Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Immunol Rev ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38873851

ABSTRACT

Systemic lupus erythematosus is a complex autoimmune disease resulting from a dysregulation of the immune system that involves gut dysbiosis and an altered host cellular metabolism. This review highlights novel insights and expands on the interactions between the gut microbiome and the host immune metabolism in lupus. Pathobionts, invasive pathogens, and even commensal microbes, when in dysbiosis, can all trigger and modulate immune responses through metabolic reprogramming. Changes in the microbiota's global composition or individual taxa may trigger a cascade of metabolic changes in immune cells that may, in turn, reprogram their functions. Factors contributing to dysbiosis include changes in intestinal hypoxia, competition for glucose, and limited availability of essential nutrients, such as tryptophan and metal ions, all of which can be driven by host metabolism changes. Conversely, the accumulation of some host metabolites, such as itaconate, succinate, and free fatty acids, could further influence the microbial composition and immune responses. Overall, mounting evidence supports a bidirectional relationship between host immunometabolism and the microbiota in lupus pathogenesis.

2.
Sci Adv ; 10(13): eadi4310, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38536923

ABSTRACT

The maintenance of regulatory T (Treg) cells critically prevents autoimmunity. Pre-B cell leukemia transcription factor 1 (Pbx1) variants are associated with lupus susceptibility, particularly through the expression of a dominant negative isoform Pbx1-d in CD4+ T cells. Pbx1-d overexpression impaired Treg cell homeostasis and promoted inflammatory CD4+ T cells. Here, we showed a high expression of Pbx1 in human and murine Treg cells, which is decreased in lupus patients and mice. Pbx1 deficiency or Pbx1-d overexpression reduced the number, stability, and suppressive activity of Treg cells, which increased murine responses to immunization and autoimmune induction. Mechanistically, Pbx1 deficiency altered the expression of genes implicated in cell cycle and apoptosis in Treg cells. Intriguingly, Rtkn2, a Rho-GTPase previously associated with Treg homeostasis, was directly transactivated by Pbx1. Our results suggest that the maintenance of Treg cell homeostasis and stability by Pbx1 through cell cycle progression prevent the expansion of inflammatory T cells that otherwise exacerbates lupus progression in the hosts.


Subject(s)
Lupus Erythematosus, Systemic , T-Lymphocytes, Regulatory , Animals , Humans , Mice , Cell Division , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Protein Isoforms/genetics , Lupus Erythematosus, Systemic/genetics
3.
bioRxiv ; 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38293097

ABSTRACT

Tryptophan modulates disease activity and the composition of microbiota in the B6.Sle1.Sle2.Sle3 (TC) mouse model of lupus. To directly test the effect of tryptophan on the gut microbiome, we transplanted fecal samples from TC and B6 control mice into germ-free or antibiotic-treated non-autoimmune B6 mice that were fed with a high or low tryptophan diet. The recipient mice with TC microbiota and high tryptophan diet had higher levels of immune activation, autoantibody production and intestinal inflammation. A bloom of Ruminococcus gnavus (Rg), a bacterium associated with disease flares in lupus patients, only emerged in the recipients of TC microbiota fed with high tryptophan. Rg depletion in TC mice decreased autoantibody production and increased the frequency of regulatory T cells. Conversely, TC mice colonized with Rg showed higher autoimmune activation. Overall, these results suggest that the interplay of genetic and tryptophan can influence the pathogenesis of lupus through the gut microbiota.

4.
Front Immunol ; 14: 1187145, 2023.
Article in English | MEDLINE | ID: mdl-37483626

ABSTRACT

Background: Mounting evidence suggests that increased gut permeability, or leaky gut, and the resulting translocation of pathobionts or their metabolites contributes to the pathogenesis of Systemic Lupus Erythematosus. However, the mechanisms underlying the induction of gut leakage remain unclear. In this study, we examined the effect of a treatment with a TLR7/8 agonist in the B6.Sle1.Sle2.Sle3 triple congenic (TC) mouse, a spontaneous mouse model of lupus without gut leakage. Materials and methods: Lupus-prone mice (TC), TC.Rag1-/- mice that lack B and T cells, and congenic B6 healthy controls were treated with R848. Gut barrier integrity was assessed by measuring FITC-dextran in the serum following oral gavage. Claudin-1 and PECAM1 expression as well as the extent of CD45+ immune cells, B220+ B cells, CD3+ T cells and CD11b+ myeloid cells were measured in the ileum by immunofluorescence. NKp46+ cells were measured in the ileum and colon by immunofluorescence. Immune cells in the ileum were also analyzed by flow cytometry. Results: R848 decreased gut barrier integrity in TC but not in congenic control B6 mice. Immunofluorescence staining of the ileum showed a reduced expression of the tight junction protein Claudin-1, endothelial cell tight junction PECAM1, as well as an increased infiltration of immune cells, including B cells and CD11b+ cells, in R848-treated TC as compared to untreated control mice. However, NKp46+ cells which play critical role in maintaining gut barrier integrity, had a lower frequency in treated TC mice. Flow cytometry showed an increased frequency of plasma cells, dendritic cells and macrophages along with a decreased frequency of NK cells in R848 treated TC mice lamina propria. In addition, we showed that the R848 treatment did not induce gut leakage in TC.Rag1-/- mice that lack mature T and B cells. Conclusions: These results demonstrate that TLR7/8 activation induces a leaky gut in lupus-prone mice, which is mediated by adaptive immune responses. TLR7/8 activation is however not sufficient to breach gut barrier integrity in non-autoimmune mice.


Subject(s)
Toll-Like Receptor 7 , Toll-Like Receptor 8 , Mice , Animals , Claudin-1 , Mice, Congenic , Mice, Inbred Strains , Homeodomain Proteins
5.
Curr Opin Immunol ; 78: 102245, 2022 10.
Article in English | MEDLINE | ID: mdl-36122544

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease in which the overactivation of the immune system has been associated with metabolic alterations. Targeting the altered immunometabolism has been proposed to treat SLE patients based on their results obtained and mouse models of the disease. Here, we review the recent literature to discuss the possible origins of the alterations in the metabolism of immune cells in lupus, the dominant role of mitochondrial defects, technological advances that may move the field forward, as well as how targeting lupus immunometabolism may have therapeutic potential.


Subject(s)
Lupus Erythematosus, Systemic , Mice , Animals , Mitochondria/metabolism , Disease Models, Animal
6.
Front Immunol ; 13: 919792, 2022.
Article in English | MEDLINE | ID: mdl-35795671

ABSTRACT

Systemic Lupus Erythematosus is a complex autoimmune disease and its etiology remains unknown. Increased gut permeability has been reported in lupus patients, yet whether it promotes or results from lupus progression is unclear. Recent studies indicate that an impaired intestinal barrier allows the translocation of bacteria and bacterial components into systemic organs, increasing immune cell activation and autoantibody generation. Indeed, induced gut leakage in a mouse model of lupus enhanced disease characteristics, including the production of anti-dsDNA antibody, serum IL-6 as well as cell apoptosis. Gut microbiota dysbiosis has been suggested to be one of the factors that decreases gut barrier integrity by outgrowing harmful bacteria and their products, or by perturbation of gut immune homeostasis, which in turn affects gut barrier integrity. The restoration of microbial balance eliminates gut leakage in mice, further confirming the role of microbiota in maintaining gut barrier integrity. In this review, we discuss recent advances on the association between microbiota dysbiosis and leaky gut, as well as their influences on the progression of lupus. The modifications on host microbiota and gut integrity may offer insights into the development of new lupus treatment.


Subject(s)
Gastrointestinal Microbiome , Lupus Erythematosus, Systemic , Microbiota , Animals , Antibodies, Antinuclear , Dysbiosis/microbiology , Humans , Lupus Erythematosus, Systemic/microbiology
7.
iScience ; 25(5): 104241, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35494242

ABSTRACT

A skewed tryptophan metabolism has been reported in patients with lupus. Here, we investigated the mechanisms by which it occurs in lupus-susceptible mice, and how tryptophan metabolites exacerbate T cell activation. Metabolomic analyses demonstrated that tryptophan is differentially catabolized in lupus mice compared to controls and that the microbiota played a role in this skewing. There was no evidence for differential expression of tryptophan catabolic enzymes in lupus mice, further supporting a major contribution of the microbiota to skewing. However, isolated lupus T cells processed tryptophan differently, suggesting a contribution of T cell intrinsic factors. Functionally, tryptophan and its microbial product tryptamine increased T cell metabolism and mTOR activation, while kynurenine promoted interferon gamma production, all of which have been associated with lupus. These results showed that a combination of microbial and T cell intrinsic factors promotes the production of tryptophan metabolites that enhance inflammatory phenotypes in lupus T cells.

8.
Front Cell Infect Microbiol ; 12: 795230, 2022.
Article in English | MEDLINE | ID: mdl-35360099

ABSTRACT

Chronic otitis media (COM) is the long-term infection and inflammation of the middle ears typically caused by upper respiratory tract pathogens that are able to ascend the Eustachian tube. Our understanding of contributing factors is limited because human otopathogens cannot naturally colonize or persist in the middle ears of mice. We recently described a natural COM in mice caused by Bordetella pseudohinzii and proposed this as an experimental system to study bacterial mechanisms of immune evasion that allow persistent infection of the middle ear. Here we describe a novel pertussis toxin (PTx)-like factor unique to B. pseudohinzii, apparently acquired horizontally, that is associated with its particularly efficient persistence and pathogenesis. The catalytic subunit of this toxin, PsxA, has conserved catalytic sites and substantial predicted structural homology to pertussis toxin catalytic subunit PtxA. Deletion of the gene predicted to encode the catalytic subunit, psxA, resulted in a significant decrease in persistence in the middle ears. The defect was not observed in mice lacking T cells, indicating that PsxA is necessary for persistence only when T cells are present. These results demonstrate the role of a novel putative toxin in the persistence of B. pseudohinzii and its generation of COM. This PsxA-mediated immune evasion strategy may similarly be utilized by human otopathogens, via other PTx-like toxins or alternative mechanisms to disrupt critical T cell functions necessary to clear bacteria from the middle ear. This work demonstrates that this experimental system can allow for the detailed study of general strategies and specific mechanisms that otopathogens use to evade host immune responses to persist in the middle ear to cause COM.


Subject(s)
Otitis Media , Animals , Bacteria , Ear, Middle/microbiology , Inflammation , Mice , Otitis Media/microbiology , Pertussis Toxin
9.
Dis Model Mech ; 15(5)2022 05 01.
Article in English | MEDLINE | ID: mdl-35311902

ABSTRACT

Pertussis (whooping cough) is a highly transmissible human respiratory disease caused by Bordetella pertussis, a human-restricted pathogen. Animal models generally involve pneumonic infections induced by depositing large numbers of bacteria in the lungs of mice. These models have informed us about the molecular pathogenesis of pertussis and guided development of vaccines that successfully protect against severe disease. However, they bypass the catarrhal stage of the disease, when bacteria first colonize and initially grow in the upper respiratory tract. This is a critical and highly transmissible stage of the infection that current vaccines do not prevent. Here, we demonstrate a model system in which B. pertussis robustly and persistently infects the nasopharynx of TLR4-deficient mice, inducing localized inflammation, neutrophil recruitment and mucus production as well as persistent shedding and occasional transmission to cage mates. This novel experimental system will allow the study of the contributions of bacterial factors to colonization of and shedding from the nasopharynx, as occurs during the catarrhal stage of pertussis, and interventions that might better control the ongoing circulation of pertussis.


Subject(s)
Respiratory Tract Infections , Whooping Cough , Animals , Bordetella pertussis , Lung/microbiology , Mice , Pertussis Vaccine , Whooping Cough/microbiology , Whooping Cough/prevention & control
10.
Front Cell Infect Microbiol ; 12: 798317, 2022.
Article in English | MEDLINE | ID: mdl-35223538

ABSTRACT

A variety of bacteria have evolved the ability to interact with environmental phagocytic predators such as amoebae, which may have facilitated their subsequent interactions with phagocytes in animal hosts. Our recent study found that the animal pathogen Bordetella bronchiseptica can evade predation by the common soil amoeba Dictyostelium discoideum, survive within, and hijack its complex life cycle as a propagation and dissemination vector. However, it is uncertain whether the mechanisms allowing interactions with predatory amoebae are conserved among Bordetella species, because divergence, evolution, and adaptation to different hosts and ecological niches was accompanied by acquisition and loss of many genes. Here we tested 9 diverse Bordetella species in three assays representing distinct aspects of their interactions with D. discoideum. Several human and animal pathogens retained the abilities to survive within single-celled amoeba, to inhibit amoebic plaque expansion, and to translocate with amoebae to the fruiting body and disseminate along with the fruiting body. In contrast, these abilities were partly degraded for the bird pathogen B. avium, and for the human-restricted species B. pertussis and B. parapertussis. Interestingly, a different lineage of B. parapertussis only known to infect sheep retained the ability to interact with D. discoideum, demonstrating that these abilities were lost in multiple lineages independently, correlating with niche specialization and recent rapid genome decay apparently mediated by insertion sequences. B. petrii has been isolated sporadically from diverse human and environmental sources, has acquired insertion sequences, undergone genome decay and has also lost the ability to interact with amoebae, suggesting some specialization to some unknown niche. A genome-wide association study (GWAS) identified a set of genes that are potentially associated with the ability to interact with D. discoideum. These results suggest that massive gene loss associated with specialization of some Bordetella species to a closed life cycle in a particular host was repeatedly and independently accompanied by loss of the ability to interact with amoebae in an environmental niche.


Subject(s)
Amoeba , Bordetella bronchiseptica , Bordetella , Dictyostelium , Amoeba/microbiology , Animals , Bordetella/genetics , Bordetella bronchiseptica/genetics , Dictyostelium/microbiology , Genome-Wide Association Study , Sheep/genetics
11.
PLoS Pathog ; 17(8): e1009735, 2021 08.
Article in English | MEDLINE | ID: mdl-34347835

ABSTRACT

Whooping cough is resurging in the United States despite high vaccine coverage. The rapid rise of Bordetella pertussis isolates lacking pertactin (PRN), a key vaccine antigen, has led to concerns about vaccine-driven evolution. Previous studies showed that pertactin can mediate binding to mammalian cells in vitro and act as an immunomodulatory factor in resisting neutrophil-mediated clearance. To further investigate the role of PRN in vivo, we examined the functions of pertactin in the context of a more naturally low dose inoculation experimental system using C3H/HeJ mice that is more sensitive to effects on colonization, growth and spread within the respiratory tract, as well as an experimental approach to measure shedding and transmission between hosts. A B. bronchiseptica pertactin deletion mutant was found to behave similarly to its wild-type (WT) parental strain in colonization of the nasal cavity, trachea, and lungs of mice. However, the pertactin-deficient strain was shed from the nares of mice in much lower numbers, resulting in a significantly lower rate of transmission between hosts. Histological examination of respiratory epithelia revealed that pertactin-deficient bacteria induced substantially less inflammation and mucus accumulation than the WT strain and in vitro assays verified the effect of PRN on the induction of TNF-α by murine macrophages. Interestingly, only WT B. bronchiseptica could be recovered from the spleen of infected mice and were further observed to be intracellular among isolated splenocytes, indicating that pertactin contributes to systemic dissemination involving intracellular survival. These results suggest that pertactin can mediate interactions with immune cells and augments inflammation that contributes to bacterial shedding and transmission between hosts. Understanding the relative contributions of various factors to inflammation, mucus production, shedding and transmission will guide novel strategies to interfere with the reemergence of pertussis.


Subject(s)
Alveolar Epithelial Cells/microbiology , Bacterial Outer Membrane Proteins/metabolism , Bacterial Shedding , Bordetella Infections/transmission , Bordetella bronchiseptica/pathogenicity , Inflammation/pathology , Virulence Factors, Bordetella/metabolism , Animals , Bacterial Adhesion , Bacterial Outer Membrane Proteins/genetics , Bordetella Infections/metabolism , Bordetella Infections/microbiology , Female , Humans , Inflammation/metabolism , Inflammation/microbiology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Virulence Factors, Bordetella/genetics
12.
Emerg Infect Dis ; 27(6): 1561-1566, 2021 06.
Article in English | MEDLINE | ID: mdl-34014152

ABSTRACT

Recent reemergence of pertussis (whooping cough) in highly vaccinated populations and rapid expansion of Bordetella pertussis strains lacking pertactin (PRN), a common acellular vaccine antigen, have raised the specter of vaccine-driven evolution and potential return of what was once the major killer of children. The discovery that most circulating B. pertussis strains in the United States have acquired new and independent disruptive mutations in PRN is compelling evidence of strong selective pressure. However, the other 4 antigens included in acellular vaccines do not appear to be selected against so rapidly. We consider 3 aspects of PRN that distinguish it from other vaccine antigens, which might, individually or collectively, explain why only this antigen is being precipitously eliminated. An understanding of the increase in PRN-deficient strains should provide useful information for the current search for new protective antigens and provide broader lessons for the design of improved subunit vaccines.


Subject(s)
Bordetella pertussis , Whooping Cough , Bacterial Outer Membrane Proteins , Child , Humans , Pertussis Vaccine , Virulence Factors, Bordetella
13.
Front Cell Infect Microbiol ; 11: 815627, 2021.
Article in English | MEDLINE | ID: mdl-35141173

ABSTRACT

Acute otitis media (AOM) is commonly caused by bacterial pathobionts of the nasopharynx that ascend the Eustachian tube to cause disease in the middle ears. To model and study the various complexities of AOM, common human otopathogens are injected directly into the middle ear bullae of rodents or are delivered with viral co-infections which contribute to the access to the middle ears in complex and partially understood ways. Here, we present the novel observation that Bordetella bronchiseptica, a well-characterized respiratory commensal/pathogen of mice, also efficiently ascends their Eustachian tubes to colonize their middle ears, providing a flexible mouse model to study naturally occurring AOM. Mice lacking T and/or B cells failed to resolve infections, highlighting the cooperative role of both in clearing middle ear infection. Adoptively transferred antibodies provided complete protection to the lungs but only partially protected the middle ears, highlighting the differences between respiratory and otoimmunology. We present this as a novel experimental system that can capitalize on the strengths of the mouse model to dissect the molecular mechanisms involved in the generation and function of immunity within the middle ear.


Subject(s)
Bordetella bronchiseptica , Eustachian Tube , Otitis Media , Animals , Ear, Middle/microbiology , Eustachian Tube/microbiology , Mice , Nasopharynx/microbiology , Otitis Media/microbiology
15.
Curr Opin Infect Dis ; 32(3): 223-230, 2019 06.
Article in English | MEDLINE | ID: mdl-30921085

ABSTRACT

PURPOSE OF REVIEW: To relate genomic changes to phenotypic adaptation and evolution from environmental bacteria to obligate human pathogens, focusing on the examples within Bordetella species. RECENT FINDINGS: Recent studies showed that animal-pathogenic and human-pathogenic Bordetella species evolved from environmental ancestors in soil. The animal-pathogenic Bordetella bronchiseptica can hijack the life cycle of the soil-living amoeba Dictyostelium discoideum, surviving inside single-celled trophozoites, translocating to the fruiting bodies and disseminating along with amoeba spores. The association with amoeba may have been a 'training ground' for bacteria during the evolution to pathogens. Adaptation to an animal-associated life style was characterized by decreasing metabolic versatility and genome size and by acquisition of 'virulence factors' mediating the interaction with the new animal hosts. Subsequent emergence of human-specific pathogens, such as Bordetella pertussis from zoonoses of broader host range progenitors, was accompanied by a dramatic reduction in genome size, marked by the loss of hundreds of genes. SUMMARY: The evolution of Bordetella from environmental microbes to animal-adapted and obligate human pathogens was accompanied by significant genome reduction with large-scale gene loss during divergence.


Subject(s)
Adaptation, Biological , Adaptation, Physiological , Biological Evolution , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/physiology , Bordetella pertussis/genetics , Bordetella pertussis/physiology , Animals , Host-Pathogen Interactions , Humans , Soil Microbiology
16.
Front Microbiol ; 10: 2839, 2019.
Article in English | MEDLINE | ID: mdl-31921025

ABSTRACT

Animal and human pathogens of the genus Bordetella are not commonly considered to be intracellular pathogens, although members of the closely related classical bordetellae are known to enter and persist within macrophages in vitro and have anecdotally been reported to be intracellular in clinical samples. B. bronchiseptica, the species closest to the ancestral lineage of the classical bordetellae, infects a wide range of mammals but is known to have an alternate life cycle, persisting, replicating and disseminating with amoeba. These observations give rise to the hypothesis that the ability for intracellular survival has an ancestral origin and is common among animal-pathogenic and environmental Bordetella species. Here we analyzed the survival of B. bronchiseptica and defined its transcriptional response to internalization by murine macrophage-like cell line RAW 264.7. Although the majority of the bacteria were killed and digested by the macrophages, a consistent fraction survived and persisted inside the phagocytes. Internalization prompted the activation of a prominent stress response characterized by upregulation of genes involved in DNA repair, oxidative stress response, pH homeostasis, chaperone functions, and activation of specific metabolic pathways. Cross species genome comparisons revealed that most of these upregulated genes are highly conserved among both the classical and non-classical Bordetella species. The diverse Bordetella species also shared the ability to survive inside RAW 264.7 cells, with the single exception being the bird pathogen B. avium, which has lost several of those genes. Knock-out mutations in genes expressed intracellularly resulted in decreased persistence inside the phagocytic cells, emphasizing the importance of these genes in this environment. These data show that the ability to persist inside macrophage-like RAW 264.7 cells is shared among nearly all Bordetella species, suggesting that resisting phagocytes may be an ancient mechanism that precedes speciation in the genus and may have facilitated the adaptation of Bordetella species from environmental bacteria to mammalian respiratory pathogens.

17.
Front Immunol ; 9: 1148, 2018.
Article in English | MEDLINE | ID: mdl-29910805

ABSTRACT

Japanese encephalitis is a neuropathological disorder caused by Japanese encephalitis virus (JEV), which is characterized by severe pathological neuroinflammation and damage to the blood-brain barrier (BBB). Inflammatory cytokines/chemokines can regulate the expression of tight junction (TJ) proteins and are believed to be a leading cause of BBB disruption, but the specific mechanisms remain unclear. IP-10 is the most abundant chemokine produced in the early stage of JEV infection, but its role in BBB disruption is unknown. The administration of IP-10-neutralizing antibody ameliorated the decrease in TJ proteins and restored BBB integrity in JEV-infected mice. In vitro study showed IP-10 and JEV treatment did not directly alter the permeability of the monolayers of endothelial cells. However, IP-10 treatment promoted tumor necrosis factor alpha (TNF-α) production and IP-10-neutralizing antibody significantly reduced the production of TNF-α. Thus, TNF-α could be a downstream cytokine of IP-10, which decreased TJ proteins and damaged BBB integrity. Further study indicated that JEV infection can stimulate upregulation of the IP-10 receptor CXCR3 on astrocytes, resulting in TNF-α production through the JNK-c-Jun signaling pathway. Consequently, TNF-α affected the expression and cellular distribution of TJs in brain microvascular endothelial cells and led to BBB damage during JEV infection. Regarding regulation of the BBB, the IP-10/TNF-α cytokine axis could be considered a potential target for the development of novel therapeutics in BBB-related neurological diseases.


Subject(s)
Blood-Brain Barrier/metabolism , Chemokine CXCL10/metabolism , Encephalitis, Japanese/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Astrocytes/metabolism , Astrocytes/virology , Cell Line , Disease Models, Animal , Encephalitis Virus, Japanese/physiology , Encephalitis, Japanese/etiology , Endothelium/metabolism , Female , Gene Expression , MAP Kinase Signaling System , Mice , Permeability , Tumor Necrosis Factor-alpha/genetics
18.
Environ Toxicol Pharmacol ; 50: 111-118, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28171822

ABSTRACT

The male reproductive toxicity of melamine (MA) has been recognized in recent years excepted for its renal toxicity. Our previous in vivo studies revealed that the damages of Sertoli cell barrier played a critical role in MA-induced testicular toxicity in mice. Herein, we performed an in vitro study to comprehensively evaluate the toxicity of MA on Sertoli cell by examining the influences of MA on the viability, morphology, mortality and intercellular junctions of mouse TM4 Sertoli cells (TM4 cells). The results showed that MA suppressed cell viability, induced obvious ultrastructural changes and cell apoptosis in concentration-dependent manner. Moreover, MA down-regulated the expressions of junction-associated proteins including occludin, N-cadherin, and vimentin, suggesting that MA disrupted the integrity of Sertoli cell barrier. Thus, these results indicated that Sertoli cell might be an important cellular target for MA-induced male reproductive toxicity.


Subject(s)
Cadherins/metabolism , Occludin/metabolism , Sertoli Cells/drug effects , Triazines/toxicity , Vimentin/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Down-Regulation , In Vitro Techniques , Male , Mice , Sertoli Cells/cytology , Toxicity Tests
19.
Toxicol Ind Health ; 33(1): 36-45, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27573348

ABSTRACT

The health effects of bisphenol A (BPA) have become a great concern in recent years. In this study, the reproductive toxicity of BPA was investigated. Male CD-1 mice were orally administrated with BPA (0, 100, 300 and 600 mg kg-1 body weight) for 56 consecutive days. Results showed that relative testis weight to total body weight was significantly lower in the high-dose group ( p < 0.01, p < 0.05). Microscopic examination under light and transmission electron microscopes showed disorders of spermatogenesis after BPA exposure, including rough basal lamina of seminiferous tubules and damage of tight junctions between Sertoli cells. Further study by terminal-deoxynucleoitidyl transferase-mediated nick end labelling assay showed a significant induction of apoptosis in the testis tissue of the BPA groups ( p < 0.01). Immunohistochemical study found that the expression of androgen-binding protein (ABP) was significantly decreased in BPA-treated mice ( p < 0.01). Our results indicated that impairment of the basal lamina of seminiferous tubules and tight junctions may contribute to BPA-induced cell injury. A decrease in the level of ABP could be the possible mechanism for the reproductive toxicity of BPA. These findings provided direct evidence and novel insight into the reproductive toxicity of BPA and may have implications for understanding the toxicity of other endocrine disruptors.


Subject(s)
Benzhydryl Compounds/toxicity , Phenols/toxicity , Testis/drug effects , Animals , Apoptosis , Endocrine Disruptors/toxicity , Male , Mice , Reproduction/drug effects , Seminiferous Tubules/drug effects , Seminiferous Tubules/pathology , Sertoli Cells/drug effects , Sertoli Cells/pathology , Spermatogenesis/drug effects , Testis/pathology , Tight Junctions/drug effects
20.
Reprod Toxicol ; 46: 1-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24607646

ABSTRACT

Eight-week-old male Kunming mice were administered either melamine (MA, 30, 140, or 700 mg/kg/day), a melamine and cyanuric acid mixture (MC, each at 15, 70, or 350 mg/kg/day), or vehicle (control) for 3 consecutive days. Testicular toxicity was evaluated on days 1 and 5 after the final exposure. The testicular and epididymal weights and serum testosterone level were significantly decreased in the highest MC group (350 mg/kg/day). Histopathologically, both MA and MC caused obvious lesions in the testis and epididymis, with significant increases in sperm abnormalities. By TEM, the blood-testis barrier was damaged dose dependently. TUNEL staining showed that both MA and MC induced increases in germ cell apoptosis. The Sertoli cell vimentin was collapsed in the treated animals as detected by immunohistochemical staining and Western blotting. This study demonstrated that both MA and MC treatments could disrupt the blood-testis barrier and cause a clear testicular toxicity.


Subject(s)
Testicular Diseases/chemically induced , Triazines/toxicity , Animals , Apoptosis/drug effects , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/ultrastructure , Body Weight/drug effects , Epididymis/pathology , Male , Mice , Organ Size/drug effects , Sertoli Cells/pathology , Spermatozoa/abnormalities , Spermatozoa/drug effects , Testicular Diseases/pathology , Testis/pathology , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...