Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Dev Biol ; 493: 29-39, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36368522

RESUMEN

A global increase in older individuals creates an increasing demand to understand numerous healthcare challenges related to aging. This population is subject to changes in tissue physiology and the immune response network. Older individuals are particularly susceptible to infectious diseases, with one of the most common being urinary tract infections (UTIs). Postmenopausal and older women have the highest risk of recurrent UTIs (rUTIs); however, why rUTIs become more frequent after menopause and during old age is incompletely understood. This increased susceptibility and severity among older individuals may involve functional changes to the immune system with age. Aging also has substantial effects on the epithelium and the immune system that led to impaired protection against pathogens, yet heightened and prolonged inflammation. How the immune system and its responses to infection changes within the bladder mucosa during aging has largely remained poorly understood. In this review, we highlight our understanding of bladder innate and adaptive immunity and the impact of aging and hormones and hormone therapy on bladder epithelial homeostasis and immunity. In particular, we elaborate on how the cellular and molecular immune landscape within the bladder can be altered during aging as aged mice develop bladder tertiary lymphoid tissues (bTLT), which are absent in young mice leading to profound age-associated change to the immune landscape in bladders that might drive the significant increase in UTI susceptibility. Knowledge of host factors that prevent or promote infection can lead to targeted treatment and prevention regimens. This review also identifies unique host factors to consider in the older, female host for improving rUTI treatment and prevention by dissecting the age-associated alteration of the bladder mucosal immune system.


Asunto(s)
Infecciones Urinarias , Sistema Urinario , Femenino , Ratones , Animales , Vejiga Urinaria , Envejecimiento , Homeostasis , Inmunidad Innata
2.
Dev Cell ; 59(1): 33-47.e5, 2024 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-38101412

RESUMEN

Aging is a risk factor for disease via increased susceptibility to infection, decreased ability to maintain homeostasis, inefficiency in combating stress, and decreased regenerative capacity. Multiple diseases, including urinary tract infection (UTI), are more prevalent with age; however, the mechanisms underlying the impact of aging on the urinary tract mucosa and the correlation between aging and disease remain poorly understood. Here, we show that, relative to young (8-12 weeks) mice, the urothelium of aged (18-24 months) female mice accumulates large lysosomes with reduced acid phosphatase activity and decreased overall autophagic flux in the aged urothelium, indicative of compromised cellular homeostasis. Aged bladders also exhibit basal accumulation of reactive oxygen species (ROS) and a dampened redox response, implying heightened oxidative stress. Furthermore, we identify a canonical senescence-associated secretory phenotype (SASP) in the aged urothelium, along with continuous NLRP3-inflammasome- and Gasdermin-D-dependent pyroptotic cell death. Consequently, aged mice chronically exfoliate urothelial cells, further exacerbating age-related urothelial dysfunction. Upon infection with uropathogenic E. coli, aged mice harbor increased bacterial reservoirs and are more prone to spontaneous recurrent UTI. Finally, we discover that treatment with D-mannose, a natural bioactive monosaccharide, rescues autophagy flux, reverses the SASP, and mitigates ROS and NLRP3/Gasdermin/interleukin (IL)-1ß-driven pyroptotic epithelial cell shedding in aged mice. Collectively, our results demonstrate that normal aging affects bladder physiology, with aging alone increasing baseline cellular stress and susceptibility to infection, and suggest that mannose supplementation could serve as a senotherapeutic to counter age-associated urothelial dysfunction.


Asunto(s)
Proteína con Dominio Pirina 3 de la Familia NLR , Infecciones Urinarias , Ratones , Femenino , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Vejiga Urinaria/metabolismo , Vejiga Urinaria/microbiología , Vejiga Urinaria/patología , Manosa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Escherichia coli/metabolismo , Urotelio/metabolismo , Urotelio/microbiología , Interleucina-1beta , Gasderminas , Infecciones Urinarias/metabolismo , Infecciones Urinarias/microbiología , Infecciones Urinarias/patología , Senescencia Celular
3.
Histochem Cell Biol ; 139(4): 535-48, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23124857

RESUMEN

Liprin α3 was reported for the first time using sperm proteomics. Present study reports its localization on sperm and immunochemical characterization. Liprin α3 is identified as a 133 kDa protein in testis and epididymal protein extracts. In testis, immunohistochemical localization was seen in pachytenes, diplotenes, round spermatids whereas it was localized in the epithelial cells and luminal sperm in all the three regions of epididymis. Protein was localized in acrosome of rat sperm, which was further confirmed by sequential treatment of sperm with hypertonic solution. In the spermatogenic cells the protein was found to be located in developing acrosome as evident by its co-localization with Golgi marker. Protein was found to be developmentally regulated. In silico analysis of Liprin α3 revealed presence of the estrogen responsive elements upstream to initiation site and its regulation by estrogen was experimentally validated using a tamoxifen treated rat model. Western blot analysis of epididymosomes showed the presence of Liprin α3, indicating its involvement in trafficking of vesicle. The protein expression was seen in both mouse and human sperm indicating conserved nature and a probable role in acrosome reaction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Estrógenos/metabolismo , Espermatozoides/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Acrosoma/química , Acrosoma/metabolismo , Reacción Acrosómica , Animales , Epididimitis/metabolismo , Antagonistas de Estrógenos/farmacología , Humanos , Masculino , Ratones , Ratas , Espermatogénesis , Espermatozoides/química , Tamoxifeno/farmacología
4.
Cell Rep ; 39(4): 110758, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35476986

RESUMEN

Urinary tract infections (UTIs) are a cause for alarm given the high rates of treatment failure. In a recent issue of Cell Reports, Pang et al. uncovered dueling molecular machinery at the host-pathogen interface in response to phosphate that points to new anti-infective strategies against UTIs.


Asunto(s)
Infecciones por Escherichia coli , Infecciones Urinarias , Escherichia coli Uropatógena , Femenino , Humanos , Masculino , Escherichia coli Uropatógena/fisiología , Urotelio
5.
Cell Rep ; 37(3): 109856, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686330

RESUMEN

Uropathogenic Escherichia coli (UPEC) cause urinary tract infections (UTIs) by invading urothelial cells. In response, the host mounts an inflammatory response to expel bacteria. Here, we show that the NF-E2-related factor 2 (NRF2) pathway is activated in response to UPEC-triggered reactive oxygen species (ROS) production. We demonstrate the molecular sequence of events wherein NRF2 activation in urothelial cells reduces ROS production, inflammation, and cell death, promotes UPEC expulsion, and reduces the bacterial load. In contrast, loss of NRF2 leads to increased ROS production, bacterial burden, and inflammation, both in vitro and in vivo. NRF2 promotes UPEC expulsion by regulating transcription of the RAB-GTPase RAB27B. Finally, dimethyl fumarate, a US Food and Administration-approved NRF2 inducer, reduces the inflammatory response, increases RAB27B expression, and lowers bacterial burden in urothelial cells and in a mouse UTI model. Our findings elucidate mechanisms underlying the host response to UPEC and provide a potential strategy to combat UTIs.


Asunto(s)
Infecciones por Escherichia coli/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Infecciones Urinarias/metabolismo , Escherichia coli Uropatógena/patogenicidad , Urotelio/metabolismo , Proteínas rab27 de Unión a GTP/metabolismo , Animales , Antiinflamatorios/farmacología , Carga Bacteriana , Línea Celular Tumoral , Dimetilfumarato/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Femenino , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/agonistas , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo , Transducción de Señal , Infecciones Urinarias/tratamiento farmacológico , Infecciones Urinarias/genética , Infecciones Urinarias/microbiología , Urotelio/efectos de los fármacos , Urotelio/microbiología , Proteínas de Unión al GTP rab , Proteínas rab27 de Unión a GTP/genética
6.
J Am Heart Assoc ; 10(2): e017995, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33440998

RESUMEN

Background Coxsackievirus B (CVB) is the most common cause of viral myocarditis. It targets cardiomyocytes through coxsackie and adenovirus receptor, which is highly expressed in the fetal heart. We hypothesized CVB3 can precipitate congenital heart defects when fetal infection occurs during critical window of gestation. Methods and Results We infected C57Bl/6 pregnant mice with CVB3 during time points in early gestation (embryonic day [E] 5, E7, E9, and E11). We used different viral titers to examine possible dose-response relationship and assessed viral loads in various fetal organs. Provided viral exposure occurred between E7 and E9, we observed characteristic features of ventricular septal defect (33.6%), abnormal myocardial architecture resembling noncompaction (23.5%), and double-outlet right ventricle (4.4%) among 209 viable fetuses examined. We observed a direct relationship between viral titers and severity of congenital heart defects, with apparent predominance among female fetuses. Infected dams remained healthy; we did not observe any maternal heart or placental injury suggestive of direct viral effects on developing heart as likely cause of congenital heart defects. We examined signaling pathways in CVB3-exposed hearts using RNA sequencing, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, and immunohistochemistry. Signaling proteins of the Hippo, tight junction, transforming growth factor-ß1, and extracellular matrix proteins were the most highly enriched in CVB3-infected fetuses with ventricular septal defects. Moreover, cardiomyocyte proliferation was 50% lower in fetuses with ventricular septal defects compared with uninfected controls. Conclusions We conclude prenatal CVB3 infection induces congenital heart defects. Alterations in myocardial proliferate capacity and consequent changes in cardiac architecture and trabeculation appear to account for most of observed phenotypes.


Asunto(s)
Infecciones por Coxsackievirus , Enterovirus Humano B/patogenicidad , Corazón Fetal , Cardiopatías Congénitas , Miocitos Cardíacos , Animales , Proliferación Celular , Correlación de Datos , Infecciones por Coxsackievirus/complicaciones , Infecciones por Coxsackievirus/virología , Femenino , Corazón Fetal/embriología , Corazón Fetal/patología , Cardiopatías Congénitas/patología , Cardiopatías Congénitas/virología , Ratones , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Miocitos Cardíacos/virología , Embarazo , Índice de Severidad de la Enfermedad , Carga Viral/métodos
7.
Med ; 2(5): 575-590.e5, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33870242

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection appears to increase the risk of adverse pregnancy outcomes, such as pre-eclampsia in pregnant women. The mechanism(s) by which this occurs remains unclear. METHODS: We investigated the pathophysiology of SARS-CoV-2 at maternal-fetal interface in pregnant women who tested positive for the virus using RNA in situ hybridization (viral RNA), immunohistochemistry, and hematoxylin and eosin staining. To investigate whether viral infection alters the renin angiotensin system (RAS) in placenta, which controls blood pressure, we treated human trophoblasts with recombinant spike protein or a live modified virus with a vesicular stomatitis viral backbone expressing spike protein (VSV-S). FINDINGS: Viral colonization was highest in maternal decidua, fetal trophoblasts, Hofbauer cells, and in placentas delivered prematurely. We localized SARS-CoV-2 to cells expressing angiotensin-converting enzyme 2 (ACE2) and demonstrate that infected placentas had significantly reduced ACE2. In response to both spike protein and VSV-S, cellular ACE2 decreased although angiotensin II receptor type 1 (AT1R) increased with concomitant increase in soluble fms-like tyrosine kinase-1 (sFlt1). Viral infection decreased pro-angiogenic factors, AT2R, and placental growth factor, which competitively binds to sFlt1. Sera from infected pregnant women had elevated levels of sFlt1 and angiotensin II type 1-receptor autoantibodies prior to delivery, both signatory markers of pre-eclampsia. CONCLUSIONS: SARS-CoV-2 colonizes ACE2-expressing maternal and fetal cells in the placenta. Infection in pregnant women correlates with alteration of placental RAS. As RAS regulates blood pressure, SARS-CoV-2 infection may thus increase adverse hemodynamic outcomes, such as pre-eclampsia in pregnant women. FUNDING: NIH/NICHD grants R01 HD091218 and 3R01HD091218-04S1 (RADx-UP Supplement).


Asunto(s)
COVID-19 , Preeclampsia , Complicaciones Infecciosas del Embarazo , Enzima Convertidora de Angiotensina 2 , Femenino , Humanos , Placenta/metabolismo , Factor de Crecimiento Placentario/metabolismo , Preeclampsia/metabolismo , Embarazo , Complicaciones Infecciosas del Embarazo/metabolismo , Sistema Renina-Angiotensina , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Cell Death Dis ; 9(7): 780, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-30006504

RESUMEN

The cellular recycling pathway of autophagy plays a fundamental role in adaptive responses to nutrient deprivation and other forms of stress under physiological and pathological conditions. However, autophagy can also be a double-edge sword during certain bacterial infections (such as urinary tract infections) and in cancer, where it can be hijacked by the pathogens and cancer cells, respectively, to promote their own survival. Thus, autophagy modulation can potentially have multiple effects in multiple contexts and this property can be leveraged to improve outcomes. In this report, we identify that a broad-spectrum antibiotic, 2-((3-(3, 6-dichloro-9H-carbazol-9-yl)-2-hydroxypropyl) amino)-2-(hydroxymethyl) propane-1, 3-diol (DCAP) modulates autophagy. We employed combined biochemical, fluorescence microscopy and correlative light electron microscopy approaches to demonstrate that DCAP treatment blocks autophagy at the late stages by preventing autophagolysosome maturation and interrupting the autophagic flux. We further show that, DCAP significantly reduces UPEC infection in urinary tract epithelial cells via inhibition of autophagy. Finally, we reveal that DCAP enhances the anticancer activity of the histone acetyltransferase (HDAC) inhibitor, vorinostat, which has been reported to increase susceptibility to bacterial infections as a common adverse effect. Collectively, our data support the concept that DCAP represents a valuable chemical scaffold for the development of an innovative class of bactericidal autophagy inhibitors for treatment of urinary tract infections and/or for adjuvant therapy in cancer treatment.


Asunto(s)
Aminofenoles/farmacología , Antibacterianos/uso terapéutico , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena/fisiología , Vorinostat/farmacología , Antibacterianos/farmacología , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Línea Celular Tumoral , Infecciones por Escherichia coli/microbiología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/efectos de los fármacos
9.
J Androl ; 33(5): 963-74, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22207704

RESUMEN

A differential proteomics approach led to the identification of several novel epididymal sperm proteins. One of the novel proteins was methylmalonate-semialdehyde dehydrogenase (MMSDH). In the present study, we carried out an in-depth characterization to study its regulation by androgen, its appearance during ontogeny, and the mechanism of its interaction with and acquisition by the sperm. Western blotting and immunohistochemical studies suggest that the protein is present in both tissue and sperm from all regions of the epididymis, indicating synthesis as well as acquisition of the protein in these regions. Androgen depletion resulted in reduction of the MMSDH protein level in the epididymis, which completely disappeared 1 week after castration. The protein reappeared after testosterone propionate injection, indicating that the protein is regulated by androgens. Ontogeny studies indicated that the protein appeared from day 10 postnatal with a gradual increase at day 30, which maximized at day 50, indicating that the protein is developmentally regulated and is probably involved in epididymal development. Sequential extraction of sperm proteins indicated that MMSDH exists both as a peripheral and integral form on the plasma membrane. We also found that the protein can be transferred from the epididymosomes to testicular sperm in vitro. The study provides evidence regarding the acquisition of this multidomain androgen and developmentally regulated protein in the epididymis via the epididymosomes. The molecule has generated enough interest and deserves to be investigated further for its physiological relevance.


Asunto(s)
Epidídimo/enzimología , Metilmalonato-Semialdehído Deshidrogenasa (Acetilante)/metabolismo , Espermatozoides/enzimología , Testosterona/metabolismo , Factores de Edad , Animales , Western Blotting , Membrana Celular/enzimología , Epidídimo/efectos de los fármacos , Epidídimo/embriología , Epidídimo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Inmunohistoquímica , Inyecciones , Masculino , Metilmalonato-Semialdehído Deshidrogenasa (Acetilante)/genética , Morfogénesis , Orquiectomía , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Espermatozoides/efectos de los fármacos , Propionato de Testosterona/administración & dosificación , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA