Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 31(20): 3458-3477, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-35652455

RESUMEN

Metabolic alterations shared between the nervous system and skin fibroblasts have emerged in amyotrophic lateral sclerosis (ALS). Recently, we found that a subgroup of sporadic ALS (sALS) fibroblasts (sALS1) is characterized by metabolic profiles distinct from other sALS cases (sALS2) and controls, suggesting that metabolic therapies could be effective in sALS. The metabolic modulators nicotinamide riboside and pterostilbene (EH301) are under clinical development for the treatment of ALS. Here, we studied the transcriptome and metabolome of sALS cells to understand the molecular bases of sALS metabotypes and the impact of EH301. Metabolomics and transcriptomics were investigated at baseline and after EH301 treatment. Moreover, weighted gene coexpression network analysis (WGCNA) was used to investigate the association of the metabolic and clinical features. We found that the sALS1 transcriptome is distinct from sALS2 and that EH301 modifies gene expression differently in sALS1, sALS2 and the controls. Furthermore, EH301 had strong protective effects against metabolic stress, an effect linked to the antiinflammatory and antioxidant pathways. WGCNA revealed that the ALS functional rating scale and metabotypes are associated with gene modules enriched for the cell cycle, immunity, autophagy and metabolic genes, which are modified by EH301. The meta-analysis of publicly available transcriptomic data from induced motor neurons by Answer ALS confirmed the functional associations of genes correlated with disease traits. A subset of genes differentially expressed in sALS fibroblasts was used in a machine learning model to predict disease progression. In conclusion, multiomic analyses highlighted the differential metabolic and transcriptomic profiles in patient-derived fibroblast sALS, which translate into differential responses to the investigational drug EH301.


Asunto(s)
Esclerosis Amiotrófica Lateral , Esclerosis Amiotrófica Lateral/metabolismo , Antioxidantes/metabolismo , Drogas en Investigación/metabolismo , Drogas en Investigación/uso terapéutico , Fibroblastos/metabolismo , Humanos , Transcriptoma/genética
2.
Blood ; 137(6): 788-800, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32785655

RESUMEN

MALT1 inhibitors are promising therapeutic agents for B-cell lymphomas that are dependent on constitutive or aberrant signaling pathways. However, a potential limitation for signal transduction-targeted therapies is the occurrence of feedback mechanisms that enable escape from the full impact of such drugs. Here, we used a functional genomics screen in activated B-cell-like (ABC) diffuse large B-cell lymphoma (DLBCL) cells treated with a small molecule irreversible inhibitor of MALT1 to identify genes that might confer resistance or enhance the activity of MALT1 inhibition (MALT1i). We find that loss of B-cell receptor (BCR)- and phosphatidylinositol 3-kinase (PI3K)-activating proteins enhanced sensitivity, whereas loss of negative regulators of these pathways (eg, TRAF2, TNFAIP3) promoted resistance. These findings were validated by knockdown of individual genes and a combinatorial drug screen focused on BCR and PI3K pathway-targeting drugs. Among these, the most potent combinatorial effect was observed with PI3Kδ inhibitors against ABC-DLBCLs in vitro and in vivo, but that led to an adaptive increase in phosphorylated S6 and eventual disease progression. Along these lines, MALT1i promoted increased MTORC1 activity and phosphorylation of S6K1-T389 and S6-S235/6, an effect that was only partially blocked by PI3Kδ inhibition in vitro and in vivo. In contrast, simultaneous inhibition of MALT1 and MTORC1 prevented S6 phosphorylation, yielded potent activity against DLBCL cell lines and primary patient specimens, and resulted in more profound tumor regression and significantly improved survival of ABC-DLBCLs in vivo compared with PI3K inhibitors. These findings provide a basis for maximal therapeutic impact of MALT1 inhibitors in the clinic, by disrupting feedback mechanisms that might otherwise limit their efficacy.


Asunto(s)
Antineoplásicos/uso terapéutico , Retroalimentación Fisiológica/efectos de los fármacos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos B/inmunología , Receptores Toll-Like/inmunología , Animales , Antineoplásicos/farmacología , Diseño de Fármacos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Femenino , Humanos , Linfoma de Células B Grandes Difuso/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos NOD , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/fisiología , Proteínas de Neoplasias/fisiología , Organoides/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , ARN Interferente Pequeño/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
EMBO Rep ; 21(4): e48978, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32090465

RESUMEN

Defects in the proteasome can result in pathological proteinopathies. However, the pathogenic role of sex- and tissue-specific sensitivity to proteotoxic stress remains elusive. Here, we map the proteasome activity across nine tissues, in male and female mice, and demonstrate strong sexual dimorphism in proteasome activity, where females have significantly higher activity in several tissues. Further, we report drastic differences in proteasome activity among tissues, independently of proteasome concentration, which are exacerbated under stress conditions. Sexual dimorphism in proteasome activity is confirmed in a SOD1 ALS mouse model, in which the spinal cord, a tissue with comparatively low proteasome activity, is severely affected. Our results offer mechanistic insight into tissue-specific sensitivities to proteostasis stress and into sex differences in the progression of neurodegenerative proteinopathies.


Asunto(s)
Esclerosis Amiotrófica Lateral , Caracteres Sexuales , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Transgénicos , Complejo de la Endopetidasa Proteasomal/genética , Agregado de Proteínas , Superóxido Dismutasa/genética , Superóxido Dismutasa-1/genética
4.
Cell Commun Signal ; 18(1): 105, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641054

RESUMEN

BACKGROUND: In the setting of diabetes mellitus, mitochondrial dysfunction and oxidative stress are important pathogenic mechanisms causing end organ damage, including diabetic kidney disease (DKD), but mechanistic understanding at a cellular level remains obscure. In mouse models of DKD, glomerular endothelial cell (GEC) dysfunction precedes albuminuria and contributes to neighboring podocyte dysfunction, implicating GECs in breakdown of the glomerular filtration barrier. In the following studies we wished to explore the cellular mechanisms by which GECs become dysfunctional in the diabetic milieu, and the impact to neighboring podocytes. METHODS: Mouse GECs were exposed to high glucose media (HG) or 2.5% v/v serum from diabetic mice or serum from non-diabetic controls, and evaluated for mitochondrial function (oxygen consumption), structure (electron microscopy), morphology (mitotracker), mitochondrial superoxide (mitoSOX), as well as accumulation of oxidized products (DNA lesion frequency (8-oxoG, endo-G), double strand breaks (γ-H2AX), endothelial function (NOS activity), autophagy (LC3) and apoptotic cell death (Annexin/PI; caspase 3). Supernatant transfer experiments from GECs to podocytes were performed to establish the effects on podocyte survival and transwell experiments were performed to determine the effects in co-culture. RESULTS: Diabetic serum specifically causes mitochondrial dysfunction and mitochondrial superoxide release in GECs. There is a rapid oxidation of mitochondrial DNA and loss of mitochondrial biogenesis without cell death. Many of these effects are blocked by mitoTEMPO a selective mitochondrial anti-oxidant. Secreted factors from dysfunctional GECs were sufficient to cause podocyte apoptosis in supernatant transfer experiments, or in co-culture but this did not occur when GECs had been previously treated with mitoTEMPO. CONCLUSION: Dissecting the impact of the diabetic environment on individual cell-types from the kidney glomerulus indicates that GECs become dysfunctional and pathological to neighboring podocytes by increased levels of mitochondrial superoxide in GEC. These studies indicate that GEC-signaling to podocytes contributes to the loss of the glomerular filtration barrier in DKD. Video abstract.


Asunto(s)
Microambiente Celular , Diabetes Mellitus Experimental/patología , Células Endoteliales/patología , Glomérulos Renales/patología , Mitocondrias/patología , Estrés Oxidativo , Podocitos/patología , Animales , Apoptosis , Autofagia , ADN Mitocondrial/genética , Endodesoxirribonucleasas/metabolismo , Células Endoteliales/ultraestructura , Masculino , Ratones , Mitocondrias/ultraestructura , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/ultraestructura , Podocitos/ultraestructura
5.
Bioorg Med Chem Lett ; 29(14): 1694-1698, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31129051

RESUMEN

Quinolines and thiazolopyridines were developed as allosteric inhibitors of MALT1, with good cellular potency and exquisite selectivity. Mouse pharmacokinetic (PK) profiling showed these to have low in vivo clearance, and moderate oral exposure. The thiazolopyridines were less lipophilic than the quinolines, and one thiazolopyridine example was active in our hIL10 mouse pharmacodynamic (PD) model upon oral dosing.


Asunto(s)
Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/antagonistas & inhibidores , Quinolinas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Humanos , Quinolinas/farmacología
6.
Nat Metab ; 5(4): 607-625, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37024752

RESUMEN

The lifetime risk of kidney disease in people with diabetes is 10-30%, implicating genetic predisposition in the cause of diabetic kidney disease (DKD). Here we identify an expression quantitative trait loci (QTLs) in the cis-acting regulatory region of the xanthine dehydrogenase, or xanthine oxidoreductase (Xor), a binding site for C/EBPß, to be associated with diabetes-induced podocyte loss in DKD in male mice. We examine mouse inbred strains that are susceptible (DBA/2J) and resistant (C57BL/6J) to DKD, as well as a panel of recombinant inbred BXD mice, to map QTLs. We also uncover promoter XOR orthologue variants in humans associated with high risk of DKD. We introduced the risk variant into the 5'-regulatory region of XOR in DKD-resistant mice, which resulted in increased Xor activity associated with podocyte depletion, albuminuria, oxidative stress and damage restricted to the glomerular endothelium, which increase further with type 1 diabetes, high-fat diet and ageing. Therefore, differential regulation of Xor contributes to phenotypic consequences with diabetes and ageing.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Humanos , Masculino , Ratones , Animales , Nefropatías Diabéticas/genética , Xantina Deshidrogenasa/genética , Xantina Deshidrogenasa/metabolismo , Predisposición Genética a la Enfermedad , Ratones Endogámicos DBA , Ratones Endogámicos C57BL
7.
J Neurochem ; 121(4): 680-92, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22394374

RESUMEN

Signal transduction and activator of transcription-3 (STAT3) plays an important role in neuronal survival, regeneration and repair after brain injury. We previously demonstrated that STAT3 is activated in brain after cerebral ischemia specifically in neurons. The effect was sex-specific and modulated by sex steroids, with higher activation in females than males. In the current study, we used a proteomics approach to identify downstream proteins affected by ischemia in male and female wild-type (WT) and neuron-specific STAT3 knockout (KO) mice. We established four comparison groups based on the transgenic condition and the hemisphere analyzed, respectively. Moreover, the sexual variable was taken into account and male and female animals were analyzed independently. Results support a role for STAT3 in metabolic, synaptic, structural and transcriptional responses to cerebral ischemia, indeed the adaptive response to ischemia/reperfusion injury is delayed in neuronal-specific STAT3 KO mice. The differences observed between males and females emphasize the importance of sex-specific neuronal survival and repair mechanisms, especially those involving antioxidant and energy-related activities, often caused by sex hormones.


Asunto(s)
Química Encefálica/genética , Isquemia Encefálica/genética , Encéfalo/fisiología , Proteoma , Daño por Reperfusión/genética , Factor de Transcripción STAT3/genética , Animales , Western Blotting , Isquemia Encefálica/fisiopatología , Mapeo Cromosómico , Colorantes , Electroforesis en Gel Bidimensional , Femenino , Lateralidad Funcional/fisiología , Procesamiento de Imagen Asistido por Computador , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Masculino , Espectrometría de Masas , Ratones , Ratones Noqueados , Ratones Transgénicos , Daño por Reperfusión/fisiopatología , Reproducibilidad de los Resultados , Caracteres Sexuales , Tripsina/química
8.
Endocrinol Diabetes Metab ; 4(1): e00191, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33532622

