Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Kidney Int ; 105(6): 1263-1278, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38286178

RESUMEN

Current classification of chronic kidney disease (CKD) into stages using indirect systemic measures (estimated glomerular filtration rate (eGFR) and albuminuria) is agnostic to the heterogeneity of underlying molecular processes in the kidney thereby limiting precision medicine approaches. To generate a novel CKD categorization that directly reflects within kidney disease drivers we analyzed publicly available transcriptomic data from kidney biopsy tissue. A Self-Organizing Maps unsupervised artificial neural network machine-learning algorithm was used to stratify a total of 369 patients with CKD and 46 living kidney donors as healthy controls. Unbiased stratification of the discovery cohort resulted in identification of four novel molecular categories of disease termed CKD-Blue, CKD-Gold, CKD-Olive, CKD-Plum that were replicated in independent CKD and diabetic kidney disease datasets and can be further tested on any external data at kidneyclass.org. Each molecular category spanned across CKD stages and histopathological diagnoses and represented transcriptional activation of distinct biological pathways. Disease progression rates were highly significantly different between the molecular categories. CKD-Gold displayed rapid progression, with significant eGFR-adjusted Cox regression hazard ratio of 5.6 [1.01-31.3] for kidney failure and hazard ratio of 4.7 [1.3-16.5] for composite of kidney failure or a 40% or more eGFR decline. Urine proteomics revealed distinct patterns between the molecular categories, and a 25-protein signature was identified to distinguish CKD-Gold from other molecular categories. Thus, patient stratification based on kidney tissue omics offers a gateway to non-invasive biomarker-driven categorization and the potential for future clinical implementation, as a key step towards precision medicine in CKD.


Asunto(s)
Progresión de la Enfermedad , Tasa de Filtración Glomerular , Riñón , Medicina de Precisión , Insuficiencia Renal Crónica , Transcriptoma , Humanos , Medicina de Precisión/métodos , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/orina , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/fisiopatología , Persona de Mediana Edad , Femenino , Masculino , Riñón/patología , Riñón/fisiopatología , Anciano , Biopsia , Adulto , Redes Neurales de la Computación , Estudios de Casos y Controles , Perfilación de la Expresión Génica , Aprendizaje Automático no Supervisado
2.
Int J Mol Sci ; 22(1)2020 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-33383945

RESUMEN

Cyclophilins have important homeostatic roles, but following tissue injury, cyclophilin A (CypA) can promote leukocyte recruitment and inflammation, while CypD can facilitate mitochondrial-dependent cell death. This study investigated the therapeutic potential of a selective cyclophilin inhibitor (GS-642362), which does not block calcineurin function, in mouse models of tubular cell necrosis and renal fibrosis. Mice underwent bilateral renal ischemia/reperfusion injury (IRI) and were killed 24 h later: treatment with 10 or 30 mg/kg/BID GS-642362 (or vehicle) began 1 h before surgery. In the second model, mice underwent unilateral ureteric obstruction (UUO) surgery and were killed 7 days later; treatment with 10 or 30 mg/kg/BID GS-642362 (or vehicle) began 1 h before surgery. GS-642362 treatment gave a profound and dose-dependent protection from acute renal failure in the IRI model. This protection was associated with reduced tubular cell death, including a dramatic reduction in neutrophil infiltration. In the UUO model, GS-642362 treatment significantly reduced tubular cell death, macrophage infiltration, and renal fibrosis. This protective effect was independent of the upregulation of IL-2 and activation of the stress-activated protein kinases (p38 and JNK). In conclusion, GS-642362 was effective in suppressing both acute kidney injury and renal fibrosis. These findings support further investigation of cyclophilin blockade in other types of acute and chronic kidney disease.


Asunto(s)
Lesión Renal Aguda/etiología , Lesión Renal Aguda/prevención & control , Ciclofilinas/farmacología , Necrosis de la Corteza Renal/etiología , Necrosis de la Corteza Renal/prevención & control , Sustancias Protectoras/farmacología , Lesión Renal Aguda/patología , Animales , Muerte Celular , Modelos Animales de Enfermedad , Fibrosis , Necrosis de la Corteza Renal/patología , Túbulos Renales/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Infiltración Neutrófila , Neutrófilos/metabolismo , Neutrófilos/patología , Oxígeno/metabolismo , Daño por Reperfusión/etiología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
3.
Kidney Int ; 92(5): 1282-1287, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28754553

RESUMEN

While increased mitochondrial reactive oxygen species have been commonly implicated in a variety of disease states, their in vivo role in the pathogenesis of diabetic nephropathy remains controversial. Using a two-photon imaging approach with a genetically encoded redox biosensor, we monitored mitochondrial redox state in the kidneys of experimental models of diabetes in real-time in vivo. Diabetic (db/db) mice that express a redox-sensitive Green Fluorescent Protein biosensor (roGFP) specifically in the mitochondrial matrix (db/dbmt-roGFP) were generated, allowing dynamic monitoring of redox changes in the kidneys. These db/dbmt-roGFP mice exhibited a marked increase in mitochondrial reactive oxygen species in the kidneys. Yeast NADH-dehydrogenase, a mammalian Complex I homolog, was ectopically expressed in cultured podocytes, and this forced expression in roGFP-expressing podocytes prevented high glucose-induced increases in mitochondrial reactive oxygen species. Thus, in vivo monitoring of mitochondrial roGFP in diabetic mice confirms increased production of mitochondrial reactive oxygen species in the kidneys.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Nefropatías Diabéticas/patología , Riñón/patología , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo , Animales , Técnicas Biosensibles , Células Cultivadas , Diabetes Mellitus Tipo 2/complicaciones , Nefropatías Diabéticas/etiología , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Endogámicos , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Oxidación-Reducción , Podocitos
4.
J Am Soc Nephrol ; 27(9): 2733-47, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26825530

RESUMEN

Mitochondrial fission has been linked to the pathogenesis of diabetic nephropathy (DN). However, how mitochondrial fission affects progression of DN in vivo is unknown. Here, we report the effect of conditional podocyte-specific deletion of dynamin-related protein 1 (Drp1), an essential component of mitochondrial fission, on the pathogenesis and progression of DN. Inducible podocyte-specific deletion of Drp1 in diabetic mice decreased albuminuria and improved mesangial matrix expansion and podocyte morphology. Ultrastructure analysis revealed a significant increase in fragmented mitochondria in the podocytes of wild-type diabetic mice but a marked improvement in mitochondrial structure in Drp1-null podocytes of diabetic mice. When isolated from diabetic mice and cultured in high glucose, Drp1-null podocytes had more elongated mitochondria and better mitochondrial fitness associated with enhanced oxygen consumption and ATP production than wild-type podocytes. Furthermore, administration of a pharmacologic inhibitor of Drp1, Mdivi1, significantly blunted mitochondrial fission and rescued key pathologic features of DN in mice. Taken together, these results provide novel correlations between mitochondrial morphology and the progression of DN and point to Drp1 as a potential therapeutic target in DN.


Asunto(s)
Nefropatías Diabéticas/etiología , Dinaminas/deficiencia , Dinámicas Mitocondriales , Animales , Nefropatías Diabéticas/prevención & control , Progresión de la Enfermedad , Dinaminas/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Podocitos
5.
Pediatr Nephrol ; 30(5): 727-40, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-24928414

RESUMEN

MicroRNAs (miRNAs) are short, non-coding RNAs that employ classic Watson-Crick base-pairing to identify their target genes, ultimately resulting in destabilization of their target mRNAs and/or inhibition of their translation. The role of miRNAs in a wide range of human diseases, including those afflicting the kidney, has been intensely investigated. However, there is still a vast dearth of knowledge regarding their specific mode of action and therapeutic effects in various kidney diseases. This review discusses the latest efforts to further our understanding of the basic biology of miRNAs, their impact on various kidney diseases and their potential as novel biomarkers and therapeutic agents. We initially provide an overview of miRNA biology and the canonical pathway implicated in their biogenesis. We then discuss commonly employed experimental strategies for miRNA research and highlight some of the newly described state-of-the-art technologies to identify miRNAs and their target genes. Finally, we carefully examine the emerging role of miRNAs in the pathogenesis of various kidney diseases.


Asunto(s)
Enfermedades Renales/genética , MicroARNs/genética , Humanos
6.
J Biol Chem ; 288(51): 36202-14, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-24163368

RESUMEN

Accumulating evidence suggests that microRNAs (miRNAs) contribute to a myriad of kidney diseases. However, the regulatory role of miRNAs on the key molecules implicated in kidney fibrosis remains poorly understood. Bone morphogenetic protein-7 (BMP-7) and its related BMP-6 have recently emerged as key regulators of kidney fibrosis. Using the established unilateral ureteral obstruction (UUO) model of kidney fibrosis as our experimental model, we examined the regulatory role of miRNAs on BMP-7/6 signaling. By analyzing the potential miRNAs that target BMP-7/6 in silica, we identified miR-22 as a potent miRNA targeting BMP-7/6. We found that expression levels of BMP-7/6 were significantly elevated in the kidneys of the miR-22 null mouse. Importantly, mice with targeted deletion of miR-22 exhibited attenuated renal fibrosis in the UUO model. Consistent with these in vivo observations, primary renal fibroblast isolated from miR-22-deficient UUO mice demonstrated a significant increase in BMP-7/6 expression and their downstream targets. This phenotype could be rescued when cells were transfected with miR-22 mimics. Interestingly, we found that miR-22 and BMP-7/6 are in a regulatory feedback circuit, whereby not only miR-22 inhibits BMP-7/6, but miR-22 by itself is induced by BMP-7/6. Finally, we identified two BMP-responsive elements in the proximal region of miR-22 promoter. These findings identify miR-22 as a critical miRNA that contributes to renal fibrosis on the basis of its pivotal role on BMP signaling cascade.


Asunto(s)
Proteína Morfogenética Ósea 6/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Riñón/metabolismo , MicroARNs/metabolismo , Animales , Secuencia de Bases , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 7/genética , Fibrosis/metabolismo , Homeostasis , Riñón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , MicroARNs/genética , Datos de Secuencia Molecular , Elementos de Respuesta , Transducción de Señal , Transcripción Genética
7.
Am J Kidney Dis ; 63(2 Suppl 2): S63-83, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24461730

RESUMEN

Diabetic kidney disease remains a major microvascular complication of diabetes and the most common cause of chronic kidney failure requiring dialysis in the United States. Medical advances over the past century have substantially improved the management of diabetes mellitus and thereby have increased patient survival. However, current standards of care reduce but do not eliminate the risk of diabetic kidney disease, and further studies are warranted to define new strategies for reducing the risk of diabetic kidney disease. In this review, we highlight some of the novel and established molecular mechanisms that contribute to the development of the disease and its outcomes. In particular, we discuss recent advances in our understanding of the molecular mechanisms implicated in the pathogenesis and progression of diabetic kidney disease, with special emphasis on the mitochondrial oxidative stress and microRNA targets. Additionally, candidate genes associated with susceptibility to diabetic kidney disease and alterations in various cytokines, chemokines, and growth factors are addressed briefly.


Asunto(s)
Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Progresión de la Enfermedad , Epigenómica , Humanos , MicroARNs/metabolismo , Mitocondrias/genética , Estrés Oxidativo/fisiología
8.
Kidney Int ; 82(11): 1151-4, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23151985

RESUMEN

Endothelial dysfunction underlies the basic pathophysiology of microvascular complications of diabetes. Endothelial dysfunction is associated with impaired nitric oxide (NO) availability. Since NO production is tightly regulated by endothelial nitric oxide synthase (eNOS), several therapeutic strategies have been investigated and proposed to improve eNOS bioavailability in the vasculature. The findings of Cheng et al. suggest that increased availability of eNOS may be an effective strategy in restoring endothelial function in patients with diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Nefropatías Diabéticas/enzimología , Endotelio Vascular/enzimología , Glomérulos Renales/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales
9.
Kidney360 ; 3(7): 1169-1182, 2022 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-35919527

RESUMEN

Background: Despite widespread use of renin-aldosterone-angiotensin system inhibitors and the benefits of lowering glomerular pressure in patients with CKD, there remains a major unmet need for therapies targeting underlying causes of CKD progression. Apoptosis signal-regulating kinase 1 (ASK1) promotes apoptosis and glomerulosclerosis, and is implicated in the progression of diabetic kidney disease (DKD), a major cause of CKD. Selonsertib is a selective ASK1 inhibitor currently in clinical development for the treatment of DKD. We examined the added benefits of selonsertib on existing glomerulosclerosis and related molecular pathways in the nondiabetic 5/6 nephrectomy (5/6 Nx) rat model in combination with the angiotensin-converting enzyme inhibitor (ACEI) enalapril. Methods: Male Sprague Dawley rats underwent 5/6 Nx with kidney biopsy 8 weeks later for assessment of glomerulosclerosis, and were randomized to four treatment groups with equal glomerulosclerosis: selonsertib, enalapril, combination (selonsertib plus enalapril), and untreated controls. Serum creatinine, systolic BP (SBP), and urinary albumin were measured at intervals. Animals were euthanized at week 12 for histologic, biochemical, and molecular analyses. Results: All rats developed hypertension, albuminuria, and glomerulosclerosis by week 8. Kidney function further declined, and glomerulosclerosis and albuminuria progressively increased in controls from week 8 to 12. Enalapril treatment alone from week 8 to 12 reduced SBP versus controls, decreased albuminuria, and resulted in numerically lower glomerulosclerosis. Selonsertib alone had no effect on SBP but preserved kidney function. Combined treatment significantly reduced glomerulosclerosis, with more regression than either monotherapy. Enalapril treatment resulted in fewer interstitial macrophages, whereas selonsertib treatment reduced apoptosis and podocyte loss. RNA-seq revealed that combined treatment influenced pathways related to extracellular matrix and wound healing. Conclusions: Selonsertib targets a novel, nonhemodynamic pathway in CKD. Our data suggest that ASK1 inhibition, when combined with ACEI, has additive effects to reduce progression of glomerulosclerosis, attenuate kidney function decline, and reduce podocyte loss.


Asunto(s)
Nefropatías Diabéticas , Hipertensión , Insuficiencia Renal Crónica , Animales , Masculino , Ratas , Albuminuria/tratamiento farmacológico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Antihipertensivos/farmacología , Benzamidas , Nefropatías Diabéticas/patología , Enalapril/farmacología , Hipertensión/patología , Imidazoles , Riñón , Piridinas , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/complicaciones , Nivel de Atención
10.
Oncogene ; 38(34): 6211-6225, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31289360

RESUMEN

One-carbon metabolism plays a central role in a broad array of metabolic processes required for the survival and growth of tumor cells. However, the molecular basis of how one-carbon metabolism may influence RNA methylation and tumorigenesis remains largely unknown. Here we show MTHFD2, a mitochondrial enzyme involved in one-carbon metabolism, contributes to the progression of renal cell carcinoma (RCC) via a novel epitranscriptomic mechanism that involves HIF-2α. We found that expression of MTHFD2 was significantly elevated in human RCC tissues, and MTHFD2 knockdown strongly reduced xenograft tumor growth. Mechanistically, using an unbiased methylated RNA immunoprecipitation sequencing (meRIP-Seq) approach, we found that MTHFD2 plays a critical role in controlling global N6-methyladenosine (m6A) methylation levels, including the m6A methylation of HIF-2α mRNA, which results in enhanced translation of HIF-2α. Enhanced HIF-2α translation, in turn, promotes the aerobic glycolysis, linking one-carbon metabolism to HIF-2α-dependent metabolic reprogramming through RNA methylation. Our findings also suggest that MTHFD2 and HIF-2α form a positive feedforward loop in RCC, promoting metabolic reprograming and tumor growth. Taken together, our results suggest that MTHFD2 links RNA methylation status to the metabolic state of tumor cells in RCC.


Asunto(s)
Aminohidrolasas/fisiología , Carcinoma de Células Renales/metabolismo , Glucólisis/genética , Neoplasias Renales/metabolismo , Metilenotetrahidrofolato Deshidrogenasa (NADP)/fisiología , Metiltransferasas/metabolismo , Enzimas Multifuncionales/fisiología , Procesamiento Postranscripcional del ARN/genética , Animales , Metabolismo de los Hidratos de Carbono/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Reprogramación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Masculino , Metilación , Ratones , Ratones Desnudos
12.
J Clin Invest ; 128(10): 4485-4500, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30024858

RESUMEN

Oxidative stress is an underlying component of acute and chronic kidney disease. Apoptosis signal-regulating kinase 1 (ASK1) is a widely expressed redox-sensitive serine threonine kinase that activates p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinase kinases, and induces apoptotic, inflammatory, and fibrotic signaling in settings of oxidative stress. We describe the discovery and characterization of a potent and selective small-molecule inhibitor of ASK1, GS-444217, and demonstrate the therapeutic potential of ASK1 inhibition to reduce kidney injury and fibrosis. Activation of the ASK1 pathway in glomerular and tubular compartments was confirmed in renal biopsies from patients with diabetic kidney disease (DKD) and was decreased by GS-444217 in several rodent models of kidney injury and fibrosis that collectively represented the hallmarks of DKD pathology. Treatment with GS-444217 reduced progressive inflammation and fibrosis in the kidney and halted glomerular filtration rate decline. Combination of GS-444217 with enalapril, an angiotensin-converting enzyme inhibitor, led to a greater reduction in proteinuria and regression of glomerulosclerosis. These results identify ASK1 as an important target for renal disease and support the clinical development of an ASK1 inhibitor for the treatment of DKD.


Asunto(s)
Nefropatías Diabéticas/enzimología , Fibroblastos/enzimología , Glomérulos Renales/enzimología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas , Animales , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Femenino , Fibroblastos/patología , Fibrosis , Humanos , Glomérulos Renales/patología , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 5/genética , Masculino , Ratones , Ratones Noqueados , Inhibidores de Proteínas Quinasas/farmacología , Distribución Aleatoria , Ratas Sprague-Dawley
13.
J Clin Invest ; 126(11): 4205-4218, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27760051

RESUMEN

The regulatory roles of long noncoding RNAs (lncRNAs) in transcriptional coactivators are still largely unknown. Here, we have shown that the peroxisome proliferator-activated receptor γ (PPARγ) coactivator α (PGC-1α, encoded by Ppargc1a) is functionally regulated by the lncRNA taurine-upregulated gene 1 (Tug1). Further, we have described a role for Tug1 in the regulation of mitochondrial function in podocytes. Using a murine model of diabetic nephropathy (DN), we performed an unbiased RNA-sequencing (RNA-seq) analysis of kidney glomeruli and identified Tug1 as a differentially expressed lncRNA in the diabetic milieu. Podocyte-specific overexpression (OE) of Tug1 in diabetic mice improved the biochemical and histological features associated with DN. Unexpectedly, we found that Tug1 OE rescued the expression of PGC-1α and its transcriptional targets. Tug1 OE was also associated with improvements in mitochondrial bioenergetics in the podocytes of diabetic mice. Mechanistically, we found that the interaction between Tug1 and PGC-1α promotes the binding of PGC-1α to its own promoter. We identified a Tug1-binding element (TBE) upstream of the Ppargc1a gene and showed that Tug1 binds with the TBE to enhance Ppargc1a promoter activity. These findings indicate that a direct interaction between PGC-1α and Tug1 modulates mitochondrial bioenergetics in podocytes in the diabetic milieu.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Metabolismo Energético , Regulación de la Expresión Génica , Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/biosíntesis , Podocitos/metabolismo , ARN Largo no Codificante/metabolismo , Animales , Línea Celular Transformada , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Podocitos/patología , ARN Largo no Codificante/genética
14.
Nat Commun ; 7: 12076, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27350436

RESUMEN

How the kidney responds to the metabolic cues from the environment remains a central question in kidney research. This question is particularly relevant to the pathogenesis of diabetic nephropathy (DN) in which evidence suggests that metabolic events in podocytes regulate chromatin structure. Here, we show that miR-93 is a critical metabolic/epigenetic switch in the diabetic milieu linking the metabolic state to chromatin remodelling. Mice with inducible overexpression of a miR-93 transgene exclusively in podocytes exhibit significant improvements in key features of DN. We identify miR-93 as a regulator of nucleosomal dynamics in podocytes. miR-93 has a critical role in chromatin reorganization and progression of DN by modulating its target Msk2, a histone kinase, and its substrate H3S10. These findings implicate a central role for miR-93 in high glucose-induced chromatin remodelling in the kidney, and provide evidence for a previously unrecognized role for Msk2 as a target for DN therapy.


Asunto(s)
Ensamble y Desensamble de Cromatina , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/etiología , MicroARNs/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Anciano , Animales , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Femenino , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Podocitos/metabolismo , Podocitos/ultraestructura
16.
Methods Enzymol ; 484: 531-48, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21036249

RESUMEN

The c-Jun N-terminal kinases (JNK) are important regulators of cell growth, proliferation, and apoptosis. JNKs are typically activated by a sequence of events that include phosphorylation of its T-P-Y motif by an upstream kinase, followed by homodimerization and translocation to the nucleus. Constitutive activation of JNK has been found in a variety of cancers including non-small cell lung carcinomas, gliomas, and mantle cell lymphoma. In vitro studies show that constitutive activation of JNK induces a transformed phenotype in fibroblasts and enhances tumorigenicity in a variety of cell lines. Interestingly, a subset of JNK isoforms was recently found to autoactivate rendering the proteins constitutively active. These constitutively active JNK proteins were found to play a pivotal role in activating transcription factors that increase cellular growth and tumor formation in mice. In this chapter, we describe techniques and methods that have been successfully used to study the three components of JNK activation. Use of these techniques may lead to a better understanding of the components of JNK pathways and how JNK is activated in cancer cells.


Asunto(s)
Pruebas de Enzimas/métodos , Isoenzimas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Animales , Línea Celular , Humanos , Isoenzimas/genética , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA