Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Development ; 144(19): 3511-3520, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28860115

RESUMEN

In many types of tubules, continuity of the lumen is paramount to tubular function, yet how tubules generate lumen continuity in vivo is not known. We recently found that the F-actin-binding protein afadin is required for lumen continuity in developing renal tubules, though its mechanism of action remains unknown. Here, we demonstrate that afadin is required for lumen continuity by orienting the mitotic spindle during cell division. Using an in vitro 3D cyst model, we find that afadin localizes to the cell cortex adjacent to the spindle poles and orients the mitotic spindle. In tubules, cell division may be oriented relative to two axes: longitudinal and apical-basal. Unexpectedly, in vivo examination of early-stage developing nephron tubules reveals that cell division is not oriented in the longitudinal (or planar-polarized) axis. However, cell division is oriented perpendicular to the apical-basal axis. Absence of afadin in vivo leads to misorientation of apical-basal cell division in nephron tubules. Together, these results support a model whereby afadin determines lumen placement by directing apical-basal spindle orientation, resulting in a continuous lumen and normal tubule morphogenesis.


Asunto(s)
División Celular , Túbulos Renales/embriología , Túbulos Renales/metabolismo , Proteínas de Microfilamentos/metabolismo , Animales , Células Cultivadas , Perros , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Enfermedades Renales Quísticas/patología , Túbulos Renales/patología , Células de Riñón Canino Madin Darby , Masculino , Ratones , Morfogénesis , Nefronas/metabolismo , Nefronas/patología , Huso Acromático/metabolismo
2.
J Biol Chem ; 293(42): 16488-16502, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30139743

RESUMEN

Up to 15% of the population have mild to moderate chronic hypomagnesemia, which is associated with type 2 diabetes mellitus, hypertension, metabolic syndrome, and chronic kidney disease. The kidney is the key organ for magnesium homeostasis, but our understanding of renal magnesium regulation is very limited. Uromodulin (UMOD) is the most abundant urinary protein in humans, and here we report that UMOD has a role in renal magnesium homeostasis. Umod-knockout (Umod-/-) mice excreted more urinary magnesium than WT mice and displayed up-regulation of genes promoting magnesium absorption. The majority of magnesium is absorbed in the thick ascending limb. However, both mouse strains responded similarly to the diuretic agent furosemide, indicating appropriate function of the thick ascending limb in the Umod-/- mice. Magnesium absorption is fine-tuned in the distal convoluted tubule (DCT) via the apical magnesium channel transient receptor potential melastatin 6 (TRPM6). We observed decreased apical Trpm6 staining in the DCT of Umod-/- mice. Applying biotinylation assays and whole-cell patch-clamp recordings, we found that UMOD enhances TRPM6 cell-surface abundance and current density from the extracellular space. UMOD physically interacted with TRPM6 and thereby impaired dynamin-dependent TRPM6 endocytosis. WT mice fed a low-magnesium diet had an increased urinary UMOD secretion compared with the same mice on a regular diet. Our results suggest that increased urinary UMOD secretion in low-magnesium states reduces TRPM6 endocytosis and thereby up-regulates TRPM6 cell-surface abundance to defend against further urinary magnesium losses.


Asunto(s)
Homeostasis , Riñón/química , Magnesio/metabolismo , Canales Catiónicos TRPM/metabolismo , Uromodulina/fisiología , Animales , Endocitosis , Furosemida/farmacología , Humanos , Túbulos Renales Distales/metabolismo , Magnesio/orina , Ratones , Ratones Noqueados , Uromodulina/genética
3.
J Am Soc Nephrol ; 29(4): 1128-1140, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29335243

RESUMEN

A critical aspect of kidney function occurs at the glomerulus, the capillary network that filters the blood. The glomerular basement membrane (GBM) is a key component of filtration, yet our understanding of GBM interactions with mesangial cells, specialized pericytes that provide structural stability to glomeruli, is limited. We investigated the role of nephronectin (Npnt), a GBM component and known ligand of α8ß1 integrin. Immunolocalization and in situ hybridization studies in kidneys of adult mice revealed that nephronectin is produced by podocytes and deposited into the GBM. Conditional deletion of Npnt from nephron progenitors caused a pronounced increase in mesangial cell number and mesangial sclerosis. Nephronectin colocalized with α8ß1 integrin to novel, specialized adhesion structures that occurred at sites of mesangial cell protrusion at the base of the capillary loops. Absence of nephronectin disrupted these adhesion structures, leading to mislocalization of α8ß1. Podocyte-specific deletion of Npnt also led to mesangial sclerosis in mice. These results demonstrate a novel role for nephronectin and α8ß1 integrin in a newly described adhesion complex and begin to uncover the molecular interactions between the GBM and mesangial cells, which govern mesangial cell behavior and may have a role in pathologic states.


Asunto(s)
Proteínas de la Matriz Extracelular/fisiología , Membrana Basal Glomerular/fisiología , Mesangio Glomerular/citología , Pericitos/citología , Podocitos/metabolismo , Animales , Adhesión Celular/fisiología , Recuento de Células , Células Epiteliales/metabolismo , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/deficiencia , Femenino , Adhesiones Focales , Eliminación de Gen , Mesangio Glomerular/anomalías , Integrinas/metabolismo , Glomérulos Renales/anomalías , Masculino , Ratones , Ratones Mutantes , Especificidad de Órganos , Pericitos/metabolismo
4.
Dev Biol ; 418(1): 66-74, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27542690

RESUMEN

Previous studies have shown CD34 family member Podocalyxin is required for epithelial lumen formation in vitro. We demonstrate that Endoglycan, a CD34 family member with homology to Podocalyxin, is produced prior to lumen formation in developing nephrons. Endoglycan localizes to Rab11-containing vesicles in nephron progenitors, and then relocalizes to the apical surface as progenitors epithelialize. Once an apical/luminal surface is formed, Endoglycan (and the actin-binding protein Ezrin) localize to large, intraluminal structures that may be vesicles/exosomes. We generated mice lacking Endoglycan and found mutants had timely initiation of lumen formation and continuous lumens, similar to controls. Mice with conditional deletion of both Endoglycan and Podocalyxin in developing nephrons also had normal tubular lumens. Despite this, Endoglycan/Podocalyxin is required for apical recruitment of the adaptor protein NHERF1, but not Ezrin, in podocyte precursors, a subset of the epithelia. In summary, while CD34 family members appear dispensable for lumen formation, our data identify Endoglycan as a novel pre-luminal marker and suggest lumen formation occurs via vesicular trafficking of apical cargo that includes Endoglycan.


Asunto(s)
Antígenos CD34/metabolismo , Mucinas/metabolismo , Nefronas/embriología , Sialoglicoproteínas/metabolismo , Animales , Proteínas del Citoesqueleto/metabolismo , Células Epiteliales/citología , Ratones , Ratones Transgénicos , Mucinas/genética , Nefronas/metabolismo , Fosfoproteínas/metabolismo , Podocitos/citología , Sialoglicoproteínas/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo
5.
J Cell Sci ; 128(23): 4293-305, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26490995

RESUMEN

The Rho GTPase Cdc42 regulates key signaling pathways required for multiple cell functions, including maintenance of shape, polarity, proliferation, migration, differentiation and morphogenesis. Although previous studies have shown that Cdc42 is required for proper epithelial development and maintenance, its exact molecular function in kidney development is not well understood. In this study, we define the specific role of Cdc42 during murine kidney epithelial tubulogenesis by deleting it selectively at the initiation of ureteric bud or metanephric mesenchyme development. Deletion in either lineage results in abnormal tubulogenesis, with profound defects in polarity, lumen formation and the actin cytoskeleton. Ultimately, these defects lead to renal failure. Additionally, in vitro analysis of Cdc42-null collecting duct cells shows that Cdc42 controls these processes by regulating the polarity Par complex (Par3-Par6-aPKC-Cdc42) and the cytoskeletal proteins N-Wasp and ezrin. Thus, we conclude that the principal role of Cdc42 in ureteric bud and metanephric mesenchyme development is to regulate epithelial cell polarity and the actin cytoskeleton.


Asunto(s)
Polaridad Celular/fisiología , Citoesqueleto/metabolismo , Células Epiteliales/metabolismo , Túbulos Renales/embriología , Proteína de Unión al GTP cdc42/metabolismo , Animales , Citoesqueleto/genética , Células Epiteliales/citología , Ratones , Proteína de Unión al GTP cdc42/genética
6.
J Am Soc Nephrol ; 27(11): 3447-3458, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27036738

RESUMEN

Hypercalciuria is a major risk factor for nephrolithiasis. We previously reported that Uromodulin (UMOD) protects against nephrolithiasis by upregulating the renal calcium channel TRPV5. This channel is crucial for calcium reabsorption in the distal convoluted tubule (DCT). Recently, mutations in the gene encoding Mucin-1 (MUC1) were found to cause autosomal dominant tubulointerstitial kidney disease, the same disease caused by UMOD mutations. Because of the similarities between UMOD and MUC1 regarding associated disease phenotype, protein structure, and function as a cellular barrier, we examined whether urinary MUC1 also enhances TRPV5 channel activity and protects against nephrolithiasis. We established a semiquantitative assay for detecting MUC1 in human urine and found that, compared with controls (n=12), patients (n=12) with hypercalciuric nephrolithiasis had significantly decreased levels of urinary MUC1. Immunofluorescence showed MUC1 in the thick ascending limb, DCT, and collecting duct. Applying whole-cell patch-clamp recording of HEK cells, we found that wild-type but not disease mutant MUC1 increased TRPV5 activity by impairing dynamin-2- and caveolin-1-mediated endocytosis of TRPV5. Coimmunoprecipitation confirmed a physical interaction between TRPV5 and MUC1. However, MUC1 did not increase the activity of N-glycan-deficient TRPV5. MUC1 is characterized by variable number tandem repeats (VNTRs) that bind the lectin galectin-3; galectin-3 siRNA but not galectin-1 siRNA prevented MUC1-induced upregulation of TRPV5 activity. Additionally, MUC1 lacking VNTRs did not increase TRPV5 activity. Our results suggest that MUC1 forms a lattice with the N-glycan of TRPV5 via galectin-3, which impairs TRPV5 endocytosis and increases urinary calcium reabsorption.


Asunto(s)
Mucina-1/fisiología , Mucina-1/orina , Nefrolitiasis/etiología , Nefrolitiasis/orina , Canales Catiónicos TRPV/fisiología , Calcio/análisis , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Regulación hacia Arriba
7.
Development ; 140(8): 1774-84, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23487309

RESUMEN

A fundamental process in biology is the de novo formation and morphogenesis of polarized tubules. Although these processes are essential for the formation of multiple metazoan organ systems, little is known about the molecular mechanisms that regulate them. In this study, we have characterized several steps in tubule formation and morphogenesis using the mouse kidney as a model system. We report that kidney mesenchymal cells contain discrete Par3-expressing membrane microdomains that become restricted to an apical domain, coinciding with lumen formation. Once lumen formation has been initiated, elongation occurs by simultaneous extension and additional de novo lumen generation. We demonstrate that lumen formation and elongation require afadin, a nectin adaptor protein implicated in adherens junction formation. Mice that lack afadin in nephron precursors show evidence of Par3-expressing membrane microdomains, but fail to develop normal apical-basal polarity and generate a continuous lumen. Absence of afadin led to delayed and diminished integration of nectin complexes and failure to recruit R-cadherin. Furthermore, we demonstrate that afadin is required for Par complex formation. Together, these results suggest that afadin acts upstream of the Par complex to regulate the integration and/or coalescence of membrane microdomains, thereby establishing apical-basal polarity and lumen formation/elongation during kidney tubulogenesis.


Asunto(s)
Polaridad Celular/fisiología , Túbulos Renales/embriología , Células Madre Mesenquimatosas/fisiología , Proteínas de Microfilamentos/metabolismo , Morfogénesis/fisiología , Proteínas Adaptadoras Transductoras de Señales , Análisis de Varianza , Animales , Moléculas de Adhesión Celular/metabolismo , Proteínas de Ciclo Celular , Técnica del Anticuerpo Fluorescente , Técnicas Histológicas , Procesamiento de Imagen Asistido por Computador , Túbulos Renales/ultraestructura , Ratones , Microscopía Confocal , Microscopía Electrónica
8.
Clin Res Hepatol Gastroenterol ; 48(3): 102290, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38311060

RESUMEN

The primary treatment for early esophageal cancer and precancerous lesions is endoscopic submucosal dissection (ESD). However, this approach leads to a high incidence of postoperative esophageal stenosis, which can significantly impact a patient's quality of life. While various methods are available to prevent post-ESD esophageal stenosis, their effectiveness varies. Therefore, this study aims to provide an overview of the currently employed methods for preventing post-ESD esophageal stenosis in clinical practice in view of assisting clinical practitioners.


Asunto(s)
Resección Endoscópica de la Mucosa , Neoplasias Esofágicas , Estenosis Esofágica , Humanos , Estenosis Esofágica/etiología , Estenosis Esofágica/prevención & control , Resección Endoscópica de la Mucosa/efectos adversos , Resección Endoscópica de la Mucosa/métodos , Calidad de Vida , Neoplasias Esofágicas/patología , Complicaciones Posoperatorias/prevención & control , Complicaciones Posoperatorias/etiología
9.
Front Oncol ; 13: 1271463, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37886180

RESUMEN

Background: Selective lateral lymph node (LLN) dissection with total mesorectal excision after neoadjuvant chemoradiotherapy (nCRT) is pointed out to reduce lateral compartment recurrence and to improve survival in patients with rectal cancer with LLN metastases. This study aimed to explore the safety, surgical indications, and survival outcomes of LLN dissection after nCRT. Methods: This multicenter retrospective study included patients with rectal cancer with clinical evidence of LLN metastases (n = 466) treated across three hospitals in China. Patients who underwent total mesorectal excision and LLN dissection were grouped into nCRT (n = 155) and non-nCRT (n = 291), respectively. Propensity score matching was used to minimize selection bias. Results: After matching, nCRT did not significantly increase the surgery duration, intraoperative blood loss or postoperative complications (P > 0.05). In a multivariate logistic regression analysis, poor/mucinous/signet adenocarcinoma (P = 0.042) and post-nCRT LLN short diameter ≥7 mm (P < 0.001) were independent risk factors for pathological LLN metastasis after nCRT. Overall survival (P < 0.001) and disease-free survival (P < 0.001) were significantly worse in patients with LLN metastasis, which was, however, not an independent risk factor for survival after eliminating confounders. Multivariate prognostic analysis of 40-patient subset with pathological LLN metastasis showed that distant metastasis, metastasis beyond the obturator or internal iliac region, and ≥2 LLN metastasis were independent predictors of poor overall survival. Conclusions: Selective LLN dissection after nCRT is safe and feasible with acceptable perioperative outcomes. Patients with a post-nCRT LLN short diameter ≥7 mm or poor/mucinous/signet adenocarcinoma should receive supplementary LLN dissection after nCRT. However, patients with distant metastasis, metastasis beyond the obturator or internal iliac region, and involvement of ≥2 LLN may not benefit from LLN dissection, and LLN dissection should be carefully considered in such patients.

10.
Mol Reprod Dev ; 79(2): 128-37, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22128093

RESUMEN

Obesity increases the risk of female reproductive tract cancers, but the underlying mechanistic link between the two is ill-defined. Thus, the objective of the current study was to identify obesity-dependent changes in the expression of immediate early (IE) genes that contribute to cell proliferation and differentiation, and epithelial-mesenchymal transition (EMT) genes that promote cell migration. When HeLa cells were treated for 0-48 hr with IGF-1, leptin, TNFα, or IL-6, each individual adipocytokine altered the abundance of IE (cJUN, cFOS, and cMYC) and EMT (SNAI1, SNAI2, and TWIST1) mRNA abundance. For example, IGF-1 increased cJUN and cFOS and decreased cMYC; leptin increased cFOS; IL-6 increased cFOS and cMYC; and TNFα increased cJUN and cFOS mRNA abundance. Likewise, EMT gene expression was altered by IGF-1, TNFα, and IL-6. SNAI1 was increased by IGF-1 and IL-6; SNAI2 was increased by IGF-1 and TNFα; and TWIST1 was increased by TNFα and IL-6. Chronic exposure to adipocytokines also altered EMT gene expression in the whole uterus of obese compared to normal-weight mice. Specifically, there was no difference in cJun, cFos, or cMyc mRNA abundance between normal-weight and obese animals. Snai1, Snai2, and Twist1 mRNA abundance, however, was increased in the uterus of obese females and correlated with increased circulating IGF-1 levels. These data indicate that obesity-dependent alterations in adipocytokine levels regulate the expression of genes associated with cell proliferation and migration, and therefore may provide a plausible mechanism for obesity-dependent increases in cancers of the female reproductive tract.


Asunto(s)
Adipoquinas/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Genes Inmediatos-Precoces/fisiología , Genitales Femeninos/efectos de los fármacos , Adipoquinas/genética , Adipoquinas/metabolismo , Animales , Diferenciación Celular/genética , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Femenino , Genes fos/fisiología , Genes jun/fisiología , Genes myc/fisiología , Genitales Femeninos/metabolismo , Genitales Femeninos/fisiología , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL
11.
Zhonghua Yi Xue Za Zhi ; 88(42): 2999-3002, 2008 Nov 18.
Artículo en Zh | MEDLINE | ID: mdl-19080080

RESUMEN

OBJECTIVE: To explore the effects of acute and chronic murine cytomegalovirus (MCMV) infections on the regulatory T cells (Treg) ratio and protein expression of the Th1/Th2 transcription factors T-bet/GATA-3. METHODS: 120 BALB/c mice were randomly divided into 2 equal groups: MCMV-infected group undergoing infra-peritoneal injection of homogenate of salivary gland containing MCMV, and mock infection group undergoing infra-peritoneal injection of normal homogenate of salivary gland 1, 3, 7, 14, 28, 45, 60, 75, 90, and 120 days after infection 6 mice from each group were killed to examine the viral load of the heart, lung, liver, and kidney by plaque assay to access the status of MCMV infection. Suspension of splenocytes was prepared. The proportion of CD4+CD25+Foxp3+Treg in the splenocytes was measured by flow cytometry. Western blotting was used to detect the protein expression of T-bet/GATA-3. RESULTS: The cutoff point between acute and chronic points was the 28th day. The CD4+CD25+Foxp3+Treg proportion in splenocytes significantly decreased during the acute infection stage and to the lowest level of (1.46+/-0.27)% at day 28, significantly lower than that of the mock infection group [(2.78+/-0.29)%, P<0.05]; then obviously increased in the chronic infection stage, increased to (4.51+/-0.24)% at day 60, significantly higher than that of the mock infection group [(2.69+/-0.12)%, P<0.05], and continued to increase still. The protein level (K value) of T-bet of the MCMV infection group peaked to the level of (0.618+/-0.053) on day 3, obviously higher than that of the mock infected group [(0.205+/-0.026)], then decreased to the level similar to that of the mock infection group on day 28, and was obviously lower than that of the mock infection group on day 75. Whereas the protein level of GATA-3 of the MCMV group increased to (0.836+/-0.061) on day 3, markedly higher than that of the mock infection group (0.398+/-0.022), peaked on day 7, then gradually decreased, and remained at the levels similar to those of the mock infection group from day 75 to day 120. CONCLUSION: In the acute infection stage, MCMV up-regulates the T-bet and GATA-3 protein expression. But during the chronic infection stage, MCMV induces a marked proliferation and activation of Treg cells which further inhibit the Th1 and Th2 reactions, especially Th1 response. Treg proliferation may be an important mechanism of chronic and persistent CMV infection in the host.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Infecciones por Herpesviridae/metabolismo , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/metabolismo , Enfermedad Aguda , Animales , Diferenciación Celular , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Infecciones por Herpesviridae/genética , Ratones , Ratones Endogámicos BALB C , Muromegalovirus/patogenicidad , Bazo/citología , Linfocitos T Reguladores/citología , Células TH1/metabolismo , Células Th2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA