Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Am J Physiol Renal Physiol ; 326(2): F178-F188, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-37994409

RESUMEN

Chronic kidney disease is increasing at an alarming rate and correlates with the increase in diabetes, obesity, and hypertension that disproportionately impact socioeconomically disadvantaged communities. Iron plays essential roles in many biological processes including oxygen transport, mitochondrial function, cell proliferation, and regeneration. However, excess iron induces the generation and propagation of reactive oxygen species, which lead to oxidative stress, cellular damage, and ferroptosis. Iron homeostasis is regulated in part by the kidney through iron resorption from the glomerular filtrate and exports into the plasma by ferroportin (FPN). Yet, the impact of iron overload in the kidney has not been addressed. To test more directly whether excess iron accumulation is toxic to kidneys, we generated a kidney proximal tubule-specific knockout of FPN. Despite significant intracellular iron accumulation in FPN mutant tubules, basal kidney function was not measurably different from wild type kidneys. However, upon induction of acute kidney injury (AKI), FPN mutant kidneys exhibited significantly more damage and failed recovery, evidence for ferroptosis, and increased fibrosis. Thus, disruption of iron export in proximal tubules, leading to iron overload, can significantly impair recovery from AKI and can contribute to progressive renal damage indicative of chronic kidney disease. Understanding the mechanisms that regulate iron homeostasis in the kidney may provide new therapeutic strategies for progressive kidney disease and other ferroptosis-associated disorders.NEW & NOTEWORTHY Physiological iron homeostasis depends in part on renal resorption and export into the plasma. We show that specific deletion of iron exporters in the proximal tubules sensitizes cells to injury and inhibits recovery. This can promote a chronic kidney disease phenotype. Our paper demonstrates the need for iron balance in the proximal tubules to maintain and promote healthy recovery after acute kidney injury.


Asunto(s)
Lesión Renal Aguda , Proteínas de Transporte de Catión , Sobrecarga de Hierro , Insuficiencia Renal Crónica , Humanos , Riñón/metabolismo , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Homeostasis/fisiología , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo
2.
Kidney Int ; 105(2): 312-327, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37977366

RESUMEN

Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part, this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Identifying the molecular and genetic regulators unique to nephron segments that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. Here, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI and found them upregulated after severe AKI and correlated with chronic injury. Surprisingly, proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to pre-conditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of proximal tubule cells in the S3 segment that displayed features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic pre-conditioning, and female sex. Thus, our results identified a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both the injury response and protection from ischemic AKI.


Asunto(s)
Lesión Renal Aguda , Túbulos Renales Proximales , Factor de Transcripción PAX2 , Factor de Transcripción PAX8 , Insuficiencia Renal Crónica , Animales , Femenino , Ratones , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/genética , Isquemia/complicaciones , Túbulos Renales Proximales/patología , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/genética , Daño por Reperfusión/genética , Factor de Transcripción PAX8/genética , Factor de Transcripción PAX8/metabolismo , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo
3.
Dev Biol ; 472: 18-29, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33428890

RESUMEN

The vertebrate eye anlage grows out of the brain and folds into bilayered optic cups. The eye is patterned along multiple axes, precisely controlled by genetic programs, to delineate neural retina, pigment epithelium, and optic stalk tissues. Pax genes encode developmental regulators of key morphogenetic events, with Pax2 being essential for interpreting inductive signals, including in the eye. PAX2 mutations cause ocular coloboma, when the ventral optic fissure fails to close. Previous studies established that Pax2 is necessary for fissure closure and to maintain the neural retina -- glial optic stalk boundary. Using a Pax2GFP/+ knock-in allele we discovered that the mutant optic nerve head (ONH) lacks molecular boundaries with the retina and RPE, rendering the ONH larger than normal. This was preceded by ventronasal cup mispatterning, a burst of overproliferation and followed by optic cup apoptosis. Our findings support the hypothesis that ONH cells are tripotential, requiring Pax2 to remain committed to glial fates. This work extends current models of ocular development, contributes to broader understanding of tissue boundary formation and informs the underlying mechanisms of human coloboma.


Asunto(s)
Ojo/embriología , Ojo/metabolismo , Disco Óptico/embriología , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Proliferación Celular/genética , Coloboma/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Disco Óptico/anomalías , Disco Óptico/citología , Retina/embriología , Células Madre/metabolismo
4.
J Am Soc Nephrol ; 31(6): 1212-1225, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32381599

RESUMEN

BACKGROUND: As the glomerular filtrate passes through the nephron and into the renal medulla, electrolytes, water, and urea are reabsorbed through the concerted actions of solute carrier channels and aquaporins at various positions along the nephron and in the outer and inner medulla. Proliferating stem cells expressing the nuclear transcription factor Pax2 give rise to renal epithelial cells. Pax2 expression ends once the epithelial cells differentiate into mature proximal and distal tubules, whereas expression of the related Pax8 protein continues. The collecting tubules and renal medulla are derived from Pax2-positive ureteric bud epithelia that continue to express Pax2 and Pax8 in adult kidneys. Despite the crucial role of Pax2 in renal development, functions for Pax2 or Pax8 in adult renal epithelia have not been established. METHODS: To examine the roles of Pax2 and Pax8 in the adult mouse kidney, we deleted either Pax2, Pax8, or both genes in adult mice and examined the resulting phenotypes and changes in gene expression patterns. We also explored the mechanism of Pax8-mediated activation of potential target genes in inner medullary collecting duct cells. RESULTS: Mice with induced deletions of both Pax2 and Pax8 exhibit severe polyuria that can be attributed to significant changes in the expression of solute carriers, such as the urea transporters encoded by Slc14a2, as well as aquaporins within the inner and outer medulla. Furthermore, Pax8 expression is induced by high-salt levels in collecting duct cells and activates the Slc14a2 gene by recruiting a histone methyltransferase complex to the promoter. CONCLUSIONS: These data reveal novel functions for Pax proteins in adult renal epithelia that are essential for retaining water and concentrating urine.


Asunto(s)
Acuaporinas/fisiología , Capacidad de Concentración Renal/fisiología , Riñón/fisiología , Proteínas de Transporte de Membrana/fisiología , Factor de Transcripción PAX2/fisiología , Factor de Transcripción PAX8/fisiología , Animales , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Osmorregulación , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX8/genética , Transportadores de Urea
5.
J Biol Chem ; 292(22): 9051-9062, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28424263

RESUMEN

Obesity and its associated complications such as insulin resistance and non-alcoholic fatty liver disease are reaching epidemic proportions. In mice, the TGF-ß superfamily is implicated in the regulation of white and brown adipose tissue differentiation. The kielin/chordin-like protein (KCP) is a secreted regulator of the TGF-ß superfamily pathways that can inhibit both TGF-ß and activin signals while enhancing bone morphogenetic protein (BMP) signaling. However, KCP's effects on metabolism and obesity have not been studied in animal models. Therefore, we examined the effects of KCP loss or gain of function in mice that were maintained on either a regular or a high-fat diet. KCP loss sensitized the mice to obesity and associated complications such as glucose intolerance and adipose tissue inflammation and fibrosis. In contrast, transgenic mice that expressed KCP in the kidney, liver, and adipose tissues were resistant to developing high-fat diet-induced obesity and had significantly reduced white adipose tissue. Moreover, KCP overexpression shifted the pattern of SMAD signaling in vivo, increasing the levels of phospho (P)-SMAD1 and decreasing P-SMAD3. Adipocytes in culture showed a cell-autonomous effect in response to added TGF-ß1 or BMP7. Metabolic profiling indicated increased energy expenditure in KCP-overexpressing mice and reduced expenditure in the KCP mutants with no effect on food intake or activity. These findings demonstrate that shifting the TGF-ß superfamily signaling with a secreted protein can alter the physiology and thermogenic properties of adipose tissue to reduce obesity even when mice are fed a high-fat diet.


Asunto(s)
Adipocitos/metabolismo , Proteínas Portadoras/metabolismo , Grasas de la Dieta/efectos adversos , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Transducción de Señal , Adipocitos/patología , Animales , Proteína Morfogenética Ósea 7/genética , Proteína Morfogenética Ósea 7/metabolismo , Proteínas Portadoras/genética , Grasas de la Dieta/farmacología , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/genética , Síndrome Metabólico/patología , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética , Obesidad/patología , Especificidad de Órganos/genética , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G587-G598, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27514479

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is a common cause of chronic liver disease and is increasing with the rising rate of obesity in the developed world. Signaling pathways known to influence the rate of lipid deposition in liver, known as hepatic steatosis, include the transforming growth factor (TGF) superfamily, which function through the SMAD second messengers. The kielin/chordin-like protein (KCP) is a large secreted protein that can enhance bone morphogenetic protein signaling while suppressing TGF-ß signaling in cells and in genetically modified mice. In this report, we show that aging KCP mutant (Kcp-/-) mice are increasingly susceptible to developing hepatic steatosis and liver fibrosis. When young mice are put on a high-fat diet, Kcp-/- mice are also more susceptible to developing liver pathology, compared with their wild-type littermates. Furthermore, mice that express a Pepck-KCP transgene (KcpTg) in the liver are resistant to developing liver pathology even when fed a high-fat diet. Analyses of liver tissues reveal a significant reduction of P-Smad3, consistent with a role for KCP in suppressing TGF-ß signaling. Transcriptome analyses show that livers from Kcp-/- mice fed a normal diet are more like wild-type livers from mice fed a high-fat diet. However, the KCP transgene can suppress many of the changes in liver gene expression that are due to a high-fat diet. These data demonstrate that shifting the TGF-ß signaling paradigm with the secreted regulatory protein KCP can significantly alter the liver pathology in aging mice and in diet-induced NAFLD.


Asunto(s)
Proteínas Portadoras/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transducción de Señal/fisiología , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Proteínas Portadoras/genética , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Hígado/patología , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Fosforilación , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
7.
J Am Soc Nephrol ; 24(6): 897-905, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23539757

RESUMEN

The secreted kielin/chordin-like (KCP) protein, one of a family of cysteine-rich proteins, suppresses TGF-ß signaling by sequestering the ligand from its receptor, but it enhances bone morphogenetic protein (BMP) signaling by promoting ligand-receptor interactions. Given the critical roles for TGF-ß and BMP proteins in enhancing or suppressing renal interstitial fibrosis, respectively, we examined whether secreted KCP could attenuate renal fibrosis in mouse models of chronic and acute disease. Transgenic mice that express KCP in adult kidneys showed significantly less expression of collagen IV, α-smooth muscle actin, and other markers of disease progression in the unilateral ureteral obstruction model of renal interstitial fibrosis. In the folic acid nephrotoxicity model of acute tubular necrosis, mice expressing KCP survived high doses of folic acid that were lethal for wild-type mice. With a lower dose of folic acid, mice expressing KCP exhibited improved renal recovery compared with wild-type mice. Thus, these data suggest that extracellular regulation of the TGF-ß/BMP signaling axis by KCP, and by extension possibly other cysteine-rich domain proteins, can attenuate both acute and chronic renal injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteínas Portadoras/metabolismo , Necrosis Tubular Aguda/metabolismo , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal/fisiología , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Animales , Proteínas Portadoras/química , Proteínas Portadoras/genética , Progresión de la Enfermedad , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Fibrosis , Regulación del Desarrollo de la Expresión Génica/fisiología , Necrosis Tubular Aguda/patología , Necrosis Tubular Aguda/fisiopatología , Ratones , Ratones Transgénicos , Cultivo Primario de Células , Estructura Terciaria de Proteína , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/fisiopatología , Factor de Crecimiento Transformador beta/metabolismo , Transgenes/fisiología
8.
Dev Biol ; 365(1): 241-50, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22410172

RESUMEN

The Pax2 gene encodes a DNA binding protein with multiple functions in the developing intermediate mesoderm and urogenital tract. Loss of Pax2 in mice results in the complete absence of kidneys, ureters, and sex specific epithelial structures derived from the intermediate mesoderm in both males and females. In this report, we describe two new alleles of Pax2 created by inserting the enhanced green fluorescent protein coding region into the 5' untranslated leader sequence. One allele is a hypomorph that generates less protein and exhibits structural defects in kidneys and ureters upon homozygosity. A second allele is a true null that can be used to image Pax2 expressing cells in a mutant background. Organ culture and embryo analyses point to a loss of epithelial cell polarity and increased mobility in cells that have deleted Pax2 function. These experiments provide new insight into the role of Pax2 protein levels in determining correct renal architecture and cell fate. These new Pax2 alleles are valuable genetic reagents for in vivo studies of urogenital development.


Asunto(s)
Alelos , Regulación del Desarrollo de la Expresión Génica , Riñón/metabolismo , Factor de Transcripción PAX2/genética , Factores de Edad , Envejecimiento/fisiología , Animales , Femenino , Proteínas Fluorescentes Verdes , Riñón/embriología , Masculino , Ratones , Técnicas de Cultivo de Órganos , Factor de Transcripción PAX2/metabolismo
9.
J Biol Chem ; 287(25): 21290-302, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22556418

RESUMEN

Transforming growth factor ß1 (TGF-ß) promotes renal interstitial fibrosis in vivo and the expression of mesenchymal genes in vitro; however, most of its direct targets in epithelial cells are still elusive. In a screen for genes directly activated by TGF-ß, we found that components of the Wnt signaling pathway, especially Wnt11, were targets of activation by TGF-ß and Smad3 in primary renal epithelial cells. In gain and loss of function experiments, Wnt11 mediated the actions of TGF-ß through enhanced activation of mesenchymal marker genes, such as Zeb1, Snail1, Pai1, and αSMA, without affecting Smad3 phosphorylation. Inhibition of Wnt11 by receptor knockdown or treatment with Wnt inhibitors limited the effects of TGF-ß on gene expression. We found no evidence that Wnt11 activated the canonical Wnt signaling pathway in renal epithelial cells; rather, the function of Wnt11 was mediated by the c-Jun N-terminal kinase (JNK) pathway. Consistent with the in vitro results, all the TGF-ß, Wnt11, and JNK targets were activated in a unilateral ureteral obstruction (UUO) model of renal fibrosis in vivo. Our findings demonstrated cooperativity among the TGF-ß, Wnt11, and JNK signaling pathways and suggest new targets for anti-fibrotic therapy in renal tissue.


Asunto(s)
Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Enfermedades Renales/metabolismo , Riñón/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/patología , Fibrosis , Técnicas de Silenciamiento del Gen , Humanos , Riñón/patología , Enfermedades Renales/genética , Enfermedades Renales/patología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Factor de Crecimiento Transformador beta1/genética , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología , Proteínas Wnt/genética
10.
Kidney Int ; 84(5): 920-30, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23677246

RESUMEN

Podocytes are highly specialized epithelial cells with complex actin cytoskeletal architecture crucial for maintenance of the glomerular filtration barrier. The mammalian Rho GTPases Rac1 and Cdc42 are molecular switches that control many cellular processes, but are best known for their roles in the regulation of actin cytoskeleton dynamics. Here, we employed podocyte-specific Cre-lox technology and found that mice with deletion of Rac1 display normal podocyte morphology without glomerular dysfunction well into adulthood. Using the protamine sulfate model of acute podocyte injury, podocyte-specific deletion of Rac1 prevented foot process effacement. In a long-term model of chronic hypertensive glomerular damage, however, loss of Rac1 led to an exacerbation of albuminuria and glomerulosclerosis. In contrast, mice with podocyte-specific deletion of Cdc42 had severe proteinuria, podocyte foot process effacement, and glomerulosclerosis beginning as early as 10 days of age. In addition, slit diaphragm proteins nephrin and podocin were redistributed, and cofilin was dephosphorylated. Cdc42 is necessary for the maintenance of podocyte structure and function, but Rac1 is entirely dispensable in physiological steady state. However, Rac1 has either beneficial or deleterious effects depending on the context of podocyte impairment. Thus, our study highlights the divergent roles of Rac1 and Cdc42 function in podocyte maintenance and injury.


Asunto(s)
Lesión Renal Aguda/enzimología , Neuropéptidos/metabolismo , Podocitos/enzimología , Insuficiencia Renal/enzimología , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Albuminuria/metabolismo , Animales , Forma de la Célula , Acetato de Desoxicorticosterona , Modelos Animales de Enfermedad , Genotipo , Hipertensión/inducido químicamente , Hipertensión/enzimología , Hipertensión/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Nefrectomía , Neuropéptidos/deficiencia , Neuropéptidos/genética , Fenotipo , Fosforilación , Podocitos/patología , Protaminas , Insuficiencia Renal/etiología , Insuficiencia Renal/genética , Insuficiencia Renal/patología , Transducción de Señal , Factores de Tiempo , Proteína de Unión al GTP cdc42/deficiencia , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/deficiencia , Proteína de Unión al GTP rac1/genética
11.
Cell Signal ; 104: 110591, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36627007

RESUMEN

The cellular trafficking protein secretory-carrier-membrane-protein 3 (SCAMP3) has been previously shown to promote hepatocellular carcinoma, melanoma, glioma and pancreatic adenocarcinoma. Moreover, previous work has shown that SCAMP3 regulates the epidermal growth factor receptor (EGFR) in triple negative breast cancer (TNBC). However, the oncogenic role of SCAMP3 in different molecular subtypes of breast cancer (BRCA) remains largely unknown. In this study, the role of SCAMP3 in different molecular subtypes of BRCA was investigated using in silico, in vitro and in vivo approaches. In silico analysis of BRCA patient samples showed that SCAMP3 is highly overexpressed in different BRCA molecular subtypes, advanced disease grades and lymph node metastatic stages. Depletion of SCAMP3 inhibited BRCA cell growth, stemness, clonogenic potential and migration and promoted autophagy and cellular senescence. The expression of stemness markers CD44 and OCT4A was reduced in SCAMP3-silenced MDA-MB-231 cells. SCAMP3 overexpression promoted cell proliferation, clonogenicity, tumor spheroid formation and migration in vitro and tumor growth in vivo. SCAMP3 promoted epithelial-mesenchymal-transition (EMT) by regulating E-cadherin expression. SCAMP3 enhanced in vivo tumor growth in MDA-MB-231 tumor xenograft mouse model. Mechanistically, SCAMP3 depletion inhibited ß-Catenin, c-MYC and SQSTM1 expression, while its overexpression increased the expression of the same oncogenic proteins. Increased SCAMP3 expression associated with increased chemoresistance in BRCA cells while its depletion associated with increased sensitivity to chemotherapy. BRCA patients with high SCAMP3 expression showed poor prognosis, decreased overall survival and relapse free survival relative to counterparts with reduced SCAMP3 expression. These findings suggest that SCAMP3 exerts a wide range of oncogenic effects in different molecular subtypes of BRCA by modulating the c-MYC-ß-Catenin-SQSTM1 axis that targets tumor growth, metastasis, stemness and chemoresistance.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , beta Catenina/metabolismo , Proteínas Portadoras/farmacología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Proteínas de la Membrana/metabolismo , Proteína Sequestosoma-1/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo
12.
Sci Rep ; 13(1): 6361, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-37076596

RESUMEN

Recovery from acute kidney injury can vary widely in patients and in animal models. Immunofluorescence staining can provide spatial information about heterogeneous injury responses, but often only a fraction of stained tissue is analyzed. Deep learning can expand analysis to larger areas and sample numbers by substituting for time-intensive manual or semi-automated quantification techniques. Here we report one approach to leverage deep learning tools to quantify heterogenous responses to kidney injury that can be deployed without specialized equipment or programming expertise. We first demonstrated that deep learning models generated from small training sets accurately identified a range of stains and structures with performance similar to that of trained human observers. We then showed this approach accurately tracks the evolution of folic acid induced kidney injury in mice and highlights spatially clustered tubules that fail to repair. We then demonstrated that this approach captures the variation in recovery across a robust sample of kidneys after ischemic injury. Finally, we showed markers of failed repair after ischemic injury were correlated both spatially within and between animals and that failed repair was inversely correlated with peritubular capillary density. Combined, we demonstrate the utility and versatility of our approach to capture spatially heterogenous responses to kidney injury.


Asunto(s)
Lesión Renal Aguda , Aprendizaje Profundo , Humanos , Ratones , Animales , Riñón/irrigación sanguínea , Modelos Animales , Ácido Fólico
13.
bioRxiv ; 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37873377

RESUMEN

Acute kidney injury (AKI) is a common condition that lacks effective treatments. In part this shortcoming is due to an incomplete understanding of the genetic mechanisms that control pathogenesis and recovery. Pax2 and Pax8 are homologous transcription factors with overlapping functions that are critical for kidney development and are re-activated in AKI. In this report, we examined the role of Pax2 and Pax8 in recovery from ischemic AKI. We found that Pax2 and Pax8 are upregulated after severe AKI and correlate with chronic injury. Surprisingly, we then discovered that proximal-tubule-selective deletion of Pax2 and Pax8 resulted in a less severe chronic injury phenotype. This effect was mediated by protection against the acute insult, similar to preconditioning. Prior to injury, Pax2 and Pax8 mutant mice develop a unique subpopulation of S3 proximal tubule cells that display features usually seen only in acute or chronic injury. The expression signature of these cells was strongly enriched with genes associated with other mechanisms of protection against ischemic AKI including caloric restriction, hypoxic preconditioning, and female sex. Taken together, our results identify a novel role for Pax2 and Pax8 in mature proximal tubules that regulates critical genes and pathways involved in both injury response and protection from ischemic AKI. TRANSLATIONAL STATEMENT: Identifying the molecular and genetic regulators unique to the nephron that dictate vulnerability to injury and regenerative potential could lead to new therapeutic targets to treat ischemic kidney injury. Pax2 and Pax8 are two homologous nephron-specific transcription factors that are critical for kidney development and physiology. Here we report that proximal-tubule-selective depletion of Pax2 and Pax8 protects against both acute and chronic injury and induces an expression profile in the S3 proximal tubule with common features shared among diverse conditions that protect against ischemia. These findings highlight a new role for Pax proteins as potential therapeutic targets to treat AKI.

14.
J Biol Chem ; 285(29): 22676-88, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20472933

RESUMEN

Actin dynamics determines podocyte morphology during development and in response to podocyte injury and might be necessary for maintaining normal podocyte morphology. Because podocyte intercellular junction receptor Nephrin plays a role in regulating actin dynamics, and given the described role of cofilin in actin filament polymerization and severing, we hypothesized that cofilin-1 activity is regulated by Nephrin and is necessary in normal podocyte actin dynamics. Nephrin activation induced cofilin dephosphorylation via intermediaries that include phosphatidylinositol 3-kinase, SSH1, 14-3-3, and LIMK in a cell culture model. This Nephrin-induced cofilin activation required a direct interaction between Nephrin and the p85 subunit of phosphatidylinositol 3-kinase. In a similar fashion, cofilin-1 dephosphorylation was observed in a rat model of podocyte injury at a time when foot process spreading is initially observed. To investigate the necessity of cofilin-1 in the glomerulus, podocyte-specific Cfl1 null mice were generated. Cfl1 null podocytes developed normally. However, these mice developed persistent proteinuria by 3 months of age, although they did not exhibit foot process spreading until 8 months, when the rate of urinary protein excretion became more exaggerated. In a mouse model of podocyte injury, protamine sulfate perfusion of the Cfl1 mutant mouse induced a broadened and flattened foot process morphology that was distinct from that observed following perfusion of control kidneys, and mutant podocytes did not recover normal structure following additional perfusion with heparin sulfate. We conclude that cofilin-1 is necessary for maintenance of normal podocyte architecture and for actin structural changes that occur during induction and recovery from podocyte injury.


Asunto(s)
Cofilina 1/metabolismo , Podocitos/metabolismo , Actinas/metabolismo , Albuminuria/metabolismo , Animales , Línea Celular , Femenino , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Quinasas Lim/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Especificidad de Órganos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Podocitos/patología , Podocitos/ultraestructura , Protaminas , Unión Proteica , Seudópodos/metabolismo , Seudópodos/ultraestructura , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley
15.
JCI Insight ; 5(3)2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-31917689

RESUMEN

A terminally differentiated cellular phenotype is thought to be maintained, at least in part, by both active and repressive histone marks. However, it is unclear whether regenerating cells after injury need to replicate such epigenetic marks to recover. To test whether renal epithelial cell regeneration is dependent on histone H3K4 methylation, we generated a mouse model that deleted the Paxip1 gene in mature renal proximal tubules. Paxip1 encodes PTIP, an essential protein in the Mll3/4 histone H3K4 methyltransferase complex. Mice with PTIP deletions in the adult kidney proximal tubules were viable and fertile. Upon acute kidney injury, such mice failed to regenerate damaged tubules, leading to scarring and interstitial fibrosis. The inability to repair damage was likely due to a failure to reenter mitosis and reactivate regulatory genes such as Sox9. PTIP deletion reduced histone H3K4 methylation in uninjured adult kidneys but did not significantly affect function or the expression of epithelial specific markers. Strikingly, cell lineage tracing revealed that surviving PTIP mutant cells could alter their phenotype and lose epithelial markers. These data demonstrate that PTIP and associated MLL3/4-mediated histone methylation are needed for regenerating proximal tubules and to maintain or reestablish the cellular epithelial phenotype.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Proteínas de Unión al ADN/fisiología , Epigénesis Genética/fisiología , Regeneración Hepática , Lesión Renal Aguda/patología , Animales , Biomarcadores/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/genética , Eliminación de Gen , Histonas/metabolismo , Metilación , Ratones
16.
J Clin Invest ; 116(5): 1346-59, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16543952

RESUMEN

A properly established and maintained podocyte intercellular junction, or slit diaphragm, is a necessary component of the selective permeability barrier of the kidney glomerulus. The observation that mutation or deletion of the slit diaphragm transmembrane protein nephrin results in failure of podocyte foot process morphogenesis and concomitant proteinuria first suggested the hypothesis that nephrin serves as a component of a signaling complex that directly integrates podocyte junctional integrity with cytoskeletal dynamics. The observations made herein provide the first direct evidence to our knowledge for a phosphorylation-mediated signaling mechanism by which this integrative function is derived. Our data support the model that during podocyte intercellular junction formation, engagement of the nephrin ectodomain induces transient Fyn catalytic activity that results in nephrin phosphorylation on specific nephrin cytoplasmic domain tyrosine residues. We found that this nephrin phosphorylation event resulted in recruitment of the SH2-SH3 domain-containing adapter protein Nck and assembly of actin filaments in an Nck-dependent fashion. Considered in the context of the role of nephrin family proteins in other organisms and the integral relationship of actin dynamics and junction formation, these observations establish a function for nephrin in regulating actin cytoskeletal dynamics.


Asunto(s)
Actinas/metabolismo , Proteínas de la Membrana/fisiología , Proteínas Oncogénicas/metabolismo , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células COS , Chlorocebus aethiops , Humanos , Glomérulos Renales/metabolismo , Proteínas de la Membrana/química , Ratones , Fosforilación , Podocitos/metabolismo , Estructura Terciaria de Proteína
17.
Stem Cell Reports ; 11(4): 912-928, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30220628

RESUMEN

The in vivo niche and basic cellular properties of nephron progenitors are poorly described. Here we studied the cellular organization and function of the MAPK/ERK pathway in nephron progenitors. Live-imaging of ERK activity by a Förster resonance energy transfer biosensor revealed a dynamic activation pattern in progenitors, whereas differentiating precursors exhibited sustained activity. Genetic experiments demonstrate that MAPK/ERK activity controls the thickness, coherence, and integrity of the nephron progenitor niche. Molecularly, MAPK/ERK activity regulates niche organization and communication with extracellular matrix through PAX2 and ITGA8, and is needed for CITED1 expression denoting undifferentiated status. MAPK/ERK activation in nephron precursors propels differentiation by priming cells for distal and proximal fates induced by the Wnt and Notch pathways. Thus, our results demonstrate a mechanism through which MAPK/ERK activity controls both progenitor maintenance and differentiation by regulating a distinct set of targets, which maintain the biomechanical milieu of tissue-residing progenitors and prime precursors for nephrogenesis.


Asunto(s)
Diferenciación Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Nefronas/citología , Nicho de Células Madre , Células Madre/citología , Animales , Apoptosis , Técnicas Biosensibles , Tipificación del Cuerpo , Proliferación Celular , Autorrenovación de las Células , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Cadenas alfa de Integrinas/metabolismo , Ratones , Organogénesis , Factor de Transcripción PAX2/metabolismo , Células Madre/metabolismo
18.
Mol Cell Biol ; 34(1): 43-56, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24164893

RESUMEN

First identified in Drosophila, the Crumbs (Crb) proteins are important in epithelial polarity, apical membrane formation, and tight junction (TJ) assembly. The conserved Crb intracellular region includes a FERM (band 4.1/ezrin/radixin/moesin) binding domain (FBD) whose mammalian binding partners are not well understood and a PDZ binding motif that interacts with mammalian Pals1 (protein associated with lin seven) (also known as MPP5). Pals1 binds Patj (Pals1-associated tight-junction protein), a multi-PDZ-domain protein that associates with many tight junction proteins. The Crb complex also binds the conserved Par3/Par6/atypical protein kinase C (aPKC) polarity cassette that restricts migration of basolateral proteins through phosphorylation. Here, we describe a Crb3 knockout mouse that demonstrates extensive defects in epithelial morphogenesis. The mice die shortly after birth, with cystic kidneys and proteinaceous debris throughout the lungs. The intestines display villus fusion, apical membrane blebs, and disrupted microvilli. These intestinal defects phenocopy those of Ezrin knockout mice, and we demonstrate an interaction between Crumbs3 and ezrin. Taken together, our data indicate that Crumbs3 is crucial for epithelial morphogenesis and plays a role in linking the apical membrane to the underlying ezrin-containing cytoskeleton.


Asunto(s)
Epitelio/metabolismo , Riñón/metabolismo , Pulmón/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Western Blotting , Línea Celular , Polaridad Celular , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Epitelio/embriología , Epitelio/ultraestructura , Femenino , Riñón/embriología , Pulmón/embriología , Masculino , Glicoproteínas de Membrana , Proteínas de la Membrana/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Análisis de Supervivencia , Uniones Estrechas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA