Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Cell ; 167(6): 1511-1524.e10, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27884405

ABSTRACT

Zika virus (ZIKV) persists in the semen of male patients, a first for flavivirus infection. Here, we demonstrate that ZIKV can induce inflammation in the testis and epididymidis, but not in the prostate or seminal vesicle, and can lead to damaged testes after 60 days post-infection in mice. ZIKV induces innate immune responses in Leydig, Sertoli, and epididymal epithelial cells, resulting in the production of pro-inflammatory cytokines/chemokines. However, ZIKV does not induce a rapid and abundant cytokine production in peritubular cell and spermatogonia, suggesting that these cells are vulnerable for ZIKV infection and could be the potential repositories for ZIKV. Our study demonstrates a correlation between ZIKV and testis infection/damage and suggests that ZIKV infection, under certain circumstances, can eventually lead to male infertility.


Subject(s)
Infertility, Male/virology , Testis/virology , Zika Virus Infection/virology , Zika Virus/physiology , Animals , Cytokines/metabolism , Epididymis/pathology , Epididymis/virology , Humans , Infertility, Male/pathology , Male , Mice , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Interferon alpha-beta/genetics , Testis/pathology , Virus Internalization , Zika Virus/isolation & purification , Zika Virus Infection/pathology , Zika Virus Infection/transmission , Axl Receptor Tyrosine Kinase
3.
J Med Virol ; 96(6): e29733, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38874268

ABSTRACT

Viruses in human semen may be sexually transmitted via free and cell-mediated viral infection. The potential effects of semen on the infection and sexual transmission of most viruses in semen remain largely unclear. The present study elucidated the inhibitory effects of human seminal plasma (SP) on Jurkat cell (JC)-mediated mumps virus (MuV) infection. We demonstrated that MuV efficiently infected JCs and that the JCs infected by MuV (JC-MuV) mediated MuV infection of HeLa cells. Remarkably, SP was highly cytotoxic to JCs and inhibited JC-MuV infection of HeLa cells. The cytotoxic factor possessed a molecular weight of less than 3 kDa, whereas that of the viricidal factor was over 100 kDa. The cooperation of cytotoxic and viricidal factors was required for the SP inhibition of JC-MuV infection, and prostatic fluid (PF) was responsible for both the cytotoxic and viricidal effects of SP. The cytotoxic effects we observed were resistant to the treatment of PF with boiling water, proteinase K, RNase A, and DNase I. Our results provide novel insights into the antiviral properties of SP, which may limit cell-mediated sexual viral transmission.


Subject(s)
Mumps virus , Semen , Humans , Mumps virus/physiology , Semen/virology , Male , HeLa Cells , Lymphocytes/virology , Jurkat Cells , Cell Survival , Molecular Weight
4.
Biol Reprod ; 105(1): 113-127, 2021 07 02.
Article in English | MEDLINE | ID: mdl-33899078

ABSTRACT

Three major pathogenic states of the prostate, including benign prostatic hyperplasia, prostate cancer, and prostatitis, are related to the local inflammation. However, the mechanisms underlying the initiation of prostate inflammation remain largely unknown. Given that the innate immune responses of the tissue-specific cells to microbial infection or autoantigens contribute to local inflammation, this study focused on pattern recognition receptor (PRR)-initiated innate immune responses in mouse prostatic epithelial cells (PECs). Primary mouse PECs abundantly expressed Toll-like receptor 3 (TLR3), TLR4, TLR5, melanoma differentiation-associated protein 5 (MDA5), and IFN-inducible protein 16 (p204 in mouse). These PRRs can be activated by their respective ligands: lipopolysaccharide (LPS) and flagellin of Gram-negative bacteria for TLR4 and TLR5, polyinosinic-polycytidylic acid (poly(I:C)) for TLR3 and MDA5, and herpes simplex virus DNA analog (HSV60) for p204. LPS and flagellin predominantly induced the expression of inflammatory cytokines, including tumor necrosis factor alpha (TNFA), interleukin 6 (IL6), chemokines monocyte chemoattractant protein-1 (MCP1), and C-X-C motif chemokine 10 (CXCL10). Poly(I:C) and HSV60 predominantly induced the expression of type 1 interferons (IFNA and IFNB) and antiviral proteins: Mx GTPase 1, 2',5'-oligoadenylate synthetase 1, and IFN-stimulated gene 15. The replication of mumps virus in PECs was inhibited by type 1 IFN signaling. These findings provide insights into the mechanisms underlying innate immune response in the prostate.


Subject(s)
Immunity, Innate/genetics , Prostate/immunology , Receptors, Pattern Recognition/genetics , Animals , Epithelial Cells/immunology , Inflammation/genetics , Male , Mice , Mice, Inbred C57BL , Receptors, Pattern Recognition/immunology
5.
J Virol ; 94(17)2020 08 17.
Article in English | MEDLINE | ID: mdl-32611752

ABSTRACT

Japanese encephalitis virus (JEV) is a flavivirus that causes Japanese encephalitis (JE), which has an unclear pathogenesis. Despite vaccination, thousands of deaths attributed to JE are reported annually. In this study, we report that mice deficient for Axl, a receptor tyrosine kinase that plays multiple roles in flaviviral infection, displayed greater mortality upon JEV infection. The effect of Axl deficiency on JEV infection was mediated by markedly elevated serum interleukin-1α (IL-1α) levels, which devastated the blood-brain-barrier and promoted viral neuroinvasion within 24 h postinfection. Using an in situ infection model, we showed that dead macrophages were the primary source of observed increased serum IL-1α levels. Axl deficiency enhanced cell death and caused pyroptosis in 80% of JEV-infected macrophages by disrupting phosphatidylinositol 3-kinase (PI3K)-Akt signaling. Intriguingly, the primary effector released by pyroptotic macrophages in our model was IL-1α rather than IL-1ß. Finally, we assessed the effect of an IL-1α antagonist and demonstrated that it effectively prevented the incidence of JE. Our results indicate that Axl plays a protective role in JEV infection, identify IL-1α released by pyroptotic macrophages as a crucial factor promoting JEV neuroinvasion, and suggest that an IL-1α antagonist may be a candidate for JE therapy.IMPORTANCE Japanese encephalitis virus (JEV) is a mosquito-borne flavivirus that causes Japanese encephalitis (JE), the most commonly diagnosed viral encephalitis worldwide. The fatality rate of JE is 20%, and nearly half of the surviving patients develop neuropsychiatric sequelae. Axl is a receptor tyrosine kinase that plays multiple roles in flaviviral infections. Currently, the involvement of Axl in JEV infection remains enigmatic. In this study, we demonstrate that Axl impedes the pathogenesis of severe JE in mice by maintaining blood-brain-barrier (BBB) integrity and restricting viral neuroinvasion. Furthermore, serum IL-1α is a key mediator of this process and is primarily released by JEV-infected pyroptotic macrophages to elicit BBB breakdown, while an IL-1α antagonist can effectively reduce the incidence of severe JE. Our work uncovers the protective role of Axl in antagonizing severe JE and shows that the use of an IL-1α antagonist may be a promising tactic to prevent severe JE.


Subject(s)
Encephalitis Virus, Japanese/physiology , Encephalitis, Japanese/virology , Interleukin-1alpha/metabolism , Macrophages/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Blood-Brain Barrier/metabolism , Brain/virology , Disease Models, Animal , Encephalitis, Viral/virology , Female , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Pyroptosis , Axl Receptor Tyrosine Kinase
6.
Adv Exp Med Biol ; 1288: 49-67, 2021.
Article in English | MEDLINE | ID: mdl-34453731

ABSTRACT

Mammalian spermatogenesis is a carefully orchestrated male germ cell differentiation process by which spermatogonia differentiate to spermatozoa in the testis. A highly organized testicular microenvironment is therefore necessary to support spermatogenesis. Regarding immunologic aspects, the testis adapts a specialized immune environment for the protection of male germ cells and testicular functions. The mammalian testis possesses two immunologic features: (1) it is an immunoprivileged organ where immunogenic germ cells do not induce deleterious immune responses under physiologic conditions; and (2) it creates its own effective innate defense system against microbial infection. Various pathologic conditions may disrupt testicular immune homeostasis, thereby resulting in a detrimental immune response and perturbing testicular functions, one of the etiologic factors of male infertility. Understanding the mechanisms underlying immunoregulation in the testis can aid in establishing strategies for the prevention and therapy of immunologic testicular dysfunction and male infertility. This chapter focuses on the mechanisms underlying immune privilege, local innate immunity, and immunologic diseases of the testis.


Subject(s)
Spermatogenesis , Testis , Animals , Humans , Immunity, Innate , Male , Spermatogonia , Spermatozoa
7.
FASEB J ; 33(11): 12528-12540, 2019 11.
Article in English | MEDLINE | ID: mdl-31450968

ABSTRACT

Mumps virus (MuV) has high tropism to the testis and may lead to male infertility. Sertoli cells are the major targets of MuV infection. However, the mechanisms by which MuV infection impairs male fertility and Sertoli cell function remain unclear. The present study elucidated the effect of MuV infection on the blood-testis barrier (BTB). The transepithelial electrical resistance of MuV-infected mouse Sertoli cells was monitored, and the expression of major proteins of the BTB was examined. We demonstrated that MuV infection disrupted the BTB by reducing the levels of occludin and zonula occludens 1. Sertoli cells derived from Tlr2-/- and Tnfa-/- mice were analyzed for mediating MuV-induced impairment. TLR2-mediated TNF-α production by Sertoli cells in response to MuV infection impaired BTB integrity. MuV-impaired BTB was not observed in Tlr2-/- and Tnfa-/- Sertoli cells. Moreover, an inhibitor of TNF-α, pomalidomide, prevents the disruption of BTB in response to MuV infection. FITC-labeled biotin tracing assay confirmed that BTB permeability and spermatogenesis were transiently impaired by MuV infection in vivo. These findings suggest that the disruption of the BTB could be one of the mechanisms underlying MuV-impaired male fertility, in which TNF-α could play a critical role.-Wu, H., Jiang, X., Gao, Y., Liu, W., Wang, F., Gong, M., Chen, R., Yu, X., Zhang, W., Gao, B., Song, C., Han, D. Mumps virus infection disrupts blood-testis barrier through the induction of TNF-α in Sertoli cells.


Subject(s)
Blood-Testis Barrier/metabolism , Mumps virus/metabolism , Mumps/metabolism , Sertoli Cells/metabolism , Spermatogenesis , Tumor Necrosis Factor-alpha/metabolism , Animals , Blood-Testis Barrier/pathology , Blood-Testis Barrier/virology , Infertility, Male/genetics , Infertility, Male/metabolism , Infertility, Male/pathology , Infertility, Male/virology , Male , Mice , Mice, Knockout , Mumps/genetics , Mumps/pathology , Mumps virus/genetics , Sertoli Cells/pathology , Sertoli Cells/virology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism
8.
Biol Reprod ; 100(3): 849-861, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30398566

ABSTRACT

Systemic inflammation may impair male fertility, and its underlying mechanisms remain poorly understood. The present study investigates the effect of lipopolysaccharide (LPS)-induced systemic inflammation on the testis and epididymis in mice. Intraperitoneal injection of LPS significantly impaired testicular functions, including testosterone production, spermatogenesis, and blood-testis barrier permeability. The epididymitis characterized by leukocyte infiltration and fibrosis was observed in the cauda epididymis after LPS injection. LPS-induced testicular dysfunction and epididymitis were abolished in tumor necrosis factor alpha (Tnfa) knockout mice. Pomalidomide, a TNFA inhibitor, blocked the detrimental effects of LPS on the testis and epididymis. The results indicate that LPS-induced systemic inflammation impairs male fertility through TNFA production, suggesting that the intervention on TNFA production would be considered for the prevention and treatment of inflammatory impairment of male fertility.


Subject(s)
Epididymitis/chemically induced , Gene Expression Regulation/drug effects , Lipopolysaccharides/toxicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Epididymitis/prevention & control , Immunologic Factors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/genetics
9.
Biol Reprod ; 101(4): 733-747, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31350848

ABSTRACT

The seminal vesicles can be infected by microorganisms, thereby resulting in vesiculitis and impairment in male fertility. Innate immune responses in seminal vesicles cells to microbial infections, which facilitate vesiculitis, have yet to be investigated. The present study aims to elucidate pattern recognition receptor-mediated innate immune responses in seminal vesicles epithelial cells. Various pattern recognition receptors, including Toll-like receptor 3, Toll-like receptor 4, cytosolic ribonucleic acid, and deoxyribonucleic acid sensors, are abundantly expressed in seminal vesicles epithelial cells. These pattern recognition receptors can recognize their respective ligands, thus activating nuclear factor kappa B and interferon regulatory factor 3. The pattern recognition receptor signaling induces expression of pro-inflammatory cytokines, such as tumor necrosis factor alpha (Tnfa) and interleukin 6 (Il6), chemokines monocyte chemoattractant protein-1 (Mcp1) and C-X-C motif chemokine 10 (Cxcl10), and type 1 interferons Ifna and Ifnb. Moreover, pattern recognition receptor-mediated innate immune responses up-regulated the expression of microsomal prostaglandin E synthase and cyclooxygenase 2, but they down-regulated semenogelin-1 expression. These results provide novel insights into the mechanism underlying vesiculitis and its impact on the functions of the seminal vesicles.


Subject(s)
Epithelial Cells/immunology , Immunity, Innate/genetics , Receptors, Pattern Recognition/physiology , Seminal Vesicles/immunology , Animals , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Poly I-C , Receptors, Pattern Recognition/genetics , Seminal Vesicles/cytology , Seminal Vesicles/metabolism , Signal Transduction
10.
Semin Cell Dev Biol ; 59: 157-165, 2016 11.
Article in English | MEDLINE | ID: mdl-26805443

ABSTRACT

The mammalian testis possesses a unique immune environment that is essential for testicular function. The testis is a remarkable immunoprivileged site that protects immunogenic germ cells from the detrimental effects of immune responses. However, the testis can be infected by various microbial pathogens. To overcome the immune privilege and enable testicular defense against microbes, the testis adopts local effective innate immune responses to microbial infections. The mechanisms underlying the testicular immune privilege have been investigated for several decades and the innate defense system in the testis is being revealed based on the identification of pattern recognition receptor-initiated innate immune responses in testicular cells. The coordination between immune privilege and local innate immune responses is critical in the maintenance of testicular immune homeostasis. Disruption of the testicular immune homeostasis may lead to orchitis and impair spermatogenesis, an etiological factor of male infertility. Dissection of the immunoregulatory mechanisms in the testis can aid in establishing preventive and therapeutic approaches for orchitis. This review discusses current understanding of the mechanisms which underlie the testicular immunoregulation and its effect on spermatogenesis.


Subject(s)
Spermatogenesis , Testis/immunology , Animals , Humans , Immunity, Innate , Leukocytes/metabolism , Male , Models, Biological , Signal Transduction
11.
Andrologia ; 50(11): e13140, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30569651

ABSTRACT

The detrimental effects of Zika virus (ZIKV) infection on mouse testicular functions have reminded a viral threat to male fertility. A broad range of virus families has tropism for male reproductive system, particularly the testes. Certain virus types of these viruses, such as mumps virus and human immunodeficiency virus (HIV), may severely damage the testes and consequently lead to male infertility. ZIKV has been recently found to damage testicular functions and lead to male infertility in mice. Many other viruses also have detrimental effects on host reproduction. Public attention has been paid to sexually transmitted viruses, such as HIV and hepatitis B and C viruses in humans and likewise in economically important farm animals. This article provides an overview on main viruses affecting the male reproductive system and their detrimental effects on fertility, and outlines some important issues for future study.


Subject(s)
Infertility, Male/immunology , Sexually Transmitted Diseases, Viral/immunology , Testis/pathology , Virus Diseases/immunology , Viruses/pathogenicity , Animals , Fertility/immunology , Humans , Infertility, Male/pathology , Infertility, Male/virology , Male , Mice , Sexually Transmitted Diseases, Viral/complications , Sexually Transmitted Diseases, Viral/pathology , Sexually Transmitted Diseases, Viral/virology , Testis/immunology , Testis/virology , Virus Diseases/complications , Virus Diseases/pathology , Virus Diseases/virology , Viruses/immunology
12.
J Gen Virol ; 98(8): 2061-2068, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28786784

ABSTRACT

Recently, Zika virus (ZIKV) outbreak has been associated with a sharp increase in cases of Guillain-Barré syndrome and severe fetal abnormalities. However, the mechanism underlying the interaction of ZIKV with host cells is not yet clear. Axl, a receptor tyrosine kinase, is postulated as a receptor for ZIKV entry; however, its in vivo role during ZIKV infection and its impact on the outcome of the disease have not been fully characterized and evaluated. Moreover, there are contradictory results on its involvement in ZIKV infection. Here we utilized Axl-deficient mice (Axl-/-) and their littermates (Axl+/-) to study the in vivo role of Axl in ZIKV infection. Our results showed that both Axl+/- and Axl-/- suckling mice supported the replication of ZIKV and presented clinical manifestations. No significant difference has been found between Axl-deficient mice and their littermates in terms of the survival rate, clinical manifestations, viral load, ZIKV distribution and histopathological changes in major organs. These results therefore indicate that Axl is not an indispensable factor for ZIKV infection in mice.


Subject(s)
Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Zika Virus Infection/enzymology , Zika Virus/physiology , Animals , Female , Host-Pathogen Interactions , Humans , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Virus Internalization , Virus Replication , Zika Virus/genetics , Zika Virus Infection/genetics , Zika Virus Infection/virology , Axl Receptor Tyrosine Kinase
13.
Reproduction ; 153(6): 821-834, 2017 06.
Article in English | MEDLINE | ID: mdl-28351933

ABSTRACT

Viral infections of the ovary may perturb ovarian functions. However, the mechanisms underlying innate immune responses in the ovary are poorly understood. The present study demonstrates that cytosolic viral DNA sensor signaling initiates the innate immune response in mouse ovarian granulosa cells and affects endocrine function. The cytosolic DNA sensors p204 and cGAS and their common signaling adaptor stimulator of interferon (IFN) genes (STING) were constitutively expressed in granulosa cells. Transfection with VACV70, a synthetic vaccinia virus (VACV) DNA analog, induced the expression of type I interferons (IFNA/B) and major inflammatory cytokines (TNFA and IL6) through IRF3 and NF-κB activation respectively. Moreover, several IFN-inducible antiviral proteins, including 2',5'-oligoadenylate synthetase, IFN-stimulating gene 15 and Mx GTPase 1, were also induced by VACV70 transfection. The innate immune responses in granulosa cells were significantly reduced by the transfection of specific small-interfering RNAs targeting p204, cGas or Sting Notably, the VACV70-triggered innate immune responses affected steroidogenesis in vivo and in vitro The data presented in this study describe the mechanism underlying ovarian immune responses to viral infection.


Subject(s)
Cytosol/metabolism , DNA, Viral/administration & dosage , Granulosa Cells/immunology , Immunity, Innate/immunology , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Phosphoproteins/metabolism , Animals , DNA, Viral/chemistry , DNA, Viral/genetics , Female , Granulosa Cells/drug effects , Immunity, Innate/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Signal Transduction
14.
FASEB J ; 30(4): 1436-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26678449

ABSTRACT

Earlier studies have shown that rats treated with an acute dose of 1-(2,4-dichlorobenzyl)-1H-indazole-3-carbohydrazide (adjudin, a male contraceptive under development) causes permanent infertility due to irreversible blood-testis barrier (BTB) disruption even though the population of undifferentiated spermatogonia remains similar to normal rat testes, because spermatogonia fail to differentiate into spermatocytes to enter meiosis. Since other studies have illustrated the significance of connexin 43 (Cx43)-based gap junction in maintaining the homeostasis of BTB in the rat testis and the phenotypes of Sertoli cell-conditional Cx43 knockout mice share many of the similarities of the adjudin-treated rats, we sought to examine if overexpression of Cx43 in these adjudin-treated rats would reseal the disrupted BTB and reinitiate spermatogenesis. A full-length Cx43 cloned into mammalian expression vector pCI-neo was used to transfect testes of adjudin-treated ratsversusempty vector. It was found that overexpression of Cx43 indeed resealed the Sertoli cell tight junction-permeability barrier based on a functionalin vivoassay in tubules displaying signs of meiosis as noted by the presence of round spermatids. Thus, these findings suggest that overexpression of Cx43 reinitiated spermatogenesis at least through the steps of meiosis to generate round spermatids in testes of rats treated with an acute dose of adjudin that led to aspermatogenesis. It was also noted that the round spermatids underwent eventual degeneration with the formation of multinucleated cells following Cx43 overexpression due to the failure of spermiogenesis because no elongating/elongated spermatids were detected in any of the tubules examined. The mechanism by which overexpression of Cx43 reboots meiosis and rescues BTB function was also examined. In summary, overexpression of Cx43 in the testis with aspermatogenesis reboots meiosis and reseals toxicant-induced BTB disruption, even though it fails to support round spermatids to enter spermiogenesis.-Li, N., Mruk, D. D., Mok, K.-W., Li, M. W. M., Wong, C. K. C., Lee, W. M., Han, D., Silvestrini, B., Cheng, C. Y. Connexin 43 reboots meiosis and reseals blood-testis barrier following toxicant-mediated aspermatogenesis and barrier disruption.


Subject(s)
Blood-Testis Barrier/metabolism , Connexin 43/genetics , Meiosis/genetics , Spermatogenesis/genetics , Animals , Blood-Testis Barrier/drug effects , Connexin 43/metabolism , Gene Expression/drug effects , Hydrazines/pharmacology , Immunoblotting , Indazoles/pharmacology , Male , Mice, Knockout , Microscopy, Fluorescence , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Spermatids/drug effects , Spermatids/metabolism , Spermatogenesis/drug effects , Spermatogonia/drug effects , Spermatogonia/metabolism , Testis/cytology , Testis/drug effects , Testis/metabolism
15.
J Immunol ; 194(10): 4825-35, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25840915

ABSTRACT

Viral infections of the epididymis may impair male fertility and spread sexually transmitted pathogens. The innate antiviral immune responses in the epididymis have yet to be intensively investigated. This study found that mouse epididymal epithelial cells (EECs) constitutively express several viral sensors, including TLR3, retinoic acid-inducible gene I, and DNA-dependent activator of IFN regulatory factors. Other DNA sensors, including p204 and cGMP-AMP synthase, can be induced by transfection of synthetic HSV genomic DNA (HSV60). TLR3 and retinoic acid-inducible gene I in EECs can be activated by their common agonist, polyinosinic-polycytidylic acid [poly(I:C)]. The signaling pathway of DNA sensors can be initiated by HSV60. Both poly(I:C) and HSV60 induced the expression of type 1 IFNs and various antiviral proteins, including IFN-stimulated gene 15, 2',5'-oligoadenylate synthetase, and myxovirus resistance 1. Poly(I:C), but not HSV60, also dramatically induced the expression of major proinflammatory cytokines, including TNF-α and MCP-1, in EECs. In vivo assay confirmed that the local injection of poly(I:C) or HSV60 induced the innate antiviral responses in EECs. This study provided novel insights into the mechanisms underlying the innate antiviral responses in the mouse epididymis.


Subject(s)
Epididymis/immunology , Epithelial Cells/immunology , Immunity, Innate/immunology , Receptors, Pattern Recognition/biosynthesis , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Virus Diseases/immunology
16.
Semin Cell Dev Biol ; 30: 75-85, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24735648

ABSTRACT

The transport of germ cells across the seminiferous epithelium is composed of a series of cellular events during the epithelial cycle essential to the completion of spermatogenesis. Without the timely transport of spermatids during spermiogenesis, spermatozoa that are transformed from step 19 spermatids in the rat testis fail to reach the luminal edge of the apical compartment and enter the tubule lumen at spermiation, thereby arriving the epididymis for further maturation. Step 19 spermatids and/or sperms that remain in the epithelium beyond stage VIII of the epithelial cycle will be removed by the Sertoli cell via phagocytosis to form phagosomes and be degraded by lysosomes, leading to subfertility and/or infertility. However, the biology of spermatid transport, in particular the final events that lead to spermiation remain elusive. Based on recent data in the field, we critically evaluate the biology of spermiation herein by focusing on the actin binding proteins (ABPs) that regulate the organization of actin microfilaments at the Sertoli-spermatid interface, which is crucial for spermatid transport during this event. The hypothesis we put forth herein also highlights some specific areas of research that can be pursued by investigators in the years to come.


Subject(s)
Microfilament Proteins/physiology , Spermatids/physiology , Actins/physiology , Animals , Humans , Male , Protein Multimerization , Signal Transduction , Sperm Transport
17.
Eur J Immunol ; 45(6): 1696-705, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25820812

ABSTRACT

Hepatitis B virus (HBV) is known to cause age-dependent infection outcomes wherein most infections during young age result in chronicity. The mechanism underlying the differential outcome remains elusive. By using hydrodynamic injection of the replication-competent pAAV-HBV, we established a mouse model in which HBV persistence was generated in 4-5 w/o C57BL/6 young mice, but not in adult mice over 10 w/o. HBV-tolerant young mice expressed higher interferon (IFN)-α/ß levels in hepatocytes and intrahepatic plasmacytoid DCs (pDCs) than adult mice after pAAV-HBV injection. Excessive IFN-α/ß expression in young mice was associated with induction of the Axl regulatory pathway and expansion of intrahepatic Treg cells. In line with these findings, augmented IFN-ß expression increased Axl expression in the liver and HBV persistence in adult mice, whereas IFN-α/ß signaling blockage decreased Axl expression and HBV persistence in young mice. Accordingly, Axl overexpression decreased HBV clearance of adult mice whereas Axl silencing enhanced HBV clearance of young mice. In vitro, IFN-ß priming of pDCs and Axl-overexpressing macrophages enhanced Treg-cell differentiation. These findings suggest that age-dependent HBV chronicity is attributed to IFN-ß-Axl immune regulation, which is selectively induced in young mice by excessive IFN-α/ß production at early stage of HBV infection.


Subject(s)
Hepatitis B virus/immunology , Hepatitis B/immunology , Hepatitis B/metabolism , Interferon-alpha/metabolism , Interferon-beta/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Age Factors , Animals , Cytokines/metabolism , Disease Models, Animal , Hepatitis B/mortality , Hepatitis B/virology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/metabolism , Hepatitis B, Chronic/virology , Humans , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Axl Receptor Tyrosine Kinase
18.
Biol Reprod ; 94(3): 58, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26819479

ABSTRACT

Uropathogenic Escherichia coli (UPEC) may cause epididymitis and impair male fertility. The mechanisms underlying the innate immune responses to UPEC infection in the epididymis are not fully understood. This study showed that UPEC induced innate immune responses in mouse epididymal epithelial cells (EECs) through the activation of Toll-like receptor 4 (TLR4) and TLR5. Infection with UPEC significantly induced the expression of proinflammatory cytokines, including tumor necrosis factor alpha, interleukin 6, and monocyte chemoattractant protein 1, in EECs through the activation of nuclear factor kappa B. Moreover, UPEC induced the production of type 1 interferons by EECs through the activation of interferon regulatory factor 3. The UPEC-induced innate immune responses were significantly reduced in the EECs of Tlr4 or Tlr5 knockout mice. The innate immune responses were further reduced in Tlr4 and Tlr5 double-knockout EECs. Furthermore, we demonstrated that TLR4 and TLR5 cooperatively initiated the epididymal innate immune responses to UPEC infection in vivo. The results provide novel insights into the mechanisms underlying the epididymal innate immune responses to UPEC infection.


Subject(s)
Epididymis/metabolism , Immunity, Innate/physiology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 5/metabolism , Uropathogenic Escherichia coli/immunology , Animals , Cells, Cultured , Epididymis/microbiology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Male , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 5/genetics
19.
FASEB J ; 29(9): 3788-805, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26048141

ABSTRACT

Ectoplasmic specialization (ES) is an actin-rich adherens junction in the seminiferous epithelium of adult mammalian testes. ES is restricted to the Sertoli-spermatid (apical ES) interface, as well as the Sertoli cell-cell (basal ES) interface at the blood-testis barrier (BTB). ES is typified by the presence of an array of bundles of actin microfilaments near the Sertoli cell plasma membrane. These actin microfilament bundles require rapid debundling to convert them from a bundled to branched/unbundled configuration and vice versa to confer plasticity to support the transport of 1) spermatids in the adluminal compartment and 2) preleptotene spermatocytes at the BTB while maintaining cell adhesion. Plastin 3 is one of the plastin family members abundantly found in yeast, plant and animal cells that confers actin microfilaments their bundled configuration. Herein, plastin 3 was shown to be a component of the apical and basal ES in the rat testis, displaying spatiotemporal expression during the epithelial cycle. A knockdown (KD) of plastin 3 in Sertoli cells by RNA interference using an in vitro model to study BTB function showed that a transient loss of plastin 3 perturbed the Sertoli cell tight junction-permeability barrier, mediated by changes in the localization of basal ES proteins N-cadherin and ß-catenin. More importantly, these changes were the result of an alteration of the actin microfilaments, converting from their bundled to branched configuration when examined microscopically, and validated by biochemical assays that quantified actin-bundling and polymerization activity. Moreover, these changes were confirmed by studies in vivo by plastin 3 KD in the testis in which mis-localization of N-cadherin and ß-catenin was also detected at the BTB, concomitant with defects in the transport of spermatids and phagosomes and a disruption of cell adhesion most notably in elongated spermatids due to a loss of actin-bundling capability at the apical ES, which in turn affected localization of adhesion protein complexes at the site. In summary, plastin 3 is a regulator of actin microfilament bundles at the ES in which it dictates the configuration of the filamentous actin network by assuming either a bundled or unbundled/branched configuration via changes in its spatiotemporal expression during the epithelial cycle.


Subject(s)
Membrane Glycoproteins/metabolism , Microfilament Proteins/metabolism , Sertoli Cells/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/physiology , Tight Junctions/metabolism , Actin Cytoskeleton/metabolism , Animals , Cadherins/metabolism , Male , Nerve Tissue Proteins/metabolism , Rats , Rats, Sprague-Dawley , Sertoli Cells/cytology , Spermatids/cytology , Spermatocytes/cytology , beta Catenin/metabolism
20.
Immunol Cell Biol ; 93(2): 147-57, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25287442

ABSTRACT

Viruses can infect adipose tissues. However, innate antiviral responses in adipose cells and their effects on adipocyte function have not yet been intensively investigated. In this study, p204-initiated innate antiviral responses in mouse adipose cells were examined. Cytosolic DNA sensor p204 and its signaling adaptor stimulator of interferon (IFN) genes (STING) were constitutively expressed in primary preadipocytes. Synthetic herpes simplex viral DNA (HSV60), a p204 ligand, induced type I IFN expression by activating IFN regulatory factor 3. Major antiviral proteins, including IFN-stimulating gene 15, 2',5'-oligoadenylate synthetase and Mx GTPase 1, in preadipocytes were upregulated by HSV60. HSV60-triggered innate antiviral responses were significantly reduced by inhibition of p204 signaling with specific small interfering RNA targeting p204 or STING. HSV60 inhibited the differentiation of preadipocytes to mature adipocytes and enhanced the proliferation of adipose cells. Moreover, HSV60 induced innate antiviral responses in mature adipocytes and inhibited expressions of several adipokines, including leptin, adiponectin and resistin. These results indicated that p204 initiated innate antiviral responses in adipose cells, thereby modulating adipocyte function.


Subject(s)
Adipocytes/immunology , Adipocytes/virology , Antiviral Agents/immunology , Immunity, Innate/immunology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Simplexvirus/immunology , Animals , Cell Differentiation/immunology , Cell Proliferation , Cytosol/metabolism , DNA/metabolism , Female , Interferon Regulatory Factor-3/metabolism , Mice, Inbred C57BL , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL