<<

Male infertility testing: reactive species and capacity

Edmund Y. Ko, M.D.,a Edmund S. Sabanegh Jr., M.D.,b and Ashok Agarwal, Ph.D.c a Department of Urology, Loma Linda University, School of Medicine, Loma Linda, California; and b Department of Urology and c Center for Reproductive Medicine, Glickman Urological and Kidney Institute, Cleveland Clinic Foundation, Cleveland, Ohio

Reactive oxygen species (ROS) are an integral component of sperm developmental , capacitation, and function. Elevated ROS levels, from processes such as infection or inflammation, can be associated with aberrations of sperm development, function, and fertil- izing capacity. We review the impact of ROS on sperm physiology, its place in infertility evaluation, the implications for reproductive outcomes, and antioxidant therapy. Our systematic review of PubMed literature from the last 3 decades focuses on the physiology and etiology of ROS and (OS), evaluation of ROS, and . ROS is normally produced physiologically and is used to maintain cellular processes such as sperm maturation, capacitation, and sperm-oocyte interaction. When ROS production exceeds the buffering capacity of antioxidants, OS occurs and can have a negative impact on sperm and fertility. ROS and antioxidant capacity testing can potentially add additional prognostic information to standard laboratory testing for the infertile male, although its role as standard part of an evaluation has yet to be determined. Elevated ROS levels have been implicated with abnormal semen parameters and , but the impact of ROS on fertilization rates and pregnancy is controversial. This is partly because of the lack of consensus on what type of patients may be suitable for ROS testing and assay standardization. Routine ROS testing for the infertile male is not currently recommended. (Fertil SterilÒ Use your smartphone 2014;102:1518–27. Ó2014 by American Society for Reproductive Medicine.) to scan this QR code Key Words: Antioxidant capacity, antioxidants, male infertility, reactive oxygen species and connect to the discussion forum for Discuss: You can discuss this article with its authors and with other ASRM members at http:// this article now.* fertstertforum.com/koe-reactive-oxygen-species-antioxidant-capacity/ * Download a free QR code scanner by searching for “QR scanner” in your smartphone’s app store or app marketplace.

nfertility, defined as the inability to described by MacLeod in 1943 (6). His- Reactive oxygen species have a sig- I achieve pregnancy after 12 months torical studies have associated elevated nificant impact on spermatogenesis as of regular intercourse, affects 15% ROS concentrations with infertility in well as on sperm function. Supraphy- of reproductive-aged couples (1).A up to 40% men (7, 8). Contemporary siologic levels of ROS can affect all as- quarter of these cases can be explained studies have also found elevated pects of the semen analysis. Multiple by male-only factors, and up to 50% ROS levels in 30% to 80% of infertile studies have demonstrated the detri- can be explained by combined male men (9–11). Oxidative stress (OS) mental effects of ROS on sperm con- and female factors (2). Many infertile in the genitourinary system can centration (13, 15, 16), motility (15, men have abnormal semen analyses, damage sperm, resulting in decreased 16), morphology (15, 17), and ROS are of which the etiology is often poorly motility, , increased implicated in sperm DNA damage (18) explained (3). These etiologies can deoxyribonucleic acid (DNA) damage, and (19). Conversely, other include environmental, dietary, medi- and decreased oocyte-sperm fusion studies have demonstrated no cal, genetic, and physiologic factors (12). An imbalance between ROS levels correlation between ROS and sperm (4, 5). and physiologic antioxidants can result motility (20, 21). Recent studies have Reactive oxygen species (ROS) in OS, with subsequent adverse effects demonstrated that ROS values have a have long been implicated as a poten- on the body's normal physiologic pro- sensitivity of 68.8% and specificity of tial cause of male infertility, as first cesses (13, 14). 93.8% in diagnosing male factor infertility (infertility due to abnormal Received July 30, 2014; revised and accepted October 14, 2014. E.Y.K. has nothing to disclose. E.S.S. has nothing to disclose. A.A. has nothing to disclose. semen parameters, not female-related Reprint requests: Edmund Y. Ko, M.D., Assistant Professor, Department of Urology, Loma Linda Uni- factors) (15). versity School of Medicine, 11234 Anderson Street, A560, Loma Linda, California 92354 (E-mail: [email protected]). Not all OS is harmful to sperm function, however. Excessive levels of Fertility and Sterility® Vol. 102, No. 6, December 2014 0015-0282/$36.00 ROS can have detrimental effects on Copyright ©2014 American Society for Reproductive Medicine, Published by Elsevier Inc. http://dx.doi.org/10.1016/j.fertnstert.2014.10.020 sperm quality, but lower physiologic

1518 VOL. 102 NO. 6 / DECEMBER 2014 Fertility and Sterility® levels are necessary for sperm capacitation, hyperactivation, FIGURE 1 and sperm-oocyte fusion, along with other critical cellular processes (22–24). We review the impact of ROS on sperm physiology and antioxidant capacity, the role of ROS testing in the male fertility evaluation, the implications for reproduction outcomes, and the potential treatments to improve antioxidant capacity.

PHYSIOLOGY OF REACTIVE OXYGEN SPECIES AND ANTIOXIDANT CAPACITY Reactive oxygen species are byproducts of normal physio- logic processes and cellular . Cellular aerobic metabolism within the mitochondria occurs via oxidative phosphorylation. Molecular oxygen (O2) normally carries two unpaired , thereby making oxygen susceptible to formation. The primary form of ROS occurs with Mechanism of oxidative stress in human semen. From: Agarwal A, the addition of an extra to O2, resulting in a super- Sekhon LH. Oxidative stress and antioxidants for idiopathic oxide anion radical (.O2 ) (25). The anion can oligoasthenoteratospermia: is it justified? Indian J Urol 2011;27:74–85. then be converted to other ROS, including hydrogen Used with permission. Ko. Male infertility and ROS and TAC. Fertil Steril 2014. peroxide (H2O2), hydroxyl (.OH ), and peroxyl (HO2) radicals (23, 24, 26). Free radicals are formed as a byproduct of the enzymatic reduction of oxygen during energy production (9). Free radicals are oxygen containing one or more unpaired electrons, making them unstable and very tem via enzymatic and nonenzymatic antioxidant pathways reactive in the presence of amino acids, lipids, and nucleic (11, 13, 29). This finely balanced oxidant-antioxidant system acids (23, 24, 27). allows the formation of beneficial oxidants for normal Reactive oxygen species are important intermediaries cellular functions and concurrently prevents the damaging involved in vascular tone and gene regulation within the effects of excess oxidative stress. testes. Normal physiologic ROS levels are required for sperm A high concentration of antioxidants and scavengers are maturation, hyperactivation, capacitation, chemotaxis, zona contained within seminal plasma. These antioxidants act to pellucida binding, acrosome reaction, and sperm-oocyte protect sperm from the deleterious effects of ROS. Enzymatic fusion (28, 29). Within normal physiologic conditions, antioxidants include , , gluta- antioxidants help maintain a low level of oxidative stress in thione peroxidase, transferase, and cerulo- the semen, thereby allowing normal signaling processes plasmin. Nonenzymatic antioxidants include albumin, and spermatic function and avoiding ROS-induced cell dam- b-carotenes, L-carnitine, glutathione, pyruvate, taurine, age (14). hypotaurine, ubiquinol, vitamins C (ascorbic acid) and E (a- The main ROS producers in semen are leukocytes and ), and zinc (3, 38–40). immature spermatozoa (Fig. 1). Leukocytes can produce up Glutathione provides the most important intracellular to 1,000 times more ROS compared with spermatozoa under protection against ROS. It contains a sulfhydryl group that physiologic conditions (30). The sperm membrane remodeling directly scavenges free radicals. The cycle is completed process during spermatogenesis may be the common origin when oxidized glutathione is reduced/regenerated by gluta- for both abnormal spermatozoa and ROS. Failures in the pro- thione reductase and nicotinamide adenine dinucleotide cess, such as head-tail attachment abnormalities, incomplete phosphate (NADPH) (41). Superoxide dismutase (SOD) is a acrosome development, or sperm cytoskeleton alterations, metal-containing that reduces two into can lead to the creation of ROS and abnormal sperm O2 and less toxic H2O2 (9). Catalase is an enzyme found within morphology (23, 24, 31, 32). that completes the reduction process by breaking Sperm membranes contain a high concentration of poly- down H2O2 to and O2. Other that also reduce unsaturated lipids, making them sensitive to ROS and oxida- H2O2 include glutathione transferase and ceruloplasmin, or tive damage (24, 33). Lipid peroxidation can result in loss of heme oxygenase (42). membrane fluidity and integrity, leading to increased Not all OS is harmful to sperm function. Within normal permeability. This can lead to impaired motility, abnormal physiologic conditions, antioxidants help maintain a low morphology, reduced sperm-oocyte fusion, DNA damage, level of oxidative stress in the semen, thereby allowing reduced acrosomal reaction, and even sperm cell death (34– normal processes and spermatic function and 36). More recent studies have demonstrated that the avoiding ROS-induced cell damage (3, 14). Normal incidence of teratozoospermia may be directly correlated to physiologic ROS levels are required for sperm maturation, excess ROS (37). hyperactivation, chemotaxis, zona pellucida binding, Although ROS are formed during normal enzymatic reac- acrosome reaction, and sperm-oocyte fusion (22–24, 29, tions, cellular damage is prevented through a scavenging sys- 43–45).

VOL. 102 NO. 6 / DECEMBER 2014 1519 VIEWS AND REVIEWS

ETIOLOGIES OF OXIDATIVE STRESS coal, has an oxidative effect in the testis (67). Exposure to Oxidative stress can arise intrinsically from sperm itself and high levels of certain metals such as cadmium (68) or lead from a variety of conditions that are known to have detri- (69) can raise testicular OS and epididymal ROS production mental effects on male fertility. These conditions can range and result in decreased sperm production, antioxidant capac- from intrinsic abnormalities to environmental exposures. ity, and motility and increase lipid peroxidation (70). Toxins Some etiologies are reviewed here. from cigarette smoking have been noted to increase ROS pro- Intrinsic ROS can originate from damaged or deficient duction in many tissues, including the testes, and have been spermatozoa (46). Impaired spermatogenesis resulting in associated with infertility (71, 72). abnormal spermatozoa can yield excess ROS (47). Other Men with cancer who have been treated with chemo- intrinsic etiologies include , cryptorchidism, testic- therapy and/or radiation therapy are at risk of infertility. ular torsion, infection/inflammation, and aging. Rapidly dividing spermatogonia are more sensitive to damage Varicocele, a condition that is caused by dilation of the compared with later stage cells. Commonly used chemo- spermatic veins, has been found to be associated with infer- therapy agents that have gonadotoxic properties include tility in approximately 30% of men (48). The increased retro- (73), cyclophosphamide (74), and doxyrubicin (75). grade flow leads to an elevation of scrotal and testicular Heat shock may facilitate the testicular OS response temperatures (49). This results in OS from rising ROS concen- (76). trations in the testes and semen of varicocele patients (50, 51). Oxidative stress is induced in the testis from ionizing ra- Varicocele is also associated with a decrease in antioxidant diation (77). This leads to germ cell apoptosis (78). Despite the capacity (50). developing spermatogonia being sensitive, Sertoli and Leydig Cryptorchidism also results in increased testicular cells are relatively resistant to the effects of radiation (79). temperature and has been associated with increased ROS This may be, in part, due to the rise of total antioxidant capac- production and OS (52, 53). The elevation of ROS levels can ity noted in these cells after irradiation. lead to germ cell apoptosis as well as alterations in genes responsible for energy and lipid metabolism, stress IMPACT OF ELEVATED ROS ON SPERM AND response, and redox reactions (54). Testicular torsion and repair can induce significant OS re- FERTILITY CONSEQUENCES sulting from an -reperfusion (55). Torsion can Reactive oxygen species have a significant impact on sper- result in disruption of the seminiferous tubules and germ cell matogenesis as well as on sperm function. Supraphysiologic loss (56). Increased OS has been found within the testis after levels of ROS have been found to potentially impact all as- testicular torsion repair (57). pects of the semen analysis (15). Pathologic damage from Localized genitourinary infections and inflammation can oxidative stress occurs with an imbalance in the oxidant- affect male fertility by causing increased OS. These conditions antioxidant system, and ROS levels increase beyond the anti- increase the leukocyte concentration in the local environment oxidant buffering capacity of the system (41). Oxidative stress (58). The inflammatory response results in the rise of testicular can occur when there is an increase in ROS production, a inducible nitric oxide synthase, interleukin-1b, and decrease in antioxidant scavenging capacity, or both (25). cyclooxygenase-2 with concomitant decline in antioxidant Infertile men are more likely to have pathologic levels of enzymes and germ cells leading to an overabundance of seminal ROS as a result of increased ROS production rather ROS (59). Testicular inflammation has also been demonstrated than low antioxidant concentrations when compared with to result in a reduction in mitochondrial membrane potential, fertile controls (80). Up to 80% of men with infertility have steroidogenesis, and ultimately germ cell apoptosis (60). elevated seminal ROS levels (40). A recent study compared Aging has been linked to elevations in testicular OS (61, the semen of infertile men to proven donors (15). The semen 62). This has been suggested to be linked to the age related of infertile men had significantly higher levels of ROS declines in steroidogenesis (63). Aging Leydig cells have compared with those of proven donors. This study found been demonstrated to have reduced expression of enzymatic that elevated ROS values had a sensitivity of 68.8% and spec- and nonenzymatic antioxidants decreasing the buffering ificity of 93.8% in correlating with poor semen parameters capacity within the testes thereby increasing the risk of such as concentration, motility, and morphology, and could damage from ROS (62, 64). result in infertility (15). Environmental or extrinsic factors can also impact testic- Multiple studies have demonstrated the association of ular ROS levels thereby raising OS levels, leading to abnor- elevated ROS levels with abnormal sperm concentration (15, malities in spermatogenesis (13). These include exposure to 16, 81), motility (16), and morphology (17), with sperm toxins, chemotherapy, and . DNA damage (18, 82, 83), and with apoptosis (19). Elevated There are many environmental and industrial toxins that ROS levels can also decrease sperm-oocyte fusion (41). cause elevated ROS levels in the testes and impact spermato- Conversely, other studies have demonstrated no correlation genesis and sperm function. Briefly, methoxyethanol, found between ROS and sperm motility (20, 21). in paints, brake fluid, and other industrial chemicals, can in- Sperm motility can be negatively impacted by elevated crease ROS levels (65). Toluene by products found in solvents ROS levels. Reactive oxygen species may result in decreased can also cause oxidative DNA damage (66). Sulfur dioxide, a axonemal phosphorylation and reduced membrane combustion byproduct of burning petroleum products and fluidity (83, 84). Certain ROS, like H2O2, can diffuse across

1520 VOL. 102 NO. 6 / DECEMBER 2014 Fertility and Sterility® cellular membranes and disrupt enzyme activity, such as with analysis, Agarwal et al. (105) reviewed nine studies that spe- G6PD (85). Reduction in the availability of NADPH results in cifically addressed the association of ROS and IVF outcomes. increased levels of oxidized glutathione, reduced levels of Only three of nine studies were included in the final meta- glutathione, decreased antioxidant capacity, and increased analysis, with selection criteria including use of density peroxidation of membrane phospholipids (45). Reactive gradient for sperm preparation and N-formylmethionyl- oxygen species have also been implicated in the underlying leucyl-phenylalanine-stimulated ROS levels with correlation mechanism of oligozoospermia; patients with values to IVF outcomes, for a total study population of n ¼ oligoasthenoteratospermia and oligoasthenospermia show 122. The estimated overall correlation for this analysis significantly higher levels of ROS compared with normal was 0.374 (95% confidence interval [CI], 0.205 to healthy donors (86). 0.520) suggesting that ROS levels negatively impact fertil- Increased OS can increase DNA damage to mitochondrial ization rates for IVF. The meta-analysis, however, was not and nuclear material of spermatozoa (87). This can occur without weaknesses, including a lack of consistent ROS on an amino acid or molecular level in the form of base testing methods, unknown male or female factor infertility, modification, production of base-free sites, deletions, DNA incomplete statistical information in the original studies, cross-links, frame shifts, or even on a higher level with rear- and inclusion of normal semen parameters into the analysis. rangement of chromosomes (13). High ROS levels are associ- The investigators proposed that if all nine studies had had ated with an increased frequency of sperm DNA strand breaks similar ROS tests with complete statistical analyses, the out- in infertile men (88, 89). Previous studies have not established comes of the meta-analysis might have been different. a cutoff value for ROS or sperm DNA fragmentation at which pregnancy would not be possible (90). ROS AND ANTIOXIDANT CAPACITY TESTING Damage to sperm DNA ultimately can be clinically man- Testing for ROS can be achieved with direct or indirect assays ifested with decreased fertilization ability, impaired embry- (Table 1). Direct assays measure the net oxidative imbalance onic development, pregnancy loss, and potentially birth between ROS production and the antioxidant concentration defects (91, 92). Mouse models have suggested that sperm in semen by measuring the amount of oxidation within the with high levels of DNA damage used for conception can sperm cell membrane (37). These tests include chemilumines- result in aberrant growth and behavior, premature cence, nitro blue tetrazolium (NBT) test, cytochrome C reduc- aging, and an increased incidence of mesenchymal tumors tion test, flow cytometry, electron spin resonance, and in offspring (93). Increased rates of aneuploidy have also xylenol orange-based assay (106). Direct ROS assays have been found to be associated with elevated sperm DNA limited widespread clinical application at this time due to fragmentation rates, potentially resulting in genetic cost and practicality issues, rendering them more suitable abnormalities (94). for research purposes (107). Elevated ROS levels have been directly correlated with Indirect assays evaluate the downstream effects of oxida- increased apoptosis in mature spermatozoa, resulting in oli- tive stress, such as DNA damage or lipid peroxidation levels gozoospermia (40, 95). Oxidative stress has also been (37). Although indirect assays do not give a direct measure implicated in abnormal apoptosis via disrupting of the level of ROS within the system, they can provide impor- mitochondrial membranes and altering the caspase activity tant information regarding the negative effects that ROS has that induces apoptosis. Although sperm apoptosis may upon the spermatozoa as well as the components within the appropriately control overproduction of male gametes, spermatozoa. Indirect testing includes the myleperoxidase abnormal apoptosis may allow the persistence of abnormal or Endtz test, redox potential, lipid peroxidation levels via thi- spermatozoa that are marked for elimination, thereby obarbituric acid-reactive substances, and the isoprostane increasing teratozoospermia (96, 97). method, chemochine, and DNA damage testing. DNA damage Elevated ROS levels can affect many aspects of the sperm levels are not solely a measure of OS or ROS, but are used as a quality, including structural integrity, motility, morphology, surrogate measurement of ROS-related damage (103). count, viability, and DNA integrity, thereby making it one Currently, there are over 30 different assays that can be of the potential etiologies of male factor infertility (13, 15– used to measure ROS and OS in semen. Most ROS testing 18, 40, 98, 99). Despite the negative effect of ROS on sperm, controversy continues on the effect of elevated ROS levels on assisted TABLE 1 reproductive technology (ART), including in vitro fertilization (IVF) and pregnancy outcomes. There have been studies that Various direct and indirect semen assays of reactive oxygen species. have demonstrated that despite the negative impact on sperm quality and function, elevated ROS levels had no negative ef- Direct assays Indirect assays fect on IVF or intracytoplasmic sperm injection (ICSI) rates Chemiluminescence Antioxidants (100, 101). Yeung et al. (100) proposed that ROS might have Cytochrome C reduction test Chemokines Electron spin resonance DNA damage a positive effect on fertilization. Flow cytometry Lipid peroxidation levels Other studies have demonstrated negative effects of ROS Nitroblue tetrazolium test Measurement of redox potential on ART outcomes (102–104), including a meta-analysis Thiobarbituric acid assay Myeloperoxidase or Endtz test demonstrating a negative correlation between ROS levels Xylenol orange-based assay and fertilization rates using IVF (105). In their meta- Ko. Male infertility and ROS and TAC. Fertil Steril 2014.

VOL. 102 NO. 6 / DECEMBER 2014 1521 VIEWS AND REVIEWS does not identify the source of ROS (leukocytes versus imma- considered in idiopathic male infertility with normal bulk ture or abnormal spermatozoa) (106). semen parameters. Unexplained idiopathic oligoasthenotera- Seminal antioxidant capacity can be tested for each indi- tospermia or abnormalities in other specialized semen testing, vidual antioxidant in the seminal system or as a cumulative such as DNA fragmentation, acrosome reaction, or sperm- sum of the antioxidants (Table 2). The total antioxidant ca- oocyte binding, may also indicate ROS testing (3, 40).As pacity (TAC) is measured by evaluating the reducing ability with any test, ROS evaluation should be considered if it of the antioxidants within the semen against an oxidative provides additional information that would potentially reagent such as , and measuring the effect change or improve the management of male infertility and on the substrate (29). Depending on the substrate, the reaction ultimately pregnancy outcomes. However, the currently can be measured with a calorimeter, spectrophotometer, available studies correlating ROS levels and pregnancy or visually. Testing of individual antioxidants within semen outcomes are limited, and some are even contradictory. is costly and time consuming (29). In addition, measurement Oxidative stress and TAC testing may provide couples of individual antioxidants does not provide the entire with additional diagnostic data as to whether the male factor antioxidant picture. Individual antioxidant capacity can be is the primary source of their infertility, what the likelihood of determined by testing for catalase, , achieving pregnancy is if given time, and whether treatment and reductase, and superoxide dismutase activity (108). with antioxidant therapy is warranted if elevated ROS levels Total antioxidant capacity assesses the cumulative effect are the only abnormality. A 4-year prospective analysis of all antioxidants present within the semen (29). Studies have comparing the diagnostic significance among conventional demonstrated that normospermia is directly correlated with semen analysis, hamster-oocyte fusion, and ROS generation higher TAC levels compared with those of patients with in 139 couples demonstrated that ROS generation had a nega- asthenoteratospermia or oligoasthenoteratospermia (109). tive impact on sperm-oocyte fusion and fertility status despite Conversely, decreased seminal TAC levels are correlated normal semen analyses (114). with impaired semen parameters (110). However, TAC levels Testing for ROS has been used in combination with other are not routinely measured as a standard part of an infertility tests, including conventional semen analysis, acrosome reac- evaluation because of the lack of a proven benefit and the cost tion, and acridine orange staining to predict IVF outcomes, and time-consuming nature of this diagnostic testing when with an overall accuracy of 83.6% (115). The utility of ROS compared with a routine semen analysis (37). testing has also been demonstrated with sperm donors and A ROS-TAC score is a parameter that is derived from the proven donors, who have considerably better semen parame- ratio between ROS concentrations and seminal TAC (111). The ters and significantly lower ROS levels compared with infer- ROS-TAC score may be more predictive of subfertility than tile men (15). ROS levels or TAC alone (112). Infertile men have lower scores Despite their potential for providing additional prognostic when compared with their fertile counterparts, with a information, ROS and TAC testing are not routinely used during score <30 predicting risk for prolonged inability to conceive the initial male fertility workup because of their high cost, (13, 33). inconvenience, and lack of efficiency. In addition, there is an Elevated seminal OS on testing is abnormal and may reflect absence of standard protocols to assess seminal oxidants an oxidative imbalance (113). The OS level is not measured despite the ample data implicating ROS in male infertility directly but is a reflection of excess ROS, or lack of adequate (37). There has been no consensus on ROS and TAC testing be- antioxidant capacity to buffer the ROS, which can create a ing included as a routine part of an initial male infertility eval- detrimental environment for sperm production as well as func- uation owing to the lack of proper studies, the poor tion. The etiologies of OS were discussed earlier in this article. understanding of the ROS measurement in multiple assays, and the confusion as to whether neat or washed seminal ROS INDICATIONS FOR ROS AND TAC TESTING should be measured and the impact on the results (113, 116). In addition, smaller laboratories may find the testing Testing of ROS or TAC can be considered when an abnormal costly as considerable capital expenditure is required to semen analysis is found in an infertile male. It can also be obtain the necessary equipment and assays. However, the economies of scale make ROS and/or TAC testing efficient and cost effective for some laboratories (such as high- TABLE 2 volume academic centers), and they potentially can provide additional clinically relevant information to the treating cli- Tests to determine antioxidant capacity. nicians and their patients (15, 29). Individual Total Currently, no literature recommends against ROS testing, Catalase Colorimetric assay but the American Urologic Association Best Practices State- Dismutase Ferric reducing ability of plasma (FRAP) ment on evaluating the infertile male does not advocate the Glutathione peroxidase Luminol routine use of ROS testing as part of managing couples (117). Oxygen radical absorbance capacity (ORAC) This is because ROS testing lacks the ability to predict preg- Lipids Phycoerythrin fluorescence-based assay nancy outside of the semen analysis, and there are no proven Proteins therapies to treat elevated ROS levels if they are found. In their Vitamins opinion statement, the American Society for Reproductive Ko. Male infertility and ROS and TAC. Fertil Steril 2014. Medicine Practice Committee supports this view as well (118).

1522 VOL. 102 NO. 6 / DECEMBER 2014 Fertility and Sterility®

ANTIOXIDANT THERAPY supplements (e.g., supplemental vitamins, minerals, or herbs) Antioxidants can be obtained through two main sources: and are thought to increase the physiologic TAC (Table 3). A physiologic or dietary. Physiologic antioxidants are present higher intake of antioxidant-rich foods can potentially in the seminal plasma and the spermatozoa (9, 24). Intrinsic improve sperm DNA integrity and overall protection against ROS cellular damage is provided by (123). This is contrasted by studies demonstrating that a lower intracellular enzymatic and nonenzymatic antioxidants. intake and subsequent lower antioxidant concentration may Sperm contain minimal intracellular antioxidants due to be associated with poor semen quality (124, 125). their lack of significant cytoplasm (24, 119, 120). The Antioxidant supplement is not without risk, however. majority of TAC is thus contained in the seminal plasma. Adverse events, although uncommon at doses at or below the The enzymatic antioxidants include catalase, glutathione recommended daily allowance, usually occur with excess peroxidase, and superoxide dismutase. The nonenzymatic intake of supplements (126). Supplement-related adverse antioxidants include carnitine, , glutathione, events can be of similar magnitude to those experienced with urate, and vitamins C and E (29, 41, 120). conventional pharmaceuticals. A link has been established be- Antioxidant supplements can further be divided into two tween plasma L-carnitine levels and atherosclerosis promotion categories: natural and synthetic antioxidants. Natural anti- resulting in an increased risk of and ma- oxidants are those that are synthesized physiologically and jor adverse cardiac events (127). Vitamin A supplementation can be isolated from food sources. Synthetic antioxidants has been linked with an increased incidence of mortality from are those that are chemically synthesized as an isolate com- lung cancer and cardiovascular disease (128, 129). pound and packaged into pill form. Several studies have sug- supplementation has been shown to increase the risk of gested that synthetic compounds have suboptimal cardiac complications, breast cancers in women, and prostate – antioxidant properties due to their chemical composition cancer men (130 132) and has been found to increase all- and having been isolated from other synergistic compounds cause mortality at doses greater than 400 IU/day (128). present in natural food sources (121, 122). Combination supplement therapy may also have addi- Dietary antioxidants are obtained through oral intake tional side effects attributed to the synergistic effects of the in- of food (e.g., fruits, vegetables, nuts, dairy, and meat) or dividual agents (133). Concurrent selenium and high-dose

TABLE 3

Mechanism of action of various antioxidants and supplements. Antioxidant/supplement Mechanism of action Arginine (139) Potent immunomodulator Role in cellular inflammatory response and protection against oxidative damage Carnitines (140) Central role in cellular energy metabolism Transporter of activated long-chain fatty acids into mitochondria through b-oxidation and buffering of acetyl-coenzyme A (CoA) to CoA ratio Coenzyme Q10 (141) Crucial component of mitochondrial oxidative phosphorylation Electron transporter in mitochondrial respiratory chain Oxidation-reduction (redox) link between flavoproteins and cytochromes Redox control of cellular signaling origin and transmission Folic acid (142) Provides carbon groups for purine and pyrimidine production Important role in DNA synthesis Important role in proper cell function Glutathione (143, 144) Maintains exogenous antioxidants in reduced states Detoxification of carcinogens and foreign compounds Lycopene (145) Protection of lipid peroxidation Gap junction communication Cell growth regulation Gene expression modulation Immune response N-acetylcysteine (146) Free radical scavenger Promotes cellular glutathione production Selenium (147) Cofactor in reduction of antioxidant enzymes as a component of glutathione peroxidase Vitamin A (carotenoids, retinoids) (148) Mechanism unknown (27, 149, 150) Important cofactor for hydroxylation and amidation reactions Assists in recycling oxidized vitamin E Neutralizes ROS in concentration-dependent manner Vitamin E () (151, 152) Prevents oxidant-induced lipid peroxidation Inhibits free-radical-induced damage to cell membranes Prevents lipid peroxidation Improves activity of other antioxidants Zinc (142, 153) Cofactor for metalloenzymes involved in DNA transcription and protein synthesis Antiapoptotic and antioxidant properties Ko. Male infertility and ROS and TAC. Fertil Steril 2014.

VOL. 102 NO. 6 / DECEMBER 2014 1523 VIEWS AND REVIEWS vitamin E has also been associated with an increased incidence What impact will treatment with antioxidants have on of prostate cancer and high-grade prostate cancer risk (134). pregnancy rates and ultimately live births? Several extensive reviews have investigated the effectiveness of single or combination antioxidant therapies. Some supple- ments, such as b-carotene, vitamins A, C, and E, and selenium, CONCLUSION fi have not been proven to have distinct bene ts and may even be Elevated ROS levels have been found to involve 30%–80% of harmful and increase all-cause mortality (135, 136). infertile males and may potentially be the cause of their infer- A recent Cochrane Collaboration that included a pooled tility. Reactive oxygen species can cause abnormalities of analysis of 34 randomized controlled trials comprising 2,876 sperm morphology, motility, concentration, and DNA integ- fi couples extensively reviewed antioxidant studies speci cally rity, resulting in difficulties achieving pregnancy. The impact for male infertility in couples undergoing ART (137).No of ROS on fertilization and pregnancy is controversial. This is conclusion could be drawn on the effects of antioxidants on partly because of the lack of consensus on what type of pa- fi sperm parameters. There was a statistically signi cant increase tients may be suitable for ROS testing and also what kind of in pregnancy and live-birth rates in the couples who had un- assay should be used. Therefore, routine ROS and TAC testing dergone treatment with antioxidant supplements. In looking is not currently recommended as a part of a standard male at the actual results, there were 96 pregnancies in 964 couples infertility evaluation. combined from 15 trials (pooled odds ratio [OR] 4.18; 95% CI, – P< 2 ¼ 2.65 6.59; .0001; I 0%). There were only 20 live births in REFERENCES the 214 antioxidant couples, obtained from three of the studies included in this review (pooled OR 4.85; 95% CI, 1.92–12.24; 1. Practice Committee of American Society for Reproductive Medicine. Defi- P¼.0008; I2 ¼ 0%) compared with a control group. That review nitions of infertility and recurrent pregnancy loss: a committee opinion. Fer- suggests that there are negative effects of elevated ROS on til Steril 2013;99:63. 2. Sharlip ID, Jarow JP, Belker AM, Lipshultz LI, Sigman M, Thomas AJ, et al. fertilization and that antioxidant therapy may be useful in Best practice policies for male infertility. Fertil Steril 2002;77:873–82. treating men with elevated levels. 3. Agarwal A, Sekhon LH. The role of antioxidant therapy in the treatment of The studies of antioxidants and supplements for infer- male infertility. Hum Fertil 2010;13:217–25. tility have had many methodologic weaknesses, including 4. Auger J, Eustache F, Anderson AG, Irvine DS, Jorgensen N, Skakkeback NE, small size and short duration, lack of randomized double- et al. Sperm morphological defects related to environment, lifestyle and blinded placebo-controlled trials, the nonstandardization of medical history of 1001 male partners of pregnant women from four Euro- – antioxidant dosing regimens, and an inability to control for pean cities. Hum Reprod 2001;16:2710 7. 5. Kenkel S, Rolf C, Nieschlag E. Occupational risks for male fertility: an anal- baseline dietary consumption (3). Establishing single antiox- ysis of patients attending a tertiary referral centre. Int J Androl 2001;24: fi fi idant ef cacy is also dif cult due to concurrent testing of 318–26. multiple supplements, resulting in potentially unrecognized 6. MacLeod J. The role of oxygen in the metabolism and motility of human synergistic or antagonist results (133). spermatozoa. Am J Physiol 1943;138:512–8. There are no existing standards as to the optimal oral 7. Iwasaki A, Gagnon C. Formation of reactive oxygen species in spermatozoa antioxidant supplement regimen, the dosing of specific of infertile patients. Fertil Steril 1992;57:409–16. agents, or the duration for administering supplements. In 8. de Lamirande E, Gagnon C. Impact of reactive oxygen species on sperma- tozoa: a balancing act between beneficial and detrimental effects. Hum Re- addition, there are no proven therapies to correct any prod 1995;10(Suppl 1):15–21. abnormal results. However, given the rather innocuous side 9. Tremellen K. Oxidative stress and male infertility: a clinical perspective. effects at or below the recommended daily allowances, oral Hum Reprod Update 2008;14:243–58. antioxidant supplementation may be a reasonable treatment 10. Makker K, Agarwal A, Sharma R. Oxidative stress and male infertility. In- regimen before proceeding with more expensive treatments dian J Med Res 2009;129:357–66. such as IVF or ICSI (138). Repeated semen analysis and ROS 11. Agarwal A, Said TM, Bedaiwy MA, Banerjee J, Alvarez JG. Oxidative stress in an assisted reproductive techniques setting. Fertil Steril 2006; testing may be considered 3 months after initiating treatment 86:503–12. to evaluate for any changes in semen parameters. 12. Rajesh Kumar T, Doreswamy K, Shrilatha B, Muralidhara. Oxidative stress associated DNA damage in testis of mice: induction of abnormal sperms LOOKING AHEAD and effects on fertility. Mutat Res 2002;513:103–11. 13. Agarwal A, Saleh RA, Bedaiwy MA. Role of reactive oxygen species in the In the future, standardization of ROS and TAC testing and re- pathophysiology of human reproduction. Fertil Steril 2003;79:829–43. porting will allow head-to-head comparison of studies and 14. Ford WC. Regulation of sperm function by reactive oxygen species. Hum the pooling of data for a more robust meta-analysis. In addi- Reprod Update 2004;10:387–99. tion, placebo-controlled, dietary-controlled, double-blind, 15. Agarwal A, Sharma RK, Sharma R, Assidi M, Abuzenadah AM, Alshahrani S, randomized-controlled, prospective studies with standardized et al. Characterizing semen parameters and their association with reactive supplement regimens are needed to answer several questions: oxygen species in infertile men. Reprod Biol Endocrinol 2014;12:33. 16. Athayde KS, Cocuzza M, Agarwal A, Krajcir N, Lucon AM, Srougi M, et al. Can antioxidant therapy improve ROS and TAC levels? Development of normal reference values for seminal reactive oxygen spe- What treatment regimens are most optimal in improving cies and their correlation with leukocytes and semen parameters in a fertile – ROS and TAC levels? population. J Androl 2007;28:613 20. 17. Aziz N, Saleh RA, Sharma RK, Lewis-Jones I, Esfandiari N, Thomas AJ Jr, What is the safe level of ROS and TAC at which there is et al. Novel association between sperm reactive oxygen species production, minimal impact upon semen parameters and ultimately sperm morphological defects, and sperm deformity index. Fertil Steril male infertility? 2004;81:349–54.

1524 VOL. 102 NO. 6 / DECEMBER 2014 Fertility and Sterility®

18. Desai N, Sharma R, Makker K, Sabanegh E, Agarwal A. Physiologic and 43. de Lamirande E, Gagnon C. A positive role for the superoxide anion in trig- pathologic levels of reactive oxygen species in neat semen of infertile gering hyperactivation and capacitation of human spermatozoa. Int J An- men. Fertil Steril 2009;92:1626–31. drol 1993;16:21–5. 19. Agarwal A, Said TM. Oxidative stress, DNA damage and apoptosis in male 44. de Lamirande E, Gagnon C. Human sperm hyperactivation and capacita- infertility: a clinical approach. BJU Int 2005;95:503–7. tion as parts of an oxidative process. Free Radic Biol Med 1993;14:157–66. 20. Whittington K, Harrison SC, Williams KM, Day JL, McLaughlin EA, Hull MG, 45. Griveau JF, Dumont E, Renard B, Callegari JP, Lannou DL. Reactive oxygen et al. Reactive oxygen species (ROS) production and the outcome of diag- species, lipid peroxidation and enzymatic defense systems in human sper- nostic tests of sperm function. Int J Androl 1999;22:236–42. matozoa. J Reprod Fertil 1995;103:17–26. 21. Pasqualotto FF, Sharma RK, Nelson DR, Thomas AJ, Agarwal A. Relation- 46. Aitken RJ, Clarkson JS. Cellular basis of defective sperm function and its as- ship between oxidative stress, semen characteristics, and clinical diagnosis sociation with the genesis of reactive oxygen species by human spermato- in men undergoing infertility investigation. Fertil Steril 2000;73:459–64. zoa. J Reprod Fertil 1987;81:459–69. 22. Guthrie HD, Welch GR. Effects of reactive oxygen species on sperm func- 47. Aitken RJ, Clarkson JS, Fishel S. Generation of reactive oxygen species, lipid tion. Theriogenology 2012;78:1700–8. peroxidation and human sperm function. Biol Reprod 1989;40:183–97. 23. Kothari S, Thompson A, Agarwal A, du Plessis SS. Free radicals: their bene- 48. Fretz PC, Sandlow JI. Varicocele: current concepts in pathophysiology, ficial and detrimental effects on sperm function. Indian J Exp Biol 2010;48: diagnosis, and treatment. Urol Clin North Am 2002;29:921–38. 425–35. 49. Goldstein M, Eid JF. Elevation of intratesticular and scrotal skin surface tem- 24. Sharma RK, Agarwal A. Role of reactive oxygen species in male infertility. perature in men with varicocele. J Urol 1989;142:743–5. Urology 1996;48:835–50. 50. Hendin BN, Kolettis PN, Sharma RK, Thomas AJ, Agarwal A. Varicocele is 25. De Lamirande E, O'Flaherty C. Sperm activation: role of reactive oxygen associated with elevated spermatozoal reactive oxygen species production species and kinases. Biochim Biophys Acta 2008;1784:106–15. and diminished seminal plasma antioxidant capacity. J Urol 1999;161: 26. Warren JS, Johnson KJ, Ward PA. Oxygen radicals in cell injury and cell 1831–4. death. Pathol Immunopathol Res 1987;6:301–15. 51. Santoro G, Romeo C. Normal and varicocele testis in adolescents. Asian J 27. Kefer JC, Agarwal A, Sabanegh E. Role of antioxidants in the treatment of Androl 2001;3:259–62. male infertility. Int J Urol 2009;16:449–57. 52. Ahotupa M, Huhtaniemi I. Impaired detoxification of reactive oxygen and 28. De Lamirande E, Lamothe G. Reactive oxygen-induced reactive oxygen for- consequent oxidative stress in experimentally cryptorchid testis. Biol Re- mation during human sperm capacitation. Free Radic Biol Med 2009;46: prod 1992;46:1114–8. 502–10. 53. Peltola V, Huhtaniemi I, Ahotupa M. Abdominal position of the rat testis is 29. Agarwal A, Virk G, Ong C, du Plessis SS. Effect of oxidative stress on male associated withhighlevel oflipidperoxidation. BiolReprod 1995;53:1146–50. reproduction. World J Mens Health 2014;32:1–17. 54. Li YC, Hu XQ, Xiao LJ, Hu ZY, Guo J, Zhang KY, et al. An oligonucleotide 30. Plante M, de Lamirande E, Gagnon C. Reactive oxygen species released by microarray study on gene expression profile in mouse testis of experimental activated neutrophis, but not by deficient spermatozoa, are sufficient to cryptorchidism. Front Biosci 2006;11:2465–82. affect normal sperm motility. Fertil Steril 1994;62:386–93. 55. Lysiak JJ, Turner SD, Nguyen QA, Singbartl K, Ley K, Turner TT. Essential 31. Platts AE, Dix DJ, Chemes HE, Thompson KE, Goodrich R, Rockett JC, et al. role of in germ cell-specific apoptosis following ischemia/perfu- Success and failure in human spermatogenesis as revealed by teratozoo- sion injury of the mouse testis. Biol Reprod 2001;65:718–25. spermia . Hum Mol Genet 2007;16:763–73. 56. Turner TT, Bang HJ, Lysiak JL. The molecular pathology of experimental 32. Venkatesh S, Singh MP, Gupta NP, Deecaraman M, Dada R. Correlation of testicular torsion suggests adjunct therapy to surgical repair. J Urol 2004; sperm morphology and oxidative stress in infertile men. Iran J Reprod Med 172:2574–8. 2009;1:29–34. 57. Da Ros CT, Teloken C, Tannhauser M, Hartmann A. Does intratesticular 33. Saleh RA, Agarwal A. Oxidative stress and male infertility: from research to testosterone administration modify the evolution of transitory testicular clinical practice. J Androl 2002;23:737–62. ischemia in pre-pubertal ? J Urol 1998;159:1752–4. 34. Aitken RJ. Free radicals, lipid peroxidation and sperm function. Reprod Fer- 58. Henkel RR. Leukocytes and oxidative stress: dilemma for sperm function til Dev 1995;7:659–68. and male fertility. Asian J Androl 2011;13:43–52. 35. Potts RJ, Notarianni LJ, Jefferies TM. Seminal plasma reduces exoge- 59. Reddy MM, Mahipal SV, Subhashini J, Reddy MC, Roy KR, Reddy GV, et al. nous oxidative damage to human sperm, determined by the measure- Bacterial -induced oxidative stress in the impairment of ment of DNA strand breaks and lipid peroxidation. Mutat Res 2000; steroidogenesis and spermatogenesis in rats. Reprod Toxicol 2006;22: 447:249–56. 493–500. 36. Jedrzejczak P, Fraczek M, Szumala-Kakol A, Taszarek-Hauke G, 60. Allen JA, Diemer T, Janus P, Hales KH, Hales DB. Bacterial endotoxin lipo- Pawelczyk L, Kurpisz M. Consequences of semen inflammation and lipid polysaccharide and reactive oxygen species inhibit Leydig cell steroidogen- peroxidation on fertilization capacity of spermatozoa in in vitro conditions. esis via perturbation of mitochondria. Endocrine 2004;25:265–75. Int J Androl 2005;28:275–83. 61. Sastre J, Pallardo FV, de la Asuncion JG, Vina J. Mitochondria, oxidative 37. Agarwal A, Tvrda E, Sharma R. Relationship amongst teratozoospermia, stress, and aging. Free Radic Res 2000;32:189–98. seminal oxidative stress and male infertility. Reprod Biol Endocrinol 2014; 62. Cao L, Leers-Sucheta S, Azhar S. Aging alters the functional expression of 12:45–52. enzymatic and non-enzymatic anti-oxidant defense systems in testicular rat 38. Sies H, Stahl W, Sundquist AR. Antioxidant functions of vitamins: vitamins E Leydig cells. J Biochem Mol Biol 2004;88:61–7. and C, beta-carotene, and other carotenoids. Ann N Y Acad Sci 1992;669: 63. Zirkin BR, Chen H. Regulation of Leydig cell steroidogenic function during 7–20. aging. Biol Reprod 2000;63:977–81. 39. Tavilani H, Goodarzi MT, Doosti M, Vaisi-Raygani A, Hassanzadeh T, 64. Luo L, Chen H, Trush MA, Show MD, Anway MD, Zirkin BR. Aging and the Salimi S, et al. Relationship between seminal antioxidant enzymes and brown Norway rat Leydig cell antioxidant defense system. J Androl 2006; the phospholipid and fatty acid composition of spermatozoa. Reprod Bio- 27:240–7. med Online 2008;16:649–56. 65. Syed V, Hecht NB. Rat pachytene spermatocytes down-regulate a polo-like 40. Agarwal A, Sekhon LH. Oxidative stress and antioxidants for idiopathic oli- kinase and up-regulate a thiol-specific antioxidant protein, whereas Sertoli goasthenoteratospermia: is it justified? Indian J Urol 2011;27:74–85. cells down-regulate a phosphodiesterase and up-regulate an oxidative 41. Agarwal A, Prabhakaran SA, Sikka SC. Clinical relevance of oxidative stress stress protein after exposure to methoxyethanol and methoxyacetic acid. in patients with male factor infertility: evidence based analysis. AUA Up- Endocrinology 1998;139:3503–11. date Series 2007;26:1–12. 66. Homma-Takeda S, Hiraku Y, Ohkuma Y, Oikawa S, Murata M, Ogawa K, 42. Agarwal A, Allamaneni SS. Free radicals and male reproduction. J Indian et al. 2,4,6-Trinitrotoluene-induced reproductive toxicity via oxidative DNA Med Assoc 2011;109:184–7. damage by its metabolite. Free Radic Res 2002;36:555–66.

VOL. 102 NO. 6 / DECEMBER 2014 1525 VIEWS AND REVIEWS

67. Ming Z, Bai W. Oxidation damage of sulfur dioxide on testicles of mice. En- 91. Tarozzi N, Bizzaro D, Flamigni C, Borini A. Clinical relevance of sperm DNA viron Res 2004;96:298–304. damage in assisted reproduction. Reprod Biomed Online 2007;14:746–57. 68. Koizumi T, Li ZG. Role of oxidative stress in single-dose, cadmium induced 92. Lewis SE, Simon L. Clinical implications of sperm DNA damage. Hum Fertil testicular cancer. J Toxicol Environ Health 1992;37:25–36. 2010;13:201–7. 69. Hsu PC, Liu MY, Hsu CC, Chen LY, Leon-Guo Y. Lead exposure causes gen- 93. Fernandez-Gonzalez R, Moreira PN, Perez-Crespo M, Sanchez-Martin M, eration of reactive oxygen species and functional impairment in rat sperm. Ramirez MA, Pericuesta E, et al. Long-term effects of mouse intracytoplas- Toxicology 1997;122:133–43. mic sperm injection with DNA-fragmented sperm on health and behavior 70. Marchlewicz M, Wiszniewska B, Gonet B, Baranowska-Bosiacka I, of adult offspring. Biol Reprod 2008;78:761–72. Safranow K, Kolasa A, et al. Increased lipid peroxidation and ascorbic 94. Enciso M, Alfarawati S, Wells D. Increased numbers of DNA-damaged sper- acid utilization in testis and epididymis of rats chronically exposed to matozoa in samples presenting an elevated rate of numerical chromosome lead. Biometals 2007;20:13–9. abnormalities. Hum Reprod 2013;28:1707–15. 71. Mattison DR. The effects of smoking on fertility from gametogenesis to im- 95. Gandini L, Lombardo F, Paoli D, Caponecchia L, Familiari G, Verlengia C, plantation. Environ Res 1982;28:410–33. et al. Study of apoptotic DNA fragmentation in human spermatozoa. 72. Peltola V, Mantyla E, Huhtaniemi I, Ahotupa M. Lipid peroxidation and Hum Reprod 2000;15:830–9. antioxidant enzyme activities in the rat testis after cigarette smoke inhala- 96. Sakkas D, Mariethoz E, Manicardi G, Bizzaro D, Bianchi P, Bianchi U. Origin of tion or administration of polychlorinated biphenyls or polychlorinated DNA damage in ejaculated human spermatozoa. Rev Reprod 1999;4:31–7. naphthalenes. J Androl 1994;25:353–61. 97. Sinha Hikim AP, Swerdloff RS. Hormonal and genetic control of germ cell 73. Santos NA, Bezerra CS, Martins NM, Curti C, Blanchi ML, Satos AC. Hy- apoptosis in the testis. Rev Reprod 1999;4:38–47. droxyl radical scavenger ameliorates cisplatin-induced nephrotoxicity by 98. Lanzafame FM, La Vignera S, Vicari E, Calogero AE. Oxidative stress and preventing oxidative stress, redox state unbalance, impairment of ener- medical antioxidant treatment in male infertility. Reprod Biomed Online getic metabolism and apoptosis in rat kidney mitochondria. Cancer Che- 2009;19:638–59. mother Pharmacol 2008;61:145–55. 99. Gharagozloo P, Aitken RJ. The role of sperm oxidative stress in male infer- 74. Sudharsan PT, Mythili Y, Selvakumar E, Varalakshmi P. Cardioprotective tility and the significance of oral antioxidant therapy. Hum Reprod 2011; effects of the pentacyclic triterpene, lupeol and its ester on 26:1628–40. cyclophosphamide-induced oxidative stress. Hum Exp Toxicol 2005;24: 100. Yeung CH, De Geyter C, De Geyter M, Nieschlag E. Production of reactive 313–8. oxygen species by and hydrogen peroxide scavenging activity of spermato- 75. Asmis R, Qiao M, Rossi RR, Cholewa J, Xu L, Asmis LM. Adriamycin pro- zoa in an IVF program. J Assist Reprod Genet 1996;13:495–500. mote macrophage dysfunction in mice. Free Radic Biol 2006;41:165–74. 101. Hammadeh ME, Al Hasani S, Rosenbaum P, Schmidt W, Fischer 76. Tilgada E. Chemotherapy: induction of stress responses. Endocr Relat Can- Hammadeh C. Reactive oxygen species, total antioxidant concentration cer 2006;13(Suppl 1):S115–24. of seminal plasma and their effect on sperm parameters and outcome of 77. Manda K, Ueno M, Moritake T, Anzai K. Alpha-lipoic acid attenuates x- IVF/ICSI patients. Arch Gynecol Obstet 2008;277:515–26. irradiation-induced oxidative stress in mice. Cell Biol Toxicol 2007;23: 102. Zorn B, Vidmar G, Meden-Vrtovec H. Seminal reactive oxygen species as 129–37. predictors of fertilization, embryo quality and pregnancy rates after con- 78. Hasegawa M, Wilson G, Russell LD, Meistrich ML. Radiation-induced cell ventional in vitro fertilization and intracytoplasmic sperm injection. Int J An- death in the mouse testis: relationship to apoptosis. Radiat Res 1997; drol 2003;26:279–85. 147:457–67. 103. Saleh RA, Agarwal A, Nada EA, El-Tonsy MH, Sharma RK, Meyer A, et al. 79. Lee K, Park JS, Kim YJ, Soo-Lee YS, Sook-Hwang TS, Kim DJ, et al. Differ- Negative effects of increased sperm DNA damage in relation to seminal ential expression of Prx I and II in mouse testis and their up-regulation by oxidative stress in men with idiopathic and male factor infertility. Fertil Steril radiation. Biochem Biophys Res Commun 2002;296:337–42. 2003;79:1597–605. 80. Zini A, de Lamirande E, Gagnon C. Reactive oxygen species in the semen of 104. Agarwal A, Aponte-Mellado A, Premkumar BJ, Shaman A, Gupta S. The ef- infertile patients: levels of superoxide dismutase- and catalase-like activities fects of oxidative stress on female reproduction: a review. Reprod Biol En- in seminal plasma. Int J Androl 1993;16:183–8. docrinol 2012;10:49. 81. Agarwal A, Said TM. Role of sperm chromatin abnormalities and DNA 105. Agarwal A, Allamaneni SS, Nallella KP, George AT, Mascha E. Correlation damage in male infertility. Hum Reprod Update 2003;9:331–45. of reactive oxygen species levels with the fertilization rate after in vitro 82. Duru NK, Morshedi M, Oehninger S. Effects of hydrogen peroxide on DNA fertilization: a qualified meta-analysis. Fertil Steril 2005;84:223–31. and plasma membrane integrity of human spermatozoa. Fertil Steril 2000; 106. Hwang K, Lipshultz LI, Lamb DJ. Use of diagnostic testing to detect infer- 74:1200–7. tility. Curr Urol Rep 2011;12:68–76. 83. De Lamirande E, Gagnon C. Reactive oxygen species and human sperma- 107. Mori R, Sabanegh E Jr. Laboratory evaluation for male infertility. In: tozoa: I. Effects on the motility of intact spermatozoa and on sperm axo- Parekattil S, Agarwal A, editors. Male infertility: contemporary clinical ap- nemes. J Androl 1992;13:368–78. proaches, andrology, ART and antioxidants. New York: Springer; 2012: 84. De Lamirande E, Gagnon C. Reactive oxygen species and human sperma- 15–26. tozoa: II. Depletion of (ATP) plays an important role 108. Wheeler CR, Salzman JA, Elsayed NM, Omaye ST, Korte DW. Automated in the inhibition of sperm motility. J Androl 1992;13:379–86. assays for superoxide dismutase, catalase, glutathione peroxidase, and 85. Aitken RJ. Molecular mechanisms regulating human sperm function. Mol glutathione reductase activity. Anal Biochem 1990;184:193–9. Hum Reprod 1997;3:169–73. 109. Hosseinzadeh Colagar A, Karimi F, Jorsaraei SG. Correlation of sperm pa- 86. Agarwal A, Mulgund A, Sharma R, Sabanegh E. Mechanisms of oligozoosper- rameters with semen lipid peroxidation and total antioxidants levels in as- mia: an oxidative stress perspective. Syst Biol Reprod Med 2014;60:206–16. theno- and oligoasheno- teratospermic men. Iran Red Crescent Med J 87. Aitken RJ, Koppers AJ. Apoptosis and DNA damage in human spermato- 2013;15:780–5. zoa. Asian J Androl 2011;13:36–42. 110. Pahune PP, Choudhari AR, Muley PA. The total antioxidant power of 88. Aitken RJ, Krausz C. Oxidative stress, DNA damage and the Y chromo- semen and its correlation with the fertility potential of human male sub- some. Reproduction 2001;122:497–506. jects. J Clin Diagn Res 2013;7:991–5. 89. Kodama H, Yamaguchi R, Fukuda J, Kasai H, Tanaka T. Increased oxidative 111. Sharma RK, Pasqualotto FF, Nelson DR, Thomas AJ, Agarwal A. The reac- deoxyribonucleic acid damage in the spermatozoa of infertile male pa- tive oxygen species-total antioxidant capacity score is a new measure of tients. Fertil Steril 1997;65:519–24. oxidative stress to predict male infertility. Hum Reprod 1999;14:2801–7. 90. Spano M, Seli E, Bizzaro D, Manicardi GC, Sakkas D. The significance of 112. Pasqualotto FF, Sharma RK, Pasqualotto EB, Agarwal A. Poor semen quality sperm nuclear DNA strand breaks on reproductive outcome. Curr Opin Ob- and ROS-TAC scores in patients with idiopathic infertility. Urol Int 2008;81: stet Gynecol 2005;17:255–60. 263–70.

1526 VOL. 102 NO. 6 / DECEMBER 2014 Fertility and Sterility®

113. Deepinder F, Cocuzza M, Agarwal A. Should seminal oxidative stress mea- 131. Lawson KA, Wright ME, Subar A, Mouw T, Hollenbeck A, Schatzkin A, surement be offered routinely to men presenting for infertility evaluation? et al. Multivitamin use and risk of prostate cancer in the National Institutes Endocr Pract 2008;14:484–91. of Health-AARP Diet and Health Study. J Natl Cancer Inst 2007;99:754–64. 114. Aitken RJ, Irvine DS, Wu FC. Prospective analysis of sperm-oocyte fusion 132. Kabat GC, Kim M, Adams-Campbell LL, Caan BJ, Chlebowski RT, and reactive oxygen species generation as criteria for the diagnosis of infer- Neuhouser ML, et al. Longitudinal study of serum , retinol, and tility. Am J Obstet Gynecol 1991;164:542–51. tocopherol concentrations in relation to breast cancer risk among postmen- 115. Sukcharoen N, Keith J, Irvine DS, Aitken RJ. Prediction of the in-vitro fertil- opausal women. WHI Investigators. Am J Clin Nutr 2009;90:162–9. ization (IVF) potential of human spermatozoa using sperm function tests: 133. Ko EY, Sabanegh ES Jr. The role of over-the-counter supplements for the the effect of the delay between testing and IVF. Hum Reprod 1996;11: treatment of male infertility—fact or fiction? J Androl 2012;33:292–308. 1030–4. 134. Albanes D, Till C, Klein EA, Goodman PJ, Mondul AM, Weinstein SJ, et al. 116. Venkatesh S, Shamsi MB, Dudeja S, Kumar R, Dada R. Reactive oxygen spe- Plasma tocopherols and risk of prostate cancer in the Selenium and Vitamin cies measurement in neat and washed semen: comparative analysis and its E Cancer Prevention Trial (SELECT). Cancer Prev Res (Phila) 2014;7:886–95. significance in male infertility assessment. Arch Gynecol Obstet 2011;283: 135. Bjelakovic G, Nikolova D, Simonetti RG, Gluud C. Antioxidant supplements 121–6. for prevention of gastrointestinal cancers: a systematic review and meta- 117. Jarow J, Sigman M, Kolettis PN, Lipshultz LR, McClure RD, Nangia AK, et al. analysis. Lancet 2004;364:1219–28. The optimal evaluation of the infertile male: best practice statement. Am 136. Bjelakovic G, Nikolova D, Simonetti RG, Gluud C. Antioxidant supplements Urol Assoc Best Pract Statements. Available at: http://www.auanet.org/ed for preventing gastrointestinal cancers. Cochrane Database Syst Rev 2008: ucation/guidelines/male-infertility-d.cfm. Accessed September 3, 2014. CD004183. 118. Practice Committee of American Society for Reproductive Medicine. Diag- 137. Showell MG, Brown J, Yazdani A, Stankiewicz MT, Hart RJ. Antioxidants for nostic evaluation of the infertile male: a committee opinion. Fertil Steril male subfertility. Cochrane Database Syst Rev 2011:CD007411. 2012;98:294–301. 138. Ko EY, Sabanegh ES. The role of nutraceuticals in male fertility. Urol Clin 119. Aitken RJ, Sawyer D. The human —not waving but North Am 2013;41:181–93. drowning. Adv Exp Med Biol 2003;518:85–98. 139. Appleton J. Arginine: clinical potential of a semi-essential amino acid. Al- 120. Aitken RJ, Roman SD. Antioxidant systems and oxidative stress in the tern Med Rev 2002;7:512–22. testes. Oxid Med Cell Longev 2008;1:15–24. 140. Palmero S, Bottazzi C, Costa M, Leone M, Fugassa E. Metabolic effects of L- 121. Herbert V. The value of antioxidant supplements vs their natural counter- carnitine on prepubertal rat Sertoli cells. Horm Metab Res 2000;32:87–90. parts. J Am Diet Assoc 1997;97:375–6. 141. Hidaka T, Fujii K, Funahashi I, Fukutomi N, Hosoe K. Safety assessment of 122. Stanner SA, Hughes J, Kelly CN, Buttriss J. A review of the epidemiolog- coenzyme Q10 (CoQ10). Biofactors 2008;32:199–208. ical evidence or the ‘antioxidant hypothesis’. Public Health Nutr 2004;7: 142. Ebisch IM, Thomas CM, Peters WH, Braat DD, Steegers-Theunissen RP. The 407–22. importance of folate, zinc and antioxidants in the pathogenesis and pre- 123. Young SS, Eskenazi B, Marchetti FM, Block G, Wyrobek AJ. The association vention of subfertility. Hum Reprod Update 2007;13:163–74. of folate, zinc, and antioxidant intake with sperm aneuploidy in healthy 143. Irvine DS. Glutathione as a treatment for male infertility. Rev Reprod 1996; non-smoking men. Hum Reprod 2008;23:1014–22. 1:6–12. 124. Mendiola J, Torres-Cantero AM, Vioque J, Moreno-Grau JM, Ten J, Roca M, 144. Pompella A, Visvikis A, Paolicchi A, De Tata V, Casini AF. The changing et al. A low intake of antioxidant nutrients is associated with poor semen faces of glutathione, a cellular protagonist. Biochem Pharmacol 2003;66: quality in patients attending fertility clinics. Fertil Steril 2010;93:1128–33. 1499–503. 125. Sharma R, Biedenharn KR, Fedor JM, Agarwal A. Lifestyle factors and 145. Rao AV, Mira MR, Rao LG. Lycopene. Adv Food Nutr Res 2006;51:99–164. reproductive health: taking control of your fertility. Reprod Biol Endocrinol 146. Zembron-Lacny A, Slowinska-Lisowska M, Szygula Z, Witkowski K, 2013;11:66. Szyszka K. The comparison of antioxidant and hematological properties 126. Mulholland CA, Benford DJ. What is known about the safety of of N-acetylcysteine and alpha-lipoic acid in physically active males. Physiol multivitamin-multimineral supplements for the generally healthy popula- Res 2009;58:855–61. tion? Theoretical basis for harm. Am J Clin Nutr 2007;85(Suppl):S318–22. 147. Brown KM, Arthur JR. Selenium, selenoproteins and human health: a re- 127. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal view. Public Health Nutr 2001;4:593–9. microbiota metabolism of l-carnitine, a nutrient in red meat, promotes 148. Kamal-Eldin A, Appelqvist LA. The chemistry and antioxidant properties of atherosclerosis. Nat Med 2013;19:576–85. tocopherols and . Lipids 1996;31:671–701. 128. Alpha-Tocopherol, Beta Carotene Cancer Prevention Study Group. The ef- 149. Greco E, Iacobelli M, Rienzi L, Ubaldi F, Ferrero S, Tesarik J. Reduction of the fect of vitamin E and beta carotene on the incidence of lung cancer and incidence of sperm DNA fragmentation by oral antioxidant treatment. other cancers in male smokers. N Engl J Med 1994;330:1029–35. J Androl 2005;26:349–53. 129. Goodman GE, Thornquist MD, Balmes J, Cullen MR, Meyskens FL Jr, 150. Linster CL, Van Schaftingen E, Vitamin C. Biosynthesis, recycling and Omenn GS, et al. The Beta-Carotene and Retinol Efficacy Trial: incidence degradation in mammals. FEBS J 2007;274:1–22. of lung cancer and cardiovascular disease mortality during 6-year follow- 151. Palamanda JR, Kehrer JR. Involvement of vitamin E and protein thiols in the up after stopping beta-carotene and retinol supplements. J Natl Cancer inhibition of microsomal lipid peroxidation by glutathione. Lipids 1993;23: Inst 2004;96:1743–50. 427–43. 130. Lonn E, Bosch J, Yusaf S, Sheridan P, Pogue J, Arnold JM, et al, HOPE and 152. Brigelius-Flohe R, Traber MG. Vitamin E: function and metabolism. FASEB J HOPE-TOO Trial Investigators. Effects of long-term vitamin E supplementa- 1999;13:1145–55. tion on cardiovascular events and cancer: a randomized controlled trial. J 153. Zago MP, Oteiza PI. The antioxidant properties of zinc: interactions with Am Med Assoc 2005;293:1338–47. and antioxidants. Free Radic Biol Med 2001;31:266–74.

VOL. 102 NO. 6 / DECEMBER 2014 1527