Next Article in Journal
Pediatric Patients with Sitosterolemia: Next-Generation Sequencing and Biochemical Examination in Clinical Practice
Previous Article in Journal
The Impact of Metabolic Factors and Lipid-Lowering Drugs on Common Bile Duct Stone Recurrence after Endoscopic Sphincterotomy with Following Cholecystectomy
Previous Article in Special Issue
Spatiotemporal Trends and Age–Period–Cohort Analysis for the Burden of Endometriosis-Related Infertility: An Analysis of the Global Burden of Disease Study 2019
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbial Dysbiosis and Male Infertility: Understanding the Impact and Exploring Therapeutic Interventions

by
Aris Kaltsas
1,
Athanasios Zachariou
1,
Eleftheria Markou
2,
Fotios Dimitriadis
3,
Nikolaos Sofikitis
1 and
Spyridon Pournaras
4,*
1
Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
2
Department of Microbiology, University Hospital of Ioannina, 45500 Ioannina, Greece
3
Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
4
Clinical Microbiology Laboratory, Attikon General University Hospital of Athens, 12462 Athens, Greece
*
Author to whom correspondence should be addressed.
J. Pers. Med. 2023, 13(10), 1491; https://doi.org/10.3390/jpm13101491
Submission received: 29 August 2023 / Revised: 10 October 2023 / Accepted: 12 October 2023 / Published: 13 October 2023
(This article belongs to the Special Issue Precision Medicine in Reproductive Health)

Abstract

:
The human microbiota in the genital tract is pivotal for maintaining fertility, but its disruption can lead to male infertility. This study examines the relationship between microbial dysbiosis and male infertility, underscoring the promise of precision medicine in this field. Through a comprehensive review, this research indicates microbial signatures associated with male infertility, such as altered bacterial diversity, the dominance of pathogenic species, and imbalances in the genital microbiome. Key mechanisms linking microbial dysbiosis to infertility include inflammation, oxidative stress, and sperm structural deterioration. Emerging strategies like targeted antimicrobial therapies, probiotics, prebiotics, and fecal microbiota transplantation have shown potential in adjusting the genital microbiota to enhance male fertility. Notably, the application of precision medicine, which customizes treatments based on individual microbial profiles and specific causes of infertility, emerges as a promising approach to enhance treatment outcomes. Ultimately, microbial dysbiosis is intricately linked to male infertility, and embracing personalized treatment strategies rooted in precision medicine principles could be the way forward in addressing infertility associated with microbial factors.

1. Introduction

Infertility, as defined by the World Health Organization, is the inability of couples to conceive after engaging in regular sexual intercourse for over one year without the use of contraception [1]. This condition, which includes instances where a full pregnancy is not sustained for at least two consecutive natural trials, has emerged as a pressing public health issue globally. Couples grappling with infertility often face emotional, social, and financial challenges, further underscoring the need for effective interventions [2].
Recent literature presents varied findings regarding male fertility trends. Some studies have documented a decline in semen parameters [3,4,5], while others have reported stable [6,7,8] or even improved semen quality over time [9,10,11]. For instance, a comprehensive study spanning from 1973 to 2011 observed a significant decline of 50–60% in sperm counts in Western nations [12]. These varied findings underscore the multifaceted nature of male fertility and the potential influence of factors such as obesity, diet patterns, and environmental toxins [13].
The human microbiome, an intricate ecosystem comprising bacteria, viruses, fungi, and protozoa, resides both internally and externally in the human body [14]. Recent advancements in the study of the human microbiome have unveiled its significant role in various physiological processes, including nutrient absorption and immune system development. Moreover, alterations in the human microbiome have been linked to the onset, progression, and management of numerous diseases [15,16,17,18].
The microbiome’s influence extends to the male reproductive system. Historically, the absence of microbial growth in male reproductive tract samples was interpreted as an absence of microbial infection. However, contemporary research has unveiled the presence of a microbiome in the male reproductive system, including seminal fluid and urine [19].
Emerging evidence suggests a pivotal role of the microbiome in male fertility [20]. Dysbiosis, an imbalance in the microbial community, has been linked to male conditions such as oligospermia [21,22], asthenospermia [22,23,24], and teratospermia [25,26]. Furthermore, recent studies have highlighted the connection between oral and gut microbiomes and their influence on systemic health, including reproductive health [27,28,29].
Precision medicine, which emphasizes tailoring medical interventions based on individual characteristics, offer a promising avenue for addressing microbial dysbiosis and its implications for infertility [30]. By integrating genetic, environmental, and microbial factors, precision medicine aims to provide more targeted diagnostics, preventions, screening, and treatments [31,32].
This review aims to provide a comprehensive understanding of the microbial factors associated with male infertility. After detailing the mechanisms and diagnostic methods, the manuscript delves into current treatment strategies and concludes with recommendations for future research directions. By exploring the role of the microbiome in male infertility, this review seeks to pave the way for innovative treatment approaches. Harnessing the insights from precision medicine and understanding the impact of microbial factors on fertility can usher in a new era in reproductive medicine, offering hope and solutions for couples facing infertility challenges.

2. Microbiome and Male Reproductive Health

2.1. Introduction to the Human Microbiome and Its Significance

The human microbiome refers to the complex ecosystem of bacteria, viruses, fungi, and protozoa living on and in the human body [33]. This intricate assembly of microorganisms, which has co-evolved alongside humans, is pivotal to our well-being, playing a crucial role in various physiological processes [34]. Each individual’s microbiota is distinct, with specific species colonizing body sites [34,35]. For instance, the gastrointestinal tract, encompassing the mouth, esophagus, stomach, small intestine, and large intestine, teems with a diverse array of microorganisms. The oral cavity, in particular, is home to a notably diverse microbiota, with hundreds of species present [36]. The skin is populated by a rich assortment of bacteria, fungi, and viruses [37]. Meanwhile, the respiratory system, which includes the nasal cavity and lungs, boasts its own unique microbiome [38]. The urogenital tract, comprising the vagina and urinary system, also harbors distinct microbial communities [39,40].
These microbial communities are integral to maintaining homeostasis in biochemical, metabolic, and immune systems [41,42]. For instance, the gastrointestinal tract, known for its rich microbial population, is essential for digestion, nutrient absorption, and immune system development [42]. Short-chained fatty acids produced in the intestines significantly contribute to these processes [43,44]. The Human Microbiome Project, initiated in 2007, aims to consolidate genetic data from diverse human microbiomes to understand the relationship between microbiome alterations and various diseases [45,46]. This project underscores the profound influence of the microbiome on human health and its role in maintaining equilibrium [34]. Research has indicated that changes in the lung microbiome might influence the progression of conditions like chronic obstructive pulmonary disease (COPD) [34]. Additionally, interactions between the microbiota and dietary components, such as polyphenols found in plant-derived foods, can impact the gut microbiota’s composition and function, further influencing human health [42].
Factors like genetics, diet, lifestyle, and environmental exposure shape the gastrointestinal microbiome [47]. Dysbiosis may lead to metabolic issues such as irritable bowel syndrome (IBS) or allergies and reproductive disorders such as polycystic ovary syndrome (PCOS) or endometriosis [48,49]. The vaginal microbiota has also been related to preterm delivery and other reproductive issues [50,51]. The male reproductive excretory ducts also have their distinct microbiota, which can influence male reproductive health [50,52]. Semen may contain microorganisms that can be transmitted between sexual partners [52].
In conclusion, the microbiota in various body sites, including gut and reproductive tracts, profoundly impacts human health and reproductive outcomes [33,53]. Further research is essential to understand the complex interactions between human microbiota and reproductive health and potential therapeutic interventions [54,55]. With the rise of precision medicine, there is potential for interventions tailored to individual microbial profiles, offering customized therapeutic approaches to male infertility issues based on a person’s unique microbiome [56].

2.2. Mechanism of Action of the Microbiome Elements in Healthy Conditions

Bacteria, a significant component of the microbiome, fulfill several functions that are essential for overall well-being. They play a pivotal role in digestion by enzymatically breaking down complex carbohydrates into more easily absorbable molecules [57,58]. This activity is particularly vital within the gastrointestinal tract, where a symbiotic relationship exists between bacteria and the fermentation of dietary fibers. This fermentation results in the production of short-chain fatty acids, which serve as a crucial energy source for the host organism [57,58]. Moreover, certain bacterial strains in the gastrointestinal tract are instrumental in the biosynthesis of essential vitamins, including vitamin K and several B vitamins, vital to human health [57]. Additionally, the gut microbiota plays an indispensable role in immune system regulation, ensuring a balanced immunological response [59].
Viruses, specifically bacteriophages, also exert a profound influence on the microbiome. They can infect and lyse specific bacterial cells, thereby regulating bacterial populations and maintaining microbial balance [57]. As natural predators of bacteria, bacteriophages are pivotal in controlling bacterial population size and diversity. Furthermore, they facilitate gene transfer between bacteria, playing a significant role in bacterial adaptation and evolution [57]. This gene transfer can introduce new functionalities, influencing the overall dynamics of the microbiome.
Fungi within the microbiome contribute significantly to overall health. Some fungi assist in breaking down complex compounds, such as certain fibers, that are indigestible by bacteria [57]. By participating in this process, fungi complement the broader digestive system. Moreover, by occupying specific ecological niches within the body, fungi can prevent the overgrowth of harmful pathogens through competitive inhibition, ensuring a balanced microbial ecosystem and thwarting the establishment of potentially harmful bacteria [57].
Protozoa, another component of the microbiome, play a role in maintaining overall health. Some protozoa in the gastrointestinal tract aid in digestion by breaking down specific compounds, enhancing nutrient absorption [57]. Additionally, the presence of protozoa can stimulate the immune system, bolstering its ability to recognize and combat foreign pathogens [57]. This immunological stimulation is vital for maintaining robust immune responses and protecting the body against various diseases.
In a state of good health, these microorganisms work in harmony, ensuring the optimal functioning of various physiological processes. They are integral to numerous physiological activities, including digestion, nutrient absorption, immune system regulation, and the prevention of pathogenic overgrowth. The balance and diversity of these microbial communities are paramount for health maintenance and disease prevention.

2.3. Microbiota in the Male Reproductive System

The male reproductive system, once perceived as largely sterile, is now recognized as a complex mosaic of microbial communities. The male genital tract, including regions like the urethra and the coronal sulcus, harbors distinct microbial communities [60]. Predominant bacterial genera in the male reproductive tract include Corynebacterium, Streptococcus, and Staphylococcus [61,62]. The microbial composition within this tract exhibits individual variations [61], influenced by factors such as sexual behavior, hygiene practices, and the presence of sexually transmitted infections (STIs) [63].
A balanced and diverse microbiota within the male genital system is essential for optimal reproductive health [61]. Bacteria like Escherichia coli and Ureaplasma urealyticum have been associated with chronic prostatitis, leading to inflammation and potential damage to the male reproductive system [64]. Furthermore, the microbial composition within the male urinary tract can influence susceptibility to STIs and the risk of transmission to sexual partners [60]. Dysbiosis in this region can be a contributing factor to various reproductive challenges [50,62].
Delving deeper, the testes, traditionally viewed as devoid of microbes, have been revealed to contain a low-abundance microbiota. The epididymis, a duct where sperm mature, also houses specific bacterial communities, the significance of which is still under exploration. The vas deferens, though less studied, might contain microbes influencing its function. Seminal vesicles and the prostate, glands vital for seminal fluid production, have their microbiota, which can impact the fluid’s composition and health. The urethra, connecting to the external environment, boasts a diverse microbiota influencing its health. Semen, beyond carrying sperm, also transports microbial communities that can affect sperm health and fertility. Lastly, the penile skin, including the glans and shaft, has its microbiota, influenced by factors like circumcision [61].
In essence, recognizing and understanding this intricate microbial landscape within the male reproductive system offers potential for therapeutic interventions and improved reproductive outcomes. The communities play pivotal roles in reproductive health, and their imbalances can lead to conditions like infertility.

3. Microbial Dysbiosis and Male Infertility

3.1. Definition and Characterization of Microbial Dysbiosis

Microbial dysbiosis refers to a disturbance in the structure and function of microbial populations within the human microbiota. It is marked by a shift from the typical or optimal microbial composition, leading the changes in the metabolic functions and distribution of the microbiota’s constituents [65]. The human microbiota’s composition has been linked to various health outcomes [66,67]. Dysbiosis in the male reproductive tract microbiota is considered a factor in male infertility. Recognizing individual microbial imbalances is crucial for precision medicine, enabling tailored interventions to address root causes of male infertility [68].
Oxidative stress (OS) significantly influences male infertility development. Elevated OS levels or DNA-damaged sperm increase infertility risk [69]. Dysbiosis of the male genital tract microbiota is associated with conditions like prostatitis, urethritis, and infertility [70]. Specific bacterial species such as Escherichia coli and Ureaplasma urealyticum are linked to chronic prostatitis and inflammation in the male reproductive system [71].
Dysbiosis in the male reproductive tract is complex, varying across diseases and individuals. It often involves a decrease in microbial diversity and an increase in facultative anaerobic species [65]. Such dysbiosis can lead to detrimental effects on the male reproductive system, impacting sperm quality and functionality [72]. Infections from specific microorganisms, like Ureaplasma urealyticum, are associated with male infertility [72].
The dysbiosis concept, especially concerning the gastrointestinal tract, is still under development. It sometimes lacks clarity and a strong scientific basis [73,74]. Yet, it remains essential in understanding the role of microbiota in health and disease [75]. Figure 1 presents an overview of the locations and components of the male microbiome, along with potential interventions.
In summary, microbial dysbiosis in the male reproductive tract correlates with male infertility. Specific bacterial species and imbalances in the male genital tract microbiota are linked to conditions, like prostatitis, inflammation, and oxidative stress, affecting sperm quality and fertility. Further research is essential to understand the mechanisms of dysbiosis in male infertility and potential therapeutic interventions.

3.2. Evidence Linking Microbial Dysbiosis to Male Infertility

1.
Altered bacterial diversity and richness: A hallmark of dysbiosis in the male reproductive tract microbiota is reduced microbial diversity and richness [65]. Such a decline can disrupt microbial community balance, affecting male infertility [76].
2.
Proliferation of pathogenic species: Dysbiosis can lead to an overgrowth of pathogenic species in the male genital tract. Bacteria like Escherichia coli and Ureaplasma urealyticum are associated with chronic prostatitis and inflammation [77]. These species can induce OS, inflammation, and damage to the male reproductive system, impacting sperm quality and fertility [69].
3.
Genital microbiome disruptions: Changes in the composition and function of the male genital tract’s microbiota are linked to male infertility. Such disruptions can cause inflammation, oxidative stress, and sperm cell damage [69].
Research supports the association between microbial dysbiosis and male infertility, highlighting changes in bacterial diversity, pathogenic species proliferation, and genital microbiome imbalances. Identifying these imbalances can guide precision medicine approaches, offering tailored treatments for male infertility [32].

3.3. The Impact of Gut Microbial Dysbiosis on Male Infertility

Research on the microbiome primarily centers on the gastrointestinal tract, which houses a significant portion of microbial organisms. Recent studies have drawn attention to the correlation between gut microbiota dysbiosis and male infertility, suggesting its potential significance in clinical practice [78,79,80].
Ding et al. [78] found a marked reduction in sperm concentration and motility in mice subjected to a high-fat diet. These mice also showed a decline in Bacteroidetes and Verrucomicrobia, with an increase in Firmicutes and Proteobacteria in their gut microbiota. In another study, Zhang et al. [81] noted a significant rise in sperm concentration and sperm motility after transferring fecal microbiota from alginate oligosaccharide-treated mice to busulfan-treated mice. This change was associated with an increase in “beneficial” bacterial populations, specifically Bacteroidales and Bifidobacteriales [81]. Zhao et al. [79] highlighted that alginate oligosaccharides could potentially counteract the spermatogenesis impairment caused by busulfan in mice. This effect was linked to an increase in beneficial bacteria like Bacteroidales and Lactobacillaceae and a decrease in harmful bacteria, notably Desulfovibrionaceae [79].
While specific research on the relationship between gut microbiota and male infertility is limited, existing studies provide valuable insights. These insights emphasize the need for further exploration into manipulating gut microbiota in infertile males. Understanding and addressing gut microbiota imbalances in infertile males can lead to innovative, tailored treatments targeting the root causes of infertility [20].

3.4. Microbial Dysbiosis in the Reproductive System and Its Association with Male Infertility

Microorganisms play a pivotal role in male fertility. The male reproductive tract microbiome, with its diverse bacterial composition, can influence various aspects of reproductive health [61]. For instance, the interactions of the male microbiome with the immune system can impact reproductive health, with dysbiosis exacerbating reproductive challenges [33]. Metabolites produced by the male microbiome can influence fertility, potentially affecting the reproductive system either immediately or over time. Such microbial imbalances can also alter sperm quality and other parameters [82]. The “seminovaginal microbiota”, transmitted between partners during sexual activity, can influence couple fertility. Comprehensive fertility assessments should consider the microbiota of both partners [83].
A myriad of factors, including STIs, immunological interactions, metabolic processes, and microbial dysbiosis can influence male fertility [61]. Understanding the intricate interactions between the male microbiota and fertility is imperative for developing effective therapeutic strategies. Precision medicine, which its focus on individualized care, can facilitate the development of tailored fertility treatment plans by identifying specific microbial imbalances, allowing for targeted interventions to enhance fertility outcomes [84].
While there has been extensive research on microbiota across various anatomical regions, the study of microbiota in the male reproductive system remains relatively limited. Historically, research primarily focused on identifying established pathogens through conventional culture-dependent techniques, microscopic examination, and targeted polymerase chain reaction (PCR) amplification. However, with the advent of next-generation sequencing, there has been a shift toward identifying the complex microbiota in the male reproductive system, offering a clearer understanding of its relationship with male infertility.
It is well-documented that disturbances in the microbiota of the female reproductive tract can lead to various reproductive disorders [85,86,87]. Yet, comprehensive research on the male reproductive tract microbiota has been lacking. Historically, the absence of microbial growth in tests on samples from the male reproductive tract, termed “culture-negative”, was seen as an indication of the absence of bacterial infection. This perspective led to a limited understanding of the microbiota composition of the male reproductive tract. However, emerging evidence suggests the presence of a microbiota in the reproductive tract and associated body fluids, such as seminal fluid and urine [88].

3.5. Dysbiosis in Specific Regions of the Male Reproductive System

3.5.1. Rete Testis, Efferent Ducts, and Epididymis

Research on the testicular microbiome is scarce. A pivotal study by Alfano et al. [89] provided initial evidence linking male infertility to changes in the testicular microbiome. Infertile men showed a lack of Bacteroidetes and Proteobacteria in the testis. Results from microsurgical testicular sperm extraction tests that yielded no sperm indicated changes in the abundance of Firmicutes and Clostridium, a complete absence of Peptoniphilus asaccharolyticus, and an elevation in Actinobacteria.

3.5.2. Deferent Duct, Seminal Vesicles, and Prostate

Lundy et al. [80] observed that post-vasectomy, there was a decrease in the presence of Collinsella (phylum Actinobacteria) and Staphylococcus (phylum Firmicutes) in semen. This observation indirectly points towards a potential link between testicular microbiota and male infertility.

3.5.3. Urethra and Coronal Sulcus

The male genital tract, encompassing regions such as the urethra and the coronal sulcus, harbors distinct microbial communities [60]. Predominant bacterial genera in the male reproductive tract include Corynebacterium, Streptococcus, and Staphylococcus [61,62].

3.5.4. Semen

Studies focusing on male infertility have primarily analyzed semen samples. These studies have consistently found variations in the microbiota composition in the semen of infertile males. Notably, there is an increased prevalence of Prevotella and Staphylococcus and a decreased presence of Lactobacillus and Pseudomonas [76,90,91]. Baud et al. [91] and Farahani et al. [76] further highlighted a negative correlation between the prevalence of Prevotella and sperm motility. In contrast, a direct correlation was observed between a decreased abundance of Lactobacillus and abnormal sperm morphology.
Comparing samples from fertile and infertile individuals, rectum samples from infertile men showed variations in the abundance of Anaerococcus, while displaying an elevated abundance of Lachnospiraceae, Collinsella, and Coprococcus. Conversely, urine samples obtained from infertile men demonstrated an increased presence of Anaerococcus. Furthermore, semen samples from infertile men exhibited a decreased presence of Collinsella and an increased presence of Aerococcus [80]. Subsequent research revealed a statistically significant inverse relationship between the abundance of Aerococcus and both leukocytospermia and semen viscosity. Additionally, a statistically significant negative correlation was observed between the abundance of Prevotella and semen concentration. Conversely, a statistically significant positive correlation was found between the abundance of Pseudomonas and sperm count, while exhibiting an inverse proportionality with the pH of semen [80]. To solidify the findings from these studies, more extensive, longitudinal research across multiple institutions is essential.

4. Mechanisms Linking Microbial Dysbiosis and Male Infertility

Male infertility is often linked with microbial dysbiosis, which is an imbalance in the composition and function of the microbiota. This imbalance is believed to influence infertility through various mechanisms. This section delves into three primary mechanisms connecting microbial dysbiosis and male infertility: inflammation and immune response, the effect of OS on sperm quality, and the implications of impaired sperm function and motility.
  • Inflammation and immune response: A significant mechanism linking microbial dysbiosis to male infertility is the activation of inflammation and immune response. For instance, the oral microbiota, known for its dynamic and polymicrobial nature, can directly lead to diseases like dental caries and periodontitis [92]. These conditions manifest an inflammatory response triggered by the interaction between the microbiota and host factors, such as inflammation and dietary sugars [92]. In the context of male infertility, inflammation and immune response can negatively influence sperm function and overall fertility.
The generation of ROS during inflammation can induce OS [93], which is known to adversely affect fertility by compromising sperm plasma membrane fluidity and DNA integrity, leading to reduced sperm counts and impaired sperm function [94].
2.
OS and its impact on sperm quality: OS, an imbalance between the ROS production and the body’s antioxidant defenses, is associated with male infertility [93]. While ROS are essential for reproduction, their overproduction can harm sperm DNA, impair sperm motility, and increase susceptibility to genetic anomalies [93]. OS can alter sperm morphology and reduce sperm concentration, affecting overall semen parameters [95]. Mechanisms through which OS affects sperm quality include lipid peroxidation, DNA damage, and compromised mitochondrial function [96].
Mitochondrial function is crucial for sperm motility, as mitochondria supply the energy required for sperm movement [96]. The motility of sperm is heavily reliant on the functionality of mitochondria, as these organelles play a crucial role in supplying the necessary energy for sperm locomotion [96]. Impaired mitochondrial function can reduce adenosine triphosphate (ATP) production, leading to decreased sperm motility and fertility [96].
3.
Impaired sperm function and motility: Dysfunction in sperm movement and function are common in male infertility cases. The gut microbiota composition, influenced by factors like diet and the immune system, can affect sperm function and motility [97]. Dysbiosis in the gut microbiota, marked by reduced microbial diversity and the growth of specific bacterial taxa, has been linked to compromised sperm function and motility [97]. Factors such as oxidative stress, bacteriophage induction, and bacterial toxin release can initiate this dysbiosis [97].
It is also worth noting that pathogenic bacteria can negatively impact sperm function and motility [98]. Bacterial infections in the male reproductive tract can induce inflammation and oxidative stress, affecting sperm quality and overall fertility [8]. The presence of intracellular bacteria within the male reproductive tract can trigger an immune response, interfering with sperm function and leading to fertility issues [98].
In summary, microbial dysbiosis can negatively influence male fertility through various mechanisms, including inflammation, oxidative stress, and impaired sperm function and motility. Understanding these mechanisms is crucial for the advancement of precision medicine, especially as findings reveal altered microbial compositions in conditions like non-obstructive azoospermia (NOA) [99]. By pinpointing specific microbial imbalances and their effects on male fertility, precision medicine can design interventions to restore microbial balance and address the root causes of infertility. This tailored approach, considering each patient’s unique microbial and genetic factors, represents the future of reproductive medicine. As research continues to unravel the complexities of microbial dysbiosis and its impact on male fertility, precision medicine remains at the forefront, offering targeted and personalized solutions for couples facing infertility challenges [19].

5. Diagnostic Approaches for Assessing Microbial Dysbiosis in Male Infertility

5.1. Current Methods and Limitations

To evaluate microbial dysbiosis in male infertility, current methodologies primarily focus on analyzing microbiota composition and diversity using techniques such as culture-based techniques, PCR, quantitative PCR (qPCR), and next-generation sequencing (NGS) [28,76,100,101,102,103,104,105].
Culture-based methods involve isolating and cultivating bacteria from clinical samples, followed by identification using biochemical tests or DNA sequencing. However, these methods might not capture the full spectrum of microbial communities, often showing a bias towards culturable bacteria [100,101].
Molecular techniques, including PCR and qPCR, allow for the identification and quantification of specific microbial taxa or genes associated with dysbiosis. They provide a broader view of microbiota composition and can identify low-abundance or non-culturable bacteria [28,76,100,102,103,104,105]. However, their specificity can sometimes limit the full coverage of microbial diversity and they might not provide insights into the functional capabilities of the microbiota [76,102].
NGS technologies, utilizing 16S rRNA gene sequencing and metagenomic sequencing, have revolutionized microbial community research. They can detect and characterize both culturable and non-culturable bacteria, shedding light on functional pathways and interactions within the microbiota [76,102,103,104,105]. Yet, they come with challenges, including high costs, complex data analysis, and the need for specialized bioinformatics knowledge [76,102,103,104,105].
Despite advancements in microbial analysis techniques, challenges persist. The lack of standardized protocols and reference databases can lead to outcome variations, making comparisons across research difficult [76,102,103,104,105]. Moreover, understanding microbial dysbiosis in male infertility is challenging due to the complex and dynamic nature of the microbiota and the lack of clear microbial indicators for infertility [76,102,103,104,105].

5.2. Advances in Molecular Techniques for Microbiota Analysis

Recent advancements in molecular techniques have enhanced the ability to study microbiota and assess microbial dysbiosis in male infertility.

5.2.1. Next-Generation Sequencing

NGS, through technologies like 16S rRNA gene sequencing and metagenomic sequencing, offers efficient DNA or RNA sequencing. This allows for a comprehensive assessment of microbial community composition and potential functionality [76,102,103,104,105].

5.2.2. Metatranscriptomics

This technique examines microbial gene expression within a community, providing insights into the functional activities of the microbiota and identifying key metabolic pathways potentially disrupted in male infertility [102].

5.2.3. Metaproteomics

Focusing on analyzing proteins expressed by the microbial community, metaproteomics can identify and quantify these proteins, offering insights into the functional behaviors of the microbiota and potential links to microbial dysbiosis in male infertility [106].

5.2.4. Shotgun Metagenomics

This comprehensive sequencing of a microbial community’s entire DNA offers insights into the genetic capabilities of microorganisms and can detect specific genes or pathways associated with dysbiosis [102].

5.2.5. Long-Read Sequencing

Technologies like PacBio and Oxford Nanopore sequencing produce extended DNA reads, resolving complex microbial communities and enhancing taxonomic and functional assignments [107].

5.3. Biomarkers for Identifying Microbial Dysbiosis in Male Infertility

Identifying reliable biomarkers for the assessment of microbial dysbiosis in male infertility is crucial for accurate diagnosis and targeted therapeutic interventions. Biomarkers can provide objective and quantifiable indicators of dysbiosis, aiding clinical decision-making. Several important biomarkers can be identified, namely:

5.3.1. Seminal Oxidative Stress Markers

OS markers, including ROS concentrations, lipid peroxidation, and antioxidant enzyme function, can provide insights into the oxidative state of seminal fluid and its association with dysbiosis [103,108,109].

5.3.2. Seminal Inflammatory Markers

Markers related to inflammation, such as cytokines, chemokines, and immune cell populations, can be quantified in seminal fluid to assess inflammation and its link to dysbiosis [109].

5.3.3. Microbial Biomarkers

Specific microbial taxa or genes can serve as indicators of dysbiosis in male infertility. Molecular techniques, like 16S rRNA gene sequencing, can help distinguish between healthy and dysbiotic microbiota [76,103].

5.3.4. Metabolomic Biomarkers

Metabolomic profiling of seminal fluid can reveal metabolic alterations linked to dysbiosis. Specific metabolites or metabolic pathways can be quantified to assess the metabolic state and its association with dysbiosis [102].

5.3.5. Epigenetic Biomarkers

Epigenetic changes, especially DNA methylation, play a role in male infertility. Microbial dysbiosis can influence these epigenetic modifications, and assessing changes linked to dysbiosis can be done by measuring epigenetic biomarkers in sperm DNA [110].
In conclusion, while current approaches for assessing microbial dysbiosis in male infertility have made significant strides, challenges remain. Molecular techniques and the identification of specific biomarkers align with precision medicine principles, aiming to provide individualized treatments based on each patient’s unique microbial and genetic factors. As the field continues to evolve, integrating these diagnostic tools with the broader precision medicine framework will be essential for delivering optimal care to patients facing infertility challenges [56,111].

6. Therapeutic Interventions for Modulating the Genital Microbiota

6.1. Targeted Antimicrobial Therapies

Addressing male infertility often necessitates treating underlying microbial infections that disrupt the genital microbiota balance. While targeted antimicrobial therapies present a potential solution, their application should be judicious to avoid exacerbating genital microbiota imbalances [35,70,94,95]. Interventions might include therapies such as probiotics, prebiotics, and synbiotics, which aim to rebalance the microbiota and enhance reproductive outcomes. Additionally, precision medicine might leverage molecular biomarkers and inflammatory mediators to modulate the immune response and bolster reproductive health [112].
Antibiotics, tailored to specific infections and their susceptibilities, are commonly employed to counter bacterial infections and diminish bacterial prevalence in the genital microbiota [35,70,94,95]. Yet, indiscriminate antibiotic use can perturb the commensal microbiota, potentially inducing further dysbiosis [35,70,94,95]. Antibiotics like quinolones, trimethoprim, tetracyclines, macrolides, and β-lactam have demonstrated efficacy in restoring semen parameters in infertile males with infections, leading to enhanced sperm motility and fertility outcomes [113,114].
Antifungal agents, such as azoles, counteract Candida species overgrowth in the genital microbiome [115]. Additionally, antiviral medications address viral infections like herpes simplex virus (HSV) and the human papillomavirus (HPV), which are associated with male infertility [116,117].
However, potential side effects warrant consideration. Some antibiotics might exhibit testicular or sperm toxicity, as suggested by rodent studies [118]. Antiviral treatment for hepatitis C virus (HCV) has been linked to adverse semen parameters in infertile males [119,120,121].
Given these considerations, targeted antimicrobial therapies should be judiciously employed, emphasizing the importance of microbiome balance and potentially integrating probiotic supplements or dietary modifications [35,70,94,95].

6.2. Probiotics and Their Potential Benefits

Probiotics, beneficial live bacteria consumed in adequate amounts, have emerged as potential agents to promote health and address male infertility. By fostering beneficial bacteria and suppressing harmful microorganisms, they can help reestablish a balanced genital microbiome [122,123].
Oral probiotics, such as Lactobacillus and Bifidobacterium species, can colonize the gut and indirectly influence the genital microbiota. They compete with pathogenic bacteria for resources, modulate the immune response, and produce antimicrobial compounds, potentially improving semen parameters [124,125,126,127,128,129].
Topical probiotics, especially Lactobacillus species, can be directly administered to the vaginal tract, aiding in reestablishing a healthy microbial balance. These probiotics are able to adhere to the vaginal epithelium, produce lactic acid to maintain an acidic pH, and inhibit the growth of harmful bacteria. This local administration could potentially be useful in improving male fertility in cases of vaginal disorders, as suggested by research that showed the protective effect of certain Lactobacillus strains on sperm in the presence of such conditions [130].
The efficacy of probiotics in male infertility might hinge on specific strains, dosage, and treatment duration. The beneficial impact of probiotics on male fertility and reproductive hormones has been indicated in both human and animal studies. Notably, improvements were observed in roosters, male mouse models, and zebrafish, along with hormonal changes in humans [125,126,127,128,129].
However, despite the promising results, it is crucial to remember that more research is needed to fully comprehend these effects and to determine the most effective probiotic strains, dosages, and treatment plans for various forms of male infertility. The potential role of probiotics in enhancing male fertility underscores the need for continued research in this intriguing area.

6.3. Prebiotics and Their Role in Restoring Microbial Balance

Prebiotics, non-digestible dietary fibers that foster beneficial bacterial growth, play a pivotal role in restoring microbial equilibrium. They support the colonization and metabolic activity of probiotic bacteria by acting as a substrate for their development [131,132,133]. Found in foods like whole grains, legumes, fruits, and vegetables, prebiotics are fermented by helpful bacteria in the upper gastrointestinal tract [7,8,9]. Short-chain fatty acids (SCFAs), including butyrate, acetate, and propionate, are created during this fermentation process and serve to maintain a balanced microbiota in the genitals and stomach [131,132,133].
By promoting beneficial bacterial growth, prebiotics can modulate the immune response, inhibit harmful bacterial proliferation, and increase antimicrobial compound production [131,132,133]. They might enhance sperm quality, reduce oxidative stress, and modulate the immune response in male infertility contexts [131,132,133]. To identify the precise prebiotic formulations, doses, and durations of therapy that are most successful in reestablishing a healthy microbial balance in male infertility, more study is required.

6.4. Fecal Microbiota Transplantation and Its Implications

Fecal microbiota transplantation (FMT) involves transferring fecal material from a healthy donor to a recipient, aiming to restore a balanced gut and potentially genital microbiota [35,95,112,115].
A disorder known as Clostridioides difficile infection (CDI), which is characterized by a dysbiosis of the gut microbiota, is predominantly treated with FMT [134]. A diversified and balanced microbial population may be restored with the transplantation of feces from a healthy donor, curing CDI symptoms [134].
FMT has attracted interest recently as a possible therapy for other ailments linked to microbial dysbiosis, such as certain gastrointestinal problems, metabolic disorders, and even diseases of the female reproductive system [35,95,115]. By altering the vaginal and intestinal microbiota, lowering inflammation, and enhancing overall reproductive health, FMT may have an impact on male infertility [35,95,115].
However, FMT remains an experimental treatment for conditions other than CDI; thus further research is needed to determine if it is safe and effective for treating male infertility [35,95,115]. For the safe and efficient use of FMT in male infertility, adequate donor selection, pathogen screening, and standardization of FMT procedures are crucial factors [35,95,115].
In conclusion, therapeutic strategies like targeted antimicrobial treatments, probiotics, prebiotics, and FMT offer potential avenues for modulating the genital microbiota in male infertility contexts. These interventions aim to enhance sperm quality, reduce OS, and regulate immune responses. However, further research is imperative to refine treatment regimens, ascertain long-term outcomes, and ensure the safety and efficacy of these therapies in clinical settings.
Aligning these therapeutic interventions with precision medicine principles offers a promising path forward. By understanding individual microbial imbalances and tailoring treatments accordingly, clinicians can provide more targeted interventions, enhancing treatment efficacy and minimizing potential side effects. As the male infertility treatment landscape evolves, integrating these therapeutic strategies within the broader precision medicine framework will be crucial for delivering optimal, individualized patient care [56].

7. Challenges and Future Perspectives

The exploration of the microbiome’s association with male infertility, while comprehensive, still presents significant limitations and offers ample opportunities for further research. While numerous randomized controlled trials have been included, the strength of the research evidence is compromised due to insufficient details on randomization methods, the frequent lack of adequate controls, and the dearth of double-blind clinical studies. Factors such as age, body mass index, lifestyle habits, and medication use can influence both the microbiome and reproductive parameters, adding layers of complexity to the research [135].
This review highlights challenges such as small sample sizes, diverse study designs, variability in antimicrobial or probiotics administration, and the limitations of 16S rRNA technology in sequencing. A significant number of studies have not delved into the impact of the microbiome on clinical fertility outcomes in infertile males. Therefore, there is a pressing need for large-scale prospective studies to ascertain the microbiome’s role as a causative factor in male infertility [76].
For microbial-based therapies for male infertility to be effectively implemented, a myriad of factors must be considered. This includes accurate diagnosis of microbial dysbiosis, selection of suitable antimicrobial agents, probiotics, or prebiotics, and considering potential interactions with the host’s immune system. Furthermore, optimizing the route of administration and treatment duration is essential for ensuring therapeutic efficacy. Long-term monitoring and follow-up are also pivotal for evaluating the success and safety of microbial-based interventions and for making necessary treatment adjustments.
Implementing microbial-based interventions in male infertility introduces ethical and safety challenges, encompassing informed consent, protection of personal health data and transparency in treatment utilization. The potential risks associated with antimicrobial agents, probiotics, or prebiotics necessitate thorough safety and efficacy evaluations through rigorous preclinical and clinical studies before broad implementation [136].
The horizon holds promise for personalized medicine in the realm of male infertility management. Personalized medicine seeks to tailor medical interventions based on individual characteristics, encompassing genetic, environmental, and microbial factors. Genetic testing can pinpoint rare genetic disorders or chromosomal abnormalities contributing to male infertility. Similarly, microbial analysis can enhance personalized medicine approaches by identifying specific dysbiotic patterns or microbial biomarkers associated with infertility [137,138,139].
Future research avenues should encompass extended clinical trials for microbial-based therapies, a deeper understanding of the interplay between microbiota, host factors, and reproductive health, and leveraging technological advancements like high-throughput sequencing and omics methodologies. This will provide a more comprehensive insight into the molecular mechanisms underpinning male infertility [118,119,121]. The development of innovative delivery mechanisms, such as nanoparticles or gene therapy techniques, can further enhance the efficacy and precision of microbial-based interventions [140,141,142]. Thus, it is imperative for upcoming research to not only deepen the understanding of the genital microbiota’s role in male infertility but also craft more precise therapeutic strategies, all while addressing the ethical and safety concerns tied to microbial-based therapies.

8. Conclusions

Grasping the impact of microbial factors on male infertility is essential for both accurate diagnosis and effective management of this intricate condition. Male infertility has been identified to have a strong correlation with microbial dysbiosis in the genital microbiota, which can directly compromise sperm health and overall fertility potential. Recent strides in microbiome research and microbial analysis techniques, such as next-generation sequencing, have shed light on the underlying causes of infertility, revealing potential etiologies behind cases previously deemed idiopathic. These insights have paved the way for the development of innovative therapeutic strategies, including microbiome therapy, aiming the reestablish a harmonious microbiome.
Disequilibrium in the human microbiome can manifest as a spectrum of diseases, many of which may benefit from interventions that aim to restore a balanced microbiome. This understanding has catalyzed the emergence of potential therapeutic modalities, including targeted antimicrobial therapies, probiotics, prebiotics, and fecal microbiota transplantation. These interventions hold promise in enhancing fertility outcomes. The increasing evidence underscoring the relationship between the human microbiome and diverse diseases has positioned it at the forefront of medical research, offering potential avenues for disease prevention, management, and prediction.
By delving deeper into the microbiome of infertile males, a holistic understanding of the etiology of male infertility can be achieved. Such knowledge is pivotal in designing microbiome-centric interventions tailored to address the unique fertility challenges faced by infertile males. Merging these interventions with personalized medicine strategies heralds a promising trajectory for addressing male infertility. As the understanding of the microbiome’s influence on male infertility deepens, the adoption of precision medicine—crafting treatments based on individual genetic, environmental, and microbial factors—emerges as a cornerstone, promising a nuanced and effective approach to this multifaceted condition.
Nevertheless, while these perspectives are promising, there remains an urgent need for further research to validate and refine these methodologies for their broader clinical application in addressing male infertility. Anticipated future research endeavors will further elucidate the role of microbial factors in male infertility, laying the foundation for the evolution of more potent treatment modalities.

Author Contributions

A.K. was involved in the conceptualization and wrote the major part of the original draft. E.M. and A.Z. contributed by searching for relevant literature and writing other parts of the manuscript. N.S., F.D. and S.P. reviewed and edited the manuscript. Both N.S. and S.P. participated in the conceptualization and provided the final editing and review of the manuscript. 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.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Zegers-Hochschild, F.; Adamson, G.D.; de Mouzon, J.; Ishihara, O.; Mansour, R.; Nygren, K.; Sullivan, E.; Vanderpoel, S.; International Committee for Monitoring Assisted Reproductive Technology; World Health Organization. International Committee for Monitoring Assisted Reproductive Technology (ICMART) and the World Health Organization (WHO) revised glossary of ART terminology, 2009. Fertil. Steril. 2009, 92, 1520–1524. [Google Scholar] [CrossRef]
  2. Dierickx, S.; De Proost, M.; Huang, A.Y.; Ceesay, S.; Clarke, E.; Balen, J. The Nairobi Summit and Reproductive Justice: Unmet Needs for People with Infertility. Am. J. Trop. Med. Hyg. 2021, 104, 812–813. [Google Scholar] [CrossRef]
  3. Mendiola, J.; Jørgensen, N.; Mínguez-Alarcón, L.; Sarabia-Cos, L.; López-Espín, J.J.; Vivero-Salmerón, G.; Ruiz-Ruiz, K.J.; Fernández, M.F.; Olea, N.; Swan, S.H.; et al. Sperm counts may have declined in young university students in Southern Spain. Andrology 2013, 1, 408–413. [Google Scholar] [CrossRef]
  4. Geoffroy-Siraudin, C.; Loundou, A.D.; Romain, F.; Achard, V.; Courbière, B.; Perrard, M.-H.; Durand, P.; Guichaoua, M.R. Decline of semen quality among 10 932 males consulting for couple infertility over a 20-year period in Marseille, France. Asian J. Androl. 2012, 14, 584–590. [Google Scholar] [CrossRef] [PubMed]
  5. Molina, R.I.; Martini, A.C.; Tissera, A.; Olmedo, J.; Senestrari, D.; De Cuneo, M.F.; Ruiz, R.D. Semen quality and aging: Analysis of 9.168 samples in Cordoba. Argentina. Arch. Esp. Urol. 2010, 63, 214–222. [Google Scholar]
  6. Axelsson, J.; Rylander, L.; Rignell-Hydbom, A.; Giwercman, A. No secular trend over the last decade in sperm counts among Swedish men from the general population. Hum. Reprod. 2011, 26, 1012–1016. [Google Scholar] [CrossRef]
  7. Esteves, S.C.; Zini, A.; Aziz, N.; Alvarez, J.G.; Sabanegh, E.S., Jr.; Agarwal, A. Critical Appraisal of World Health Organization’s New Reference Values for Human Semen Characteristics and Effect on Diagnosis and Treatment of Subfertile Men. Urology 2012, 79, 16–22. [Google Scholar] [CrossRef] [PubMed]
  8. Itoh, N.; Kayama, F.; Tatsuki, T.J.; Tsukamoto, T. Have sperm counts deteriorated over the past 20 years in healthy, young Japanese men? Results from the Sapporo area. J. Androl. 2001, 22, 40–44. [Google Scholar] [PubMed]
  9. Jensen, T.K.; Sobotka, T.; Hansen, M.A.; Pedersen, A.T.; Lutz, W.; Skakkebæk, N.E. Declining trends in conception rates in recent birth cohorts of native Danish women: A possible role of deteriorating male reproductive health. Int. J. Androl. 2008, 31, 81–92. [Google Scholar] [CrossRef]
  10. Carlsen, E.; Swan, S.H.; Petersen, J.H.; Skakkebæk, N.E. Longitudinal changes in semen parameters in young Danish men from the Copenhagen area. Hum. Reprod. 2005, 20, 942–949. [Google Scholar] [CrossRef]
  11. Gyllenborg, J.; Skakkebæk, N.E.; Nielsen, N.C.; Keiding, N.; Giwercman, A. Secular and seasonal changes in semen quality among young Danish men: A statistical analysis of semen samples from 1927 donor candidates during 1977–1995. Int. J. Androl. 1999, 22, 28–36. [Google Scholar] [CrossRef]
  12. Levine, H.; Jørgensen, N.; Martino-Andrade, A.; Mendiola, J.; Weksler-Derri, D.; Mindlis, I.; Pinotti, R.; Swan, S.H. Temporal trends in sperm count: A systematic review and meta-regression analysis. Hum. Reprod. Update 2017, 23, 646–659. [Google Scholar] [CrossRef]
  13. Mann, U.; Shiff, B.; Patel, P. Reasons for worldwide decline in male fertility. Curr. Opin. Urol. 2020, 30, 296–301. [Google Scholar] [CrossRef]
  14. Aggarwal, N.; Kitano, S.; Puah, G.R.Y.; Kittelmann, S.; Hwang, I.Y.; Chang, M.W. Microbiome and Human Health: Current Understanding, Engineering, and Enabling Technologies. Chem. Rev. 2023, 123, 31–72. [Google Scholar] [CrossRef]
  15. Baothman, O.A.; Zamzami, M.A.; Taher, I.; Abubaker, J.; Abu-Farha, M. The role of Gut Microbiota in the development of obesity and Diabetes. Lipids Health Dis. 2016, 15, 108. [Google Scholar] [CrossRef] [PubMed]
  16. Scheithauer, T.P.M.; Rampanelli, E.; Nieuwdorp, M.; Vallance, B.A.; Verchere, C.B.; van Raalte, D.H.; Herrema, H. Gut Microbiota as a Trigger for Metabolic Inflammation in Obesity and Type 2 Diabetes. Front. Immunol. 2020, 11, 571731. [Google Scholar] [CrossRef] [PubMed]
  17. Ouabbou, S.; He, Y.; Butler, K.; Tsuang, M. Inflammation in Mental Disorders: Is the Microbiota the Missing Link? Neurosci. Bull. 2020, 36, 1071–1084. [Google Scholar] [CrossRef] [PubMed]
  18. Cani, P.D. Human gut microbiome: Hopes, threats and promises. Gut 2018, 67, 1716–1725. [Google Scholar] [CrossRef]
  19. Wang, H.; Xu, A.; Gong, L.; Chen, Z.; Zhang, B.; Li, X. The Microbiome, an Important Factor That Is Easily Overlooked in Male Infertility. Front. Microbiol. 2022, 13, 831272. [Google Scholar] [CrossRef] [PubMed]
  20. Magill, R.G.; MacDonald, S.M. Male infertility and the human microbiome. Front. Reprod. Health 2023, 5, 1166201. [Google Scholar] [CrossRef]
  21. Potts, J.M.; Sharma, R.; Pasqualotto, F.; Nelson, D.; Hall, G.; Agarwal, A. Association of ureaplasma urealyticum with abnormal reactive oxygen species levels and absence of leukocytospermia. J. Urol. 2000, 163, 1775–1778. [Google Scholar] [CrossRef] [PubMed]
  22. Zeyad, A.; Hamad, M.; Amor, H.; Hammadeh, M.E. Relationships between bacteriospermia, DNA integrity, nuclear protamine alteration, sperm quality and ICSI outcome. Reprod. Biol. 2018, 18, 115–121. [Google Scholar] [CrossRef] [PubMed]
  23. Merino, G.; Carranza-Lira, S.; Murrieta, S.; Rodriguez, L.; Cuevas, E.; Moran, C. Bacterial Infection and Semen Characteristics in Infertile Men. Arch. Androl. 1995, 35, 43–47. [Google Scholar] [CrossRef] [PubMed]
  24. Schulz, M.; Sánchez, R.; Soto, L.; Risopatrón, J.; Villegas, J. Effect of Escherichia coli and its soluble factors on mitochondrial membrane potential, phosphatidylserine translocation, viability, and motility of human spermatozoa. Fertil. Steril. 2010, 94, 619–623. [Google Scholar] [CrossRef] [PubMed]
  25. Berjis, K.; Ghiasi, M.; Sangy, S. Study of seminal infection among an infertile male population in Qom, Iran, and its effect on sperm quality. Iran. J. Microbiol. 2018, 10, 111–116. [Google Scholar]
  26. Ricci, S.; De Giorgi, S.; Lazzeri, E.; Luddi, A.; Rossi, S.; Piomboni, P.; De Leo, V.; Pozzi, G. Impact of asymptomatic genital tract infections on in vitro Fertilization (IVF) outcome. PLoS ONE 2018, 13, e0207684. [Google Scholar] [CrossRef]
  27. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
  28. Khor, B.; Snow, M.; Herrman, E.; Ray, N.; Mansukhani, K.; Patel, K.A.; Said-Al-Naief, N.; Maier, T.R.; Machida, C.A. Interconnections between the Oral and Gut Microbiomes: Reversal of Microbial Dysbiosis and the Balance between Systemic Health and Disease. Microorganisms 2021, 9, 496. [Google Scholar] [CrossRef]
  29. Wang, Y.; Xie, Z. Exploring the role of gut microbiome in male reproduction. Andrology 2022, 10, 441–450. [Google Scholar] [CrossRef]
  30. Velez, D.; Hwang, K. Personalized Medicine for the Infertile Male. Urol. Clin. N. Am. 2020, 47, 523–536. [Google Scholar] [CrossRef]
  31. Petrosino, J.F. The microbiome in precision medicine: The way forward. Genome Med. 2018, 10, 12. [Google Scholar] [CrossRef] [PubMed]
  32. Agharezaee, N.; Hashemi, M.; Shahani, M.; Gilany, K. Male Infertility, Precision Medicine and Systems Proteomics. J. Reprod. Infertil. 2018, 19, 185–192. [Google Scholar]
  33. Al-Nasiry, S.; Ambrosino, E.; Schlaepfer, M.; Morré, S.A.; Wieten, L.; Voncken, J.W.; Spinelli, M.; Mueller, M.; Kramer, B.W. The Interplay Between Reproductive Tract Microbiota and Immunological System in Human Reproduction. Front. Immunol. 2020, 11, 378. [Google Scholar] [CrossRef] [PubMed]
  34. Mammen, M.J.; Sethi, S. COPD and the microbiome. Respirology 2016, 21, 590–599. [Google Scholar] [CrossRef] [PubMed]
  35. Smith, S.B.; Ravel, J. The vaginal microbiota, host defence and reproductive physiology. J. Physiol. 2016, 595, 451–463. [Google Scholar] [CrossRef] [PubMed]
  36. Gao, L.; Xu, T.; Huang, G.; Jiang, S.; Gu, Y.; Chen, F. Oral microbiomes: More and more importance in oral cavity and whole body. Protein Cell 2018, 9, 488–500. [Google Scholar] [CrossRef]
  37. Khadka, V.D.; Key, F.M.; Romo-González, C.; Martínez-Gayosso, A.; Campos-Cabrera, B.L.; Gerónimo-Gallegos, A.; Lynn, T.C.; Durán-McKinster, C.; Coria-Jiménez, R.; Lieberman, T.D.; et al. The Skin Microbiome of Patients With Atopic Dermatitis Normalizes Gradually During Treatment. Front. Cell. Infect. Microbiol. 2021, 11, 720674. [Google Scholar] [CrossRef]
  38. Dickson, R.P.; Singer, B.H.; Newstead, M.W.; Falkowski, N.R.; Erb-Downward, J.R.; Standiford, T.J.; Huffnagle, G.B. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat. Microbiol. 2016, 1, 16113. [Google Scholar] [CrossRef]
  39. Gudnadottir, U.; Debelius, J.W.; Du, J.; Hugerth, L.W.; Danielsson, H.; Schuppe-Koistinen, I.; Fransson, E.; Brusselaers, N. The vaginal microbiome and the risk of preterm birth: A systematic review and network meta-analysis. Sci. Rep. 2022, 12, 7926. [Google Scholar] [CrossRef]
  40. Hurst, R.; Meader, E.; Gihawi, A.; Rallapalli, G.; Clark, J.; Kay, G.L.; Webb, M.; Manley, K.; Curley, H.; Walker, H.; et al. Microbiomes of Urine and the Prostate Are Linked to Human Prostate Cancer Risk Groups. Eur. Urol. Oncol. 2022, 5, 412–419. [Google Scholar] [CrossRef]
  41. Coscia, A.; Bardanzellu, F.; Caboni, E.; Fanos, V.; Peroni, D.G. When a Neonate Is Born, So Is a Microbiota. Life 2021, 11, 148. [Google Scholar] [CrossRef] [PubMed]
  42. Duda-Chodak, A.; Tarko, T.; Satora, P.; Sroka, P. Interaction of dietary compounds, especially polyphenols, with the intestinal microbiota: A review. Eur. J. Nutr. 2015, 54, 325–341. [Google Scholar] [CrossRef]
  43. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef]
  44. Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. BioMed Res. Int. 2018, 2018, 9478630. [Google Scholar] [CrossRef] [PubMed]
  45. O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. EMBO Rep. 2006, 7, 688–693. [Google Scholar] [CrossRef]
  46. Hattori, M.; Taylor, T.D. The Human Intestinal Microbiome: A New Frontier of Human Biology. DNA Res. 2009, 16, 1–12. [Google Scholar] [CrossRef] [PubMed]
  47. Moossavi, S.; Bishehsari, F. Microbes: Possible link between modern lifestyle transition and the rise of metabolic syndrome. Obes. Rev. 2018, 20, 407–419. [Google Scholar] [CrossRef] [PubMed]
  48. Qi, X.; Yun, C.; Pang, Y.; Qiao, J. The impact of the gut microbiota on the reproductive and metabolic endocrine system. Gut Microbes 2021, 13, 1894070. [Google Scholar] [CrossRef] [PubMed]
  49. Khan, I.; Ullah, N.; Zha, L.; Bai, Y.; Khan, A.; Zhao, T.; Che, T.; Zhang, C. Alteration of Gut Microbiota in Inflammatory Bowel Disease (IBD): Cause or Consequence? IBD Treatment Targeting the Gut Microbiome. Pathogens 2019, 8, 126. [Google Scholar] [CrossRef]
  50. Tomaiuolo, R.; Veneruso, I.; Cariati, F.; D’argenio, V. Microbiota and Human Reproduction: The Case of Female Infertility. BioTech 2020, 9, 12. [Google Scholar] [CrossRef]
  51. Barrientos-Durán, A.; Fuentes-López, A.; de Salazar, A.; Plaza-Díaz, J.; García, F. Reviewing the Composition of Vaginal Microbiota: Inclusion of Nutrition and Probiotic Factors in the Maintenance of Eubiosis. Nutrients 2020, 12, 419. [Google Scholar] [CrossRef] [PubMed]
  52. García-Velasco, J.A.; Menabrito, M.; Catalán, I.B. What fertility specialists should know about the vaginal microbiome: A review. Reprod. Biomed. Online 2017, 35, 103–112. [Google Scholar] [CrossRef]
  53. Punzón-Jiménez, P.; Labarta, E. The impact of the female genital tract microbiome in women health and reproduction: A review. J. Assist. Reprod. Genet. 2021, 38, 2519–2541. [Google Scholar] [CrossRef] [PubMed]
  54. López-Moreno, A.; Aguilera, M. Probiotics Dietary Supplementation for Modulating Endocrine and Fertility Microbiota Dysbiosis. Nutrients 2020, 12, 757. [Google Scholar] [CrossRef]
  55. Quaranta, G.; Sanguinetti, M.; Masucci, L. Fecal Microbiota Transplantation: A Potential Tool for Treatment of Human Female Reproductive Tract Diseases. Front. Immunol. 2019, 10, 2653. [Google Scholar] [CrossRef]
  56. Kashyap, P.C.; Chia, N.; Nelson, H.; Segal, E.; Elinav, E. Microbiome at the Frontier of Personalized Medicine. Mayo Clin. Proc. 2017, 92, 1855–1864. [Google Scholar] [CrossRef]
  57. Barko, P.C.; McMichael, M.A.; Swanson, K.S.; Williams, D.A. The Gastrointestinal Microbiome: A Review. J. Vet. Intern. Med. 2018, 32, 9–25. [Google Scholar] [CrossRef]
  58. 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] [PubMed]
  59. Wu, H.J.; Wu, E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes 2012, 3, 4–14. [Google Scholar] [CrossRef] [PubMed]
  60. Petrova, M.I.; Broek, M.V.D.; Balzarini, J.; Vanderleyden, J.; Lebeer, S. Vaginal microbiota and its role in HIV transmission and infection. FEMS Microbiol. Rev. 2013, 37, 762–792. [Google Scholar] [CrossRef]
  61. Tomaiuolo, R.; Veneruso, I.; Cariati, F.; D’argenio, V. Microbiota and Human Reproduction: The Case of Male Infertility. BioTech 2020, 9, 10. [Google Scholar] [CrossRef] [PubMed]
  62. Zuber, A.; Peric, A.; Pluchino, N.; Baud, D.; Stojanov, M. Human Male Genital Tract Microbiota. Int. J. Mol. Sci. 2023, 24, 6939. [Google Scholar] [CrossRef]
  63. Tuddenham, S.; Ravel, J.; Marrazzo, J.M. Protection and Risk: Male and Female Genital Microbiota and Sexually Transmitted Infections. J. Infect. Dis. 2021, 223, S222–S235. [Google Scholar] [CrossRef]
  64. Motrich, R.D.; Salazar, F.C.; Breser, M.L.; Mackern-Oberti, J.P.; Godoy, G.J.; Olivera, C.; Paira, D.A.; Rivero, V.E. Implications of prostate inflammation on male fertility. Andrologia 2018, 50, e13093. [Google Scholar] [CrossRef]
  65. Kriss, M.; Hazleton, K.Z.; Nusbacher, N.M.; Martin, C.G.; A Lozupone, C. Low diversity gut microbiota dysbiosis: Drivers, functional implications and recovery. Curr. Opin. Microbiol. 2018, 44, 34–40. [Google Scholar] [CrossRef] [PubMed]
  66. Natarajan, A.; Bhatt, A.S. Microbes and microbiomes in 2020 and beyond. Nat. Commun. 2020, 11, 4988. [Google Scholar] [CrossRef] [PubMed]
  67. Tett, A.; Pasolli, E.; Masetti, G.; Ercolini, D.; Segata, N. Prevotella diversity, niches and interactions with the human host. Nat. Rev. Microbiol. 2021, 19, 585–599. [Google Scholar] [CrossRef]
  68. DeGruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current Understanding of Dysbiosis in Disease in Human and Animal Models. Inflamm. Bowel Dis. 2016, 22, 1137–1150. [Google Scholar] [CrossRef] [PubMed]
  69. Agarwal, A.; Rana, M.; Qiu, E.; Albunni, H.; Bui, A.D.; Henkel, R. Role of oxidative stress, infection and inflammation in male infertility. Andrologia 2018, 50, e13126. [Google Scholar] [CrossRef]
  70. Suárez, J.P.; Maya, W.D.C. Microbiota, Prostatitis, and Fertility: Bacterial Diversity as a Possible Health Ally. Adv. Urol. 2021, 2021, 1007366. [Google Scholar] [CrossRef]
  71. 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]
  72. Fraczek, M.; Kurpisz, M. Inflammatory mediators exert toxic effects of oxidative stress on human spermatozoa. J. Androl. 2006, 28, 325–333. [Google Scholar] [CrossRef]
  73. Riccio, P.; Rossano, R. The human gut microbiota is neither an organ nor a commensal. FEBS Lett. 2020, 594, 3262–3271. [Google Scholar] [CrossRef]
  74. Brüssow, H. Problems with the concept of gut microbiota dysbiosis. Microb. Biotechnol. 2019, 13, 423–434. [Google Scholar] [CrossRef]
  75. Tiffany, C.R.; Bäumler, A.J. Dysbiosis: From fiction to function. Am. J. Physiol. Liver Physiol. 2019, 317, G602–G608. [Google Scholar] [CrossRef]
  76. Farahani, L.; Tharakan, T.; Yap, T.; Ramsay, J.W.; Jayasena, C.N.; Minhas, S. The semen microbiome and its impact on sperm function and male fertility: A systematic review and meta-analysis. Andrology 2021, 9, 115–144. [Google Scholar] [CrossRef]
  77. Huang, C.-P.; Zhu, H.; Xu, K.; Wang, S.; Fan, L.T.; Zhu, W. Mycoplasma and ureaplasma infection and male infertility: A systematic review and meta-analysis. Andrology 2015, 3, 809–816. [Google Scholar] [CrossRef] [PubMed]
  78. Ding, N.; Zhang, X.D.; Zhang, X.; 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] [PubMed]
  79. Zhao, Y.; Zhang, P.; Ge, W.; Feng, Y.; Li, L.; Sun, Z.; Zhang, H.; Shen, W. Alginate oligosaccharides improve germ cell development and testicular microenvironment to rescue busulfan disrupted spermatogenesis. Theranostics 2020, 10, 3308–3324. [Google Scholar] [CrossRef]
  80. 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]
  81. Zhang, P.; Feng, Y.; Li, L.; Ge, W.; Yu, S.; Hao, Y.; Shen, W.; Han, X.; Ma, D.; Yin, S.; et al. Improvement in sperm quality and spermatogenesis following faecal microbiota transplantation from alginate oligosaccharide dosed mice. Gut 2021, 70, 222–225. [Google Scholar] [CrossRef]
  82. Verspoor, R.L.; Price, T.A.R.; Wedell, N. Selfish genetic elements and male fertility. Philos. Trans. R. Soc. B Biol. Sci. 2020, 375, 20200067. [Google Scholar] [CrossRef] [PubMed]
  83. D’argenio, V.; Dittfeld, L.; Lazzeri, P.; Tomaiuolo, R.; Tasciotti, E. Unraveling the Balance between Genes, Microbes, Lifestyle and the Environment to Improve Healthy Reproduction. Genes 2021, 12, 605. [Google Scholar] [CrossRef] [PubMed]
  84. Matthewman, C.; Narin, A.; Huston, H.; Hopkins, C.E. Systems to model the personalized aspects of microbiome health and gut dysbiosis. Mol. Asp. Med. 2023, 91, 101115. [Google Scholar] [CrossRef]
  85. Ravel, J.; Gajer, P.; Abdo, Z.; Schneider, G.M.; Koenig, S.S.K.; McCulle, S.L.; Karlebach, S.; Gorle, R.; Russell, J.; Tacket, C.O.; et al. Vaginal microbiome of reproductive-age women. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4680–4687. [Google Scholar] [CrossRef] [PubMed]
  86. Phukan, N.; Parsamand, T.; Brooks, A.E.S.; Nguyen, T.N.M.; Simoes-Barbosa, A. The adherence of Trichomonas vaginalis to host ectocervical cells is influenced by lactobacilli. Sex. Transm. Infect. 2013, 89, 455–459. [Google Scholar] [CrossRef]
  87. Petrova, M.I.; Lievens, E.; Malik, S.; Imholz, N.; Lebeer, S. Lactobacillus species as biomarkers and agents that can promote various aspects of vaginal health. Front. Physiol. 2015, 6, 81. [Google Scholar] [CrossRef]
  88. Allen-Vercoe, E. Bringing the gut microbiota into focus through microbial culture: Recent progress and future perspective. Curr. Opin. Microbiol. 2013, 16, 625–629. [Google Scholar] [CrossRef] [PubMed]
  89. 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]
  90. 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]
  91. Baud, D.; Pattaroni, C.; Vulliemoz, N.; Castella, V.; Marsland, B.J.; Stojanov, M. Sperm Microbiota and Its Impact on Semen Parameters. Front. Microbiol. 2019, 10, 234. [Google Scholar] [CrossRef]
  92. Lamont, R.J.; Koo, H.; Hajishengallis, G. The oral microbiota: Dynamic communities and host interactions. Nat. Rev. Microbiol. 2018, 16, 745–759. [Google Scholar] [CrossRef] [PubMed]
  93. Alahmar, A.T. Role of Oxidative Stress in Male Infertility: An Updated Review. J. Hum. Reprod. Sci. 2019, 12, 4–18. [Google Scholar] [CrossRef] [PubMed]
  94. Agarwal, A.; Saleh, R.A.; Bedaiwy, M.A. Role of reactive oxygen species in the pathophysiology of human reproduction. Fertil. Steril. 2003, 79, 829–843. [Google Scholar] [CrossRef]
  95. Kumar, N.; Singh, A.K. Trends of male factor infertility, an important cause of infertility: A review of literature. J. Hum. Reprod. Sci. 2015, 8, 191–196. [Google Scholar] [CrossRef] [PubMed]
  96. Barbagallo, F.; La Vignera, S.; Cannarella, R.; Aversa, A.; Calogero, A.E.; Condorelli, R.A. Evaluation of Sperm Mitochondrial Function: A Key Organelle for Sperm Motility. J. Clin. Med. 2020, 9, 363. [Google Scholar] [CrossRef] [PubMed]
  97. Weiss, G.A.; Hennet, T. Mechanisms and consequences of intestinal dysbiosis. Cell. Mol. Life Sci. 2017, 74, 2959–2977. [Google Scholar] [CrossRef] [PubMed]
  98. Stojanov, M.; Baud, D.; Greub, G.; Vulliemoz, N. Male infertility: The intracellular bacterial hypothesis. New Microbes New Infect. 2018, 26, 37–41. [Google Scholar] [CrossRef]
  99. Cao, Y.; Wang, H.; Jin, Z.; Hang, J.; Jiang, H.; Wu, H.; Zhang, Z. Characterization of Non-Obstructive Azoospermia in Men Using Gut Microbial Profiling. J. Clin. Med. 2023, 12, 701. [Google Scholar] [CrossRef]
  100. Dutta, S.; Majzoub, A.; Agarwal, A. Oxidative stress and sperm function: A systematic review on evaluation and management. Arab. J. Urol. 2019, 17, 87–97. [Google Scholar] [CrossRef]
  101. Schulte, R.T.; Ohl, D.A.; Sigman, M.; Smith, G.A. Sperm DNA damage in male infertility: Etiologies, assays, and outcomes. J. Assist. Reprod. Genet. 2009, 27, 3–12. [Google Scholar] [CrossRef]
  102. Ruiz, L.; García-Carral, C.; Rodriguez, J.M. Unfolding the Human Milk Microbiome Landscape in the Omics Era. Front. Microbiol. 2019, 10, 1378. [Google Scholar] [CrossRef] [PubMed]
  103. Agarwal, A.; Bui, A.D. Oxidation-reduction potential as a new marker for oxidative stress: Correlation to male infertility. Investig. Clin. Urol. 2017, 58, 385–399. [Google Scholar] [CrossRef] [PubMed]
  104. Bajinka, O.; Tan, Y.; Abdelhalim, K.A.; Özdemir, G.; Qiu, X. Extrinsic factors influencing gut microbes, the immediate consequences and restoring eubiosis. AMB Express 2020, 10, 130. [Google Scholar] [CrossRef]
  105. Douglas, C.J.; Parekh, N.; Kahn, L.S.; Henkel, R.; Agarwal, A. A Novel Approach to Improving the Reliability of Manual Semen Analysis: A Paradigm Shift in the Workup of Infertile Men. World J. Men’s Health 2021, 39, 172–185. [Google Scholar] [CrossRef] [PubMed]
  106. Agarwal, A.; Selvam, M.K.P.; Baskaran, S. Proteomic Analyses of Human Sperm Cells: Understanding the Role of Proteins and Molecular Pathways Affecting Male Reproductive Health. Int. J. Mol. Sci. 2020, 21, 1621. [Google Scholar] [CrossRef] [PubMed]
  107. Wei, S.; Bahl, M.I.; Baunwall, S.M.D.; Hvas, C.L.; Licht, T.R. Determining Gut Microbial Dysbiosis: A Review of Applied Indexes for Assessment of Intestinal Microbiota Imbalances. Appl. Environ. Microbiol. 2021, 87, e00395-21. [Google Scholar] [CrossRef]
  108. Agarwal, A.; Roychoudhury, S.; Bjugstad, K.B.; Cho, C.L. Oxidation-reduction potential of semen: What is its role in the treatment of male infertility? Ther. Adv. Urol. 2016, 8, 302–318. [Google Scholar] [CrossRef] [PubMed]
  109. Agarwal, A.; Parekh, N.; Selvam, M.K.P.; 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]
  110. Rotondo, J.C.; Lanzillotti, C.; Mazziotta, C.; Tognon, M.; Martini, F. Epigenetics of Male Infertility: The Role of DNA Methylation. Front. Cell Dev. Biol. 2021, 9, 689624. [Google Scholar] [CrossRef]
  111. Behrouzi, A.; Nafari, A.H.; Siadat, S.D. The significance of microbiome in personalized medicine. Clin. Transl. Med. 2019, 8, 16. [Google Scholar] [CrossRef] [PubMed]
  112. Hashem, N.M.; Gonzalez-Bulnes, A. The Use of Probiotics for Management and Improvement of Reproductive Eubiosis and Function. Nutrients 2022, 14, 902. [Google Scholar] [CrossRef] [PubMed]
  113. Farsimadan, M.; Motamedifar, M. Bacterial infection of the male reproductive system causing infertility. J. Reprod. Immunol. 2020, 142, 103183. [Google Scholar] [CrossRef]
  114. Ahmadi, M.H.; Mirsalehian, A.; Gilani, M.A.S.; Bahador, A.; Afraz, K. Association of asymptomatic Chlamydia trachomatis infection with male infertility and the effect of antibiotic therapy in improvement of semen quality in infected infertile men. Andrologia 2018, 50, e12944. [Google Scholar] [CrossRef] [PubMed]
  115. Bondaryk, M.; Kurzątkowski, W.; Staniszewska, M. Antifungal agents commonly used in the superficial and mucosal candidiasis treatment: Mode of action and resistance development. Adv. Dermatol. Allergol. 2013, 5, 293–301. [Google Scholar] [CrossRef] [PubMed]
  116. Akhigbe, R.E.; Dutta, S.; Hamed, M.A.; Ajayi, A.F.; Sengupta, P.; Ahmad, G. Viral Infections and Male Infertility: A Comprehensive Review of the Role of Oxidative Stress. Front. Reprod. Health 2022, 4, 782915. [Google Scholar] [CrossRef] [PubMed]
  117. Drobnis, E.Z.; Nangia, A.K. Antivirals and Male Reproduction. Adv. Exp. Med. Biol. 2017, 1034, 163–178. [Google Scholar] [CrossRef] [PubMed]
  118. Calogero, A.E.; Condorelli, R.A.; Russo, G.I.; La Vignera, S. Conservative Nonhormonal Options for the Treatment of Male Infertility: Antibiotics, Anti-Inflammatory Drugs, and Antioxidants. BioMed Res. Int. 2017, 2017, 4650182. [Google Scholar] [CrossRef] [PubMed]
  119. Lorusso, F.; Palmisano, M.; Chironna, M.; Vacca, M.; Masciandaro, P.; Bassi, E.; Luigi, L.S.; Depalo, R. Impact of chronic viral diseases on semen parameters. Andrologia 2010, 42, 121–126. [Google Scholar] [CrossRef] [PubMed]
  120. Hofer, H.; Donnerer, J.; Sator, K.; Staufer, K.; Scherzer, T.-M.; Dejaco, C.; Sator, M.; Kessler, H.; Ferenci, P. Seminal fluid ribavirin level and functional semen parameters in patients with chronic hepatitis C on antiviral combination therapy. J. Hepatol. 2010, 52, 812–816. [Google Scholar] [CrossRef]
  121. Bukhari, S.A.; Ahmed, M.M.; Anjum, F.; Anwar, H.; Naqvi, S.A.R.; Zahra, T.; Batool, U. Post interferon therapy decreases male fertility through gonadotoxic effect. Pak. J. Pharm. Sci. 2018, 31, 1565–1570. [Google Scholar] [PubMed]
  122. Kechagia, M.; Basoulis, D.; Konstantopoulou, S.; Dimitriadi, D.; Gyftopoulou, K.; Skarmoutsou, N.; Fakiri, E.M. Health Benefits of Probiotics: A Review. Int. Sch. Res. Not. 2013, 2013, 481651. [Google Scholar] [CrossRef] [PubMed]
  123. Younis, N.; Mahasneh, A. Probiotics and the envisaged role in treating human infertility. Middle East Fertil. Soc. J. 2020, 25, 33. [Google Scholar] [CrossRef]
  124. Unno, T.; Choi, J.-H.; Hur, H.-G.; Sadowsky, M.J.; Ahn, Y.-T.; Huh, C.-S.; Kim, G.-B.; Cha, C.-J. Changes in human gut microbiota influenced by probiotic fermented milk ingestion. J. Dairy Sci. 2015, 98, 3568–3576. [Google Scholar] [CrossRef] [PubMed]
  125. Maretti, C.; Cavallini, G. The association of a probiotic with a prebiotic (Flortec, Bracco) to improve the quality/quantity of spermatozoa in infertile patients with idiopathic oligoasthenoteratospermia: A pilot study. Andrology 2017, 5, 439–444. [Google Scholar] [CrossRef]
  126. Mazanko, M.S.; Gorlov, I.F.; Prazdnova, E.V.; Makarenko, M.S.; Usatov, A.V.; Bren, A.B.; Chistyakov, V.A.; Tutelyan, A.V.; Komarova, Z.B.; Mosolova, N.I.; et al. Bacillus Probiotic Supplementations Improve Laying Performance, Egg Quality, Hatching of Laying Hens, and Sperm Quality of Roosters. Probiotics Antimicrob. Proteins 2018, 10, 367–373. [Google Scholar] [CrossRef] [PubMed]
  127. Valcarce, D.G.; Riesco, M.F.; Martínez-Vázquez, J.M.; Robles, V. Diet Supplemented with Antioxidant and Anti-Inflammatory Probiotics Improves Sperm Quality after Only One Spermatogenic Cycle in Zebrafish Model. Nutrients 2019, 11, 843. [Google Scholar] [CrossRef]
  128. Keshtmand, Z.; Akbaribazm, M.; Bagheri, Y.; Oliaei, R. The ameliorative effects of Lactobacillus coagulans and Lactobacillus casei probiotics on CCl4-induced testicular toxicity based on biochemical, histological and molecular analyses in rat. Andrologia 2021, 53, e13908. [Google Scholar] [CrossRef]
  129. Helli, B.; Kavianpour, M.; Ghaedi, E.; Dadfar, M.; Haghighian, H.K. Probiotic effects on sperm parameters, oxidative stress index, inflammatory factors and sex hormones in infertile men. Hum. Fertil. 2022, 25, 499–507. [Google Scholar] [CrossRef]
  130. Barbonetti, A.; Cinque, B.; Vassallo, M.R.C.; Mineo, S.; Francavilla, S.; Cifone, M.G.; Francavilla, F. Effect of vaginal probiotic lactobacilli on in vitro–induced sperm lipid peroxidation and its impact on sperm motility and viability. Fertil. Steril. 2011, 95, 2485–2488. [Google Scholar] [CrossRef]
  131. Kaźmierczak-Siedlecka, K.; Daca, A.; Fic, M.; van de Wetering, T.; Folwarski, M.; Makarewicz, W. Therapeutic methods of gut microbiota modification in colorectal cancer management–fecal microbiota transplantation, prebiotics, probiotics, and synbiotics. Gut Microbes 2020, 11, 1518–1530. [Google Scholar] [CrossRef] [PubMed]
  132. Molina, N.M.; Sola-Leyva, A.; Saez-Lara, M.J.; Plaza-Diaz, J.; Tubić-Pavlović, A.; Romero, B.; Clavero, A.; Mozas-Moreno, J.; Fontes, J.; Altmäe, S. New Opportunities for Endometrial Health by Modifying Uterine Microbial Composition: Present or Future? Biomolecules 2020, 10, 593. [Google Scholar] [CrossRef]
  133. Fooks, L.J.; Gibson, G.R. Probiotics as modulators of the gut flora. Br. J. Nutr. 2002, 88, s39–s49. [Google Scholar] [CrossRef] [PubMed]
  134. van Nood, E.; Speelman, P.; Nieuwdorp, M.; Keller, J.J. Fecal microbiota transplantation. Curr. Opin. Gastroenterol. 2014, 30, 34–39. [Google Scholar] [CrossRef]
  135. Vujkovic-Cvijin, I.; Sklar, J.; Jiang, L.; Natarajan, L.; Knight, R.; Belkaid, Y. Host variables confound gut microbiota studies of human disease. Nature 2020, 587, 448–454. [Google Scholar] [CrossRef] [PubMed]
  136. Bordalo Tonucci, L.; Dos Santos, K.M.; De Luces Fortes Ferreira, C.L.; Ribeiro, S.M.; De Oliveira, L.L.; Martino, H.S. Gut microbiota and probiotics: Focus on diabetes mellitus. Crit. Rev. Food Sci. Nutr. 2017, 57, 2296–2309. [Google Scholar] [CrossRef]
  137. Xavier, M.J.; Salas-Huetos, A.; Oud, M.S.; Aston, K.I.; Veltman, J.A. Disease gene discovery in male infertility: Past, present and future. Hum. Genet. 2020, 140, 7–19. [Google Scholar] [CrossRef] [PubMed]
  138. Lotti, F.; Bertolotto, M.; Maggi, M. Historical trends for the standards in scrotal ultrasonography: What was, what is and what will be normal. Andrology 2021, 9, 1331–1355. [Google Scholar] [CrossRef]
  139. Egle, U.T.; Egloff, N.; von Känel, R. Stressinduzierte Hyperalgesie (SIH) als Folge von emotionaler Deprivation und psychischer Traumatisierung in der Kindheit. Schmerz 2016, 30, 526–536. [Google Scholar] [CrossRef] [PubMed]
  140. Tijani, H.A.; Bhattacharya, S. The role of intrauterine insemination in male infertility. Hum. Fertil. 2010, 13, 226–232. [Google Scholar] [CrossRef]
  141. Wang, Y.; Wang, Y.; Wang, Y. Apoplastic Proteases: Powerful Weapons against Pathogen Infection in Plants. Plant Commun. 2020, 1, 100085. [Google Scholar] [CrossRef] [PubMed]
  142. Sheng, H.; Sun, X.; Yan, Y.; Yuan, Q.; Wang, J.; Shen, X. Metabolic Engineering of Microorganisms for the Production of Flavonoids. Front. Bioeng. Biotechnol. 2020, 8, 589069. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Locations, components, and potential intervention of the male microbiome.
Figure 1. Locations, components, and potential intervention of the male microbiome.
Jpm 13 01491 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kaltsas, A.; Zachariou, A.; Markou, E.; Dimitriadis, F.; Sofikitis, N.; Pournaras, S. Microbial Dysbiosis and Male Infertility: Understanding the Impact and Exploring Therapeutic Interventions. J. Pers. Med. 2023, 13, 1491. https://doi.org/10.3390/jpm13101491

AMA Style

Kaltsas A, Zachariou A, Markou E, Dimitriadis F, Sofikitis N, Pournaras S. Microbial Dysbiosis and Male Infertility: Understanding the Impact and Exploring Therapeutic Interventions. Journal of Personalized Medicine. 2023; 13(10):1491. https://doi.org/10.3390/jpm13101491

Chicago/Turabian Style

Kaltsas, Aris, Athanasios Zachariou, Eleftheria Markou, Fotios Dimitriadis, Nikolaos Sofikitis, and Spyridon Pournaras. 2023. "Microbial Dysbiosis and Male Infertility: Understanding the Impact and Exploring Therapeutic Interventions" Journal of Personalized Medicine 13, no. 10: 1491. https://doi.org/10.3390/jpm13101491

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