Next Article in Journal
Efficacy of Intravenous Elosulfase Alfa for Mucopolysaccharidosis Type IVA: A Systematic Review and Meta-Analysis
Previous Article in Journal
History of Endometriosis Is Independently Associated with an Increased Risk of Ovarian Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Testicular Immunity and Its Connection with the Microbiota. Physiological and Clinical Implications in the Light of Personalized Medicine

1
Interdisciplinary Department of Medicine, Section of Microbiology and Virology, University of Bari “Aldo Moro”, 70124 Bari, Italy
2
Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples “Federico II”, 80131Naples, Italy
3
Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
4
Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy
5
Independent Researcher, Sorriso & Benessere—Ricerca e Clinica, 70129 Bari, Italy
6
Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy
*
Authors to whom correspondence should be addressed.
J. Pers. Med. 2022, 12(8), 1335; https://doi.org/10.3390/jpm12081335
Submission received: 22 July 2022 / Revised: 17 August 2022 / Accepted: 18 August 2022 / Published: 20 August 2022
(This article belongs to the Section Omics/Informatics)

Abstract

:
Reproduction is a complex process, which is based on the cooperation between the endocrine–immune system and the microbiota. Testicular immunity is characterized by the so-called immune privilege, a mechanism that avoids autoimmune attacks against proteins expressed by spermatozoa. Testicular microbiota is connected with the gut microbiota, the most prevalent site of commensals inthe body. Both microbiotas take part inthe development of the immune system and protection againstpathogen invasion. Dysbiosis is caused by concurrent pathologies, such as obesity, diabetes, infections and trauma. The substitution of beneficial bacteria with pathogens may lead to destruction of spermatozoa directly or indirectly and, ultimately, to male infertility. Novel therapeutic interventions, i.e., nutritional interventions and supplementation of natural products, such as, probiotics, prebiotics, antioxidants and polyphenols, may lead to the restoration of the otherwise-impaired male reproductive potential, even if experimental and clinical results are not always concordant. In this review, the structure and immune function of the testis will be described with special reference to the blood–testisbarrier. The regulatory role of both the gut and testicular microbiota will be illustrated in health and disease, also emphasizing therapeutic attempts with natural products for the correction of male infertility, in the era of personalized medicine.

1. Introduction

The testis is a continuous source of germ cells as the first step of male reproduction, followed by the transport of sperm to the fallopian tube sperm–egg binding sites [1]. In general terms, reproduction is a complex process, which requires a strict collaboration between the endocrine and the immune system. In fact, spermatogenesis is regulated by the hypothalamic–pituitary–testicular axis for the gonadal steroid hormone to occur [2,3]. On the other hand, the testis is endowed with a specialized immune system that becomes tolerogenic towards the antigenic proteins expressed by spermatozoa [4]. Such a characteristic of the testis is known as “immune privilege” in the sense that spermatozoa are protected from autoimmune attacks by gonadal immune cells [5,6]. Furthermore, the testis owns a physical barrier, the so-called blood–testis barrier (BTB), which protects germ cells from noxious immune responses [7]. Alterations of the above-described protective homeostasis by metabolic disorders, infectious events, inflammation and trauma may lead to autoimmunity and infertility [8,9,10].
Microbiota is the assembly of commensal microorganisms located in different districts of the body, which contribute to the health of the host [11]. Particularly, gut microbiota, the most abundant in the body (80%), neutralizes pathogen colonization, exerts metabolic and nutritional activities and takes part in the development of the immune system [12,13,14,15]. Of note, only 9% of the human microbiota is harbored in the urogenital tract, but it is mostly gut microbiota that takes part in male and female sexual maturation. In fact, intestinal metabolites, such as secondary bile acids as well as indole and soybean, regulate male and female sexual organs [16,17,18].
The gut microbiota is composed offour major phyla, i.e., Bacteroidota, Bacillota, Actinomycetota and Pseudomonadota with Bacteroidota and Bacillota representing the 90% of intestinal bacteria contingent. The imbalance of gut microbiota, also in relation to high-fat and calorie-rich diets, may lead to a condition of dysbiosis, which with the time canresult in disease status [19]. Moving to the male microbiota, Lactobacillus, Pseudomonas and Prevotella represent the main bacteria contained in the seminal fluid and their replacement by other pathogens may cause dysbiosis, which, in turn, leads to infertility [20].
In this review, special emphasis will be placed on the description of the immune system of the testis, as well as to the components of BTB. Then, the influence of the gut microbiota and male microbiota on the testicular immune system will be illustrated, pointing out all those conditions of dysbiosis which may alter gonadal function and fertility. Novel therapeutic attempts with natural products will also be discussed. All needs must be considered when determining the optimal way to treat an individual patient in the emerging era of personalized medicine.

1.1. The Immune Environment of the Testis

Immune privilege is confined to a few districts of the body, and, among them, the testicular environment is included. In fact, the proteins expressed on the spermatozoa membrane can elicit a robust immune attack, which may destroy spermatozoa, ultimately leading to infertility. Immune privilege relies on two major mechanisms: (I) the physical shield represented by the BTB, mainly consisting of Sertoli cells (SCs); (II) the tolerogenic response mounted by the immune armamentarium of the testis [21]. In this framework, it is important to briefly describe the structure of the testis for a better comprehension of its function. The epithelium of the seminiferous tubules holds SCs, which provide nutrition and growth factors to germ cells [22,23]. On the other hand, Leydig cells are interspersed between the tubules and secrete testosterone (T) for spermatogenesis to take place [24,25]. The interstitial space of the testis harbors lymphatic vessels, which permit access to afferent lymph nodes [26]. Peritubular lymphatic sinusoids surround the seminiferous tubules with lymphatic capillaries ubicated beneath the tunica albuginea [27]. Macrophages, dendritic cells (DCs), mast cells and T cells are contained in the interstitium, and their function will be described in a specific section of this review. The structure of the testis is depicted in Figure 1.

1.2. Blood–Testis Barrier Function

The BTB is constituted by tight junctions (TJs), gap junctions (GJs), desmosome-like junctions and SCs. BTB is maintained by the N-cadherin/beta-catenin of the GJs and occluding/Z0-1 of the TJs all anchored in F-actin bundles [28].Junctions prevent haploid germ cells fromreaching the blood [29]. SCs supply support to germ cells, giving glucose, fatty acids and growth factors, as well asmaintaining an appropriate ionic and metabolic milieu in the testis [30,31]. Moreover, SCs secrete factors which keep an immunoprotected environment in the testis, such as transforming growth factor (TGF)-beta 1 in order to avoid autoimmune destruction of sperm cells [32].
Another mechanism of protection elicited by SCs is their ability to phagocyte apoptotic germ cells and residual bodies [33]. In order to accomplish their phagocyitc activity, SCs utilize Axl, tyro3 and Mer tyrosine kinase receptors, as well as growth-arrest-specific gene 6 (GAS6). SC-mediated phagocytosis supports spermatogenesis through various mechanisms: (1) making room for the germ cell differentiation process; (2) eliminating harmful substances derived from necrotic germ cells; (3) clearing autoantigens, which may trigger autoimmune responses; (4) providing energy sources to other SCs via the recycle of apoptotic germ cell components [34]. Lastly, SCs switchoff the inflammatory responses of T cells in the testicular interstitium [35]. The BTB is illustrated in Figure 2.

1.3. The Immune Arsenal of the Testis

The BTB via SCs constitutes a physical barrier devoted to the protection of germ cells from a destructive immune attack. Besides that, testicular immune cells maintain either a tolerogenic milieu or protect spermatogenesis from pathogen invasion; thus, theycontrol inflammatory processes, which very often are responsible for male infertility [36,37,38]. In fact, evidence has been provided that infections or inflammatory states inhibit steroidogenesis, cause apoptosis of germ cells and destroy testicular epithelial cells, thus provoking infertility [39].
Immune response relies on two major arms, the innate immune system and the adaptive immune system, respectively [40]. Phagocytes(granulocytes and macrophages), natural killer cells and dendritic cells(DCs) [majorantigen-presenting cells (APCs)] are involved in the innate immuneresponse. On the other hand, T and B lymphocytes recognize their specific antigensand maintain immunological memory. Mostly, T cells are divided into differentsubsets, such as T helper (h), T cytotoxic (Tc) and T regulatory (Treg) cells [41].
In the next paragraphs, the functions of testicular macrophages, DCs and lymphocytes will be discussed under both steady state and inflammatory conditions.
(a) 
Macrophages
Testicular macrophages derive from three distinct sources: (1) early yolk sac macrophages; (2) fetal livermonocytes; (3) bone-marrow-derived monocytes [42,43]. Experimental studies have reported that testicular macrophages are able to preserve the local immune privilege, as observed in the testis of rats where these activated phagocytes produce the anti-inflammatory cytokine, interleukin (IL)-10, also expanding T regulatory (Treg) cells [44,45].
Testicular inflammation is caused either by bacteria such as Escherichia (E.) coli and Klebsiella spp. or viruses (HIV-1, Zika and Mumps orthorubulavirus) [46,47]. Furthermore, in this instance, animal experiments have clarified the detrimental role of infiltrating monocyte-derived macrophages in the promotion of local inflammation, even if the differentiation of peripheral monocytes into testicular macrophages needs further demonstration [48]. Moreover, infected testicular macrophages have been shown to alter SC TJ and interrupt the BTB [49].
Testicular macrophages have been divided into three groups: (1) ED-1 recognizing macrophages, a class of pro-inflammatory cells, which produce tumor necrosis factor-alpha and interferon-gamma; (2) ED-2 macrophages, which exert anti-inflammatory activities by release of IL-10; (3) ED1+ED-2 macrophages, which are a source of nitric oxidase synthase (NOS) [46,47]. ED2+ cells are the majority of the testis macrophages that support a tolerogenic milieu in this organ [50].
(b) 
Dendritic Cells
Dendritic cells (DCs), as professional antigen-presenting cells (APCs), play a tolerogenic effect in the testis, principally leading to Treg cell activation in response to normal sperm antigens [51]. Furthermore, indoleamine 2,3-dioxygenase (IDO), whichcatalyzes the tryptophan metabolism and generates kynurerine, has been found in activated DCs, thus contributing to immune privilege [52]. In fact, kynurerine, acting as a ligand for aryl hydrocarbon receptors on T cells, induces the generation of Foxp3+ Treg cells [51]. Of note, IDO has been shown to induce Treg cell activation in tumors and pregnant uterus, which are also privileged sites, like the testis [53,54].
Under pathological circumstances, in azoospermic humans testicular DCs are able to activate autoreactive T cells, upregulating co-stimulatory molecules, proinflammatory cytokines and major histocompatibility complex class-II (MHC-II), thus leading to male infertility [55,56].
(c) 
T Cells
Treg cells (T cells) have been detected in the mouse, rat and human testis, where they reside in the draining lymph nodes, thus interacting with tissue-specificautoantigens [57]. Located in such a strategic position, Treg cells exert their suppressive function, thus protecting spermatozoa from autoimmune attacks. In this respect, patients with autoimmune regulator gene mutation associated to a defect of Treg cells undergo a chronic testicular inflammation [58]. In chronic inflamed human azoospermic testis, evidence has been provided that Foxp3+ Treg cells are decreased with an increase in the proinflammatory T cell subset, T helper (h) 17 cells [59]. In rat experimental autoimmune orchitis (EAO), CD8+, CD25+, Foxp3+ and CD4+, CD25+, Foxp3+ and Treg cells are increased in the early phase, while the latter subset decreases in the chronic phase [60].
All the above evidence suggests that Treg cells are overly critical in the prevention of organ-specific autoimmunity and maintenance of the immune privilege in the testis. Testicular Th1 cells seem to be necessary for supporting immune homeostasis in this organ. However, an excessive activation of these cells may contribute to autoimmune orchitis [61]. Further studies have proven the intervention of Th17 cells in the later phase of autoimmune orchitis, thus hampering the function of Treg cells, also contributing to the subversion of the testis structure and spermatogenesis [62].
T cytotoxic (c) lymphocytes (CD8+ cells) harbor the testis in a percentage which is 2-fold higher than that of Th cells [63]. Testicular CD8+ cells are functionally associated with resident macrophages or Leydig cells and take part in graft survival [64]. In this respect, pancreatic and islet transplantation in the testis undergoes a lower rate of rejection with an elevated induction of Treg cells [65,66]. This may depend on the SC-mediated activation of Treg cells or on the less potent cytotoxic activity of testicular Tc lymphocytes [67]. To complete the above issue, it is worth mentioning the relationship between T lymphocytes, Leydig cells and SCs, respectively. Leydig cells harbor the interstitial region between seminiferous tubules and represent themajor source of T [68]. Co-cultures of Leydig cells and T cellshave revealed the suppressive effect of the former on the latter, also in view of the binding of Leydig cells to T cells viavascular adhesion molecules [69,70].Androgen receptors are expressed on T cells and, therefore, Leydig cells can modulate their function through androgen secretion.
Experimentally, depletion of T by ethane dimethane sulphonate gives rise to an epididymal sperm granuloma and accumulation of CD4+ and CD8+ T cells, which can be abrogated by supplementation of T [71]. Furthermore, in the EAO rat model, T replacement inhibits the development of autoimmune orchitis through the expansion of Treg cells [72]. Conclusively, Leydig cells are able to limit the infiltration of T cells within the testis, directly and indirectly. SCs are devoted to the protection of spermatogenesis, acting as immunological sentinels. In this regard, it appears that SCs promote the differentiation of tolerogenic DCs and Treg cells [73]. Of note, SCs behave as nonprofessional APCs, expressing MHC-II molecules, thus mediating the expansion of Foxp3+ Treg cells [74].
In this direction, transplanted SCs protect syngeneic islet grafts, generating Treg cells and decreasing release of IL-17 by T helper (h)17 cells [75]. In sum, SCs not only contribute to the BTB composition but also keep on check detrimental T cell responses. The testicular immune cells are expressed in Figure 3.

1.4. Composition and Function of the Testicular Microbiota

The dogma according to which the testis is an immune privileged site has been contradicted by the evidence that a few bacteria are able to colonize the gonad milieu. In fact, the phyla Actinomycetota, Bacteroidota, Bacillota and Pseudomonadota have been detected in testicular biopsies of azoospermic patients [76]. Moreover, the phyla Bacillota, Actinomycetota, Bacteroidota and the genera Blautia, Clostridium and Prevotella were found in testicular specimens of infertile men [77]. In another report, in dyspermic patients and healthy donors Lactobacillus, Pseudomonas, Prevotella and the phyla Pseudomonadota, Bacillota, Actinomycetota, Bacteroidota and Fusobacteria were identified, with the genus Prevotella being inversely associated with sperm concentration, while the Pseudomonas genus was correlated with sperm motility [78,79].
Despite the detection of the testicular microbiota, its role in the testis is still debated. According to a recent report, testicular microbiota seems to expand IL-17, producing gamma-delta T cells during puberty, promoting gonadal immune surveillance [80]. It is noteworthy that current research in this specific field has been focused on the link between gut microbiota and testicular microbiota. In the zebrafish model, the genera Vibrio, Aeromonas, Pseudomonas and Plesiomonas spp. have been detected in both gut andtestis [81,82]. In the same model, excessive fat intake led to a dramatic reduction of the genus Vibrio and Plesiomonas spp., with a subversion of signal transduction mechanisms, amino acid transport and metabolism. Furthermore, testicular microbiota regulates the signaling mechanisms of vitamin K and vitamin A and its alteration may change the composition of the extracellular matrix, ultimately leading to male infertility [83,84].
In this direction, evidence has been provided that in a metabolic syndrome model, vitamin A deficit alters the gut–testis axis, finally resulting in an impaired spermatogenesis [85,86]. The gut–testis axis is supported by experimental evidence. Transplantation of fecal flora from high-fat diet (HFD) to normal mice caused an increase in Bacteroidota phylum and Prevotella genus in normal mice followed by intestinal inflammation and endotoxemia, but mostly by an impaired spermatogenesis [82,87]. In the human counterpart, male infertility is characterized by a negative correlation between Bacteroidota phylum and Prevotella genus with sperm viability as a result of the “leaky gut hypothesis”. Thus, intestinal endotoxins may impede the T synthesis in Leydig cells, thus provoking a decrease in spermatozoa [88]. More precisely, endotoxins via binding to the TLR-4 expressed on immune cells and epithelial cells can activate the NF-kB pathway with massive release of proinflammatory cytokines [89,90]. In turn, cytokines activate the xanthine oxidase system, thus generating, elevated levels of reactive oxygen species (ROS) and oxidative stress [91].
Conclusively, the bacterial translocation-mediated inflammation can account for endothelial damage, subversion of the BTB and alteration of the spermatogenesis and spermatozoa viability [92]. Additionally, DCs and macrophages, which infiltrate the epididymis, are able to capture spermatozoa, thus, contributing, to the impairment of spermatogenesis [93]. Another link between gut microbiota and male reproduction is represented by the endotoxin-mediated insulin resistance (IR), as an expression of altered intestinal permeability [94,95,96]. IR stands for an event of pathogenetic relevance since it alters both gut microbiota and spermatogenesis. In fact, in infertility models with IR, higher levels of Saccharibacteria phylum and lower levels of the phyla Actinomycetota and Verrucomicrobia have been observed in comparison to controls without IR [97]. Parallelly, increased IR is associated with a decreased secretion of T by Leydig cells also in view of a reduced gonadotropin release [98]. In Figure 4 the gut–testis axis is described.

1.5. Seminal Dysbiosis with Particular Reference to Male Infertility

The influence of seminal dysbiosis is an issue of current interest. Dyspermic conditions, i.e., oligo-azoospermia, asteno-azoospermia and azoospermia, have been investigated in terms of microbial composition of seminal fluid. For instance, in azoospermic individuals, Bacteroidota and Bacillota phyla are increased, while the phyla Pseudomonadota and Actinomycetota are reduced [98]. In the oligo-asteno-teratozoospermic patients, instead, the genera Neisseria, Klebsiella and Pseudomonas and the phylum Bacillota are very abundant, but there is a decrease in Lactobacillus [99]. In idiopathic non-obstructive azoospermic patients, the Clostridium genus was decreased [100].
Quite interestingly, over the past few years, the influence of female microbiota on the male microbiota has intensively been investigated. For instance, Gardnerella vaginalis and the genus Lactobacillus have been detected in younger men’s seminal microbiota, while the genera Pseudomonas, Flavobacterium and Acidovorax have been found in seminal fluid of older individuals [101,102]. On the other hand, inflammatory seminal fluid is associated with Streptococcus agalactiae, Gardnerella vaginalis and bacterial vaginosis-related bacteria [103].

1.6. Microbial-Mediated Male Infertily

Despite the presence of the BTB, the testicular immune arsenal and the local microbiota, the testis can be invaded by urethral pathogens and sexually transmitted bacteria [104]. Acute epididymitis is a very frequent infection of the male reproductive tract, even if this organ has a structure quite overlapping that of the testis [105]. More exactly, epididymis is a less immunologically privileged site in comparison to the testis with a certain degree of immune responsiveness in the caput and an inflammatory profile in the cauda [106,107]. In epididymitis patients, the quality of semen is very low, with an alteration of the protein composition of the sperm, thus contributing to male infertility [108,109]. Persistent pathogen damage leads to fibrotic transformation and epithelial degeneration of the epididymis [110].
Due to the scarcity of human epididymal specimens, research has mainly been conducted on rodent tissue samples. Experimental Gram-negative and Gram-positive infections in the mouse testis have revealed a strong proinflammatory cytokine response with upregulation of NOS-2 [111]. Uropathogenic Escherichia coli (UPEC) infections in the mouse are characterized by an activation of TLR4and TLR5 in the epididymis caput with liberation of proinflammatory cytokines and type 1 interferon [112]. On the other hand, epididymal cells respond to UPEC challenge with the production of the antimicrobial peptide defensin b2, which is more effective than gentamycin in reducing bacterial load in both epididymis and testis [113].
Chlamydia trachomatis (Ct) is the most frequent sexually transmitted pathogen in males, leading to chronic inflammation and scarring of the male genital tract [114]. Ct antigens bind to TLR2 and TLR4 and pathogen recognition receptors with massive liberation of proinflammatory cytokines, which account for chronicity of inflammation [115]. As far as viral diseases are concerned, mumps virus, an RNA virus, is the most frequent cause of epididymitis and orchitis, which in turn cause male infertility [116]. COVID-19 has been reported to infect the testis, impairing T secretion, thus inducing primary hypogonadism or aggravating a preexistent status of hypogonadism [117]. In particular, a reduced number of Leydig cells have been detected in COVID-19 patients along with a high expression of angiotensin-converting enzyme 2 in the testis [118,119,120,121]. Additionally, involvement of testicular T and B lymphocytes in COVID-19 infection has been reported [122]. Infections of the male reproductive tract are illustrated in Figure 5.

1.7. Therapeutic Correction of Testicular Dysbiosis with Natural Products

In view of the connection between microbiota and male reproduction new therapies have been explored. With special reference to personalized medicine, there is a large body of evidence that nutrition can influence the composition of the microbiota, the quality of sperm in terms of caloric content of food components, as well as fatty acid, carbohydrate and protein profiles. In this regard, a high intake of saturated fatty acids may impair male fertility, while a healthy dietary regimen, i.e., the Mediterranean diet (MED) contributes to the preservation of the microbiota and sperm quality [123,124,125]. Conversely, the Western diet causes the rapid spread of obesity associated with hyperinsulinemia and hyperglycemia, which lead to an altered sperm function [126,127].
On these grounds, nutritional interventions to protect male reproduction have been adopted. For instance, MED has been shown to positively affect male reproductive performance, especially through the consumption of extra virgin olive oil (EVOO). According to [128], EVOO is able to change the sperm membrane lipid composition, reducing oxidative stress and enhancing mitochondrial function. Furthermore, MED exerts a homeostatic function in the endocrine–metabolic–immune axis, also shifting the gut microbiota towards an anti-inflammatory profile [129,130]. It is likely that testicular microbiota may be positively affected by MED, but such an assumption needs scientific demonstration. Certain natural products potentially effective in the restoration of testicular microbiota will be illustrated in the following paragraphs.
(a) 
Probiotics
Probiotics by definition are “live microorganisms which when administered in adequate amounts confer a health benefit on the host” [131,132]. They have been used to enhance male reproduction, owing to their ability to protect the intestinal barrier, inhibit pathogen growth and activate the immune response [133,134]. In astheno-azoospermic human donors, 3 weeks’ supplementation of Lactobacillus (L.) rhamnosus and Bifidobacterium longum improved sperm motility while reducing DNA fragmentation [135,136]. In another study, administration of a symbiotic, Familact®, composed by Lactobacillus strains and oligo-fructosaccharides, to idiopathic male infertility could enhance sperm quality and DNA integrity, while reducing free radicals in the semen [137].
Experimentally, in HFD obese mice, supplementation of L. rhamnosus improved spermatozoa motility, increasing the number of Leydig cells [138]. In infertile mice, administration of Lactobacillus spp., Bacillus spp., Saccharomyces cerevisiae (beer yeast) and photosynthetic bacteria cultures reduced sperm damage and improved motility [139].
(b) 
Prebiotics
Oligofructose, galacto-oligosaccharides and breast-milk oligosaccharides are the most representative prebiotics, endowed with the ability to increase levels of Bifidobacterium and Lactobacillus, as well as of SCFAs [140,141]. In a preclinical study, evidence has been provided that manno-oligosaccharides were able to accelerate sexual maturation in rats [142]. In particular, the decrease in blood corticosterone observed in this study could account for the elevated levels of T and maturation of seminiferous tubules. To the best of our knowledge, no clinical trials have been conducted to treat male infertility with prebiotics.
(c) 
Antioxidants
Vitamin C and vitamin E can exert especially beneficial effects in infertile men, reducing ROS levels, improving sperm motility and maintaining DNA integrity [143]. Among other antioxidants, lycopene, present in tomatoes and red fruits, seems to display positive effects on the testicular mitochondria by modulating lipid peroxidation within the mitochondrial membrane [144]. Conversely, other studies based on theadministration of antioxidants did not show any improvement of semen biomarkers and DNA integrity in infertile men [145,146].
(d) 
Polyphenols
Polyphenols are natural compounds, mainly contained in fruits, vegetables, oil, wine and cocoa [147,148]. They exert potent anti-inflammatory and antioxidant activities on different cell types, even including spermcells [149,150,151]. Experimental and human studies have been undertaken with quercetin and resveratrol; however, results have been quite controversial, since both polyphenols are endowed with antioxidant and pro-oxidant activities [152,153].
Table 1 shows the main natural products putatively involved in the treatment of male genital tract infections.

2. Conclusions

A mutual cooperation between testicular immunity and microbiota contribute to normal spermatogenesis and sperm maturation. Such an equilibrium may be subverted by a range of factors, even including concurrent pathologies, e.g., obesity, diabetes, infections and trauma. Among novel therapeutic approaches to restore male infertility, a proper nutritional regimen, as in the case of MED, may be useful in male infertility associated to obesity and diabetes. Furthermore, supplementation of natural products, such as probiotics, prebiotics, antioxidants and polyphenols has been demonstrated to enhance male reproductive function either in animal models or clinical trials. However, also, in view of conflicting results, more clinical attempts are needed to establish the actual effectiveness of natural products for the correction of testicular microbiota and immune function in the era of personalized medicine.

Author Contributions

Conceptualization, E.J., A.B. and L.S.; supervision and project administration, C.I., F.C., E.D.Z., M.C. and S.C.; revising the work critically for important intellectual content, E.J., A.B., S.C. and L.S.; final approval of the version to be published, E.J., C.I., A.B. and L.S.; validation and bibliographic research, S.C. and S.S.; acquisition and interpretation of data for the work, S.C. and E.D.Z.; agreement to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved, all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Acknowledgments

The authors thank Ioannis A. Charitos for his valuable help in preparing the figures of the text.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Agarwal, A.; Barbăroșie, C.; Ambar, R.; Finelli, R. The Impact of Single- and Double-Strand DNA Breaks in Human Spermatozoa on Assisted Reproduction. Int. J. Mol. Sci. 2020, 21, 3882. [Google Scholar] [CrossRef] [PubMed]
  2. Hermann, B.P.; Cheng, K.; Singh, A.; Roa-De La Cruz, L.; Mutoji, K.N.; Chen, I.C.; Gildersleeve, H.; Lehle, J.D.; Mayo, M.; Westernströer, B.; et al. The Mammalian Spermatogenesis Single-Cell Transcriptome, from Spermatogonial Stem Cells to Spermatids. Cell Rep. 2018, 25, 1650–1667.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ye, L.; Zeng, Q.; Ling, M.; Ma, R.; Chen, H.; Lin, F.; Li, Z.; Pan, L. Inhibition of IP3R/Ca2+ Dysregulation Protects Mice from Ventilator-Induced Lung Injury via Endoplasmic Reticulum and Mitochondrial Pathways. Front. Immunol. 2021, 12, 729094. [Google Scholar] [CrossRef] [PubMed]
  4. Meinhardt, A.; Wang, M.; Schulz, C.; Bhushan, S. Microenvironmental signals govern the cellular identity of testicular macrophages. J. Leukoc. Biol. 2018, 104, 757–766. [Google Scholar] [CrossRef]
  5. Forrester, J.V.; Xu, H.; Lambe, T.; Cornall, R. Immune privilege or privileged immunity? Mucosal Immunol. 2008, 1, 372–381. [Google Scholar] [CrossRef]
  6. O’Donnell, L.; Smith, L.B.; Rebourcet, D. Sperm-specific proteins: New implications for diagnostic development and cancer immunotherapy. Curr. Opin. Cell Biol. 2022, 77, 102104. [Google Scholar] [CrossRef]
  7. Stanton, P.G. Regulation of the blood-testis barrier. Semin. Cell Dev. Biol. 2016, 59, 166–173. [Google Scholar] [CrossRef]
  8. Schuppe, H.C.; Meinhardt, A. Immune privilege and inflammation of the testis. Immunol. Gametes Embryo Implant. 2005, 88, 1–14. [Google Scholar] [CrossRef]
  9. Chen, Q.; Deng, T.; Han, D. Testicular immunoregulation and spermatogenesis. Semin. Cell Dev. Biol. 2016, 59, 157–165. [Google Scholar] [CrossRef]
  10. Loveland, K.L.; Klein, B.; Pueschl, D.; Indumathy, S.; Bergmann, M.; Loveland, B.E.; Hedger, M.P.; Schuppe, H.C. Cytokines in Male Fertility and Reproductive Pathologies: Immunoregulation and Beyond. Front. Endocrinol. 2017, 8, 307. [Google Scholar] [CrossRef]
  11. Proctor, L. Priorities for the next 10 years of human microbiome research. Nature 2019, 569, 623–625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 2006, 7, 688–693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Belkaid, Y.; Harrison, O.J. Homeostatic Immunity and the Microbiota. Immunity 2017, 46, 562–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Desmet, L.; Thijs, T.; Segers, A.; Verbeke, K.; Depoortere, I. Chronodisruption by chronic jetlag impacts metabolic and gastrointestinal homeostasis in male mice. Acta Physiol. 2021, 233, e13703. [Google Scholar] [CrossRef]
  15. Zhang, Z.J.; Lehmann, C.J.; Cole, C.G.; Pamer, E.G. Translating Microbiome Research from and to the Clinic. Annu. Rev. Microbiol. 2022. [Google Scholar] [CrossRef]
  16. Baptissart, M.; Vega, A.; Martinot, E.; Pommier, A.J.; Houten, S.M.; Marceau, G.; de Haze, A.; Baron, S.; Schoonjans, K.; Lobaccaro, J.M.; et al. Bile acids alter male fertility through G-protein-coupled bile acid receptor 1 signaling pathways in mice. Hepatology 2014, 60, 1054–1065. [Google Scholar] [CrossRef]
  17. Sonowal, R.; Swimm, A.; Sahoo, A.; Luo, L.; Matsunaga, Y.; Wu, Z.; Bhingarde, J.A.; Ejzak, E.A.; Ranawade, A.; Qadota, H.; et al. Indoles from commensal bacteria extend healthspan. Proc. Natl. Acad. Sci. USA 2017, 114, E7506–E7515. [Google Scholar] [CrossRef] [Green Version]
  18. Selvaraj, V.; Zakroczymski, M.A.; Naaz, A.; Mukai, M.; Ju, Y.H.; Doerge, D.R.; Katzenellenbogen, J.A.; Helferich, W.G.; Cooke, P.S. Estrogenicity of the isoflavone metabolite equol on reproductive and non-reproductive organs in mice. Biol. Reprod. 2004, 71, 966–972. [Google Scholar] [CrossRef] [Green Version]
  19. Nobs, S.P.; Zmora, N.; Elinav, E. Nutrition Regulates Innate Immunity in Health and Disease. Annu. Rev. Nutr. 2020, 40, 189–219. [Google Scholar] [CrossRef]
  20. Lundy, S.D.; Vij, S.C.; Eng, C. Reply to Eugenio Ventimiglia, EdoardoPozzi, Massimo Alfano, Francesco Montorsi, and Andrea Salonia’s Letter to the Editor re: Scott D. Lundy, Naseer Sangwan, Neel V. Parekh; et al. Functional and Taxonomic Dysbiosis of the Gut, Urine, and Semen Microbiomes in Male Infertility. Eur Urol 2021;79:826–36. Eur. Urol. 2021, 80, e55–e56. [Google Scholar] [CrossRef]
  21. Kaur, G.; Thompson, L.A.; Dufour, J.M. Sertoli cells—Immunological sentinels of spermatogenesis. Semin. Cell Dev. Biol. 2014, 30, 36–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Li, N.; Wang, T.; Han, D. Structural, cellular and molecular aspects of immune privilege in the testis. Front. Immunol. 2012, 3, 152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ruthig, V.A.; Lamb, D.J. Updates in Sertoli Cell-Mediated Signaling During Spermatogenesis and Advances in Restoring Sertoli Cell Function. Front. Endocrinol. 2022, 13, 897196. [Google Scholar] [CrossRef] [PubMed]
  24. Meinhardt, A.; Hedger, M.P. Immunological, paracrine and endocrine aspects of testicular immune privilege. Mol. Cell. Endocrinol. 2011, 335, 60–68. [Google Scholar] [CrossRef]
  25. Gu, X.; Li, S.Y.; Matsuyama, S.; DeFalco, T. Immune Cells as Critical Regulators of Steroidogenesis in the Testis and Beyond. Front. Endocrinol. 2022, 13, 894437. [Google Scholar] [CrossRef]
  26. Fijak, M.; Bhushan, S.; Meinhardt, A. Immunoprivileged sites: The testis. Methods Mol. Biol. 2011, 677, 459–470. [Google Scholar] [CrossRef]
  27. Hirai, S.; Naito, M.; Terayama, H.; Qu, N.; Kuerban, M.; Musha, M.; Ikeda, A.; Miura, M.; Itoh, M. The origin of lymphatic capillaries in murine testes. J. Androl. 2012, 33, 745–751. [Google Scholar] [CrossRef]
  28. Su, L.; Wang, Z.; Xie, S.; Hu, D.; Cheng, Y.C.; Mruk, D.D.; Guan, Y. Testin regulates the blood-testis barrier via disturbing occludin/ZO-1 association and actin organization. J. Cell. Physiol. 2020, 235, 6127–6138. [Google Scholar] [CrossRef]
  29. Mital, P.; Hinton, B.T.; Dufour, J.M. The blood-testis and blood-epididymis barriers are more than just their tight junctions. Biol. Reprod. 2011, 84, 851–858. [Google Scholar] [CrossRef] [Green Version]
  30. Rato, L.; Alves, M.G.; Socorro, S.; Duarte, A.I.; Cavaco, J.E.; Oliveira, P.F. Metabolic regulation is important for spermatogenesis. Nat. Rev. Urol. 2012, 9, 330–338. [Google Scholar] [CrossRef]
  31. Ye, L.; Huang, W.; Liu, S.; Cai, S.; Hong, L.; Xiao, W.; Thiele, K.; Zeng, Y.; Song, M.; Diao, L. Impacts of Immunometabolism on Male Reproduction. Front. Immunol. 2021, 12, 658432. [Google Scholar] [CrossRef] [PubMed]
  32. Suarez-Pinzon, W.; Korbutt, G.S.; Power, R.; Hooton, J.; Rajotte, R.V.; Rabinovitch, A. Testicular sertoli cells protect islet beta-cells from autoimmune destruction in NOD mice by a transforming growth factor-beta1-dependent mechanism. Diabetes 2000, 49, 1810–1818. [Google Scholar] [CrossRef] [Green Version]
  33. Nakanishi, Y.; Shiratsuchi, A. Phagocytic removal of apoptotic spermatogenic cells by Sertoli cells: Mechanisms and consequences. Biol. Pharm. Bull. 2004, 27, 13–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Dutta, S.; Sengupta, P.; Slama, P.; Roychoudhury, S. Oxidative Stress, Testicular Inflammatory Pathways, and Male Reproduction. Int. J. Mol. Sci. 2021, 22, 10043. [Google Scholar] [CrossRef] [PubMed]
  35. Zhao, S.; Zhu, W.; Xue, S.; Han, D. Testicular defense systems: Immune privilege and innate immunity. Cell. Mol. Immunol. 2014, 11, 428–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Punab, M.; Poolamets, O.; Paju, P.; Vihljajev, V.; Pomm, K.; Ladva, R.; Korrovits, P.; Laan, M. Causes of male infertility: A 9-year prospective monocentre study on 1737 patients with reduced total sperm counts. Hum. Reprod. 2017, 32, 18–31. [Google Scholar] [CrossRef]
  37. Michel, V.; Duan, Y.; Stoschek, E.; Bhushan, S.; Middendorff, R.; Young, J.M.; Loveland, K.L.; Kretser, D.M.; Hedger, M.P.; Meinhardt, A. Uropathogenic Escherichia coli causes fibrotic remodelling of the epididymis. J. Pathol. 2016, 240, 15–24. [Google Scholar] [CrossRef] [PubMed]
  38. Schuppe, H.C.; Pilatz, A.; Hossain, H.; Diemer, T.; Wagenlehner, F.; Weidner, W. Urogenital Infection as a Risk Factor for Male Infertility. Dtsch. Ärzteblatt Int. 2017, 114, 339–346. [Google Scholar] [CrossRef] [Green Version]
  39. Hedger, M.P. Immunophysiology and pathology of inflammation in the testis and epididymis. J. Androl. 2011, 32, 625–640. [Google Scholar] [CrossRef]
  40. Gray, J.I.; Farber, D.L. Tissue-Resident Immune Cells in Humans. Annu. Rev. Immunol. 2022, 40, 195–220. [Google Scholar] [CrossRef]
  41. Ivanov, I.I.; Tuganbaev, T.; Skelly, A.N.; Honda, K. T Cell Responses to the Microbiota. Annu. Rev. Immunol. 2022, 40, 559–587. [Google Scholar] [CrossRef] [PubMed]
  42. Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Gomez Perdiguero, E.; Klapproth, K.; Schulz, C.; Busch, K.; Azzoni, E.; Crozet, L.; Garner, H.; Trouillet, C.; de Bruijn, M.F.; Geissmann, F.; et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 2015, 518, 547–551. [Google Scholar] [CrossRef]
  44. Meinhardt, A.; Dejucq-Rainsford, N.; Bhushan, S. Testicular macrophages: Development and function in health and disease. Trends Immunol. 2022, 43, 51–62. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, M.; Fijak, M.; Hossain, H.; Markmann, M.; Nüsing, R.M.; Lochnit, G.; Hartmann, M.F.; Wudy, S.A.; Zhang, L.; Gu, H.; et al. Characterization of the Micro-Environment of the Testis that Shapes the Phenotype and Function of Testicular Macrophages. J. Immunol. 2017, 198, 4327–4340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Pleuger, C.; Silva, E.J.R.; Pilatz, A.; Bhushan, S.; Meinhardt, A. Differential Immune Response to Infection and Acute Inflammation Along the Epididymis. Front. Immunol. 2020, 11, 599594. [Google Scholar] [CrossRef] [PubMed]
  47. Ali, B.R.; Atiyah, S.A.; Yser, H.T.; Khelewe, A.M.; Hameed, H.N. The influence of SARS-CoV-2 on semen parameters of infected infertile male in comparison with those that noninfected. J. Clin. Lab. Anal. 2022, e24568. [Google Scholar] [CrossRef]
  48. Tsetsarkin, K.A.; Acklin, J.A.; Liu, G.; Kenney, H.; Teterina, N.L.; Pletnev, A.G.; Lim, J.K. Zika virus tropism during early infection of the testicular interstitium and its role in viral pathogenesis in the testes. PLoS Pathog. 2020, 16, e1008601. [Google Scholar] [CrossRef]
  49. Bhushan, S.; Meinhardt, A. The macrophages in testis function. J. Reprod. Immunol. 2017, 119, 107–112. [Google Scholar] [CrossRef]
  50. Jarazo-Dietrich, S.; Jacobo, P.; Pérez, C.V.; Guazzone, V.A.; Lustig, L.; Theas, M.S. Up regulation of nitric oxide synthase-nitric oxide system in the testis of rats undergoing autoimmune orchitis. Immunobiology 2012, 217, 778–787. [Google Scholar] [CrossRef]
  51. De Rose, R.; Fernandez, C.S.; Hedger, M.P.; Kent, S.J.; Winnall, W.R. Characterisation of macaque testicular leucocyte populations and T-lymphocyte immunity. J. Reprod. Immunol. 2013, 100, 146–156. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, P.; Duan, Y.G. The role of dendritic cells in male reproductive tract. Am. J. Reprod. Immunol. 2016, 76, 186–192. [Google Scholar] [CrossRef] [PubMed]
  53. Gualdoni, G.S.; Jacobo, P.V.; Sobarzo, C.M.; Pérez, C.V.; Matzkin, M.E.; Höcht, C.; Frungieri, M.B.; Hill, M.; Anegon, I.; Lustig, L.; et al. Role of indoleamine 2,3-dioxygenase in testicular immune-privilege. Sci. Rep. 2019, 9, 15919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Mezrich, J.D.; Fechner, J.H.; Zhang, X.; Johnson, B.P.; Burlingham, W.J.; Bradfield, C.A. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J. Immunol. 2010, 185, 3190–3198. [Google Scholar] [CrossRef] [Green Version]
  55. Pallotta, M.T.; Orabona, C.; Volpi, C.; Vacca, C.; Belladonna, M.L.; Bianchi, R.; Servillo, G.; Brunacci, C.; Calvitti, M.; Bicciato, S.; et al. Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells. Nat. Immunol. 2011, 12, 870–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Munn, D.H.; Mellor, A.L. Indoleamine 2,3-dioxygenase and tumor-induced tolerance. J. Clin. Investig. 2007, 117, 1147–1154. [Google Scholar] [CrossRef] [Green Version]
  57. Fijak, M.; Meinhardt, A. The testis in immune privilege. Immunol. Rev. 2006, 213, 66–81. [Google Scholar] [CrossRef]
  58. Jacobo, P.; Guazzone, V.A.; Theas, M.S.; Lustig, L. Testicular autoimmunity. Autoimmun. Rev. 2011, 10, 201–204. [Google Scholar] [CrossRef]
  59. Garza, K.M.; Agersborg, S.S.; Baker, E.; Tung, K.S. Persistence of physiological self antigen is required for the regulation of self tolerance. J. Immunol. 2000, 164, 3982–3989. [Google Scholar] [CrossRef] [Green Version]
  60. Kekäläinen, E.; Tuovinen, H.; Joensuu, J.; Gylling, M.; Franssila, R.; Pöntynen, N.; Talvensaari, K.; Perheentupa, J.; Miettinen, A.; Arstila, T.P. A defect of regulatory T cells in patients with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J. Immunol. 2007, 178, 1208–1215. [Google Scholar] [CrossRef] [Green Version]
  61. Duan, Y.G.; Yu, C.F.; Novak, N.; Bieber, T.; Zhu, C.H.; Schuppe, H.C.; Haidl, G.; Allam, J.P. Immunodeviation towards a Th17 immune response associated with testicular damage in azoospermic men. Int. J. Androl. 2011, 34 Pt 2, e536–e545. [Google Scholar] [CrossRef] [PubMed]
  62. Jacobo, P.; Guazzone, V.A.; Jarazo-Dietrich, S.; Theas, M.S.; Lustig, L. Differential changes in CD4+ and CD8+ effector and regulatory T lymphocyte subsets in the testis of rats undergoing autoimmune orchitis. J. Reprod. Immunol. 2009, 81, 44–54. [Google Scholar] [CrossRef] [PubMed]
  63. Gong, J.; Zeng, Q.; Yu, D.; Duan, Y.G. T Lymphocytes and Testicular Immunity: A New Insight into Immune Regulation in Testes. Int. J. Mol. Sci. 2020, 22, 57. [Google Scholar] [CrossRef] [PubMed]
  64. Bhati, M.; Llamosas, E.; Jacques, D.A.; Jeffries, C.M.; Dastmalchi, S.; Ripin, N.; Nicholas, H.R.; Matthews, J.M. Interactions between LHX3- and ISL1-family LIM-homeodomain transcription factors are conserved in Caenorhabditis elegans. Sci. Rep. 2017, 7, 4579. [Google Scholar] [CrossRef] [Green Version]
  65. Bhushan, S.; Theas, M.S.; Guazzone, V.A.; Jacobo, P.; Wang, M.; Fijak, M.; Meinhardt, A.; Lustig, L. Immune Cell Subtypes and Their Function in the Testis. Front. Immunol. 2020, 11, 583304. [Google Scholar] [CrossRef]
  66. Hedger, M.P.; Meinhardt, A. Local regulation of T cell numbers and lymphocyte-inhibiting activity in the interstitial tissue of the adult rat testis. J. Reprod. Immunol. 2000, 48, 69–80. [Google Scholar] [CrossRef]
  67. Dai, Z.; Nasr, I.W.; Reel, M.; Deng, S.; Diggs, L.; Larsen, C.P.; Rothstein, D.M.; Lakkis, F.G. Impaired recall of CD8 memory T cells in immunologically privileged tissue. J. Immunol. 2005, 174, 1165–1170. [Google Scholar] [CrossRef] [Green Version]
  68. Nasr, I.W.; Wang, Y.; Gao, G.; Deng, S.; Diggs, L.; Rothstein, D.M.; Tellides, G.; Lakkis, F.G.; Dai, Z. Testicular immune privilege promotes transplantation tolerance by altering the balance between memory and regulatory T cells. J. Immunol. 2005, 174, 6161–6168. [Google Scholar] [CrossRef] [Green Version]
  69. Cheng, X.; Dai, H.; Wan, N.; Moore, Y.; Vankayalapati, R.; Dai, Z. Interaction of programmed death-1 and programmed death-1 ligand-1 contributes to testicular immune privilege. Transplantation 2009, 87, 1778–1786. [Google Scholar] [CrossRef]
  70. Zhou, R.; Wu, J.; Liu, B.; Jiang, Y.; Chen, W.; Li, J.; He, Q.; He, Z. The roles and mechanisms of Leydig cells and myoid cells in regulating spermatogenesis. Cell. Mol. Life Sci. 2019, 76, 2681–2695. [Google Scholar] [CrossRef]
  71. Jahnukainen, K.; Saari, T.; Salmi, T.T.; Pöllänen, P.; Pelliniemi, L.J. Reactions of Leydig cells and blood vessels to lymphoblastic leukemia in the rat testis. Leukemia 1995, 9, 908–914. [Google Scholar] [PubMed]
  72. Sainio-Pöllänen, S.; Sundström, J.; Erkkilä, S.; Hänninen, A.; Vainiopää, M.; Martikainen, M.; Salminen, E.; Veräjänkorva, E.; Antola, H.; Nikula, H.; et al. CD106 (VCAM-1) in testicular immunoregulation. J. Reprod. Immunol. 1997, 33, 221–238. [Google Scholar] [CrossRef]
  73. Dutta, D.; Park, I.; Guililat, H.; Sang, S.; Talapatra, A.; Hanson, L.; Mills, N.C. Ethylene dimethane sulfonate (EDS) ablation of Leydig cells in adult rat depletes testosterone resulting in epididymal sperm granuloma: Testosterone replacement prevents granuloma formation. Reprod. Biol. 2019, 19, 89–99. [Google Scholar] [CrossRef] [PubMed]
  74. Fijak, M.; Schneider, E.; Klug, J.; Bhushan, S.; Hackstein, H.; Schuler, G.; Wygrecka, M.; Gromoll, J.; Meinhardt, A. Testosterone replacement effectively inhibits the development of experimental autoimmune orchitis in rats: Evidence for a direct role of testosterone on regulatory T cell expansion. J. Immunol. 2011, 186, 5162–5172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Monsivais, D.; Matzuk, M.M.; Pangas, S.A. The TGF-β Family in the Reproductive Tract. Cold Spring Harb. Perspect. Biol. 2017, 9, a022251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Dal Secco, V.; Riccioli, A.; Padula, F.; Ziparo, E.; Filippini, A. Mouse Sertoli cells display phenotypical and functional traits of antigen-presenting cells in response to interferon gamma. Biol. Reprod. 2008, 78, 234–242. [Google Scholar] [CrossRef] [Green Version]
  77. Fallarino, F.; Luca, G.; Calvitti, M.; Mancuso, F.; Nastruzzi, C.; Fioretti, M.C.; Grohmann, U.; Becchetti, E.; Burgevin, A.; Kratzer, R.; et al. Therapy of experimental type 1 diabetes by isolated Sertoli cell xenografts alone. J. Exp. Med. 2009, 206, 2511–2526. [Google Scholar] [CrossRef] [Green Version]
  78. Alfano, M.; Ferrarese, R.; Locatelli, I.; Ventimiglia, E.; Ippolito, S.; Gallina, P.; Cesana, D.; Canducci, F.; Pagliardini, L.; Viganò, P.; et al. Testicular microbiome in azoospermic men-first evidence of the impact of an altered microenvironment. Hum. Reprod. 2018, 33, 1212–1217. [Google Scholar] [CrossRef]
  79. Molina, N.M.; Plaza-Díaz, J.; Vilchez-Vargas, R.; Sola-Leyva, A.; Vargas, E.; Mendoza-Tesarik, R.; Galán-Lázaro, M.; Mendoza-Ladrón de Guevara, N.; Tesarik, J.; Altmäe, S. Assessing the testicular sperm microbiome: A low-biomass site with abundant contamination. Reprod. Biomed. Online 2021, 43, 523–531. [Google Scholar] [CrossRef]
  80. Yang, H.; Büttner, A.; Albiol, L.; Julien, C.; Thiele, T.; Figge, C.; Kramer, I.; Kneissel, M.; Duda, G.N.; Checa, S.; et al. Cortical bone adaptation to a moderate level of mechanical loading in male Sost deficient mice. Sci. Rep. 2020, 10, 22299. [Google Scholar] [CrossRef]
  81. Lundy, S.D.; Sangwan, N.; Parekh, N.V.; Selvam, M.K.P.; Gupta, S.; McCaffrey, P.; Bessoff, K.; Vala, A.; Agarwal, A.; Sabanegh, E.S.; et al. Functional and Taxonomic Dysbiosis of the Gut, Urine, and Semen Microbiomes in Male Infertility. Eur. Urol. 2021, 79, 826–836. [Google Scholar] [CrossRef] [PubMed]
  82. Wilharm, A.; Brigas, H.C.; Sandrock, I.; Ribeiro, M.; Amado, T.; Reinhardt, A.; Demera, A.; Hoenicke, L.; Strowig, T.; Carvalho, T.; et al. Microbiota-dependent expansion of testicular IL-17-producing Vγ6+γδ T cells upon puberty promotes local tissue immune surveillance. Mucosal Immunol. 2021, 14, 242–252. [Google Scholar] [CrossRef] [PubMed]
  83. Gachet, C.; Prat, M.; Burucoa, C.; Grivard, P.; Pichon, M. Spermatic Microbiome Characteristics in Infertile Patients: Impact on Sperm Count, Mobility, and Morphology. J. Clin. Med. 2022, 11, 1505. [Google Scholar] [CrossRef] [PubMed]
  84. Charitos, I.A.; Topi, S.; Gagliano-Candela, R.; De Nitto, E.; Polimeno, L.; Montagnani, M.; Santacroce, L. The toxic effects of endocrine disrupting chemicals (EDCs) on gut microbiota: Bisphenol A (BPA). A review. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 716–727. [Google Scholar] [CrossRef]
  85. Santacroce, L.; Bottalico, L.; Topi, S.; Castellaneta, F.; Charitos, I.A. The “Scourge of the Renaissance”. A Short Review about Treponema pallidum infection. Endocr. Metab. Immune Disord. Drug Targets 2020, 20, 335–343. [Google Scholar] [CrossRef]
  86. Alfano, M.; Pederzoli, F.; Locatelli, I.; Ippolito, S.; Longhi, E.; Zerbi, P.; Ferrari, M.; Brendolan, A.; Montorsi, F.; Drago, D.; et al. Impaired testicular signaling of vitamin A and vitamin K contributes to the aberrant composition of the extracellular matrix in idiopathic germ cell aplasia. Fertil. Steril. 2019, 111, 687–698. [Google Scholar] [CrossRef] [Green Version]
  87. Zhang, S.Y.; Li, R.J.W.; Lim, Y.M.; Batchuluun, B.; Liu, H.; Waise, T.M.Z.; Lam, T.K.T. FXR in the dorsal vagal complex is sufficient and necessary for upper small intestinal microbiome-mediated changes of TCDCA to alter insulin action in rats. Gut 2021, 70, 1675–1683. [Google Scholar] [CrossRef]
  88. Santacroce, L.; Man, A.; Charitos, I.A.; Haxhirexha, K.; Topi, S. Current knowledge about the connection between health status and gut microbiota from birth to elderly. A narrative review. Front. Biosci. 2021, 26, 135–148. [Google Scholar] [CrossRef]
  89. Ding, N.; Zhang, X.; Zhang, X.D.; Jing, J.; Liu, S.S.; Mu, Y.P.; Peng, L.L.; Yan, Y.J.; Xiao, G.M.; Bi, X.Y.; et al. Impairment of spermatogenesis and sperm motility by the high-fat diet-induced dysbiosis of gut microbes. Gut 2020, 69, 1608–1619. [Google Scholar] [CrossRef] [Green Version]
  90. Tremellen, K. Gut Endotoxin Leading to a Decline IN Gonadal function (GELDING)—A novel theory for the development of late onset hypogonadism in obese men. Basic Clin. Androl. 2016, 26, 7. [Google Scholar] [CrossRef] [Green Version]
  91. Sonnex, C. Toll-like receptors and genital tract infection. Int. J. STD AIDS 2010, 21, 153–157. [Google Scholar] [CrossRef] [PubMed]
  92. Wegierska, A.E.; Charitos, I.A.; Topi, S.; Potenza, M.A.; Montagnani, M.; Santacroce, L. The Connection between Physical Exercise and Gut Microbiota, Implications for Competitive Sports Athletes. Sports Med. 2022; ahead of print. [Google Scholar] [CrossRef]
  93. Sarkar, O.; Bahrainwala, J.; Chandrasekaran, S.; Kothari, S.; Mathur, P.P.; Agarwal, A. Impact of inflammation on male fertility. Front. Biosci. 2011, 3, 89–95. [Google Scholar] [CrossRef] [Green Version]
  94. Alvarez, J.G.; Sharma, R.K.; Ollero, M.; Saleh, R.A.; Lopez, M.C.; Thomas, A.J., Jr.; Evenson, D.P.; Agarwal, A. Increased DNA damage in sperm from leukocytospermic semen samples as determined by the sperm chromatin structure assay. Fertil. Steril. 2002, 78, 319–329. [Google Scholar] [CrossRef]
  95. Zheng, W.; Wu, H.; Liu, C.; Yan, Q.; Wang, T.; Wu, P.; Liu, X.; Jiang, Y.; Zhan, S. Identification of COVID-19 and Dengue Host Factor Interaction Networks Based on Integrative Bioinformatics Analyses. Front. Immunol. 2021, 12, 707287. [Google Scholar] [CrossRef] [PubMed]
  96. Andreasen, A.S.; Larsen, N.; Pedersen-Skovsgaard, T.; Berg, R.M.; Møller, K.; Svendsen, K.D.; Jakobsen, M.; Pedersen, B.K. Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic inflammatory response in human subjects. Br. J. Nutr. 2010, 104, 1831–1838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Hawkesworth, S.; Moore, S.E.; Fulford, A.J.; Barclay, G.R.; Darboe, A.A.; Mark, H.; Nyan, O.A.; Prentice, A.M. Evidence for metabolic endotoxemia in obese and diabetic Gambian women. Nutr. Diabetes 2013, 3, e83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Man, A.; Ciurea, C.N.; Pasaroiu, D.; Savin, A.I.; Toma, F.; Sular, F.; Santacroce, L.; Mare, A. New perspectives on the nutritional factors influencing growth rate of Candida albicans in diabetics. An in vitro study. MemóriasDo Inst. Oswaldo Cruz 2017, 112, 587–592. [Google Scholar] [CrossRef]
  99. Zhu, Y.Z.; Sun, H.; Fu, Y.; Wang, J.; Song, M.; Li, M.; Li, Y.F.; Miao, L.G. Effects of sub-chronic aluminum chloride on spermatogenesis and testicular enzymatic activity in male rats. Life Sci. 2014, 102, 36–40. [Google Scholar] [CrossRef]
  100. Pitteloud, N.; Hardin, M.; Dwyer, A.A.; Valassi, E.; Yialamas, M.; Elahi, D.; Hayes, F.J. Increasing insulin resistance is associated with a decrease in Leydig cell testosterone secretion in men. J. Clin. Endocrinol. Metab. 2005, 90, 2636–2641. [Google Scholar] [CrossRef] [Green Version]
  101. Monteiro, C.; Marques, P.I.; Cavadas, B.; Damião, I.; Almeida, V.; Barros, N.; Barros, A.; Carvalho, F.; Gomes, S.; Seixas, S. Characterization of microbiota in male infertility cases uncovers differences in seminal hyperviscosity and oligoasthenoteratozoospermia possibly correlated with increased prevalence of infectious bacteria. Am. J. Reprod. Immunol. 2018, 79, e12838. [Google Scholar] [CrossRef]
  102. Zhao, L.; Yao, C.; Xing, X.; Jing, T.; Li, P.; Zhu, Z.; Yang, C.; Zhai, J.; Tian, R.; Chen, H.; et al. Single-cell analysis of developing and azoospermia human testicles reveals central role of Sertoli cells. Nat. Commun. 2020, 11, 5683. [Google Scholar] [CrossRef] [PubMed]
  103. Mändar, R.; Türk, S.; Korrovits, P.; Ausmees, K.; Punab, M. Impact of sexual debut on culturable human seminal microbiota. Andrology 2018, 6, 510–512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Weng, S.L.; Chiu, C.M.; Lin, F.M.; Huang, W.C.; Liang, C.; Yang, T.; Yang, T.L.; Liu, C.Y.; Wu, W.Y.; Chang, Y.A.; et al. Bacterial communities in semen from men of infertile couples: Metagenomic sequencing reveals relationships of seminal microbiota to semen quality. PLoS ONE 2014, 9, e110152. [Google Scholar] [CrossRef] [Green Version]
  105. Mändar, R.; Punab, M.; Korrovits, P.; Türk, S.; Ausmees, K.; Lapp, E.; Preem, J.K.; Oopkaup, K.; Salumets, A.; Truu, J. Seminal microbiome in men with and without prostatitis. Int. J. Urol. 2017, 24, 211–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. McConaghy, J.R.; Panchal, B. Epididymitis: An Overview. Am. Fam. Physician 2016, 94, 723–726. [Google Scholar] [PubMed]
  107. Fijak, M.; Pilatz, A.; Hedger, M.P.; Nicolas, N.; Bhushan, S.; Michel, V.; Tung, K.S.K.; Schuppe, H.C.; Meinhardt, A. Infectious, inflammatory and ‘autoimmune’ male factor infertility: How do rodent models inform clinical practice? Hum. Reprod. Update 2018, 24, 416–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. de Kretser, D.M.; Huidobro, C.; Southwick, G.J.; Temple-Smith, P.D. The role of the epididymis in human infertility. J. Reprod. Fertil. Suppl. 1998, 53, 271–275. [Google Scholar] [PubMed]
  109. Guiton, R.; Henry-Berger, J.; Drevet, J.R. The immunobiology of the mammalian epididymis: The black box is now open! Basic Clin. Androl. 2013, 23, 8. [Google Scholar] [CrossRef] [Green Version]
  110. Pilatz, A.; Lochnit, G.; Karnati, S.; Paradowska-Dogan, A.; Lang, T.; Schultheiss, D.; Schuppe, H.C.; Hossain, H.; Baumgart-Vogt, E.; Weidner, W.; et al. Acute epididymitis induces alterations in sperm protein composition. Fertil. Steril. 2014, 101, 1609–1617.e5. [Google Scholar] [CrossRef]
  111. Schirinzi, A.; Cazzolla, A.P.; Mascolo, E.; Palmieri, G.; Pesce, F.; Gesualdo, L.; Santacroce, L.; Ballini, A.; Lovero, R.; Di Serio, F. Determination of the Upper Reference Limit of Human Epididymis Secretory Protein 4 (HE4) in Healthy Male Individuals and Correlation with Renal and Fertility Markers. Endocr. Metab. Immune Disord. Drug Targets 2021, 21, 912–918. [Google Scholar] [CrossRef]
  112. Stammler, A.; Hau, T.; Bhushan, S.; Meinhardt, A.; Jonigk, D.; Lippmann, T.; Pilatz, A.; Schneider-Hüther, I.; Middendorff, R. Epididymitis: Ascending infection restricted by segmental boundaries. Hum. Reprod. 2015, 30, 1557–1565. [Google Scholar] [CrossRef] [Green Version]
  113. Silva, E.J.R.; Ribeiro, C.M.; Mirim, A.F.M.; Silva, A.A.S.; Romano, R.M.; Hallak, J.; Avellar, M.C.W. Lipopolysaccharide and lipotheicoic acid differentially modulate epididymal cytokine and chemokine profiles and sperm parameters in experimental acute epididymitis. Sci. Rep. 2018, 8, 103. [Google Scholar] [CrossRef] [PubMed]
  114. Mutoji, K.; Singh, A.; Nguyen, T.; Gildersleeve, H.; Kaucher, A.V.; Oatley, M.J.; Oatley, J.M.; Velte, E.K.; Geyer, C.B.; Cheng, K.; et al. TSPAN8 Expression Distinguishes Spermatogonial Stem Cells in the Prepubertal Mouse Testis. Biol. Reprod. 2016, 95, 117. [Google Scholar] [CrossRef] [PubMed]
  115. Biswas, B.; Bhushan, S.; Rajesh, A.; Suraj, S.K.; Lu, Y.; Meinhardt, A.; Yenugu, S. Uropathogenic Escherichia coli (UPEC) induced antimicrobial gene expression in the male reproductive tract of rat: Evaluation of the potential of Defensin 21 to limit infection. Andrology 2015, 3, 368–375. [Google Scholar] [CrossRef] [PubMed]
  116. Bryan, E.R.; Kollipara, A.; Trim, L.K.; Armitage, C.W.; Carey, A.J.; Mihalas, B.; Redgrove, K.A.; McLaughlin, E.A.; Beagley, K.W. Hematogenous dissemination of Chlamydia muridarum from the urethra in macrophages causes testicular infection and sperm DNA damage. Biol. Reprod. 2019, 101, 748–759. [Google Scholar] [CrossRef] [PubMed]
  117. Mackern-Oberti, J.P.; Motrich, R.D.; Breser, M.L.; Sánchez, L.R.; Cuffini, C.; Rivero, V.E. Chlamydia trachomatis infection of the male genital tract: An update. J. Reprod. Immunol. 2013, 100, 37–53. [Google Scholar] [CrossRef] [PubMed]
  118. Emerson, C.; Dinsmore, W.W.; Quah, S.P. Are we missing mumps epididymo-orchitis? Int. J. STD AIDS 2007, 18, 341–342. [Google Scholar] [CrossRef]
  119. Giagulli, V.A.; Guastamacchia, E.; Magrone, T.; Jirillo, E.; Lisco, G.; De Pergola, G.; Triggiani, V. Worse progression of COVID-19 in men: Is testosterone a key factor? Andrology 2021, 9, 53–64. [Google Scholar] [CrossRef]
  120. Santacroce, L.; Charitos, I.A.; Carretta, D.M.; De Nitto, E.; Lovero, R. The human coronaviruses (HCoVs) and the molecular mechanisms of SARS-CoV-2 infection. J. Mol. Med. 2021, 99, 93–106. [Google Scholar] [CrossRef]
  121. Schirinzi, A.; Cazzolla, A.P.; Lovero, R.; Lo Muzio, L.; Testa, N.F.; Ciavarella, D.; Palmieri, G.; Pozzessere, P.; Procacci, V.; Di Serio, F.; et al. New Insights in Laboratory Testing for COVID-19 Patients: Looking for the Role and Predictive Value of Human epididymis secretory protein 4 (HE4) and the Innate Immunity of the Oral Cavity and Respiratory Tract. Microorganisms 2020, 8, 1718. [Google Scholar] [CrossRef]
  122. Vabret, N.; Britton, G.J.; Gruber, C.; Hegde, S.; Kim, J.; Kuksin, M.; Levantovsky, R.; Malle, L.; Moreira, A.; Park, M.D.; et al. Immunology of COVID-19: Current State of the Science. Immunity 2020, 52, 910–941. [Google Scholar] [CrossRef] [PubMed]
  123. Yang, M.; Chen, S.; Huang, B.; Zhong, J.M.; Su, H.; Chen, Y.J.; Cao, Q.; Ma, L.; He, J.; Li, X.F.; et al. Pathological Findings in the Testes of COVID-19 Patients: Clinical Implications. Eur. Urol. Focus 2020, 6, 1124–1129. [Google Scholar] [CrossRef]
  124. Ferramosca, A.; Zara, V. Diet and Male Fertility: The Impact of Nutrients and Antioxidants on Sperm Energetic Metabolism. Int. J. Mol. Sci. 2022, 23, 2542. [Google Scholar] [CrossRef]
  125. Suliga, E.; Głuszek, S. The relationship between diet, energy balance and fertility in men. Int. J. Vitam. Nutr. Res. 2020, 90, 514–526. [Google Scholar] [CrossRef] [PubMed]
  126. Guasch-Ferré, M.; Willett, W.C. The Mediterranean diet and health: A comprehensive overview. J. Intern. Med. 2021, 290, 549–566. [Google Scholar] [CrossRef] [PubMed]
  127. Bachir, B.G.; Jarvi, K. Infectious, inflammatory, and immunologic conditions resulting in male infertility. Urol. Clin. N. Am. 2014, 41, 67–81. [Google Scholar] [CrossRef]
  128. Morielli, T.; O’Flaherty, C. Oxidative stress impairs function and increases redox protein modifications in human spermatozoa. Reproduction 2015, 149, 113–123. [Google Scholar] [CrossRef] [Green Version]
  129. Ferramosca, A.; Conte, A.; Moscatelli, N.; Zara, V. A high-fat diet negatively affects rat sperm mitochondrial respiration. Andrology 2016, 4, 520–525. [Google Scholar] [CrossRef] [Green Version]
  130. Casas, R.; Estruch, R.; Sacanella, E. The Protective Effects of Extra Virgin Olive Oil on Immune-mediated Inflammatory Responses. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 23–35. [Google Scholar] [CrossRef]
  131. Magrone, T.; Spagnoletta, A.; Salvatore, R.; Magrone, M.; Dentamaro, F.; Russo, M.A.; Difonzo, G.; Summo, C.; Caponio, F.; Jirillo, E. Olive Leaf Extracts Act as Modulators of the Human Immune Response. Endocr. Metab. Immune Disord. Drug Targets 2018, 18, 85–93. [Google Scholar] [CrossRef]
  132. Müller, A.K.; Albrecht, F.; Rohrer, C.; Koeberle, A.; Werz, O.; Schlörmann, W.; Glei, M.; Lorkowski, S.; Wallert, M. Olive Oil Extracts and Oleic Acid Attenuate the LPS-Induced Inflammatory Response in Murine RAW264.7 Macrophages but Induce the Release of Prostaglandin E2. Nutrients. 2021, 13, 4437. [Google Scholar] [CrossRef] [PubMed]
  133. Santacroce, L.; Inchingolo, F.; Topi, S.; Del Prete, R.; Di Cosola, M.; Charitos, I.A.; Montagnani, M. Potential beneficial role of probiotics on the outcome of COVID-19 patients: An evolving perspective. Diabetes Metab. Syndr. 2021, 15, 295–301. [Google Scholar] [CrossRef] [PubMed]
  134. Jones, S.E.; Versalovic, J. Probiotic Lactobacillus reuteri biofilms produce antimicrobial and anti-inflammatory factors. BMC Microbiol. 2009, 9, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Yan, F.; Cao, H.; Cover, T.L.; Whitehead, R.; Washington, M.K.; Polk, D.B. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 2007, 132, 562–575. [Google Scholar] [CrossRef] [Green Version]
  136. Valcarce, D.G.; Genovés, S.; Riesco, M.F.; Martorell, P.; Herráez, M.P.; Ramón, D.; Robles, V. Probiotic administration improves sperm quality in asthenozoospermic human donors. Benef. Microbes 2017, 8, 193–206. [Google Scholar] [CrossRef]
  137. Pacifici, L.; Santacroce, L.; Dipalma, G.; Haxhirexha, K.; Topi, S.; Cantore, S.; Altini, V.; Pacifici, A.; De Vito, D.; Pettini, F.; et al. Gender medicine: The impact of probiotics on male patients. Clin. Ter. 2021, 171, e8–e15. [Google Scholar] [CrossRef]
  138. Abbasi, B.; Abbasi, H.; Niroumand, H. Synbiotic (FamiLact) administration in idiopathic male infertility enhances sperm quality, DNA integrity, and chromatin status: A triple-blinded randomized clinical trial. Int. J. Reprod. Biomed. 2021, 19, 235–244. [Google Scholar] [CrossRef]
  139. Dardmeh, F.; Alipour, H.; Gazerani, P.; van der Horst, G.; Brandsborg, E.; Nielsen, H.I. Lactobacillus rhamnosus PB01 (DSM 14870) supplementation affects markers of sperm kinematic parameters in a diet-induced obesity mice model. PLoS ONE 2017, 12, e0185964. [Google Scholar] [CrossRef] [Green Version]
  140. Chen, X.L.; Gong, L.Z.; Xu, J.X. Antioxidative activity and protective effect of probiotics against high-fat diet-induced sperm damage in rats. Animal 2013, 7, 287–292. [Google Scholar] [CrossRef] [Green Version]
  141. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the gut microbiota in nutrition and health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef] [Green Version]
  142. Di Domenico, M.; Ballini, A.; Boccellino, M.; Scacco, S.; Lovero, R.; Charitos, I.A.; Santacroce, L. The Intestinal Microbiota May Be a Potential Theranostic Tool for Personalized Medicine. J. Pers. Med. 2022, 12, 523. [Google Scholar] [CrossRef] [PubMed]
  143. Rodrigues, L.E.; Kishibe, M.M.; Keller, R.; Caetano, H.R.D.S.; Rufino, M.N.; Sanches, O.C.; Giometti, I.C.; Giuffrida, R.; Bremer-Neto, H. Prebiotics mannan-oligosaccharides accelerate sexual maturity in rats: A randomized preclinical study. Vet. World 2021, 14, 1210–1219. [Google Scholar] [CrossRef] [PubMed]
  144. Agarwal, A.; Parekh, N.; Panner Selvam, M.K.; Henkel, R.; Shah, R.; Homa, S.T.; Ramasamy, R.; Ko, E.; Tremellen, K.; Esteves, S.; et al. Male Oxidative Stress Infertility (MOSI): Proposed Terminology and Clinical Practice Guidelines for Management of Idiopathic Male Infertility. World J. Men’s Health 2019, 37, 296–312. [Google Scholar] [CrossRef] [PubMed]
  145. Aly, H.A.; El-Beshbishy, H.A.; Banjar, Z.M. Mitochondrial dysfunction induced impairment of spermatogenesis in LPS-treated rats: Modulatory role of lycopene. Eur. J. Pharmacol. 2012, 677, 31–38. [Google Scholar] [CrossRef] [PubMed]
  146. Schisterman, E.F.; Sjaarda, L.A.; Clemons, T.; Carrell, D.T.; Perkins, N.J.; Johnstone, E.; Lamb, D.; Chaney, K.; Van Voorhis, B.J.; Ryan, G.; et al. Effect of Folic Acid and Zinc Supplementation in Men on Semen Quality and Live Birth among Couples Undergoing Infertility Treatment: A Randomized Clinical Trial. JAMA 2020, 323, 35–48. [Google Scholar] [CrossRef]
  147. Steiner, A.Z.; Hansen, K.R.; Barnhart, K.T.; Cedars, M.I.; Legro, R.S.; Diamond, M.P.; Krawetz, S.A.; Usadi, R.; Baker, V.L.; Coward, R.M.; et al. The effect of antioxidants on male factor infertility: The Males, Antioxidants, and Infertility (MOXI) randomized clinical trial. Fertil. Steril. 2020, 113, 552–560.e3. [Google Scholar] [CrossRef]
  148. Magrone, T.; Magrone, M.; Russo, M.A.; Jirillo, E. Recent Advances on the Anti-Inflammatory and Antioxidant Properties of Red Grape Polyphenols: In Vitro and In Vivo Studies. Antioxidants 2019, 9, 35. [Google Scholar] [CrossRef] [Green Version]
  149. Ibrahim, M.A.A.; Abdeljawaad, K.A.A.; Abdelrahman, A.H.M.; Jaragh-Alhadad, L.A.; Oraby, H.F.; Elkaeed, E.B.; Mekhemer, G.A.H.; Gabr, G.A.; Shawky, A.M.; Sidhom, P.A.; et al. Exploring Natural Product Activity and Species Source Candidates for Hunting ABCB1 Transporter Inhibitors: An In Silico Drug Discovery Study. Molecules 2022, 27, 3104. [Google Scholar] [CrossRef]
  150. Juan, M.E.; González-Pons, E.; Munuera, T.; Ballester, J.; Rodríguez-Gil, J.E.; Planas, J.M. trans-Resveratrol, a natural antioxidant from grapes, increases sperm output in healthy rats. J. Nutr. 2005, 135, 757–760. [Google Scholar] [CrossRef]
  151. Pasquariello, R.; Verdile, N.; Brevini, T.A.L.; Gandolfi, F.; Boiti, C.; Zerani, M.; Maranesi, M. The Role of Resveratrol in Mammalian Reproduction. Molecules 2020, 25, 4554. [Google Scholar] [CrossRef]
  152. Magrone, T.; Jirillo, E. The New Era of Nutraceuticals: Beneficial Effects of Polyphenols in Various Experimental and Clinical Settings. Curr. Pharm. Des. 2018, 24, 5229–5231. [Google Scholar] [CrossRef] [PubMed]
  153. Ranawat, P.; Pathak, C.M.; Khanduja, K.L. A new perspective on the quercetin paradox in male reproductive dysfunction. Phytother. Res. 2013, 27, 802–810. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structure of the testis. The structure of the testis consists of the epithelium of the seminiferous tubules and the intestitium. In turn, the interstium harbors Leydig cells, immune cells and lymphatic vessels.
Figure 1. Structure of the testis. The structure of the testis consists of the epithelium of the seminiferous tubules and the intestitium. In turn, the interstium harbors Leydig cells, immune cells and lymphatic vessels.
Jpm 12 01335 g001
Figure 2. Blood–testis barrier composition. The blood–testis barrier, besides in gap junctions, desmosome-like junctions and tight junctions, has Sertoli cells (SCs). SCs participate in the nutrition and growth of germ cells, maintenance of the immune privilege and clearance of autoantigens and apoptotic cells via phagocytosis.
Figure 2. Blood–testis barrier composition. The blood–testis barrier, besides in gap junctions, desmosome-like junctions and tight junctions, has Sertoli cells (SCs). SCs participate in the nutrition and growth of germ cells, maintenance of the immune privilege and clearance of autoantigens and apoptotic cells via phagocytosis.
Jpm 12 01335 g002
Figure 3. Macrophages, Dendritic cells and T cells sustain the testicular immune response. ED-2 macrophages, DCs and Treg cells keep immune privilege. Conversely, ED-1 macrophages and Th17 cells take part inchronic orchitis progression. Leydig cells attenuate autoimmune orchitis via release oftestosterone. SCs maintain the immune privilege via activation of tolerogenic DCs and Treg cells.
Figure 3. Macrophages, Dendritic cells and T cells sustain the testicular immune response. ED-2 macrophages, DCs and Treg cells keep immune privilege. Conversely, ED-1 macrophages and Th17 cells take part inchronic orchitis progression. Leydig cells attenuate autoimmune orchitis via release oftestosterone. SCs maintain the immune privilege via activation of tolerogenic DCs and Treg cells.
Jpm 12 01335 g003
Figure 4. The gut–testis axis. The existence of the gut–testis axis is supported by different evidence. Fecal transplantation from high-fat diet mice to normal mice accounts for endotoxemia occurrence and altered spermatogenesis. In turn, endotoxemia abrogates synthesis of testosterone from Leydig cells, thus reducing the number of spermatozoa with increased release of pro-inflammatory cytokines. Insulin resistance as a result of an altered intestinal permeability leads to a reduced spermatogenesis.
Figure 4. The gut–testis axis. The existence of the gut–testis axis is supported by different evidence. Fecal transplantation from high-fat diet mice to normal mice accounts for endotoxemia occurrence and altered spermatogenesis. In turn, endotoxemia abrogates synthesis of testosterone from Leydig cells, thus reducing the number of spermatozoa with increased release of pro-inflammatory cytokines. Insulin resistance as a result of an altered intestinal permeability leads to a reduced spermatogenesis.
Jpm 12 01335 g004
Figure 5. Microbial invasion of the testis. UPEC and C. trachomatis damage the male genital tract via production of pro-inflammatory cytokines and nitric oxide, with tissue scarring. COVID-19 infection is characterized by a reduction of Leydig cells, infiltration of T and B cells and elevated expression of ACE-2.
Figure 5. Microbial invasion of the testis. UPEC and C. trachomatis damage the male genital tract via production of pro-inflammatory cytokines and nitric oxide, with tissue scarring. COVID-19 infection is characterized by a reduction of Leydig cells, infiltration of T and B cells and elevated expression of ACE-2.
Jpm 12 01335 g005
Table 1. Natural products for the correction of testicular dysbiosis. Both probiotics and synbiotics are able to improve sperm quality and motility in male infertility. In rats, manno-oligosaccharides (prebiotics) promote sexual maturity. Polyphenols exert both antioxidant and pro-oxidant activities. Among antioxidants, vitamin C and vitamin E are able to reduce ROS generation and improve sperm mobility and DNA integrity. Lycopene enhances sperm performance via lipid peroxidation on mitochondrial membranes.
Table 1. Natural products for the correction of testicular dysbiosis. Both probiotics and synbiotics are able to improve sperm quality and motility in male infertility. In rats, manno-oligosaccharides (prebiotics) promote sexual maturity. Polyphenols exert both antioxidant and pro-oxidant activities. Among antioxidants, vitamin C and vitamin E are able to reduce ROS generation and improve sperm mobility and DNA integrity. Lycopene enhances sperm performance via lipid peroxidation on mitochondrial membranes.
Natural Products against Testicular Dysbiosis
PROBIOTICSPREBIOTICSPOLYPHENOLSANTI-OXIDANTS
  • In astenoazoospermic human donors
  • In idiopathic male infertility symbiotic-induced
  • Lactobacillus-mediated improvement of enhancement of sperm quality and reduction of sperm motility and DNA fragmentation free radicals in the semen
Mann-oligosaccharides acceleration of sexual maturity in ratsControversial results with quercetin and resveratrol since they are endowed with both anti-oxidants and pro-oxidants activities
  • Vitamin C and vitamin E -> reduction of ROS and improvement of sperm mobility and DNA integrity
  • Lycopene -> modulation of lipid peroxidation on mitochondrial membrane
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Santacroce, L.; Imbimbo, C.; Ballini, A.; Crocetto, F.; Scacco, S.; Cantore, S.; Di Zazzo, E.; Colella, M.; Jirillo, E. Testicular Immunity and Its Connection with the Microbiota. Physiological and Clinical Implications in the Light of Personalized Medicine. J. Pers. Med. 2022, 12, 1335. https://doi.org/10.3390/jpm12081335

AMA Style

Santacroce L, Imbimbo C, Ballini A, Crocetto F, Scacco S, Cantore S, Di Zazzo E, Colella M, Jirillo E. Testicular Immunity and Its Connection with the Microbiota. Physiological and Clinical Implications in the Light of Personalized Medicine. Journal of Personalized Medicine. 2022; 12(8):1335. https://doi.org/10.3390/jpm12081335

Chicago/Turabian Style

Santacroce, Luigi, Ciro Imbimbo, Andrea Ballini, Felice Crocetto, Salvatore Scacco, Stefania Cantore, Erika Di Zazzo, Marica Colella, and Emilio Jirillo. 2022. "Testicular Immunity and Its Connection with the Microbiota. Physiological and Clinical Implications in the Light of Personalized Medicine" Journal of Personalized Medicine 12, no. 8: 1335. https://doi.org/10.3390/jpm12081335

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

Article Metrics

Back to TopTop