] [CrossRef]
  215. Yang, Y.; Lu, L.; Zhang, J.; Yang, Y.; Wu, Y.; Shao, B. Simultaneous determination of seven bisphenols in environmental water and solid samples by liquid chromatography-electrospray tandem mass spectrometry. J. Chromatogr. A 2014, 1328, 26–34. [Google Scholar] [CrossRef] [PubMed]
  216. Bókony, V.; Üveges, B.; Ujhegyi, N.; Verebélyi, V.; Nemesházi, E.; Csíkvári, O.; Hettyey, A. Endocrine disruptors in breeding ponds and reproductive health of toads in agricultural, urban and natural landscapes. Sci. Total Environ. 2018, 634, 1335–1345. [Google Scholar] [CrossRef]
  217. Wen, Z.; Huang, X.; Gao, D.; Liu, G.; Fang, C.; Shang, Y.; Du, J.; Zhao, Y.; Lv, L.; Song, K. Phthalate esters in surface water of Songhua River watershed associated with land use types, Northeast China. Environ. Sci. Pollut. Res. Int. 2018, 25, 7688–7698. [Google Scholar] [CrossRef] [PubMed]
  218. Zhang, Z.M.; Zhang, H.H.; Zou, Y.W.; Yang, G.P. Distribution and ecotoxicological state of phthalate esters in the sea-surface microlayer, seawater and sediment of the Bohai Sea and the Yellow Sea. Environ. Pollut. 2018, 240, 235–247. [Google Scholar] [CrossRef] [PubMed]
  219. Readman, J.W.; Albanis, T.A.; Barcelo, D.; Galassi, S.; Tronczynski, J.; Gabrielides, G.P. Fungicide contamination of Mediterranean estuarine waters: Results from a MED POL pilot survey. Mar. Poll. Bull. 1997, 34, 259–263. [Google Scholar] [CrossRef]
  220. Kreuger, J.; Graaf, S.; Patring, J.; Adielsson, S. Pesticides in Surface Water in Areas with Open Ground and Greenhouse Horticultural Crops in Sweden 2008; Swedish University of Agricultural Sciences: Uppsala, Sweden, 2010. [Google Scholar]
  221. Zheng, S.; Chen, B.; Qiu, X.; Chen, M.; Ma, Z.; Yu, X. Distribution and risk assessment of 82 pesticides in Jiulong River and estuary in South China. Chemosphere 2016, 144, 1177–1192. [Google Scholar] [CrossRef] [PubMed]
  222. Wang, L.; Ying, G.G.; Zhao, J.L.; Liu, S.; Yang, B.; Zhou, L.J.; Tao, R.; Su, H.C. Assessing estrogenic activity in surface water and sediment of the Liao River system in northeast China using combined chemical and biological tools. Environ. Pollut. 2011, 159, 148–156. [Google Scholar] [CrossRef]
  223. Chen, T.S.; Chen, T.C.; Yeh, K.J.C.; Chao, H.R.; Liaw, E.T.; Hsieh, C.Y.; Chen, K.C.; Hsieh, L.T.; Yeh, Y.L. High estrogen concentrations in receiving river discharge from a concentrated livestock feedlot. Sci. Total Environ. 2010, 408, 3223–3230. [Google Scholar] [CrossRef] [PubMed]
  224. Lei, B.; Huang, S.; Zhou, Y.; Wang, D.; Wang, Z. Levels of six estrogens in water and sediment from three rivers in Tianjin area, China. Chemosphere 2009, 76, 36–42. [Google Scholar] [CrossRef]
  225. Pojana, G.; Gomiero, A.; Jonkers, N.; Marcomini, A. Natural and synthetic endocrine disrupting compounds (EDCs) in water, sediment and biota of a coastal lagoon. Environ. Int. 2007, 33, 929–936. [Google Scholar] [CrossRef]
  226. Wu, M.; Xiang, J.; Que, C.; Chen, F.; Xu, G. Occurrence and fate of psychiatric pharmaceuticals in the urban water system of Shanghai, China. Chemosphere 2015, 138, 486–493. [Google Scholar] [CrossRef]
  227. Paíga, P.; Santos, L.H.; Ramos, S.; Jorge, S.; Silva, J.G.; Delerue-Matos, C. Presence of pharmaceuticals in the Lis river (Portugal): Sources, fate and seasonal variation. Sci. Total Environ. 2016, 573, 164–177. [Google Scholar] [CrossRef]
  228. Lindim, C.; van Gils, J.; Georgieva, D.; Mekenyan, O.; Cousins, I.T. Evaluation of human pharmaceutical emissions and concentrations in Swedish river basins. Sci. Total Environ. 2016, 572, 508–519. [Google Scholar] [CrossRef]
  229. Santos, L.H.; Gros, M.; Rodriguez-Mozaz, S.; Delerue-Matos, C.; Pena, A.; Barceló, D.; Montenegro, M.C.B. Contribution of hospital effluents to the load of pharmaceuticals in urban wastewaters: Identification of ecologically relevant pharmaceuticals. Sci. Total Environ. 2013, 461, 302–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Yan, Q.; Zhang, Y.X.; Kang, J.; Gan, X.M.; Xu, Y.P.; Guo, J.S.; Gao, X. A preliminary study on the occurrence of pharmaceutically active compounds in the river basins and their removal in two conventional drinking water treatment plants in Chongqing, China. Clean Soil Air Water 2015, 43, 794–803. [Google Scholar] [CrossRef]
  231. Nentwig, G.; Oetken, M.; Oehlmann, J. Effects of Pharmaceuticals on Aquatic Invertebrates—The Example of Carbamazepine and Clofibric Acid. In Pharmaceuticals in the Environment. Sources, Fate, Effects and Risks; Kümmerer, K., Ed.; Springer: Heidelberg, Germany, 2004; pp. 195–208. [Google Scholar] [CrossRef] [Green Version]
  232. Anjum, R.; Malik, A. Evaluation of mutagenicity of wastewater in the vicinity of pesticide industry. Environ. Toxicol. Pharmacol. 2013, 35, 284–291. [Google Scholar] [CrossRef] [PubMed]
  233. Kang, Y.; Zhang, G. Analysis of organophosphorous pesticides from source water using solid-phase extraction technique. China Environ. Sci. 2000, 20, 1–4. [Google Scholar]
  234. Kumari, B.; Madan, V.K.; Kathpal, T.S. Pesticide residues in rain water from Hisar, India. Environ. Monit. Assess. 2007, 133, 467–471. [Google Scholar] [CrossRef]
  235. Canty, M.N.; Hutchinson, T.H.; Brown, R.J.; Jones, M.B.; Jha, A.N. Linking genotoxic responses with cytotoxic and behavioural or physiological consequences: Differential sensitivity of echinoderms (Asterias rubens) and marine molluscs (Mytilus edulis). Aquat. Toxicol. 2009, 94, 68–76. [Google Scholar] [CrossRef]
  236. Guo, S.; Qian, L.; Shi, H.; Barry, T.; Cao, Q.; Liu, J. Effects of tributyltin (TBT) on Xenopus tropicalis embryos at environmentally relevant concentrations. Chemosphere 2010, 79, 529–533. [Google Scholar] [CrossRef] [PubMed]
  237. Li, X.; Yin, P.; Zhao, L. Phthalate esters in water and surface sediments of the Pearl River Estuary: Distribution, ecological, and human health risks. Environ. Sci. Pollut. Res. 2016, 23, 19341–19349. [Google Scholar] [CrossRef]
  238. Nibamureke, U.M.C.; Barnhoorn, I.E.J.; Wagenaar, G.M. Health assessment of freshwater fish species from Albasini Dam, outside a DDT-sprayed area in Limpopo province, South Africa: A preliminary study. Afr. J. Aquat. Sci. 2016, 41, 1–12. [Google Scholar] [CrossRef]
  239. Song, S.; Ruan, T.; Wang, T.; Liu, R.; Jiang, G. Distribution and preliminary exposure assessment of bisphenol AF (BPAF) in various environmental matrices around a manufacturing plant in China. Environ. Sci. Technol. 2012, 46, 13136–13143. [Google Scholar] [CrossRef]
  240. Corcoran, J.; Winter, M.J.; Tyler, C.R. Pharmaceuticals in the aquatic environment: A critical review of the evidence for health effects in fish. Crit. Rev. Toxicol. 2010, 40, 287–304. [Google Scholar] [CrossRef]
  241. Patel, M.; Kumar, R.; Kishor, K.; Mlsna, T.; Pittman, C.U., Jr.; Mohan, D. Pharmaceuticals of emerging concern in aquatic systems: Chemistry, occurrence, effects, and removal methods. Chem. Rev. 2019, 119, 3510–3673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Dsikowitzky, L.; Schwarzbauer, J. Industrial organic contaminants: Identification, toxicity and fate in the environment. Environ. Chem. Lett. 2014, 12, 371–386. [Google Scholar] [CrossRef]
  243. Petrie, B.; Barden, R.; Kasprzyk-Hordern, B. A review on emerging contaminants in wastewaters and the environment: Current knowledge, understudied areas and recommendations for future monitoring. Water Res. 2015, 72, 3–27. [Google Scholar] [CrossRef] [PubMed]
  244. Schwarzenbach, R.P.; Egli, T.; Hofstetter, T.B.; von Gunten, U.; Wehrli, B. Global water pollution and human health. Ann. Rev. Environ. Resour. 2010, 35, 109–136. [Google Scholar] [CrossRef]
  245. Denslow, N.; Sepulveda, M. Ecotoxicological effects of endocrine disrupting compounds on fish reproduction. In The Fish Oocyte: From Basic Studies to Biotechnological Applications; Babin, P.J., Cerdà, J., Lubzens, E., Eds.; Springer: Dordrecht, The Netherlands, 2008; pp. 255–322. [Google Scholar] [CrossRef]
  246. Dietrich, M.A.; Nynca, J.; Ciereszko, A. Proteomic and metabolomic insights into the functions of the male reproductive system in fishes. Theriogenology 2019, 132, 182–200. [Google Scholar] [CrossRef]
  247. Hatef, A.; Alavi, S.M.H.; Butts, I.A.E.; Policar, T.; Linhart, O. The mechanisms of action of mercury on sperm morphology, adenosine-5′-triphosphate content and motility in Perca fluviatilis (Percidae; Teleostei). Environ. Toxicol. Chem. 2011, 30, 905–914. [Google Scholar] [CrossRef]
  248. Schiffer, C.; Müller, A.; Egeberg, D.L.; Alvarez, L.; Brenker, C.; Rehfeld, A.; Frederiksen, H.; Wäschle, B.; Kaupp, U.B.; Balbach, M.; et al. Direct action of endocrine disrupting chemicals on human sperm. EMBO Rep. 2014, 15, 758–765. [Google Scholar] [CrossRef] [Green Version]
  249. De la Fuente, I.M.; Cortés, J.M.; Valero, E.; Desroches, M.; Rodrigues, S.; Malaina, I.; Martínez, L. On the dynamics of the adenylate energy system: Homeorhesis vs. homeostasis. PLoS ONE 2014, 9, e108676. [Google Scholar] [CrossRef] [Green Version]
  250. Mills, L.J.; Chichester, C. Review of evidence: Are endocrine-disrupting chemicals in the aquatic environment impacting fish populations? Sci. Total Environ. 2005, 343, 1–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Zhou, J.; Zhu, X.S.; Cai, Z.H. Endocrine disruptors: An overview and discussion on issues surrounding their impact on marine animals. J. Mar. Anim. Ecol. 2009, 2, 7–17. [Google Scholar]
  252. Hamilton, P.B.; Cowx, I.G.; Oleksiak, M.F.; Griffiths, A.M.; Grahn, M.; Stevens, J.R.; Carvalho, G.R.; Nicol, E.; Tyler, C.R. Population-level consequences for wild fish exposed to sublethal concentrations of chemicals—A critical review. Fish Fish. 2016, 17, 545–566. [Google Scholar] [CrossRef]
  253. Boryshpolets, S.; Kowalski, R.K.; Dietrich, G.J.; Dzyuba, B.; Ciereszko, A. Different computer-assisted sperm analysis (CASA) systems highly influence sperm motility parameters. Theriogenology 2013, 80, 758–765. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Reproductive system in male fishes. Panels (A,C) show the anatomy of the reproductive system in primitive fishes (sturgeons). Sperm is released from the testes into the testicular ducts, which pass the kidney. At spawning, semen is released into the aquatic environment through the urinary ducts opened into the urogenital opening (UO). Panels (B,D) show the anatomy of the reproductive system in bony fishes (teleosts). Sperm is released from the testes into the testicular ducts. At spawning, semen is released into the aquatic environment through the sperm ducts opened into the UO. Panels (E,F) are schematic of the tubular testis and lobular testis, respectively. Panel G shows testicular compartments in fishes. K, kidneys; SD, sperm duct; SC-I, primary spermatocyte, SC-II, secondary spermatocyte; SG, spermatogonia; SP, spermatid; SZ, sperm; T, testis; TD, testicular duct; UB, Urinary bladder; UD (WD), urinary duct (Wolffian duct). The panels are modified from Grier [30], Nagahama [32]; Alavi et al. [28] and Dzyuba et al. [29]. The photo of panel A is courtesy of Associate Professor Borys Dzyuba from the sterlet (Acipenser ruthenus). The photo of panel B is from S. M. H. Alavi from the Northern pike (Esox Lucius). Panels C-G credits: © S. Barzegar-Fallah.
Figure 1. Reproductive system in male fishes. Panels (A,C) show the anatomy of the reproductive system in primitive fishes (sturgeons). Sperm is released from the testes into the testicular ducts, which pass the kidney. At spawning, semen is released into the aquatic environment through the urinary ducts opened into the urogenital opening (UO). Panels (B,D) show the anatomy of the reproductive system in bony fishes (teleosts). Sperm is released from the testes into the testicular ducts. At spawning, semen is released into the aquatic environment through the sperm ducts opened into the UO. Panels (E,F) are schematic of the tubular testis and lobular testis, respectively. Panel G shows testicular compartments in fishes. K, kidneys; SD, sperm duct; SC-I, primary spermatocyte, SC-II, secondary spermatocyte; SG, spermatogonia; SP, spermatid; SZ, sperm; T, testis; TD, testicular duct; UB, Urinary bladder; UD (WD), urinary duct (Wolffian duct). The panels are modified from Grier [30], Nagahama [32]; Alavi et al. [28] and Dzyuba et al. [29]. The photo of panel A is courtesy of Associate Professor Borys Dzyuba from the sterlet (Acipenser ruthenus). The photo of panel B is from S. M. H. Alavi from the Northern pike (Esox Lucius). Panels C-G credits: © S. Barzegar-Fallah.
Animals 11 02817 g001
Figure 2. Sperm morphology in primitive (chondrostei) and bony (teleostei) fishes. Sperm is composed of a head (nucleus, N), midpiece (M) and flagellum (F). In chondrostei fishes (such as sturgeons), there is an acrosome (A) at the top of the head of sperm. The ultrastructure compartments of sperm are similar between chondrostei and teleostei fishes: DC, distal centriole; PC, proximal centriole; Mt, mitochondria. The structure of the motility apparatus called “axoneme” is highly conserved, and possesses the typical 9 + 2 microtubule structure of cilia surrounded by plasma membrane. The electron micrographs are selected from the Russian sturgeon (Acipenser gueldenstaedtii) [47], and Atlantic halibut (Hippoglossus hippoglossus) sperm [48]. The schematic of the axoneme is from Inaba [49].
Figure 2. Sperm morphology in primitive (chondrostei) and bony (teleostei) fishes. Sperm is composed of a head (nucleus, N), midpiece (M) and flagellum (F). In chondrostei fishes (such as sturgeons), there is an acrosome (A) at the top of the head of sperm. The ultrastructure compartments of sperm are similar between chondrostei and teleostei fishes: DC, distal centriole; PC, proximal centriole; Mt, mitochondria. The structure of the motility apparatus called “axoneme” is highly conserved, and possesses the typical 9 + 2 microtubule structure of cilia surrounded by plasma membrane. The electron micrographs are selected from the Russian sturgeon (Acipenser gueldenstaedtii) [47], and Atlantic halibut (Hippoglossus hippoglossus) sperm [48]. The schematic of the axoneme is from Inaba [49].
Animals 11 02817 g002
Figure 3. Sperm motility signaling and kinetics in fishes. Panel (A) summarizes sperm motility signaling in fishes. Sperm is immotile in the sperm ducts and seminal plasma. At spawning, a hypo- osmolality accompanied by K+ efflux or hyper-osmolality accompanied by water efflux trigger sperm motility activation in freshwater and marine fish species, respectively. Activation of ATP- dependent sperm motility initiation is associated with an increase in intracellular calcium ([Ca2+]i) ions in all fish species and an increase in intracellular potassium ([K+]i) ions in marine species, while cyclic adenosine monophosphate (cAMP) remains unchanged. However, demembranated sperm of salmonids requires cAMP for axonemal beating. Panel (B) shows sperm motility kinetics in fishes. After initiation of sperm motility, percentage of motile sperm and sperm velocity decrease rapidly in both freshwater and marine fishes due to depletion of adenosine triphosphate (ATP) content. Panel (C) is a schematic representing various sperm velocity parameters analyzed by a computer-assisted sperm analysis. The curvilinear velocity (VCL) is the velocity along the trajectory of sperm head. The straight line velocity (VSL) is the straight line distance between the start and end points of the track divided by the duration of the movement. The angular path velocity (VAP) is the velocity along a derived smoothed path.
Figure 3. Sperm motility signaling and kinetics in fishes. Panel (A) summarizes sperm motility signaling in fishes. Sperm is immotile in the sperm ducts and seminal plasma. At spawning, a hypo- osmolality accompanied by K+ efflux or hyper-osmolality accompanied by water efflux trigger sperm motility activation in freshwater and marine fish species, respectively. Activation of ATP- dependent sperm motility initiation is associated with an increase in intracellular calcium ([Ca2+]i) ions in all fish species and an increase in intracellular potassium ([K+]i) ions in marine species, while cyclic adenosine monophosphate (cAMP) remains unchanged. However, demembranated sperm of salmonids requires cAMP for axonemal beating. Panel (B) shows sperm motility kinetics in fishes. After initiation of sperm motility, percentage of motile sperm and sperm velocity decrease rapidly in both freshwater and marine fishes due to depletion of adenosine triphosphate (ATP) content. Panel (C) is a schematic representing various sperm velocity parameters analyzed by a computer-assisted sperm analysis. The curvilinear velocity (VCL) is the velocity along the trajectory of sperm head. The straight line velocity (VSL) is the straight line distance between the start and end points of the track divided by the duration of the movement. The angular path velocity (VAP) is the velocity along a derived smoothed path.
Animals 11 02817 g003
Figure 4. Indicators and determinants of fertility in male fishes. Fertilization success is assessed by fertilization rate or hatching rate (A). Sperm production, morphology, genome and motility are key determinants of fertility in male fishes (B).
Figure 4. Indicators and determinants of fertility in male fishes. Fertilization success is assessed by fertilization rate or hatching rate (A). Sperm production, morphology, genome and motility are key determinants of fertility in male fishes (B).
Animals 11 02817 g004
Figure 5. The adverse effects of environmental contaminants (ECs) on fertility in male fishes. The aquatic environments are the final repository of the municipal wastewater, and industrial, agri- cultural, and treatment plants effluents that contain various natural and man-made contaminants including metals, pesticides, pharmaceuticals, and organic compounds, as well as compounds used in personal care products. Evidences from wildlife and results of laboratory studies reveal adverse effects of ECs on sperm production, morphology, genome, and motility to cause fertility threat at the level of the individual. Contribution of ECs to declining fish populations is largely unknown and needs to be elucidated.
Figure 5. The adverse effects of environmental contaminants (ECs) on fertility in male fishes. The aquatic environments are the final repository of the municipal wastewater, and industrial, agri- cultural, and treatment plants effluents that contain various natural and man-made contaminants including metals, pesticides, pharmaceuticals, and organic compounds, as well as compounds used in personal care products. Evidences from wildlife and results of laboratory studies reveal adverse effects of ECs on sperm production, morphology, genome, and motility to cause fertility threat at the level of the individual. Contribution of ECs to declining fish populations is largely unknown and needs to be elucidated.
Animals 11 02817 g005
Figure 6. Effects and mechanisms of action of environmental contaminants (ECs) on the reproductive system and fertility in male fishes. In the left, hypothalamus–pituitary–testis (HPT) regulation of sperm production and maturation is shown. The follicle-stimulating hormone (FSH) regulates 11-ketotestosterone (11-KT)-stimulated spermatogenesis. At spawning, luteinizing hormone (LH) regulates 17α,20β-dihydroxy-4-pregnen-3-one (17,20β-P)-stimulated sperm maturation. Sperm are released into the sperm ducts to acquire potential for the motility initiation. In the right, lessons from wildlife and laboratory studies on the reproductive system in male fishes are shown. ECs cause phenotypic changes in the reproductive system and affect determinants of fertility. It has been reported that adverse effects of ECs on sperm production, genome, and motility kinetics are associated with generation of reaction oxygen species (ROS) and inhibition of testicular testosterone (T), 11-KT, and 17,20β-P. GnRH, gonadotropin-releasing hormone; E2, 17β-estradiol; Vtg, vitellogenin.
Figure 6. Effects and mechanisms of action of environmental contaminants (ECs) on the reproductive system and fertility in male fishes. In the left, hypothalamus–pituitary–testis (HPT) regulation of sperm production and maturation is shown. The follicle-stimulating hormone (FSH) regulates 11-ketotestosterone (11-KT)-stimulated spermatogenesis. At spawning, luteinizing hormone (LH) regulates 17α,20β-dihydroxy-4-pregnen-3-one (17,20β-P)-stimulated sperm maturation. Sperm are released into the sperm ducts to acquire potential for the motility initiation. In the right, lessons from wildlife and laboratory studies on the reproductive system in male fishes are shown. ECs cause phenotypic changes in the reproductive system and affect determinants of fertility. It has been reported that adverse effects of ECs on sperm production, genome, and motility kinetics are associated with generation of reaction oxygen species (ROS) and inhibition of testicular testosterone (T), 11-KT, and 17,20β-P. GnRH, gonadotropin-releasing hormone; E2, 17β-estradiol; Vtg, vitellogenin.
Animals 11 02817 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alavi, S.M.H.; Barzegar-Fallah, S.; Rahdar, P.; Ahmadi, M.M.; Yavari, M.; Hatef, A.; Golshan, M.; Linhart, O. A Review on Environmental Contaminants-Related Fertility Threat in Male Fishes: Effects and Possible Mechanisms of Action Learned from Wildlife and Laboratory Studies. Animals 2021, 11, 2817. https://doi.org/10.3390/ani11102817

AMA Style

Alavi SMH, Barzegar-Fallah S, Rahdar P, Ahmadi MM, Yavari M, Hatef A, Golshan M, Linhart O. A Review on Environmental Contaminants-Related Fertility Threat in Male Fishes: Effects and Possible Mechanisms of Action Learned from Wildlife and Laboratory Studies. Animals. 2021; 11(10):2817. https://doi.org/10.3390/ani11102817

Chicago/Turabian Style

Alavi, Sayyed Mohammad Hadi, Sepideh Barzegar-Fallah, Parastoo Rahdar, Mohammad Mahdi Ahmadi, Mina Yavari, Azadeh Hatef, Mahdi Golshan, and Otomar Linhart. 2021. "A Review on Environmental Contaminants-Related Fertility Threat in Male Fishes: Effects and Possible Mechanisms of Action Learned from Wildlife and Laboratory Studies" Animals 11, no. 10: 2817. https://doi.org/10.3390/ani11102817

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop