Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 326(2): F202-F218, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38059296

RESUMEN

Angiotensin II increases apical plasma membrane pendrin abundance and function. This study explored the role of the small GTPase Rac1 in the regulation of pendrin by angiotensin II. To do this, we generated intercalated cell (IC) Rac1 knockout mice and observed that IC Rac1 gene ablation reduced the relative abundance of pendrin in the apical region of intercalated cells in angiotensin II-treated mice but not vehicle-treated mice. Similarly, the Rac1 inhibitor EHT 1864 reduced apical pendrin abundance in angiotensin II-treated mice, through a mechanism that does not require aldosterone. This IC angiotensin II-Rac1 signaling cascade modulates pendrin subcellular distribution without significantly changing actin organization. However, NADPH oxidase inhibition with APX 115 reduced apical pendrin abundance in vivo in angiotensin II-treated mice. Moreover, superoxide dismutase mimetics reduced Cl- absorption in angiotensin II-treated cortical collecting ducts perfused in vitro. Since Rac1 is an NADPH subunit, Rac1 may modulate pendrin through NADPH oxidase-mediated reactive oxygen species production. Because pendrin gene ablation blunts the pressor response to angiotensin II, we asked if pendrin blunts the angiotensin II-induced increase in kidney superoxide. Although kidney superoxide was similar in vehicle-treated wild-type and pendrin knockout mice, it was lower in angiotensin II-treated pendrin-null kidneys than in wild-type kidneys. We conclude that angiotensin II acts through Rac1, independently of aldosterone, to increase apical pendrin abundance. Rac1 may stimulate pendrin, at least partly, through NADPH oxidase. This increase in pendrin abundance contributes to the increment in blood pressure and kidney superoxide content seen in angiotensin II-treated mice.NEW & NOTEWORTHY This study defines a new signaling mechanism by which angiotensin II modulates oxidative stress and blood pressure.


Asunto(s)
Angiotensina II , Transportadores de Sulfato , Proteína de Unión al GTP rac1 , Animales , Ratones , Aldosterona/farmacología , Aldosterona/metabolismo , Angiotensina II/farmacología , Angiotensina II/metabolismo , Ratones Noqueados , NADPH Oxidasas/metabolismo , Transportadores de Sulfato/genética , Superóxidos/metabolismo , Proteína de Unión al GTP rac1/metabolismo
2.
Am J Physiol Renal Physiol ; 326(6): F1091-F1100, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38695074

RESUMEN

We have previously shown that kidney collecting ducts make vasopressin. However, the physiological role of collecting duct-derived vasopressin is uncertain. We hypothesized that collecting duct-derived vasopressin is required for the appropriate concentration of urine. We developed a vasopressin conditional knockout (KO) mouse model wherein Cre recombinase expression induces deletion of arginine vasopressin (Avp) exon 1 in the distal nephron. We then used age-matched 8- to 12-wk-old Avp fl/fl;Ksp-Cre(-) [wild type (WT)] and Avp fl/fl;Ksp-Cre(+) mice for all experiments. We collected urine, serum, and kidney lysates at baseline. We then challenged both WT and knockout (KO) mice with 24-h water restriction, water loading, and administration of the vasopressin type 2 receptor agonist desmopressin (1 µg/kg ip) followed by the vasopressin type 2 receptor antagonist OPC-31260 (10 mg/kg ip). We performed immunofluorescence and immunoblot analysis at baseline and confirmed vasopressin KO in the collecting duct. We found that urinary osmolality (UOsm), plasma Na+, K+, Cl-, blood urea nitrogen, and copeptin were similar in WT vs. KO mice at baseline. Immunoblots of the vasopressin-regulated proteins Na+-K+-2Cl- cotransporter, NaCl cotransporter, and water channel aquaporin-2 showed no difference in expression or phosphorylation at baseline. Following 24-h water restriction, WT and KO mice had no differences in UOsm, plasma Na+, K+, Cl-, blood urea nitrogen, or copeptin. In addition, there were no differences in the rate of urinary concentration or dilution as in WT and KO mice UOsm was nearly identical after desmopressin and OPC-31260 administration. We conclude that collecting duct-derived vasopressin is not essential to appropriately concentrate or dilute urine.NEW & NOTEWORTHY Hypothalamic vasopressin is required for appropriate urinary concentration. However, whether collecting duct-derived vasopressin is involved remains unknown. We developed a novel transgenic mouse model to induce tissue-specific deletion of vasopressin and showed that collecting duct-derived vasopressin is not required to concentrate or dilute urine.


Asunto(s)
Desamino Arginina Vasopresina , Túbulos Renales Colectores , Ratones Noqueados , Animales , Túbulos Renales Colectores/metabolismo , Túbulos Renales Colectores/efectos de los fármacos , Desamino Arginina Vasopresina/farmacología , Capacidad de Concentración Renal/efectos de los fármacos , Arginina Vasopresina/metabolismo , Masculino , Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Ratones , Acuaporina 2/metabolismo , Acuaporina 2/genética , Fármacos Antidiuréticos/farmacología , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo , Ratones Endogámicos C57BL , Privación de Agua , Concentración Osmolar , Sodio/orina , Sodio/metabolismo , Vasopresinas/metabolismo , Benzazepinas
3.
Am J Physiol Renal Physiol ; 324(6): F521-F531, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36995926

RESUMEN

The objective of this study was to understand the response of mice lacking insulin-regulated aminopeptidase (IRAP) to an acute water load. For mammals to respond appropriately to acute water loading, vasopressin activity needs to decrease. IRAP degrades vasopressin in vivo. Therefore, we hypothesized that mice lacking IRAP have an impaired ability to degrade vasopressin and, thus, have persistent urinary concentration. Age-matched 8- to 12-wk-old IRAP wild-type (WT) and knockout (KO) male mice were used for all experiments. Blood electrolytes and urine osmolality were measured before and 1 h after water load (∼2 mL sterile water via intraperitoneal injection). Urine was collected from IRAP WT and KO mice for urine osmolality measurements at baseline and after 1 h administration of the vasopressin type 2 receptor antagonist OPC-31260 (10 mg/kg ip). Immunofluorescence and immunoblot analysis were performed on kidneys at baseline and after 1 h acute water load. IRAP was expressed in the glomerulus, thick ascending loop of Henle, distal tubule, connecting duct, and collecting duct. IRAP KO mice had elevated urine osmolality compared with WT mice due to higher membrane expression of aquaporin 2 (AQP2), which was restored to that of controls after administration of OPC-31260. IRAP KO mice developed hyponatremia after an acute water load because they were unable to increase free water excretion due to increased surface expression of AQP2. In conclusion, IRAP is required to increase water excretion in response to an acute water load due to persistent vasopressin stimulation of AQP2.NEW & NOTEWORTHY Insulin-regulated aminopeptidase (IRAP) degrades vasopressin, but its role in urinary concentration and dilution is unknown. Here, we show that IRAP-deficient mice have a high urinary osmolality at baseline and are unable to excrete free water in response to water loading. These results reveal a novel regulatory role for IRAP in urine concentration and dilution.


Asunto(s)
Acuaporina 2 , Insulina , Animales , Masculino , Ratones , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Acuaporina 2/genética , Acuaporina 2/metabolismo , Insulina/metabolismo , Mamíferos/metabolismo , Presión Osmótica , Vasopresinas/farmacología , Vasopresinas/metabolismo , Agua/metabolismo
4.
J Cell Sci ; 134(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34841431

RESUMEN

The main laminin-binding integrins α3ß1, α6ß1 and α6ß4 are co-expressed in the developing kidney collecting duct system. We previously showed that deleting the integrin α3 or α6 subunit in the ureteric bud, which gives rise to the kidney collecting system, caused either a mild or no branching morphogenesis phenotype, respectively. To determine whether these two integrin subunits cooperate in kidney collecting duct development, we deleted α3 and α6 in the developing ureteric bud. The collecting system of the double knockout phenocopied the α3 integrin conditional knockout. However, with age, the mice developed severe inflammation and fibrosis around the collecting ducts, resulting in kidney failure. Integrin α3α6-null collecting duct epithelial cells showed increased secretion of pro-inflammatory cytokines and displayed mesenchymal characteristics, causing loss of barrier function. These features resulted from increased nuclear factor kappa-B (NF-κB) activity, which regulated the Snail and Slug (also known as Snai1 and Snai2, respectively) transcription factors and their downstream targets. These data suggest that laminin-binding integrins play a key role in the maintenance of kidney tubule epithelial cell polarity and decrease pro-inflammatory cytokine secretion by regulating NF-κB-dependent signaling.


Asunto(s)
Integrinas , Túbulos Renales Colectores , Animales , Células Epiteliales , Inflamación/genética , Integrina alfa3beta1 , Integrinas/genética , Laminina/genética , Ratones , FN-kappa B/genética
5.
Am J Physiol Heart Circ Physiol ; 322(5): H857-H866, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35333120

RESUMEN

Pressure overload of the heart is characterized by concentric hypertrophy and interstitial fibrosis. Cardiac fibroblasts (CFs) in the ventricular wall become activated during injury and synthesize and compact the extracellular matrix, which causes interstitial fibrosis and stiffening of the ventricular heart walls. Talin1 (Tln1) and Talin2 (Tln2) are mechanosensitive proteins that participate in focal adhesion transmission of signals from the extracellular environment to the actin cytoskeleton of CFs. The aim of the present study was to determine whether the removal of Tln1 and Tln2 from CFs would reduce interstitial fibrosis and cardiac hypertrophy. Twelve-week-old male and female Tln2-null (Tln2-/-) and Tln2-null, CF-specific Tln1 knockout (Tln2-/-;Tln1CF-/-) mice were given angiotensin-II (ANG II) (1.5 mg/kg/day) or saline through osmotic pumps for 8 wk. Cardiomyocyte area and measures of heart thickness were increased in the male ANG II-infused Tln2-/-;Tln1CF-/- mice, whereas there was no increase in interstitial fibrosis. Systolic blood pressure was increased in the female Tln2-/-;Tln1CF-/- mice after ANG II infusion compared with the Tln2-/- mice. However, there was no increase in cardiac hypertrophy in the Tln2-/-;Tln1CF-/- mice, which was seen in the Tln2-/- mice. Collectively, these data indicate that in male mice, the absence of Tln1 and Tln2 in CFs leads to cardiomyocyte hypertrophy in response to ANG II, whereas it results in a hypertrophy-resistant phenotype in female mice. These findings have important implications for the role of mechanosensitive proteins in CFs and their impact on cardiomyocyte function in the pathogenesis of hypertension and cardiac hypertrophy.NEW & NOTEWORTHY The role of talins has been previously studied in cardiomyocytes; however, these mechanotransductive proteins that are members of the focal adhesion complex have not been examined in cardiac fibroblasts previously. We hypothesized that loss of talins in cardiac fibroblasts would reduce interstitial fibrosis in the heart with a pressure overload model. However, we found that although loss of talins did not alter fibrosis, it did result in cardiomyocyte and ventricular hypertrophy.


Asunto(s)
Miocitos Cardíacos , Talina , Angiotensina II/farmacología , Animales , Cardiomegalia/metabolismo , Femenino , Fibroblastos/metabolismo , Fibrosis , Masculino , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Talina/genética , Talina/metabolismo
6.
Am J Pathol ; 191(2): 309-319, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33159885

RESUMEN

A unique and complex microstructure underlies the diverse functions of the liver. Breakdown of this organization, as occurs in fibrosis and cirrhosis, impairs liver function and leads to disease. The role of integrin ß1 was examined both in establishing liver microstructure and recreating it after injury. Embryonic deletion of integrin ß1 in the liver disrupts the normal development of hepatocyte polarity, specification of cell-cell junctions, and canalicular formation. This in turn leads to the expression of transforming growth factor ß (TGF-ß) and widespread fibrosis. Targeted deletion of integrin ß1 in adult hepatocytes prevents recreation of normal hepatocyte architecture after liver injury, with resultant fibrosis. In vitro, integrin ß1 is essential for canalicular formation and is needed to prevent stellate cell activation by modulating TGF-ß. Taken together, these findings identify integrin ß1 as a key determinant of liver architecture with a critical role as a regulator of TGF-ß secretion. These results suggest that disrupting the hepatocyte-extracellular matrix interaction is sufficient to drive fibrosis.


Asunto(s)
Integrina beta1/metabolismo , Regeneración Hepática/fisiología , Hígado/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Matriz Extracelular/metabolismo , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Ratones , Ratones Transgénicos
7.
J Cell Sci ; 132(20)2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31558679

RESUMEN

Synthetic hydrogels with controlled physicochemical matrix properties serve as powerful in vitro tools to dissect cell-extracellular matrix (ECM) interactions that regulate epithelial morphogenesis in 3D microenvironments. In addition, these fully defined matrices overcome the lot-to-lot variability of naturally derived materials and have provided insights into the formation of rudimentary epithelial organs. Therefore, we engineered a fully defined synthetic hydrogel with independent control over proteolytic degradation, mechanical properties, and adhesive ligand type and density to study the impact of ECM properties on epithelial tubulogenesis for inner medullary collecting duct (IMCD) cells. Protease sensitivity of the synthetic material for membrane-type matrix metalloproteinase-1 (MT1-MMP, also known as MMP14) was required for tubulogenesis. Additionally, a defined range of matrix elasticity and presentation of RGD adhesive peptide at a threshold level of 2 mM ligand density were required for epithelial tubulogenesis. Finally, we demonstrated that the engineered hydrogel supported organization of epithelial tubules with a lumen and secreted laminin. This synthetic hydrogel serves as a platform that supports epithelial tubular morphogenetic programs and can be tuned to identify ECM biophysical and biochemical properties required for epithelial tubulogenesis.


Asunto(s)
Microambiente Celular , Células Epiteliales/metabolismo , Matriz Extracelular/química , Hidrogeles/química , Túbulos Renales Colectores/metabolismo , Túbulos Renales/metabolismo , Animales , Línea Celular Transformada , Células Epiteliales/citología , Túbulos Renales/citología , Túbulos Renales Colectores/citología , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones , Oligopéptidos/química
8.
FASEB J ; 34(6): 7885-7904, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32297675

RESUMEN

Nearly 6 million Americans suffer from heart failure. Increased fibrosis contributes to functional decline of the heart that leads to heart failure. Previously, we identified a mechanosensitive protein, small proline-rich repeat 3 (SPRR3), in vascular smooth muscle cells of atheromas. In this study, we demonstrate SPRR3 expression in cardiac fibroblasts which is induced in activated fibroblasts following pressure-induced heart failure. Sprr3 deletion in mice showed preserved cardiac function and reduced interstitial fibrosis in vivo and reduced fibroblast proliferation and collagen expression in vitro. SPRR3 loss resulted in reduced activation of Akt, FAK, ERK, and p38 signaling pathways, which are coordinately regulated by integrins and growth factors. SPRR3 deletion did not impede integrin-associated functions including cell adhesion, migration, or contraction. SPRR3 loss resulted in reduced activation of PDGFRß in fibroblasts. This was not due to the reduced PDGFRß expression levels or decreased binding of the PDGF ligand to PDGFRß. SPRR3 facilitated the association of integrin ß1 with PDGFRß and subsequently fibroblast proliferation, suggesting a role in PDGFRß-Integrin synergy. We postulate that SPRR3 may function as a conduit for the coordinated activation of PDGFRß by integrin ß1, leading to augmentation of fibroblast proliferation and matrix synthesis downstream of biomechanical and growth factor signals.


Asunto(s)
Proliferación Celular/fisiología , Proteínas Ricas en Prolina del Estrato Córneo/metabolismo , Fibroblastos/metabolismo , Corazón/fisiología , Integrina beta1/metabolismo , Miocardio/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Adhesión Celular/fisiología , Colágeno/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/metabolismo , Transducción de Señal/fisiología
9.
J Cell Sci ; 131(22)2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30333137

RESUMEN

Integrins are α/ß heterodimers that interconvert between inactive and active states. In the active state the α/ß cytoplasmic domains recruit integrin-activating proteins and separate the transmembrane and cytoplasmic (TMcyto) domains (unclasped TMcyto). Conversely, in the inactive state the α/ß TMcyto domains bind integrin-inactivating proteins, resulting in the association of the TMcyto domains (clasped TMcyto). Here, we report the isolation of integrin cytoplasmic tail interactors using either lipid bicelle-incorporated integrin TMcyto domains (α5, αM, αIIb, ß1, ß2 and ß3 integrin TMcyto) or a clasped, lipid bicelle-incorporated αMß2 TMcyto. Among the proteins found to preferentially bind clasped rather than the isolated αM and ß2 subunits was L-plastin (LCP1, also known as plastin-2), which binds to and maintains the inactive state of αMß2 integrin in vivo and thereby regulates leukocyte adhesion to integrin ligands under flow. Our findings offer a global view on cytoplasmic proteins interacting with different integrins and provide evidence for the existence of conformation-specific integrin interactors.


Asunto(s)
Adhesión Celular/fisiología , Leucocitos/citología , Leucocitos/metabolismo , Antígeno de Macrófago-1/metabolismo , Proteínas de Microfilamentos/metabolismo , Animales , Membrana Celular/metabolismo , Citoplasma/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Conformación Proteica , Células RAW 264.7
10.
Development ; 144(22): 4148-4158, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28993400

RESUMEN

Kidney collecting system development requires integrin-dependent cell-extracellular matrix interactions. Integrins are heterodimeric transmembrane receptors consisting of α and ß subunits; crucial integrins in the kidney collecting system express the ß1 subunit. The ß1 cytoplasmic tail has two NPxY motifs that mediate functions by binding to cytoplasmic signaling and scaffolding molecules. Talins, scaffolding proteins that bind to the membrane proximal NPxY motif, are proposed to activate integrins and to link them to the actin cytoskeleton. We have defined the role of talin binding to the ß1 proximal NPxY motif in the developing kidney collecting system in mice that selectively express a Y-to-A mutation in this motif. The mice developed a hypoplastic dysplastic collecting system. Collecting duct cells expressing this mutation had moderate abnormalities in cell adhesion, migration, proliferation and growth factor-dependent signaling. In contrast, mice lacking talins in the developing ureteric bud developed kidney agenesis and collecting duct cells had severe cytoskeletal, adhesion and polarity defects. Thus, talins are essential for kidney collecting duct development through mechanisms that extend beyond those requiring binding to the ß1 integrin subunit NPxY motif.


Asunto(s)
Integrina beta1/metabolismo , Morfogénesis , Talina/metabolismo , Uréter/citología , Uréter/embriología , Uniones Adherentes/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Adhesión Celular , Membrana Celular/metabolismo , Polaridad Celular , Regulación del Desarrollo de la Expresión Génica , Integrina beta1/química , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/embriología , Ratones Endogámicos C57BL , Mutación/genética , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo , Uréter/metabolismo
11.
J Am Soc Nephrol ; 30(9): 1605-1624, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31383731

RESUMEN

BACKGROUND: The discoidin domain receptor 1 (DDR1) is activated by collagens, upregulated in injured and fibrotic kidneys, and contributes to fibrosis by regulating extracellular matrix production, but how DDR1 controls fibrosis is poorly understood. DDR1 is a receptor tyrosine kinase (RTK). RTKs can translocate to the nucleus via a nuclear localization sequence (NLS) present on the receptor itself or a ligand it is bound to. In the nucleus, RTKs regulate gene expression by binding chromatin directly or by interacting with transcription factors. METHODS: To determine whether DDR1 translocates to the nucleus and whether this event is mediated by collagen-induced DDR1 activation, we generated renal cells expressing wild-type or mutant forms of DDR1 no longer able to bind collagen. Then, we determined the location of the DDR1 upon collagen stimulation. Using both biochemical assays and immunofluorescence, we analyzed the steps involved in DDR1 nuclear translocation. RESULTS: We show that although DDR1 and its natural ligand, collagen, lack an NLS, DDR1 is present in the nucleus of injured human and mouse kidney proximal tubules. We show that DDR1 nuclear translocation requires collagen-mediated receptor activation and interaction of DDR1 with SEC61B, a component of the Sec61 translocon, and nonmuscle myosin IIA and ß-actin. Once in the nucleus, DDR1 binds to chromatin to increase the transcription of collagen IV, a major collagen upregulated in fibrosis. CONCLUSIONS: These findings reveal a novel mechanism whereby activated DDR1 translates to the nucleus to regulate synthesis of profibrotic molecules.


Asunto(s)
Colágeno Tipo IV/genética , Colágeno Tipo I/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Túbulos Renales Proximales/metabolismo , Actinas/metabolismo , Lesión Renal Aguda/metabolismo , Animales , Transporte Biológico , Línea Celular , Núcleo Celular , Cromatina/metabolismo , Colágeno Tipo I/farmacología , Colágeno Tipo IV/metabolismo , Receptor con Dominio Discoidina 1/genética , Humanos , Túbulos Renales Proximales/patología , Masculino , Ratones , Cadenas Pesadas de Miosina/metabolismo , Señales de Localización Nuclear , Proteína 4 de Unión a Retinoblastoma/metabolismo , Canales de Translocación SEC/metabolismo , Transcripción Genética
12.
Am J Physiol Renal Physiol ; 316(2): F382-F395, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30207166

RESUMEN

Proteinuria, the most common symptom of renal injury, is an independent factor for renal tubular injury. However, the underlying mechanism remains to be fully elucidated. Mitochondrion is an important target for proteinuria-induced renal tubular cell injury. Insufficient mitophagy exacerbates cell injury by initiating mitochondrial dysfunction-related cell apoptosis. In the experiment, the role of NIP3-like protein X (NIX)-mediated mitophagy was investigated in proteinuria-induced renal injury. In this study, we demonstrated that NIX expression was reduced in renal tubules and correlated with the decline of estimated glomerular filtration rate and increase of the proteinuria in patients. In proteinuric mice, NIX-mediated mitophagy was significantly suppressed. Meanwhile, the proteinuric mice exhibited renal dysfunction, increased mitochondrial fragmentation, and tubular cell apoptosis. Overexpression of NIX attenuated those disruptions in proteinuric mice. In cultured renal tubular epithelial cells, albumin induced a decrease in NIX-mediated mitophagy and an increase in cell apoptosis. Overexpression of NIX attenuated albumin-induced cell apoptosis, whereas NIX siRNA aggravated these perturbations. These results indicate that proteinuria suppresses NIX-mediated mitophagy in the renal tubular epithelial cell, which triggers the cell undergoing mitochondria-dependent cell apoptosis. Collectively, our finding suggests that restoration of NIX-mediated mitophagy might be a novel therapeutic target for alleviating proteinuria-induced kidney injury.


Asunto(s)
Albuminuria/metabolismo , Apoptosis , Células Epiteliales/metabolismo , Túbulos Renales/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Mitofagia , Nefrosis/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Albuminuria/genética , Albuminuria/patología , Albuminuria/fisiopatología , Animales , Estudios de Casos y Controles , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Tasa de Filtración Glomerular , Humanos , Túbulos Renales/patología , Túbulos Renales/fisiopatología , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/patología , Proteínas Mitocondriales/genética , Nefrosis/genética , Nefrosis/patología , Nefrosis/fisiopatología , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Adulto Joven
13.
Am J Physiol Endocrinol Metab ; 316(6): E1118-E1135, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30835508

RESUMEN

Integrin-linked kinase (ILK) is a critical intracellular signaling node for integrin receptors. Its role in liver development is complex, as ILK deletion at E10.5 (before hepatocyte differentiation) results in biochemical and morphological differences that resolve as mice age. Nevertheless, mice with ILK depleted specifically in hepatocytes are protected from the hepatic insulin resistance during obesity. Despite the potential importance of hepatocyte ILK to metabolic health, it is unknown how ILK controls hepatic metabolism or glucoregulation. The present study tested the role of ILK in hepatic metabolism and glucoregulation by deleting it specifically in hepatocytes, using a cre-lox system that begins expression at E15.5 (after initiation of hepatocyte differentiation). These mice develop the most severe morphological and glucoregulatory abnormalities at 6 wk, but these gradually resolve with age. After identifying when the deletion of ILK caused a severe metabolic phenotype, in depth studies were performed at this time point to define the metabolic programs that coordinate control of glucoregulation that are regulated by ILK. We show that 6-wk-old ILK-deficient mice have higher glucose tolerance and decreased net glycogen synthesis. Additionally, ILK was shown to be necessary for transcription of mitochondrial-related genes, oxidative metabolism, and maintenance of cellular energy status. Thus, ILK is required for maintaining hepatic transcriptional and metabolic programs that sustain oxidative metabolism, which are required for hepatic maintenance of glucose homeostasis.


Asunto(s)
Glucemia/metabolismo , Hepatocitos/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Hígado/metabolismo , Obesidad/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Factores de Edad , Animales , Diferenciación Celular , Respiración de la Célula , Metabolismo Energético , Técnicas de Inactivación de Genes , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis , Inflamación , Hígado/embriología , Hígado/patología , Cirrosis Hepática , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo
14.
Kidney Int ; 93(3): 643-655, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29241625

RESUMEN

Cell-matrix interactions and podocyte intercellular junctions are key for maintaining the glomerular filtration barrier. Vinculin, a cytoplasmic protein, couples actin filaments to integrin-mediated cell-matrix adhesions and to cadherin-based intercellular junctions. Here, we examined the role of vinculin in podocytes by the generation of a podocyte-specific knockout mouse. Mice lacking podocyte vinculin had increased albuminuria and foot process effacement following injury in vivo. Analysis of primary podocytes isolated from the mutant mice revealed defects in cell protrusions, altered focal adhesion size and signaling, as well as impaired cell migration. Furthermore, we found a marked mislocalization of the intercellular junction protein zonula occludens-1. In kidney sections from patients with focal segmental glomerulosclerosis, minimal change disease and membranous nephropathy, we observed dramatic differences in the expression levels and localization of vinculin. Thus, our results suggest that vinculin is necessary to maintain the integrity of the glomerular filtration barrier by modulating podocyte foot processes and stabilizing intercellular junctions.


Asunto(s)
Glomerulonefritis Membranosa/metabolismo , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Nefrosis Lipoidea/metabolismo , Podocitos/metabolismo , Vinculina/metabolismo , Albuminuria/genética , Albuminuria/metabolismo , Animales , Movimiento Celular , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/patología , Células Cultivadas , Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Adhesiones Focales/patología , Glomerulonefritis Membranosa/patología , Glomeruloesclerosis Focal y Segmentaria/patología , Mecanotransducción Celular , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrosis Lipoidea/patología , Fosforilación , Podocitos/patología , Vinculina/deficiencia , Vinculina/genética , Proteína de la Zonula Occludens-1/metabolismo
16.
Kidney Int ; 91(3): 552-560, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27773427

RESUMEN

Tubulointerstitial fibrosis, tubular atrophy, and peritubular capillary rarefaction are major hallmarks of chronic kidney disease. The tubulointerstitium consists of multiple cell components including tubular epithelial, mesenchymal (fibroblasts and pericytes), endothelial, and inflammatory cells. Crosstalk among these cell components is a key component in the pathogenesis of this complex disease. After severe or recurrent injury, the renal tubular epithelial cells undergo changes in structure and cell cycle that are accompanied by altered expression and production of cytokines. These cytokines contribute to the initiation of the fibrotic response by favoring activation of fibroblasts, recruitment of inflammatory cells, and loss of endothelial cells. This review focuses on how augmented growth factor and cytokine production induces epithelial crosstalk with cells in the interstitium to promote progressive tubulointerstitial fibrosis after renal injury.


Asunto(s)
Comunicación Celular , Citocinas/metabolismo , Riñón/metabolismo , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Animales , Citocinas/inmunología , Fibrosis , Humanos , Riñón/inmunología , Riñón/patología , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/patología
17.
J Cell Sci ; 128(5): 1023-33, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25609703

RESUMEN

PINCH-1 is a LIM-only domain protein that forms a ternary complex with integrin-linked kinase (ILK) and parvin (to form the IPP complex) downstream of integrins. Here, we demonstrate that PINCH-1 (also known as Lims1) gene ablation in the epidermis of mice caused epidermal detachment from the basement membrane, epidermal hyperthickening and progressive hair loss. PINCH-1-deficient keratinocytes also displayed profound adhesion, spreading and migration defects in vitro that were substantially more severe than those of ILK-deficient keratinocytes indicating that PINCH-1 also exerts functions in an ILK-independent manner. By isolating the PINCH-1 interactome, the LIM-domain-containing and actin-binding protein epithelial protein lost in neoplasm (EPLIN, also known as LIMA1) was identified as a new PINCH-1-associated protein. EPLIN localized, in a PINCH-1-dependent manner, to integrin adhesion sites of keratinocytes in vivo and in vitro and its depletion severely attenuated keratinocyte spreading and migration on collagen and fibronectin without affecting PINCH-1 levels in focal adhesions. Given that the low PINCH-1 levels in ILK-deficient keratinocytes were sufficient to recruit EPLIN to integrin adhesions, our findings suggest that PINCH-1 regulates integrin-mediated adhesion of keratinocytes through the interactions with ILK as well as EPLIN.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Adhesiones Focales/metabolismo , Integrinas/metabolismo , Queratinocitos/metabolismo , Proteínas con Dominio LIM/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Proteínas del Citoesqueleto/genética , Adhesiones Focales/genética , Integrinas/genética , Queratinocitos/citología , Proteínas con Dominio LIM/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos
18.
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
19.
Development ; 141(24): 4751-62, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25395457

RESUMEN

Integrin-dependent interactions between cells and extracellular matrix regulate lung development; however, specific roles for ß1-containing integrins in individual cell types, including epithelial cells, remain incompletely understood. In this study, the functional importance of ß1 integrin in lung epithelium during mouse lung development was investigated by deleting the integrin from E10.5 onwards using surfactant protein C promoter-driven Cre. These mutant mice appeared normal at birth but failed to gain weight appropriately and died by 4 months of age with severe hypoxemia. Defects in airway branching morphogenesis in association with impaired epithelial cell adhesion and migration, as well as alveolarization defects and persistent macrophage-mediated inflammation were identified. Using an inducible system to delete ß1 integrin after completion of airway branching, we showed that alveolarization defects, characterized by disrupted secondary septation, abnormal alveolar epithelial cell differentiation, excessive collagen I and elastin deposition, and hypercellularity of the mesenchyme occurred independently of airway branching defects. By depleting macrophages using liposomal clodronate, we found that alveolarization defects were secondary to persistent alveolar inflammation. ß1 integrin-deficient alveolar epithelial cells produced excessive monocyte chemoattractant protein 1 and reactive oxygen species, suggesting a direct role for ß1 integrin in regulating alveolar homeostasis. Taken together, these studies define distinct functions of epithelial ß1 integrin during both early and late lung development that affect airway branching morphogenesis, epithelial cell differentiation, alveolar septation and regulation of alveolar homeostasis.


Asunto(s)
Células Epiteliales/metabolismo , Integrina beta1/metabolismo , Pulmón/embriología , Organogénesis/fisiología , Alveolos Pulmonares/embriología , Animales , Lavado Broncoalveolar , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Quimiocina CCL2/metabolismo , Ensayo de Inmunoadsorción Enzimática , Matriz Extracelular/metabolismo , Integrasas/metabolismo , Ratones , Microscopía Confocal , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico
20.
Am J Physiol Renal Physiol ; 310(6): F499-510, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26739889

RESUMEN

Transforming growth factor-ß (TGF-ß) and hepatocyte growth factor (HGF) play key roles in regulating the response to renal injury but are thought to mediate divergent effects on cell behavior. However, how TGF-ß signaling alters the response to HGF in epithelia, the key site of HGF signaling in the injured kidney, is not well studied. Contrary to our expectation, we showed that deletion of the TGF-ß type II receptor in conditionally immortalized proximal tubule (PT) cells impaired HGF-dependent signaling. This reduced signaling was due to decreased transcription of c-Met, the HGF receptor, and the TGF-ß-dependent c-Met transcription and increased response to HGF in PT cells were mediated by the Notch pathway. The interactions of TGF-ß, HGF, and Notch pathways had biologically significant effects on branching morphogenesis, cell morphology, migration, and proliferation. In conclusion, epithelial TGF-ß signaling promotes HGF signaling in a Notch-dependent pathway. These findings suggest that TGF-ß modulates PT responses not only by direct effects, but also by affecting other growth factor signaling pathways.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Túbulos Renales Proximales/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores Notch/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Células Cultivadas , Ratones , Proteínas Serina-Treonina Quinasas/deficiencia , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Transducción de Señal , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA