Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell Rep Med ; 5(5): 101554, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38729157

RESUMEN

The axons of retinal ganglion cells (RGCs) form the optic nerve, transmitting visual information from the eye to the brain. Damage or loss of RGCs and their axons is the leading cause of visual functional defects in traumatic injury and degenerative diseases such as glaucoma. However, there are no effective clinical treatments for nerve damage in these neurodegenerative diseases. Here, we report that LIM homeodomain transcription factor Lhx2 promotes RGC survival and axon regeneration in multiple animal models mimicking glaucoma disease. Furthermore, following N-methyl-D-aspartate (NMDA)-induced excitotoxicity damage of RGCs, Lhx2 mitigates the loss of visual signal transduction. Mechanistic analysis revealed that overexpression of Lhx2 supports axon regeneration by systematically regulating the transcription of regeneration-related genes and inhibiting transcription of Semaphorin 3C (Sema3C). Collectively, our studies identify a critical role of Lhx2 in promoting RGC survival and axon regeneration, providing a promising neural repair strategy for glaucomatous neurodegeneration.


Asunto(s)
Axones , Modelos Animales de Enfermedad , Glaucoma , Proteínas con Homeodominio LIM , Regeneración Nerviosa , Células Ganglionares de la Retina , Factores de Transcripción , Animales , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Proteínas con Homeodominio LIM/metabolismo , Proteínas con Homeodominio LIM/genética , Glaucoma/genética , Glaucoma/patología , Glaucoma/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Axones/metabolismo , Axones/patología , Ratones , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Ratones Endogámicos C57BL , Supervivencia Celular/genética , Semaforinas/metabolismo , Semaforinas/genética , N-Metilaspartato/metabolismo
2.
Cell Prolif ; 55(11): e13314, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35854653

RESUMEN

OBJECTIVE: Microglia, the prototypical innate immune cells of the central nervous system (CNS), are highly plastic and assume their phenotypes dependent on intrinsically genetic, epigenetic regulation or extrinsically microenvironmental cues. Microglia has been recognized as key regulators of neural stem/progenitor cells (NSPCs) and brain functions. Chromatin accessibility is implicated in immune cell development and functional regulation. However, it is still unknown whether and how chromatin remodelling regulates the phenotypic plasticity of microglia and exerts what kind of effects on NSPCs. METHODS: We investigated the role of chromatin accessibility in microglia by deleting chromatin remodelling gene Arid1a using microglia-specific Cx3cr1-cre and Cx3cr1-CreERT2 mice. RNA-seq and ATAC-seq were performed to dissect the molecular mechanisms. In addition, we examined postnatal M1/M2 microglia polarization and analysed neuronal differentiation of NSPCs. Finally, we tested the effects of microglial Arid1a deletion on mouse behaviours. RESULTS: Increased chromatin accessibility upon Arid1a ablation resulted in enhanced M1 microglial polarization and weakened M2 polarization, which led to abnormal neurogenesis and anxiety-like behaviours. Switching the polarization state under IL4 stimulation could rescue abnormal neurogenesis, supporting an essential role for chromatin remodeler ARID1A in balancing microglial polarization and brain functions. CONCLUSIONS: Our study identifies ARID1A as a central regulator of microglia polarization, establishing a mechanistic link between chromatin remodelling, neurogenesis and mouse behaviours, and highlights the potential development of innovative therapeutics exploiting the innate regenerative capacity of the nervous system.


Asunto(s)
Microglía , Células-Madre Neurales , Ratones , Animales , Epigénesis Genética , Neurogénesis , Células-Madre Neurales/fisiología , Cromatina , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética
3.
Adv Exp Med Biol ; 1288: 49-67, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34453731

RESUMEN

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.


Asunto(s)
Espermatogénesis , Testículo , Animales , Humanos , Inmunidad Innata , Masculino , Espermatogonias , Espermatozoides
4.
Biol Reprod ; 105(1): 113-127, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-33899078

RESUMEN

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.


Asunto(s)
Inmunidad Innata/genética , Próstata/inmunología , Receptores de Reconocimiento de Patrones/genética , Animales , Células Epiteliales/inmunología , Inflamación/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Reconocimiento de Patrones/inmunología
5.
Front Immunol ; 12: 580454, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679733

RESUMEN

Numerous types of viruses have been found in human semen, which raises concerns about the sexual transmission of these viruses. The overall effect of semen on viral infection and transmission have yet to be fully investigated. In the present study, we aimed at the effect of seminal plasma (SP) on viral infection by focusing on the mumps viral (MuV) infection of HeLa cells. MuV efficiently infected HeLa cells in vitro. MuV infection was strongly inhibited by the pre-treatment of viruses with SP. SP inhibited MuV infection through the impairment of the virus's attachment to cells. The antiviral activity of SP was resistant to the treatment of SP with boiling water, Proteinase K, RNase A, and DNase I, suggesting that the antiviral factor would not be proteins and nucleic acids. PNGase or PLA2 treatments did not abrogate the antiviral effect of SP against MuV. Further, we showed that the prostatic fluid (PF) showed similar inhibition as SP, whereas the epididymal fluid and seminal vesicle extract did not inhibit MuV infection. Both SP and PF also inhibited MuV infection of other cell types, including another human cervical carcinoma cell line C33a, mouse primary epididymal epithelial cells, and Sertoli cell line 15P1. Moreover, this inhibitory effect was not specific to MuV, as the herpes simplex virus 1, dengue virus 2, and adenovirus 5 infections were also inhibited by SP and PF. Our findings suggest that SP contains a prostate-derived pan-antiviral factor that may limit the sexual transmission of various viruses.


Asunto(s)
Antivirales/inmunología , Células Epiteliales/inmunología , Virus de la Parotiditis/inmunología , Semen/inmunología , Virus/inmunología , Animales , Antivirales/metabolismo , Línea Celular Tumoral , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/metabolismo , Células Epiteliales/virología , Células HeLa , Interacciones Huésped-Patógeno/inmunología , Humanos , Masculino , Ratones Endogámicos C57BL , Virus de la Parotiditis/fisiología , Semen/metabolismo , Semen/virología , Células Vero
6.
Am J Physiol Heart Circ Physiol ; 320(3): H969-H979, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33164579

RESUMEN

Myocardial infarction (MI) is recognized as a major cause of death and disability around the world. Macrophage-derived extracellular vesicles (EVs) have been reportedly involved in the regulation of cellular responses to MI. Thus, we sought to clarify the mechanism by which macrophage-derived EVs regulate this process. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) was performed to determine microRNA-150 (miR-150) expression in an MI mouse model with ligation of the left anterior descending coronary artery (LAD) and in hypoxia/reoxygenation (H/R)-exposed cardiomyocytes. Bioinformatics analysis and dual luciferase reporter gene assay were adopted to identify the correlation of miR-150 with tumor protein 53 (TP53) expression in cardiomyocytes. Gain- and loss-of-function experiments were conducted in H/R-induced cardiomyocytes, cardiomyocytes incubated with EVs from miR-150 mimic-transfected macrophages, or MI-model mice treated with EVs from miR-150 mimic-transfected macrophages. hematoxylin-eosin (HE) and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining assays were used for detecting inflammatory infiltration and cell apoptosis. The release of lactate dehydrogenase (LDH) by dead cardiomyocytes was measured with an LDH kit, and the apoptosis-related proteins, Bax, and cleaved-caspase 3 were determined by Western blot analysis. miR-150 expression was downregulated in the infarcted cardiac tissues of MI mice. Macrophage-derived EVs could transfer miR-150 into cardiomyocytes, where it directly targeted and suppressed TP53. Furthermore, miR-150 suppressed phosphatase and tensin homology (PTEN) and activated p-Akt to upregulate IGF-1 expression. Furthermore, increased expression of EV-derived miR-150 prevented cardiomyocyte apoptosis in vitro, as evidenced by downregulated Bax and cleaved-caspase 3 and upregulated Bcl2 and alleviated MI in vivo. In conclusion, our study demonstrates the cardioprotective effect of macrophage-derived EV-miR-150 on MI-induced heart injury through negatively regulating the TP53-IGF-1 signaling pathway.NEW & NOTEWORTHY miR-150 is expressed at a low level in cardiac tissues after myocardial infarction. Macrophages-derived EVs transfer miR-150 to cardiomyocytes. miR-150 directly targets TP53. miR-150 elevation regulates TP53-IGF-1 axis to reduce cardiomyocyte apoptosis. EV-derived miR-150 could be a potential therapeutic target for myocardial infarction.


Asunto(s)
Vesículas Extracelulares/trasplante , Factor I del Crecimiento Similar a la Insulina/metabolismo , Macrófagos/trasplante , MicroARNs/metabolismo , Infarto del Miocardio/terapia , Miocardio/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Línea Celular , Modelos Animales de Enfermedad , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/genética , Preparación de Corazón Aislado , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Miocardio/patología , Células RAW 264.7 , Ratas , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
7.
Front Microbiol ; 11: 1292, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32695074

RESUMEN

The mumps virus (MuV) causes epidemic parotitis. MuV also frequently infects the testis and induces orchitis, an important etiological factor contributing to male infertility. However, mechanisms underlying MuV infection of the testis remain unknown. Here, we describe that sialic acid, AXL, and MER receptor tyrosine kinases regulate MuV entry and replication in mouse major testicular cells, including Sertoli and Leydig cells. Sialic acid, AXL, and MER were present in Sertoli and Leydig cells. Sialic acid specifically mediated MuV entry into Sertoli and Leydig cells, whereas both AXL and MER facilitated MuV replication within cells through the inhibition of cellular innate antiviral responses. Mechanistically, the inhibition of type 1 interferon signaling by AXL and MER is essential for MuV replication in Sertoli and Leydig cells. Our findings provide novel insights into the mechanisms behind MuV infection and replication in the testis.

8.
Aging (Albany NY) ; 12(11): 10441-10456, 2020 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-32507765

RESUMEN

Previous studies have shown that lncRNA small nuclear RNA host gene 7 (lncRNA SNHG7) played an important role in cancer progression. However, the role of lncRNA SNHG7 in cardiac fibrosis is still poorly understood. In this study, the results of quantitative real time polymerase chain reaction (qRT-PCR) analysis showed that lncRNA SNHG7 was over expressed in the infarcted and peri-infarcted area in the left ventricle after MI in mice. Western blot analysis showed that knockdown of SNHG7 decreased the expression of collagen type 1 (Col1)and α-smooth muscle actin (α-SMA). Echocardiographic study suggested that inhibition of SNHG7 improved cardiac function after MI in mice. Luciferase assay indicated SNHG7 could act as a competing endogenous RNA (ceRNA) by sponging miR-34-5p. The MTT cell proliferation assay and 5-ethynyl-2'-deoxyuridine (EdU) labelling assay revealed that co-transfection of SNHG7 and miR-34-5p inhibited cell viability and proliferation of cardiac fibroblasts (CF). All the results indicated that lncRNA SNHG7 could promote cardiac fibrosis via targeting miR-34-5p through acting as a ceRNA in mice after MI. Silencing of SNHG7 could attenuate deposition of collagens and improve cardiac function. miR-34-5p could suppress the fibrogenesis of CF by targeting ROCK1 and abolish SNHG7-induced CF proliferation and fibroblast-to-myofibroblast transition.


Asunto(s)
Ventrículos Cardíacos/patología , MicroARNs/metabolismo , Infarto del Miocardio/patología , ARN Largo no Codificante/metabolismo , Remodelación Ventricular/genética , Quinasas Asociadas a rho/genética , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Ecocardiografía , Fibroblastos , Fibrosis , Técnicas de Silenciamiento del Gen , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/diagnóstico por imagen , Humanos , Masculino , Ratones , Infarto del Miocardio/diagnóstico , Cultivo Primario de Células , ARN Largo no Codificante/genética
9.
Asian J Androl ; 22(5): 472-480, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31696835

RESUMEN

Epididymitis can be caused by infectious and noninfectious etiological factors. While microbial infections are responsible for infectious epididymitis, the etiological factors contributing to noninfectious epididymitis remain to be defined. The present study demonstrated that damaged male germ cells (DMGCs) induce epididymitis in mice. Intraperitoneal injection of the alkylating agent busulfan damaged murine male germ cells. Epididymitis was observed in mice 4 weeks after the injection of busulfan and was characterized by massive macrophage infiltration. Epididymitis was coincident with an accumulation of DMGCs in the epididymis. In contrast, busulfan injection into mice lacking male germ cells did not induce epididymitis. DMGCs induced innate immune responses in epididymal epithelial cells (EECs), thereby upregulating the pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß), as well as the chemokines such as monocyte chemotactic protein-1 (MCP-1), monocyte chemotactic protein-5 (MCP-5), and chemokine ligand-10 (CXCL10). These results suggest that male germ cell damage may induce noninfectious epididymitis through the induction of innate immune responses in EECs. These findings provide novel insights into the mechanisms underlying noninfectious epididymitis, which might aid in the diagnosis and treatment of the disease.


Asunto(s)
Citocinas/metabolismo , Epididimitis/inmunología , Epididimitis/patología , Células Germinativas/inmunología , Células Germinativas/metabolismo , Animales , Busulfano , Movimiento Celular , Quimiocina CCL2/metabolismo , Quimiocina CXCL10/metabolismo , Células Germinativas/patología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quimioatrayentes de Monocitos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
FASEB J ; 33(11): 12528-12540, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31450968

RESUMEN

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.


Asunto(s)
Barrera Hematotesticular/metabolismo , Virus de la Parotiditis/metabolismo , Paperas/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Barrera Hematotesticular/patología , Barrera Hematotesticular/virología , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Infertilidad Masculina/patología , Infertilidad Masculina/virología , Masculino , Ratones , Ratones Noqueados , Paperas/genética , Paperas/patología , Virus de la Parotiditis/genética , Células de Sertoli/patología , Células de Sertoli/virología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Factor de Necrosis Tumoral alfa/genética , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo
11.
Biol Reprod ; 101(4): 733-747, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31350848

RESUMEN

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.


Asunto(s)
Células Epiteliales/inmunología , Inmunidad Innata/genética , Receptores de Reconocimiento de Patrones/fisiología , Vesículas Seminales/inmunología , Animales , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C , Receptores de Reconocimiento de Patrones/genética , Vesículas Seminales/citología , Vesículas Seminales/metabolismo , Transducción de Señal
12.
Biol Reprod ; 100(3): 849-861, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30398566

RESUMEN

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.


Asunto(s)
Epididimitis/inducido químicamente , Regulación de la Expresión Génica/efectos de los fármacos , Lipopolisacáridos/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Epididimitis/prevención & control , Factores Inmunológicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Talidomida/análogos & derivados , Talidomida/farmacología , Factor de Necrosis Tumoral alfa/genética
13.
Cell Death Dis ; 9(8): 789, 2018 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-30013140

RESUMEN

The PDF and HTML versions of the article have been updated to include the Creative Commons Attribution 4.0 International License information.

14.
Cell Death Dis ; 8(10): e3146, 2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29072682

RESUMEN

Mumps virus (MuV) infection usually results in germ cell degeneration in the testis, which is an etiological factor for male infertility. However, the mechanisms by which MuV infection damages male germ cells remain unclear. The present study showed that C-X-C motif chemokine ligand 10 (CXCL10) is produced by mouse Sertoli cells in response to MuV infection, which induces germ cell apoptosis through the activation of caspase-3. CXC chemokine receptor 3 (CXCR3), a functional receptor of CXCL10, is constitutively expressed in male germ cells. Neutralizing antibodies against CXCR3 and an inhibitor of caspase-3 activation significantly inhibited CXCL10-induced male germ cell apoptosis. Furthermore, the tumor necrosis factor-α (TNF-α) upregulated CXCL10 production in Sertoli cells after MuV infection. The knockout of either CXCL10 or TNF-α reduced germ cell apoptosis in the co-cultures of germ cells and Sertoli cells in response to MuV infection. Local injection of MuV into the testes of mice confirmed the involvement of CXCL10 in germ cell apoptosis in vivo. These results provide novel insights into MuV-induced germ cell apoptosis in the testis.


Asunto(s)
Quimiocina CXCL10/biosíntesis , Células Germinativas/metabolismo , Virus de la Parotiditis/fisiología , Paperas/metabolismo , Células de Sertoli/metabolismo , Animales , Apoptosis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Paperas/patología , Paperas/virología , Células de Sertoli/virología
15.
Front Immunol ; 8: 117, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28239382

RESUMEN

Mumps virus (MuV) infection has high tropism to the testis and usually leads to orchitis, an etiological factor in male infertility. However, MuV replication in testicular cells and the cellular antiviral responses against MuV are not fully understood. The present study showed that MuV infected the majority of testicular cells, including Leydig cells (LC), testicular macrophages, Sertoli cells (SC), and male germ cells (GC). MuV was replicated at relatively high efficiencies in SC compared with LC and testicular macrophages. In contrast, MuV did not replicate in male GC. Notably, testicular cells exhibited different innate antiviral responses against MuV replication. We showed that interferon ß (IFN-ß) inhibited MuV replication in LC, macrophages, and SC, which were associated with the upregulation of major antiviral proteins. We provided primary evidence that autophagy plays a role in blocking MuV replication in male GC. Autophagy was also involved in limiting MuV replication in testicular macrophages but not in Leydig and SC. These findings indicate the involvement of the innate defense against MuV replication in testicular cells.

16.
Mol Cell Biochem ; 391(1-2): 59-66, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24522555

RESUMEN

Tumor necrosis factor alpha (TNFα) is an adipokine involved in the regulation of cell differentiation and lipid metabolism, but its specific role has not been clearly understood. We validated a hypothesis that loss of TNFα function would inhibit Wnt/ß-catenin signaling and accelerate adipogenesis in adolescent genetic obese mice. Epididymal white adipose tissues (eWAT) from TNFα deficient (TNFα(-/-)), leptin receptor deficient (db/db) and double gene mutant (db/db/TNFα(-/-), DT) male mice were used for comparative analysis of key molecules in Wnt/ß-catenin signaling and adipogenic markers by qRT-PCR and western blot techniques. Compared with TNFα(-/-) and WT mice of 28 days old, an obese trait was observed in both db/db and DT mice, while the latter showed more significant body weight gain and eWAT hypertrophy. The mRNA level of key molecules in Wnt/ß-catenin pathway was reduced in both obese groups, while the DT group was the lowest. Expression of adipocyte-specific genes was up-regulated during obese development in the two obese groups, while the DT group revealed more correlation than that of db/db group. At the protein level, a down regulation of Wnt10b and ß-catenin in obese eWAT showed similar tendency with that of mRNA level. Compared with the lean groups, the levels of adiponectin and PPARγ2 for the obese groups were down-regulated at 21-day-old age, while they were elevated at older age. Our results suggested that deficiency in TNFα inhibited Wnt/ß-catenin signaling of the obese eWAT and up-regulated expression of adipokines, and accelerated adipogenesis in genetic obese mice on a chow diet.


Asunto(s)
Envejecimiento/patología , Obesidad/metabolismo , Obesidad/patología , Factor de Necrosis Tumoral alfa/deficiencia , Vía de Señalización Wnt , Adipogénesis/genética , Tejido Adiposo Blanco/patología , Animales , Glucemia/metabolismo , Peso Corporal , Epidídimo/patología , Regulación de la Expresión Génica , Genotipo , Homeostasis , Insulina/sangre , Lípidos/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/sangre , Obesidad/genética , Tamaño de los Órganos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Vía de Señalización Wnt/genética
17.
Zhonghua Yi Xue Za Zhi ; 93(17): 1305-8, 2013 May 07.
Artículo en Chino | MEDLINE | ID: mdl-24029477

RESUMEN

OBJECTIVE: To explore the value of dynamic arterial elastance (Eadyn) in the predication of arterial pressure response to volume loading in shock patients. METHODS: A total of 32 patients with pulse indicator continuous cardiac output (PICCO) monitoring at our intensive care unit from January 2011 to December 2012 were retrospectively studied. The decision of fluid replacement was based upon the presence of shock (mean arterial pressure (MAP) ≤ 65 mm Hg, systolic arterial pressure <90 mm Hg or a decrease of 40 mm Hg from baseline) and preserved volume responsiveness condition with a stroke volume variation (SVV) value ≥ 10%. According to the MAP increase after volume loading, they were classified into MAP responders (≥ 15%) and MAP nonresponders (<15%) respectively. The goal was to investigate the influencing factors of the changes of MAP after volume loading and predict the arterial pressure response to volume loading. RESULTS: Significantly different between MAP responders and MAP nonresponders, baseline Eadyn was an effective predictor of MAP increase after volume loading. The area under the ROC curve was 0.95 for the prediction of volume loading on MAP for Eadyn at baseline (P < 0.01). A baseline Eadyn value >0.85 predicted a MAP increase after volume administration with a sensitivity of 89.5% and a specificity of 92.3%. CONCLUSION: Baseline Eadyn may predict accurately arterial pressure response in MAP to volume loading in shock patients.


Asunto(s)
Arterias/fisiopatología , Choque/fisiopatología , Choque/terapia , Anciano , Anciano de 80 o más Años , Presión Sanguínea , Elasticidad , Fluidoterapia , Humanos , Persona de Mediana Edad , Estudios Retrospectivos , Capacitancia Vascular
18.
J Cell Mol Med ; 13(11-12): 4444-52, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18754816

RESUMEN

Although peroxisome proliferator-activated receptor alpha (PPARalpha) is highly expressed in the heart, the effects of PPARalpha on cardiac remodelling and the underlying mechanisms are unclear. The present study was undertaken to test the hypothesis that PPARalpha activator fenofibrate plays a key role in left ventricular hypertrophic remodelling via the formation of c-fos/c-jun heterodimers in spontaneous hypertensive rats (SHRs). Twenty-four male 8-week-old SHRs were randomly divided into two groups, one group treated with oral saline (n= 10) and another treated with oral fenofibrate (60 mg.kg-1.d-1, n= 14). Ten same-aged Wistar-Kyoto (WKY) rats were selected as a normal control group. Using echocardiography, immunohistochemistry, co-immunoprecipitation, Western blot analysis and real-time RT-PCR, we showed that the left ventricular wall thickness and significantly reduced and left ventricular diastolic function improved in SHRs treated with fenofibrate compared with SHRs treated with saline. Similarly, the excessive collagen deposition and the up-regulation of collagen I, collagen III, c-fos and c-jun seen in SHRs receiving saline were significantly attenuated in SHRs receiving fenofibrate. In addition, fenofibrate markedly decreased the expression of AP-1 and c-fos/c-jun heterodimers (P < 0.01). These results demonstrated that PPARalpha activator fenofibrate may exert a protective effect on cardiac remodelling in SHRs by decreasing the expression of c-fos and c-jun and suppressing the formation of c-fos/c-jun heterodimers, which may further inhibit transcription of the downstream genes involved in the pathogenesis of left ventricular hypertrophy induced by hypertension.


Asunto(s)
Fenofibrato/uso terapéutico , Hipertensión/tratamiento farmacológico , Hipertensión/fisiopatología , Miocardio/patología , PPAR alfa/metabolismo , Remodelación Ventricular/fisiología , Animales , Colágeno/genética , Colágeno/metabolismo , Fenofibrato/farmacología , Hemodinámica/efectos de los fármacos , Hipertensión/diagnóstico por imagen , Inmunohistoquímica , Masculino , Miocardio/ultraestructura , PPAR alfa/genética , Multimerización de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ultrasonografía , Remodelación Ventricular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA