(1)
Department of Anatomy, Tokyo Medical University, Shinjyuku-ku, Tokyo, Japan
5.1 Introduction
Experimental autoimmune orchitis (EAO) can be induced by testicular antigens’ immunization with simultaneous treatments with adjuvants such as complete Freund’s adjuvant (CFA), Bordetella pertussis antigens (BP), other microbial agents, and some immune-stimulating chemical (Fig. 5.1). These immuno-potentiating agents may easily overcome the testicular immune privilege systemically and/or locally.
Fig. 5.1
Immuno-potentiating agents for stimulation of testicular inflammation. CFA complete Freund’s adjuvant. BP Bordetella pertussigens
5.2 Various EAO Models with the Aid of Immunopotentiators
5.2.1 EAO Induced by Treatment with Cyclophosphamide and the Following Immunization with TGC
Cyclophosphamide is an anticancer chemotherapy drug, which is classified as an alkylating agent. It has been used for therapy of Hodgkin’s and non-Hodgkin’s lymphoma, Burkitt lymphoma, chronic lymphocytic leukemia, chronic myelocytic leukemia, acute myelocytic leukemia, acute lymphocytic leukemia, T cell lymphoma (mycosis fungoides), and multiple myeloma. Its common side effect is low blood counts with increased risk for infection, anemia, and bleeding. Moreover, cyclophosphamide is also known to be toxic to male reproductive organs (Rezvangfar et al. 2008; Drumond et al. 2011). On the other hand, it appears that cyclophosphamide can eliminate CD4+CD25+Foxp3+ Treg in experimental and clinical medicine (Yoshida et al. 1979; Brode et al. 2006; Audia et al. 2007; Cao et al. 2010) (Fig. 5.1). Indeed, cyclophosphamide-treated mice that received a single subcutaneous injection with TGC 2 days later developed EAO, whereas no such lesions were seen in TGC-immunized but cyclophosphamide-untreated mice (Sakamoto et al. 1985). Significant DTH against TGC and anti-TGC antibodies in sera were detected in EAO-affected mice. It is suggested that the mechanism that facilitated the EAO induction might be attributable to the elimination of endogenous cyclophosphamide-sensitive Treg (Sakamoto and Nomoto 1986). In mice that received cyclophosphamide on day 0 and TGC immunization on day 2, anti-TGC DTH is thought to play a key role in the induction of EAO. Actually, DTH against TGC was mildly induced by only one immunization with viable syngeneic TGC and was significantly augmented by cyclophosphamide pretreatment, and, furthermore, the DTH was suppressed by administration of cyclosporine A in a dose-dependent manner in mice that were cyclophosphamide pretreated and then immunized with TGC (Sakamoto et al. 1994). However, anti-TGC DTH was rather enhanced significantly in mice pretreated with cyclosporine A alone without any TGC immunization. Therefore, even though administration of cyclosporine A suppressed the specific DTH in TGC-immunized mice, administration of cyclosporine A alone rather eliminates some suppressive mechanism resulting in augmentation of anti-TGC DTH.
5.2.2 EAO Induced by Immunization with Testicular Antigens or Homogenate Emulsified in Complete Freund’s Adjuvant (CFA)
CFA is most commonly used adjuvant in experimental research (Fig. 5.1). It is prepared from non-metabolizable oil containing killed Mycobacterium tuberculosis and designed to provide continuous release of antigens necessary for stimulating a strong, persistent immune response. CFA-induced peripheral inflammation evokes pro-inflammatory cytokines such as IL-1-beta, IL-6 TNF-alpha, and IFN-gamma (Lapchak et al. 1992; Chuang et al. 1997; Raghavendra et al. 2004). The mycobacterial components in CFA activate CD4+ T cells and signal them to assume a Th1 profile so that strong DTH against autoantigens develops (Billiau and Matthys 2001). On the other hand, immune effector cells induced by CFA exert an inhibitory effect on antigen-specific Th2 responses (Chuang et al. 1997).
Eyquem and Krieg (1965) produced bilateral EAO by injecting a small amount of CFA alone into the left testis in rats, guinea pigs, and monkeys. In left testes, infiltration of mononuclear cells, resorption of the inoculated product in the granulomata, spermatogenic disturbance, and exudation of fibrinoid products were induced. On the other hand, in the right testes, the spermatogenic disturbance was seen, but there was no inflammatory cell response and granulomata, implying the induction of sympathetic lesion.
Freund et al. (1953) discovered that subcutaneous injection of homologous testicular homogenate emulsified in CFA into guinea pigs is followed by inflammatory lesions of the testes and, finally, by the spermatogenic disturbance. It is generally assumed that CFA acts by prolonging the lifetime of injected autoantigen, by stimulating its effective delivery to the immune system, and by providing a complex set of signals to the innate compartment of the immune system, resulting in altered leukocyte proliferation and differentiation (Tung et al. 1971c; Billiau and Matthys 2001). Early events include rapid uptake of adjuvant components by macrophages and dendritic cells, enhanced phagocytosis, secretion of cytokines by mononuclear phagocytes, and transient activation and proliferation of CD4+ T cells. The mycobacterial component in CFA also remodels the hemopoietic system, leading to a drastic expansion of Mac-1+ immature myeloid cells. The effects of CFA cause the so-called ASIA (autoimmune/autoinflammatory syndrome induced by adjuvants), in which the injection of CFA accelerates various autoimmune manifestations (Bassi et al. 2012).
Studies on this EAO induced by testicular homogenate+CFA-immunization had been carried out using the guinea pigs because of the ease of disease induction in this species (Freund et al. 1953, Tung et al. 1971a, b, 1977; Hojo et al. 1980). Katsh (1964) found that testes from prepubertal guinea pigs failed to induce EAO lesions when incorporated in CFA, indicating that mature rather than immature TGC autoantigens is critical for EAO induction. In EAO-induced rats, both DTH and autoantibodies against testicular antigens were detected at different times with maximum levels at 50 days with significant decrease in CD8+ T cells (Doncel et al. 1991; Doncel and Lustig 1991). Typical EAO was successfully transferred to naïve recipients with peritoneal exudate and lymph node cells from male and female donor guinea pigs preciously immunized with testicular antigens+CFA (Carlo et al. 1976). However, attempts to transfer EAO with circulating antibody from the immunized animals were not successful, supporting a cell-mediated basis for the immunologic events in EAO (Carlo et al. 1976). An important role of DTH to testicular antigen in both the induction and suppression of EAO has been also described (Tung et al. 1977; Hojo et al. 1980).
Bernard et al. (1975) found that immunization with homogenate of testicular antigens+CFA+saline induces EAO, while that with homogenate of testicular antigens+CFA+normal human serum failed to induce EAO, although the inhibitory components of normal serum have not yet been identified. They demonstrated that the capacity of antigen P purified from guinea pigs’ spermatozoa to produce lesions of EAO could be inhibited by mixing antigen P with a small amount of normal human serum before immunization of animals. Characteristics of the serum inhibitory factors remain unsolved.
Chutna and Rychlikova (1964) reported that guinea pigs pretreated with testis antigens in incomplete Freund’s adjuvant (non-metabolizable oils without killed Mycobacterium tuberculosis) were unresponsive to the sensitization for EAO. In the absence of Mycobacterium tuberculosis, T cell differentiation tends to assume a Th2 profile with strong antibody production only (Billiau and Matthys 2001). Later, Hojo and Hiramine (1982) and Hiramine and Hojo (1984a) investigated “antigen-specific” suppression of EAO in guinea pigs in a model made by subcutaneous injections with testicular antigens in incomplete Freund’s adjuvant prior to EAO sensitization with testicular antigens in CFA. This suppressive state for EAO induction was adoptively transferable to normal syngeneic recipients with lymph node T cells taken from animals pretreated with testicular antigens+incomplete Freund’s adjuvant, indicating the presence of specific Treg. The Treg suppressed DTH reaction to testicular antigens, but not anti-sperm antibody production in their recipients. Cyclophosphamide treatment 3 days before EAO sensitization abolished the preventive effect of pretreatment with testicular antigens+incomplete Freund’s adjuvant. In another study, it was demonstrated that cyclosporine A administration for 2 weeks, starting on the day of testicular antigens+CFA-immunization, almost completely abrogated the EAO induction, DTH to testicular antigens, and also anti-sperm antibody response in guinea pigs (Hojo and Hiramine 1985). Transfer of lymphocytes taken from cyclosporine A-treated, EAO-suppressed guinea pigs into normal syngeneic recipients inhibited the EAO induction in an antigen-specific manner, indicating the induction of specific Treg by cyclosporine A. However, as far as guinea pig EAO is employed, it is impossible to describe in detail the suppression mechanism, such as the phenotype of Treg and related cytokines.
In the rat, EAO is also inducible by immunization with Sertoli cells+CFA (Tung and Fritz 1987), by immunization with basal lamina of the seminiferous tubules+CFA (Denduchis et al. 1975, 1979; Lustig et al. 1977, 1982, 1986), and by immunization with pachytene spermatocytes+CFA (Tung and Fritz 1987) (Fig. 5.2). This indicates that testicular cells other than spermatids and spermatozoa have a potential to induce EAO with the aid of CFA. It is also noted that male rats treated with CFA alone have reduced serum testosterone and elevated serum luteinizing hormone concentrations, showing the induction of testicular dysfunction (Clemens and Bruot 1989) (Fig. 5.3).
Fig. 5.2
Various sensitizing antigens by which EAO is inducible with the aid adjuvants
Fig. 5.3
Effects of adjuvants on the testis. BTB, blood-testis barrier. D, tubuli recti. TGC, testicular germ cells
In the mouse, thymectomy in adult life prevented EAO induction or interfered with further development of established EAO (Vojtiskova and Pokorna 1964). On the contrary, neonatal thymectomy further enhanced severity of EAO by subsequent immunization with testicular antigens+CFA in the rat (Lipscomb et al. 1979). These two studies suggest an involvement of the thymus for both induction and suppression of EAO. In general, mice have been considered relatively resistant to testicular antigens+CFA-induced EAO in comparison with guinea pigs, but Feng et al. (1990) developed a protocol for transferring EAO to naïve recipient mice in this disease model. In that study, cell donors were balb/c mice immunized 12 days earlier with epididymal spermatozoa+CFA+bovine serum albumin (Figs. 5.1 and 5.2). As few as 3 × 106 sperm-specific T lymphocytes obtained from the immunized mice were able to transfer EAO. Later, Li et al. (2014) aimed to determine the role of Axl and Mer receptor tyrosine kinases in maintaining the systemic tolerance to TGC antigens. Axl and Mer double knockout (Axl−/−Mer−/−) mice developed severe EAO after a single immunization with TGC homogenates+CFA. This immunization protocol also induced mild EAO in Axl or Mer single-gene knockout mice. By contrast, the TGC homogenate+CFA-immunization failed to induce EAO in wild-type mice. These results indicate that Axl and Mer receptors cooperatively regulate the systemic immune tolerance to TGC antigens. More recently, the roles of TLR2 and TLR4 in mediating the EAO induction were investigated in mice (Liu et al. 2015). Wild-type mice developed severe EAO after three immunizations with TGC antigens+CFA; however, TLR2- or TLR4-deficient mice showed relatively low susceptibility to EAO induction. It was demonstrated that TLR2 was crucial in mediating autoantibody production in response to the immunization. Notably, TLR2 and TLR4 double knockout mice were almost completely protected from EAO induction, implying that TLR2 and TLR4 cooperatively mediate EAO induction. However, it remains unclear whether TLR2 and TLR4 affect EAO induction locally in the testis or through systemic immunity.
Significant spermatogenic disturbance and appearance of anti-TGC autoantibodies were also observed in mice and guinea pigs after syngeneic, allogeneic, and xenogeneic immunization with mouse F9 embryonic carcinoma cells+CFA (Vojtiskova et al. 1983) (Fig. 5.2). This indicates that common cancer antigens between mice and guinea pigs may play a critical role for EAO induction in the both species.
Although immune T cells are most importantly required for EAO induction, Hiramine and Hojo (1984b) elucidated the participation of B cells in the local transfer system of EAO in the guinea pig. Proliferative response of EAO-inducing (orchitogenic) T cells was enhanced by the presence of B cells as well as macrophages (Hojo et al. 1980). Their study showed the requirement of B cells in the local adoptive transfer of EAO and suggested the possibility of cooperative interactions among T cells, B cells, and macrophages involved in the generation of the transferred EAO lesions. From this study, it was first demonstrated that B cells possess the antigen-presenting capability like macrophages and dendritic cells in the guinea pig EAO. However, the participation of humoral antibodies themselves has been the subject of controversy. Although typical EAO was successfully transferred to naïve recipients with peritoneal exudate and lymph node cells from male and female donor guinea pigs previously immunized with testicular antigens+CFA, attempts to transfer EAO with circulating antibody from the immunized animals were not successful (Carlo et al. 1976). In contrast, in testicular antigens+CFA-immunized guinea pigs and rats, passive transfer of antiserum against testicular antigens induced EAO (Tung et al. 1971b; Toullet and Voisin 1976; Lustig et al. 1977, 1978; Denduchis et al. 1979, 1985). In guinea pigs that had been injected into the recipient testis with immune sera from donors immunized with testicular homogenate+CFA, congestive vessels and exudation of polymorphonuclear cells were seen in the testis during the first week after the local injection. Leukocytes also appeared in the vicinity of the canaliculi of the rete testis (Mancini et al. 1974). This picture was accompanied by vacuolization of Sertoli cells and sloughing of TGC. Later, marked disturbance of spermatogenesis with a moderate infiltration of mononuclear cells between the seminiferous tubules and also in the interstitium of the rete testis and the epididymis was found (Chattopadhyay 1979). Injection of normal sera did evoke any histological changes in the recipient testis. The autoantibodies induced by testicular homogenate+CFA-immunization should include not only anti-TGC but also anti-Sertoli cell and anti-basal lamina of the seminiferous tubules antibodies (Fig. 5.4). It is known that the laminins constitute the major non-collagenous components of basal lamina of the seminiferous tubules. When rabbit antibodies to laminin1(IgG fraction) were injected into adult guinea pigs, thickening of the tubular limiting membrane, infolding in the basal lamina, deposits of immune complexes coincident with sloughing of pachytene spermatocytes and spermatids, and vacuolization of the Sertoli cells were induced. However, Sertoli cell tight junctions remained impermeable, and mononuclear cell infiltration in the testis was rare (Lustig et al. 2000). This indicates a possible participation of B cell lineage for EAO induction as well as T cells.
Fig. 5.4
Various immune responses induced by sensitization with testicular homogenates+adjuvants. D, ductuli efferentes. E, epididymis. T, testis. V, vas deferens. Dotted areas indicate the presence of the lymphocytic inflammation
In EAO using animals immunized with testicular antigens+CFA, transfer of EAO with “immune RNA” was reported in guinea pigs. “Immune RNA” obtained from the lymphocytes of donor guinea pigs, which were immunized with testis antigens+CFA, was injected intraperitoneally into normal recipient guinea pigs (Fainboim et al. 1978). The transferred guinea pigs developed DTH to sperm antigens and EAO as seen in the “immune RNA” donor guinea pigs. In contrast, when the transfer was performed with RNA extracted from guinea pigs that were immunized with CFA alone, no specific DTH and EAO were observed. Furthermore, in cases of transfer with ribonuclease-treated “immune RNA,” the recipient animals failed to have EAO lesions. “Immune RNA” may convert non-sensitized mouse peritoneal cells to a state of specific immunologic reactivity to testicular antigens. In an in vitro study, the “immune RNA” was incubated with normal guinea pig peritoneal exudate cells, which showed specific inhibition of migration when tested with the TGC antigen (Fainboim et al. 1979). In similar experiments, normal guinea pig peritoneal exudate cells, when incubated in vitro with “immune RNA” from “rats” immunized with testicular homogenate+CFA, recognized and responded to the TGC antigens specifically as assessed by the direct migration inhibition reaction (Sztein et al. 1980). This indicates the success of transfer of cell migration inhibition in vitro with not only syngeneic but also xenogeneic RNA in EAO. Therefore, “immune RNA” is able to cross the species barrier and sensitive to xenogeneic lymphoid cells. Sztein et al. (1980) also showed that the transferred antigen reactivity is directed against common testicular antigens present in rats, mice, and guinea pigs. The reaction would appear to be organ specific, kidney homogenate+CFA being unable to cause migration inhibition. EAO can be transferred by other “noncellular materials” (Pokorna 1969). In his study, male donor mice of C57BL/10 strain were immunized with testicular homogenate+CFA. On day 14, spleens and lymph nodes of the donors were taken, and their cell suspensions were prepared. The cell suspension was incubated at 56 C for 1 h, and cellular sediment was then centrifuged and the supernatant was used. EAO was induced in recipient mice that were intraperitoneally injected with “the prepared supernatant” in amount of 0.5 ml (Pokorna 1969). Furthermore, it was found that spleen cells and lymph node cells from non-immunized normal mice can also transfer EAO when the cells were incubated with “the prepared supernatant.” The incubated cells from normal donors were washed three times with Hanks solution and injected intraperitoneally in amount of 0.5 ml containing 2 × 108 cells. The EAO-inducing supernatant exhibited cytotoxic complement-dependent activity on testicular cells and was partly inactivated by trypsin, whereas RNase and DNase did not influence their activity. Since the EAO transfer induced by “immune RNA” or the “noncellular supernatant” obtained from immune cells has not been further reexamined, the minute pathophysiology remains unclear.
In regard to testicular autoantigens, four glycoproteins (GP1, GP2, GP3, and GP4) rich in carbohydrate were isolated from guinea pig testes (Hagopian et al. 1975). The molecular weights of the glycoproteins were estimated to be 47,000, 105,000, and 13,000, respectively, for GP1, GP2, and GP4. GP3 showed two major bands, with molecular weights of 41,500 and 22,800. GP1, GP2, and GP4 were localized in the sperm acrosome (Fig. 5.2). Approximately, 5 mg each purified GP1, GP3, and GP4, and 3 mg of GP2 were isolated from 1000 g of wet guinea pig testes. In particular, both GP1 and GP4 are strongly orchitogenic (EAO-inducing) antigens, while GP2 and GP3 were inactive (Hagopian et al. 1975, 1976). A unique highly soluble aspermatogenic protein, AP1, was also isolated from guinea pig testes. Approximately 20 mg of AP1 were obtained from 5000 g of wet guinea pig testes (Jackson et al. 1975, 1976). This protein is localized on the outer surface of sperm acrosome and is a potent inducer of EAO (Fig. 5.2). Toullet and Voisin (1976) reported that three different autoantigens (S, P, and T), extracted and separated from guinea pig spermatozoa, give rise to EAO when injected with CFA. They also induce specific antibodies, such as anaphylactic (with S and P), complement-fixing (with P and T), spermotoxic (only with T), and precipitating and Arthus-inducing antibodies (only with P). Autoantigens S is present on proacrosomal granules, acrosomal granules, and acrosome and head cap of spermatozoa. Autoantigen P is present on the same formations except for the head cap. Autoantigen T is also localized on acrosomes and head caps but on their membranes as well as on the cytoplasmic membranes of spermatozoa and spermatids (Toullet et al. 1973). In particular, autoantigen S was detected on the acrosomal apparatus in not only guinea pigs but also rabbits, rats, and mice. Later, aspermatogenic polypeptides, AP2 and AP3, capable of inducing EAO in the guinea pig were purified from their testes and epididymides by sequential biochemical methods (Teuscher et al. 1983a, b). Approximately 80 mg of AP2 was obtained from 1010 cauda epididymal spermatozoa. AP2 has a molecular weight of 9500. Approximately 250 mg of AP3 were obtained from 500 g wet weight of the testes. AP3 appeared as a single band with a molecular weight of 12,500. Immunization with AP2+CFA or AP3+CFA induced severe EAO in 100% of the guinea pigs tested. Guinea pigs were immunized with a defined and highly potent aspermatogenic antigen, G75 m, and the following occurrence of EAO was correlated with cell-mediated immune response to G75 m (Meng and Tung 1983). Later, it was reported that the immunization of male guinea pigs with PH-20+CFA also reproducibly results in EAO induction (Tung et al. 1997). PH-20, a testis-specific protein first expressed in haploid TGC, is present on the posterior head plasma membrane and inner acrosomal membrane of mature guinea pig spermatozoa (Fig. 5.2). PH-20 is bifunctional, having a hyaluronidase activity that allows sperm to penetrate the cumulus layer and a separate activity required for binding of acrosome-reacted sperm to the zona pellucida. Remarkably, PH-20+CFA-induced EAO differed from testicular homogenate+CFA-induced EAO in two respects: (1) an absence of epididymitis with abscess and granulomata and (2) the deposit of antibodies on TGC within the seminiferous tubules and abnormal spermatozoa inside the cauda epididymis in PH-20-induced EAO lesions. The former suggests that crude testis antigens other than PH-20 are responsible for autoimmune epididymitis in testicular homogenate+CFA-immunized animals, and the latter suggests an active role of humoral immunity in PH-20-induced EAO pathogenesis. In addition, guinea pigs immunized with testicular acrosin with molecular weight of 34,000 + CFA also exhibited typical EAO (Falase et al. 1989).
5.2.3 EAO Induced by Immunization with Mixture Containing Testicular Homogenate and Bordetella pertussis (BP)
In vivo intoxication with BP elicits a variety of physiological responses including a marked leukocytosis, disruption of glucose regulation, adjuvant activity, alterations in vascular function, hypersensitivity to vasoactive agents, and upregulation of MHC class II molecules (Tonon et al. 2002; Gao et al. 2003) (Figs. 5.1 and 5.3). BP-induced lymphocytosis is associated with alteration in thymocyte subpopulations (Person et al. 1992b). BP primarily affects and depletes thymic T cells with an immature phenotype. However, in the periphery of BP-treated mice, the relative increase in the number of CD4+ T cells is more than that of CD8+ T cells. In regard to pro-inflammatory cytokines, BP treatment induced the release of IL-6, IL-12, and TNF-alpha (Mielcarek et al. 2001; Tonon et al. 2002).
EAO was induced in mice immunized with testicular homogenate+BP on day 0 and day 34 (Hargis et al. 1968). On day 50, the immunological aspermatogenesis was characterized by degenerating TGC, signet ring nuclei, abnormal mitotic figures, karyorrhexis, the presence of an eosinophilic coagulum, and multinucleate giant cells. There was moderate interstitial edema, but it is noted that accumulations and invasion of mononuclear cells were rare. Both Sertoli cells and Leydig cells appeared normal. BP enhances of the DTH as well as hypersensitivity of the immediate type. BP treatment alone in the absence of immunization with testicular homogenate induced systemic leukocytosis in mice with significant lymphocytic inflammation in the ductuli efferentes, epididymis, and prostate, but not in the testes (Itoh et al. 1995).
In vivo intoxication with BP elicits a variety of inflammatory responses, including vasoactive amine sensitization to histamine, serotonin, and bradykinin (Diehl et al. 2014). Histamine is genetically controlled by a locus controlling BP-induced histamine sensitization (Bphs). Furthermore, it was found that BP sensitizes mice to histamine independently of TLR4, a purported receptor for BP (Diehl et al. 2014). Therefore, Bphs on chromosome 6 may be critical in susceptibility to testicular homogenate+BP-induced EAO (Teuscher 1985, 1986; Sudweeks et al. 1993; Meeker et al. 1999; Ma et al. 2002; Gao et al. 2003).
5.2.4 EAO Induced by Immunization with Testicular Antigens or Homogenate Emulsified in CFA and the Following Intravenous Administration of BP
5.2.4.1 Induction of EAO
Previously, many investigators had a difficulty to reproduce murine EAO model by immunization with testis antigens+CFA or with testis antigens+BP. Bernard et al. (1978) were the first to develop active EAO model in mice by immunization with testicular homogenate emulsified in CFA followed by intravenous injection with BP. This EAO using both CFA and BP is reproducible and consistently accompanied by severe inflammation in the epididymis and the vas deferens (=epididymo-vasitis). They also first succeeded in adoptive transfer of murine EAO (=passive EAO) with immune T cells into T cell-deficient athymic nude recipient mice treated with BP. Therefore, an important role for T cells in mediation of murine EAO was demonstrated by this adoptive transfer experiment (Bernard et al. 1978). The inflammatory lesions composed of macrophages, lymphocytes, neutrophils, and eosinophils are mainly found in association with the spermatogenic disturbance and Leydig cell hyperplasia (Sato et al. 1981; Kohno et al. 1983). DTH against testicular antigens was detected early at day 7; it increased with time, reaching a maximum at day 80; and a good temporal relationship between DTH and histopathology was found (Doncel et al. 1989). In contrast, circulating specific autoantibodies were only present in approximately 60% of the animals with EAO, and no deposits of IgG or C3 in the seminiferous tubules were seen (Doncel et al. 1989). Later, Mahi-Brown et al. (1987, 1988) and Mahi-Brown and Tung (1989, 1990) have shown that activated T cells can successfully transfer of the EAO to thymus-bearing normal recipient mice. The donor mice used in the adoptive transfer experiment received an immunization schedule consisting of subcutaneous injection with whole testicular homogenate+CFA+BP. A successful transfer of EAO to naive recipient mice was done by performing an in vitro challenge of the donor CD4+ T cells with testis antigens before the cell transfer (Tung et al. 1989). Later, EAO was shown to be induced by testis and sperm antigen-specific T cell clone that had been derived from testicular homogenate+CFA+BP-immunized mice (Yule and Tung 1993). Similar to other EAO models, MHC class II antigen-restricted CD4+ T cells are the primary effectors; however, recent evidence suggests the involvement of CD8+ T cells during the onset and maintenance of chronic EAO, in which Th17 cytokines are also involved in the pathogenesis of EAO (Jacobo et al. 2009, 2011a, b). At EAO onset, the number of CD4+ and CD8+ effector T cells dramatically increased in the testis, with the CD4+ T cell subset predominating. As the severity of EAO progressed, CD4+ effector T cells declined in number, while the CD8+ effector T cell subset remained unchanged, suggesting their involvement in maintenance of the chronic phase of EAO (Doncel and Lustig 1991; Lustig et al. 1993; Jacobo et al. 2009). Mast cells increased tenfold in number, were more widely distributed throughout the interstitial tissue, and were partially degranulated (Iosub et al. 2006). Mielcarek et al. (2001) demonstrated that mast cells phagocytose BP and can process and present BP molecules to T lymphocytes. Furthermore, exposure of mast cells to BP induced the release of pro-inflammatory cytokines such as TNF-alpha and IL-6. In the seminiferous tubules, TGC death occurs through an apoptotic mechanism preceding TGC sloughing in an autocrine and/or paracrine way. Later lesions of EAO included granulomata formation and necrosis in the testis (Zhou et al. 1989).
EAO was also induced by immunization with laminin+CFA+BP in the rat (Lustig et al. 1977, 1982, 1986, 1987, 1989) (Fig. 5.2). Laminin has been characterized as the main non-collagenous glycoprotein of basal lamina of the seminiferous tubules and has been detected in a variety of cells, including Sertoli cells (Denduchis et al. 1975). The main components of basal lamina are type IV collagens, laminins, entactin/nidogen, and heparin sulfate proteoglycans. In this EAO, interstitial mononuclear cell infiltration was also seen in the epididymis. The testicular lesions were characterized by multiple foci of the seminiferous tubules with different degrees of sloughing of the germinal epithelium. In the basal lamina of the seminiferous tubules, splitting and focal thickenings of knob-like projections toward the epithelium was found (Lustig et al. 1982, 1987). The thickening and delamination of the basement membrane was consistently accompanied by vacuolization of the Sertoli cell cytoplasm. In vivo-bound rat IgG was detected along the walls of the seminiferous tubules as bright linear and dense reaction products on the basal lamina. High titers of circulating anti-laminin antibodies and a significant DTH to laminin were detected in the immunized rats. Leukocyte migration was also inhibited when the spleen cells of the immunized rats were incubated with laminins.
5.2.4.2 The Effects of CFA+BP Sensitization
The mycobacterial components within CFA signal T cells to assume a Th1 profile so that strong DTH against testicular autoantigens develops. In the absence of mycobacteria, T cell differentiation tends to assume a Th2 profile with strong antibody production only (Billiau and Matthys 2001). In the periphery of BP-treated mice, the relative increase in the number of CD4+ T cells is more than that of CD8+ T cells. As mentioned earlier, treatment with CFA and BP releases various pro-inflammatory cytokines involving IL-1-beta, IL-6, IL-12, TNF-alpha, and IFN-gamma in vivo (Lapchak et al. 1992; Chuang et al. 1997; Mielcarek et al. 2001; Tonon et al. 2002; Raghavendra et al. 2004). Considering that severe splenomegaly and lymphadenopathy was induced by CFA+BP treatment alone, various lymphocytic clones that are not specific to the testicular antigens should be also activated, resulting in enhancement of the immune responses and the breakdown of testicular immune privilege. Indeed, the employment of CFA and BP has proved to be instrumental in altering the completeness of BTB indirectly (Fig. 5.3), as well as in augmenting of DTH (Pelletier et al. 1981; Sewell et al. 1986; Adekunle et al. 1987). Foci of tubules with exfoliated TGC into the seminiferous tubular lumen or sloughing of the germinal epithelium was found in CFA+BP-injected mice and rats (Adekunle et al. 1987; Lustig et al. 1987). In particular, TNF at high concentrations are toxic to the spermatogenesis (Mealy et al. 1990). The presence of IL-6 made Sertoli cells in vitro to exhibit a redistribution of tight junction proteins, resulting in reduction of transepithelial electrical resistance (Perez et al. 2011, 2012). This indicates that IL-6 is also involved in downregulating BTB permeability.
The number of MHC class II antigen-bearing cells increased after the CFA+BP treatment even when immunization with testicular antigens was absent (Tung et al. 1987) (Fig. 5.3). Therefore, both CFA and BP also may affect histopathological pattern of autoimmune inflammation against the testicular antigens. Tung et al. (1987) have demonstrated that the tubuli recti, rete testis, and ductuli efferentes were predominant sites of inflammation in passive EAO (induced by transferring CD4+ T cells isolated from the immunized donors into non-immunized, naïve recipients) but that active EAO (induced by immunization with testicular homogenate+CFA+BP) affected the peripheral seminiferous tubules under the tunica albuginea away from the tubuli recti and rete testis. The reason for the first involvement of the tubuli recti in passive model of EAO may be that the recipient mice were free from CFA+BP-induced upregulation of the distribution of antigen-presenting macrophages/dendritic cells throughout the testes (Fig. 5.2).
Sato et al. (1981) and Kohno et al. (1983) demonstrated minute histopathology of this EAO. In the EAO lesion, granular deposits of IgG were identified around seminiferous tubules. A focal degeneration and desquamation of both spermatogonia and Sertoli cells was induced prior to inflammatory cell responses. Yule et al. (1988) showed immune sera from mice immunized with testicular homogenate+CFA+BP and had reactivity with testicular cells of mice younger than 2 weeks of age. The autoantibodies bound to spermatogonia and spermatocytes were IgG1 but not IgG2 isotypes. Therefore, immunization with testicular homogenate+CFA+BP, but not with testicular homogenate alone, produced autoantibodies against diploid germ cells (spermatogonia and preleptotene spermatocytes) outside the BTB (Yule et al. 1988) (Fig. 5.4). Furthermore, it also became evident that immunization with testis homogenate+CFA+BP elicits not only immune responses against germ cells but also the responses against other testicular components such as Leydig cells, Sertoli cells, and basal lamina of the seminiferous tubules (Sato et al. 1981; Lustig et al. 1982) (Fig. 5.4). Namely, immunization with testicular homogenate+CFA+BP includes immune responses against spermatozoa, spermatids, spermatocytes, spermatogonia, Sertoli cells, Leydig cells, and basal lamina of the seminiferous tubules.
Recently, a new syndrome, namely, the “autoimmune/autoinflammatory syndrome induced by adjuvants” (ASIA), has been defined (Shoenfeld and Agmon-Levin 2011; Colafrancesco et al. 2014). In this syndrome, different conditions induced by various adjuvants such as infectious fragments, hormones, aluminum, silicone, and metal are included, and the syndrome is characterized by common signs and symptoms, resulting in boosting the immune response and triggering the development of autoimmune phenomena (Loyo et al. 2012; Cruz-Tapias et al. 2013; Lujan et al. 2013). Therefore, CFA+BP treatment also may induce ASIA-like condition. Indeed, in CFA+BP-injected mice, both production of anti-TGC autoantibodies and development of cytolytic activity of T cells against TGC are inducible without the use of testicular antigens for sensitization (Ben et al. 1986; Musha et al. 2013) (Fig. 5.3). Moreover, reversibility of disease resistance to EAO was found by treatment with BP in EAO-resistant rats (Teuscher et al. 1989). Therefore, there is a possibility that EAO may be inducible with CFA+BP treatment alone if some optimal conditions such as longer injection periods and/or more frequent injections are devised.
5.2.4.3 Role of Antibodies
Demonstration of IgG in the testis after immunization with testicular homogenate+CFA+BP or in the recipient testis after transfer of sera from orchiectomized donor mice immunized with testicular homogenate+CFA+BP shows that autoantigens are present on TGC outside the Sertoli cell barrier. It suggests the existence of dynamic protective mechanisms against immune responses to the non-sequestered TGC in normal individuals (Yule et al. 1990). The IgG deposits in EAO lesions were characterized as antibodies bound to the spermatogonia and preleptotene spermatocytes and were detected as early as day 7 after immunization, 5–6 days before the onset of EAO (Tung and Teuscher 1995) (Fig. 5.4). Testis IgG deposits were also elicited by immunization with testis homogenate in incomplete Freund’s adjuvant which does not elicit EAO. This IgG is absorbed from circulation by the testis, because the IgG is found only in the serum of mice orchiectomized before the immunization (Mahi-Brown et al. 1988). Thus the immune deposits alone are not sufficient to cause active EAO. The lack of pathogenicity by immunization with testis homogenate in incomplete Freund’s adjuvant is probably related to the unique immunoglobulin subclass of the IgG deposits. Only IgG1 and IgG3 but not IgG2a or IgG2b were detected. This finding can explain lack of association of complement components in the deposits in testis. Actually, in the testis of mice immunized with testis homogenate, autoantibodies were detected as immune complexes around the damaged seminiferous tubules free of inflammatory cell response, although the pathogenic role of testicular immune complexes has not yet been explored. There is a possibility that binding of autoantibodies to the target antigen-bearing cells and tissues such as spermatogonia, spermatocytes, and basal lamina of the seminiferous tubular wall affects the physiological function of the seminiferous epithelium. To investigate it, autoantibodies against Sertoli cells and basal lamina of the seminiferous tubules can be obtained from animals immunized with testicular homogenate+CFA+BP. Lustig et al. (1978) found that EAO can be induced by passive transfer of an antiserum to seminiferous tubular basal lamina. The damage was characterized by foci of perivascular and peritubular infiltrates of mononuclear cells. Moreover, infolding, thickening, and rupture of the seminiferous tubular wall and vacuolization of Sertoli cells and spermatogenic disturbance were also seen. A linear deposit of immunoglobulins was detected along the basal lamina of seminiferous tubules and blood capillaries. Additionally, in the kidneys of recipients, a deposit of immunoglobulins along glomerular basement membrane, focal areas of mononuclear cell infiltrates, hypercellularity of glomeruli, and thickening of both glomerular capillary wall and Bowman’s capsule were also found. Moreover, by using two kinds of monoclonal antibodies (IgM) against Sertoli cell and basal lamina of the seminiferous tubule that were derived from testicular antigens+CFA+BP-immunized mice, the spermatogenic disturbance was induced experimentally by intra-testicular injection of a set of these two monoclonal antibodies in mice (Ichinohasama et al. 1986) (Figs. 5.2 and 5.4). However, single injection of either monoclonal antibody failed to induce the lesion. This fact indicated that monoclonal antibody against Sertoli cells could reach the Sertoli cell to impair its function, which was otherwise inaccessible without coincidental action of monoclonal antibody against the basal lamina of the seminiferous tubules. The monoclonal antibody against the basal lamina appeared to alter the permeability of the BTB and lower its barrier effect. The most depressed spermatogenesis was frequently observed in testes 1 week after the second injection in mice that had been injected with a mixture of the two kinds of monoclonal antibodies twice at a 1-week interval. Immunohistochemically, the percentage of seminiferous tubules positive for anti-mouse IgM and anti-mouse complement appeared to parallel the impaired degree of spermatogenesis. It is important to note that there was no inflammatory cell infiltration. Neither congestion nor thrombosis of vessels nor ischemic changes was seen. The chronology of changes in the testes revealed that the spermatogenesis gradually recovered and became almost normal at 8 weeks after the second monoclonal antibody treatment. This suggests that humoral immunity is not so critical for producing chronic EAO.
5.2.4.4 Relevant Autoantigens
Immunization with allogeneic murine tissue homogenate of various organs emulsified in CFA accompanied by the injection of BP revealed that only testicular and epididymal homogenates of adult but not prepubertal mice are capable of eliciting EAO (Adekunle et al. 1987). Immunization of mice with xenogeneic testicular antigens+CFA+BP failed to elicit significant EAO, indicating that the major target autoantigens are highly species specific (Adekunle et al. 1987). Moreover, the use of allogeneic mouse testicular homogenate from EAO-resistant strains as sensitizing antigens exhibited similar potential for inducing significant EAO like in cases of the use of allogeneic testicular homogenates from EAO-susceptible strains. It suggests that immunoregulation against testicular antigens, but not autoimmunogenicity of testicular antigens, affect EAO sensitivity. However, testicular homogenates from NZB/B1NJ and MRL/MpJ−/+ mice were significantly less potent at inducing autoimmune epididymitis as compared to other strains, indicating possible interstrain differences of relevant autoantigens for EAO induction (Adekunle et al. 1987).
By using SDS-polyacrylamide gel electrophoresis and immunoblotting by reaction of normal sera with normal testicular homogenates in mice, two immunoreactive bands corresponding to approximately 45 and 100 kDa were detected (Musha et al. 2013). These two natural autoantibodies were more definitely detected in CFA+BP-immunized mice with no use of testicular antigens. In immunohistochemical staining by reacting normal testicular sections with immune sera from CFA+BP-injected group, haploid TGC and other immature TGC near the basement membrane of seminiferous tubules were finely stained. Moreover, compared with the normal controls, one band of approximately 40 kDa was additionally detected by immune sera obtained from the CFA+BP-injected mice, indicating that treatment with CFA+BP alone can evoke autoimmune reactions against some testicular autoantigens despite the use of no testicular homogenate (Fig. 5.3). It may be due to CFA+BP-induced ASIA (autoimmune/autoinflammatory syndrome induced by adjuvants). In testicular homogenate+CFA+BP-induced EAO, two natural autoantibodies against testicular antigens corresponding to approximately 45 and 100 kDa were further definitely detected like in CFA+BP-immunized mice. Moreover, the serum samples of testicular homogenate+CFA+BP-immunized mice also reacted with eight additional testicular autoantigens of approximately 15, 22, 25, 40, 60, 75, 120, and 250 kDa.
Sperm-specific zonadhesin is a target autoantigen with an EAO-inducing (orchitogenic) polypeptide domain (Hardy and Garbers 1995; Wheeler et al. 2011) (Fig. 5.2). Zonadhesin is expressed on the acrosomal membrane of spermatid and sperm and can bind zona pellucida and inhibit interspecies gamete interaction (Tardif et al. 2010). Serum antibody reacted with the 340 kDa protein in sperm of wild type but not zonadhesin-null mice. It was next shown that mice immunized with recombinant zonadhesin+CFA+BP developed EAO. Two other murine testis-specific autoantigens (mAP1 and mAP2) for EAO induction were partially purified from mouse testis acetone powder (Teuscher et al. 1994). Dose-response bioassays revealed that mAP1 and mAP2 are most effective at eliciting active EAO by mixing with CFA+BP and passive EAO induced by transferring CD4+ T cells isolated from the immunized donors into non-immunized, naïve recipients, respectively.
A proteomic approach using 2D SDS-PAGE and immunoblotting analysis with immune sera obtained from testicular homogenate+CFA+BP-injected mice identified 12 spots. Seven were subsequently identified by mass spectrometry as heat shock proteins (HSPs), including HSP60 and HSP70, disulfide isomerase ER-60, alpha-1 antitrypsin, heterogeneous nuclear ribonucleoprotein H1, sperm outer dense fiber major protein 2, and phosphoglycerate kinase 1, which are abundant in male TGC and characterized as testicular autoantigens for EAO (Fijak et al. 2005). HSP70 has been reported to promote antigen-presenting cell function and converts T cell tolerance to autoimmunity in vivo (Millar et al. 2003).
Laminin, a component of the basal lamina, was also shown to be important autoantigens for EAO induction, because immunization with laminin+CFA+BP induced EAO (Lustig et al. 1987) (Fig. 5.2). This indicates that immunologic injury specific to the basal lamina induces TGC depletion from the seminiferous epithelium in an antigen-nonspecific manner. Indeed, morphological alteration of the basal lamina of the seminiferous tubules in various pathological conditions including a varicocele, cryptorchid testes, hypogonadotropic hypogonadism, and irradiated testes have been reported. It has also been demonstrated that the basal lamina of the seminiferous tubules in the Japanese monkey is histologically changed when spermatogenetic activity decreases in the nonbearing season. These findings suggest a possibility that physiological condition of the basal lamina of the seminiferous tubules influences the activity of seminiferous tubules (Tainosho et al. 2011; Naito et al. 2012).
5.2.4.5 Pathophysiology of Leukocytic Infiltration and the Following Spermatogenic Disturbance
For EAO induction, interaction between blood capillary endothelial cells and circulating leukocytes are important, because the capillary endothelial cells play an essential role by facilitating leukocyte recruitment via their ability to express cell surface adhesion molecules. Rats with EAO showed a significant increase in the percentage of CD31+ endothelial cells that upregulate the expression of CD106. The percentage of leukocytes expressing CD49d (CD106 ligand) also increases during EAO (Guazzone et al. 2012). The CD44 antigen is a cell surface glycoprotein involved in cell-cell interactions, cell adhesion, and migration. It functions as a hyaluronic acid receptor, and both lipopolysaccharide and TNF-alpha upregulate CD44-mediated hyaluronic acid binding. It is known that activated lymphocytes bind hyaluronic acid present on the endothelium, and this specific binding facilitates the rolling and extravasation of leukocytes into the inflammation site (Guazzone et al. 2005). By in vitro hyaluronic acid-binding assay, the level of peripheral blood mononuclear cell adhesion was higher in testicular antigens+CFA+BP-immunized rats compared to normal controls. By immunohistochemistry, a significant increase in the number of CD44+ cells was detected in the testicular interstitium of rats with severe EAO, indicating that the CD44 molecules are involved in the homing of lymphocytes into the testis of rats with EAO (Guazzone et al. 2005). In the testis under physiological conditions, IL-1-beta is feebly produced; however, an upregulation of IL-1-beta, which may promote adhesion and migration of leukocytes, was also observed in the testis of rats immunized with testicular antigens+CFA+BP (Guazzone et al. 2009). Chemokines such as monocyte chemoattractant protein-1 and macrophage inflammatory protein-1 were also upregulated in the EAO lesion and therefore might induce attraction and extravasation of immune cells into the testicular interstitium. A significant increase of monocyte chemoattractant protein-1 was observed in the testicular fluid and in the conditioned medium obtained from cultures of testicular macrophages of rats with EAO (Guazzone et al. 2003). An increase in monocyte chemoattractant protein-1 expression was immunohistochemically observed in mononuclear cells, endothelial cells, Leydig cells, peritubular myoid cells, and Sertoli cells of rats with severe EAO. This indicates that this chemokine has an important role in recruiting immune cells to the testis in rats undergoing EAO.
CD4+ T cells that secrete Th1 cytokines in vitro play the most important role for EAO induction (Yule and Tung 1993; Tung 1998). All orchitogenic T cell clones, derived from mice immunized with testicular homogenate+CFA+BP, expressed both CD4 and the alpha-beta TCR. When activated, they produced IL-2, IFN-gamma, and TNF-alpha, but not IL-4. EAO transfer was significantly and reproducibly attenuated when recipients were given neutralizing antibody to TNF-alpha but not IFN-gamma in vivo. Hence, TNF-alpha rather than IFN-gamma was shown to be required for amplification of the pathogenic T cell response (Tung and Teuscher 1995). Although TNF-alpha protects TGC from apoptosis at physiologically low concentrations in normal testes, it behaves as an apoptotic factor that induces TGC death under inflammatory conditions (Theas et al. 2008). Actually, spermatogenic disturbance was experimentally induced by injection of TNF into the rat testis through a direct effect on TGC and an indirect mechanism involving Leydig cells or Sertoli cell dysfunction (Mealy et al. 1990). On the contrary, there is a study demonstrating that TNF does not appear to be the principal cytokine involved in the pathogenesis of actively induced EAO (Teuscher et al. 1990c). In that study, the ability of TNF to function as a coadjuvant in eliciting active EAO was examined by treating the EAO-immunized mice with recombinant murine TNF or anti-TNF antibodies at various time points throughout both the induction and effector phase of the EAO process; however, the all treated mice failed to exhibit a markedly significant change in EAO outcome in comparison with EAO-sensitized mice without TNF or anti-TNF antibodies.
Ben et al. (1986) have shown a good correlation between the severity of testicular pathological changes and cytolytic activity of immune lymphocytes against TGC. They concluded that the cytolytic activity against TGC might be a major factor in the development of EAO, but they did not determine the phenotype of the lymphocytes responsible for the cytolytic function. They also demonstrated that spleen cells from CFA+BP-injected mice kill TGC in vitro in spite of no immunization with testicular homogenate+CFA+BP (Fig. 5.3). They hypothesized that CFA+BP treatment nonspecifically damages the BTB by which testis-specific cytotoxic T cell clones are stimulated and expanded. However, there is another possibility that the adjuvant treatment nonspecifically stimulates all immune cells to secrete various kinds of cytokines, which are toxic and injurious to the seminiferous epithelium. Ben et al. (1986) also suggested that EAO may be induced in mice with adjuvants alone if the optimal experimental procedures are used. If this hypothesis is correct, an anamnesis of tuberculosis or whooping cough could be one of causes for immunologic orchitis, like mumps orchitis.
Testicular macrophages in rats immunized with testicular homogenate+CFA+BP also secreted both IFN-gamma and TNF-alpha, like CD4+ T cells (Suescun et al. 2003; Rival et al. 2008; Theas et al. 2008). Furthermore, IL-6 expression in testicular macrophages is also significantly increased in EAO (Rival et al. 2006a). In the rat EAO, the balance between ED1+ macrophage (circulating monocytes arriving at the testis) and ED2+ macrophage (resident macrophages in the testis) subsets can be disrupted under EAO conditions (Rival et al. 2008). The increased number of ED1+ cells is maintained for long periods of time during chronic inflammation in EAO. What causes the persistent elevated macrophage numbers in the testis with EAO is still unclear. ED1+ cells notably express IL-6 at high levels compared with ED2+, suggesting the distinct roles of the two types of macrophages in mediating inflammatory responses. An in vitro study has shown that exogenous IL-6 induces TGC apoptosis (Theas et al. 2003). Therefore, the high production of IL-6 by testicular ED1+ macrophages, the increased expression of IL-6 receptor in TGC, and the involvement of this cytokine in TGC apoptosis suggest a pathogenic role of IL-6 in EAO. Actually, IL-6 mRNA expression in murine testes also dramatically increased in testicular homogenate+CFA+BP-induced EAO (Musha et al. 2013).
Dendritic cells bearing CD80, CD86, and MHC class II antigens in EAO lesions have a mature immunogenic status and are able to induce immune responses to testicular antigens (Jacobo et al. 2011b). The mRNA expression level of IL-10 and IL-12p35 was significantly upregulated in enriched dendritic cell fraction in draining lymph nodes from the EAO-affected testis (Guazzone et al. 2011a). Furthermore, mRNA level of chemokine receptor for monocyte chemoattractant protein 3 was significantly increased, and the expression of chemokine receptor for monocyte chemoattractant protein-1 was decreased in isolated dendritic cells from rats with EAO (Rival et al. 2006b, 2007). In co-culture experiments, testicular dendritic cells isolated from EAO lesion significantly enhanced naïve T cell proliferation compared with control testicular dendritic cells (Rival et al. 2007). Therefore, testicular dendritic cells in control testis is not mature and functionally tolerogenic, while they reach a mature immunogenic state by IL-12 expression in EAO-affected testes and stimulate T cell proliferation prior imminent migration to the draining lymph nodes to amplify immune responses against testicular antigens (Rival et al. 2007). During EAO, elevated levels of high-mobility group box protein 1 were found. High-mobility group box protein 1 is a pro-inflammatory cytokine released from both monocytes and macrophages and functions as an immunostimulatory signal that induces dendritic cell maturation. This protein appears to be involved in regulating inflammatory reactions in EAO-affected testes, as blocking its action by ethyl pyruvate reduces the disease progression (Aslani et al. 2014). Therefore, high-mobility group box protein 1 could be one of promising targets in attenuating testicular damage caused by inflammatory reactions.
Mast cells are also involved in this EAO. They increased tenfold in number throughout the interstitial tissue and were partially degradated (Iosub et al. 2006). Protease-activated receptor 2 is known to be activated by mast cell tryptase. In EAO, protease-activated receptor 2 on testicular macrophages and peritubular cells was strongly upregulated. Isolated peritubular-like cells responded to protease-activated receptor 2 activation by increased mRNA expressions of monocyte chemoattractant protein-1, transforming growth factor-beta-2, and cyclooxygenase-2 in vitro. Indeed, expression of these three inflammatory mediators, together with nitric oxide-nitric oxide synthase, increased significantly in EAO-affected testes (Iosub et al. 2006; Jarazo-Dietrich et al. 2012). Furthermore, expression of these cytokines was upregulated after injection of recombinant tryptase in vivo. This suggests that mast cell tryptase contributes to pathogenic mechanisms of EAO.
The spermatogenic disturbance is characterized by apoptosis of TGC, which is mediated by the Fas/Fas ligand and Bax/Bcl-2 systems during EAO (Theas et al. 2003, 2006; Jacobo et al. 2012). Fas/Fas ligand system works through activation of caspase 8, whereas the Bax/Bcl-2 system works through activation of caspase 9; these activations eventually lead to activation of caspase 3, thereby resulting in TGC apoptosis. In testicular antigens+CFA+BP-immunized rats, real-time RT-PCR analyses revealed that Fas mRNA expression significantly increased compared with the control group. Most spermatocytes expressing Fas were apoptotic (Theas et al. 2003). The numbers of membrane Fas ligand-expressing CD4+ and CD8+ T cells were increased in the testis during EAO, but no expression of Fas ligand by macrophages was found (Jacobo et al. 2012). By Western blotting, an increase in soluble form of Fas ligand content was detected in the testicular fluid in EAO-affected testis (Jacobo et al. 2012). Since the soluble form of Fas ligand is able to enter the adluminal compartment of the seminiferous tubules, this molecule induces apoptosis of Fas-bearing TGC. Indeed, many Fas-bearing TGC were also immunoreactive for Fas ligand (Theas et al. 2003). On the other hand, Bax was mainly expressed in spermatocytes and spermatids and Bcl-2 in TGC at the basal compartment of the seminiferous tubules. Bax-beta isoform content increased in EAO rat testis compared with controls, whereas content of Bax-alpha remained unchanged. However, Bax-alpha content decreased in the cytosol and increased in the mitochondrial and endoplasmic reticulum-enriched fractions of testis from EAO rats compared with controls. Bcl-2 content also increased in the EAO-affected testes. Therefore, extrinsic, mitochondrial and possibly endoplasmic reticulum pathways are inducers of TGC apoptosis in EAO and Bax and Bcl-2 proteins modulate this process (Theas et al. 2006).