RESUMEN

Introduction: Sex differences in brain cortical function affect cognition, behaviour and susceptibility to neural diseases, but the molecular basis of sexual dimorphism in cortical function is still largely unknown. Oestrogen and oestrogen receptors (ERs), specifically ERß, the most abundant ER in the cortex, may play a role in determining sex differences in gene expression, which could underlie functional sex differences. However, further investigation is needed to address brain region specificity of the effects of sex and ERß on gene expression. The goal of this study was to investigate sex differences in gene expression in the mouse posterior cortex, where sex differences in transcription have never been examined, and to determine how genetic ablation of ERß affects transcription. Methods: In this study, we performed unbiased transcriptomics on RNA from the posterior cortex of adult wild-type and ERß knockout mice (n = 4/sex/genotype). We used unbiased clustering to analyse whole-transcriptome changes between the groups. We also performed differential expression analysis on the data using DESeq2 to identify specific changes in gene expression. Results: We found only 27 significantly differentially expressed genes (DEGs) in wild-type (WT) males vs females, of which 17 were autosomal genes. Interestingly, in ERßKO males vs females all the autosomal DEGs were lost. Gene Ontology analysis of the subset of DEGs with sex differences only in the WT cortex revealed a significant enrichment of genes annotated with the function 'cation channel activity'. Moreover, within each sex we found only a few DEGs in ERßKO vs WT mice (8 and 5 in males and females, respectively). Conclusions: Overall, our results suggest that in the adult mouse posterior cortex there are surprisingly few sex differences in gene expression, and those that exist are mainly related to cation channel activity. Additionally, they indicate that brain region-specific functional effects of ERß may be largely post-transcriptional.


Asunto(s)
Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Receptor beta de Estrógeno/fisiología , Expresión Génica/genética , Caracteres Sexuales , Animales , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos/genética , Transcripción Genética/genética
9.
Endocrinology ; 162(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33269387

RESUMEN

Several neurodegenerative disorders are characterized by proteasome dysfunctions leading to protein aggregations and pathogenesis. Since we showed that estrogen receptor alpha (ERα) activates the proteasome, drugs able to stimulate ERα in the central nervous system (CNS) could hold potential for therapeutic intervention. However, the transcriptional effects of selective estrogen receptor modulators (SERMs), such as tamoxifen and raloxifene, can be tissue specific. A direct comparison of the effects of different SERMs on gene transcription in the CNS has never been performed. Here, we report an RNA-seq analysis of the spinal cord treated with estrogen, tamoxifen, or raloxifene. We find stark SERM and sex-specific differences in gene expression profiles in the spinal cord. Notably, raloxifene, but not estrogen or tamoxifen, modulates numerous deubiquitinating enzymes, proteasome subunits and assembly factors, and these effects translate into decreased protein aggregates. In the SOD1-G93A mouse model of amyotrophic lateral sclerosis, we found that even a low dose of raloxifene causes a significant decrease in mutant SOD1 aggregates in the spinal cord, accompanied by a delay in the decline of muscle strength in females, but not in males. These results strongly indicate SERM-selective as well as sex-specific effects, and emphasize the importance of sex as a biological variable to be considered for the careful selection of specific SERM for use in clinical trials for neurodegenerative diseases.


Asunto(s)
Enfermedades Neurodegenerativas/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Clorhidrato de Raloxifeno/uso terapéutico , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Médula Espinal/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Clorhidrato de Raloxifeno/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Caracteres Sexuales , Médula Espinal/enzimología , Ubiquitinación/efectos de los fármacos
10.
Hum Mol Genet ; 17(21): 3291-302, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18678599

RESUMEN

Autosomal dominant optic atrophy (ADOA), the commonest cause of inherited optic atrophy, is caused by mutations in the ubiquitously expressed gene optic atrophy 1 (OPA1), involved in fusion and biogenesis of the inner membrane of mitochondria. Bioenergetic failure, mitochondrial network abnormalities and increased apoptosis have all been proposed as possible causal factors. However, their relative contribution to pathogenesis as well as the prominent susceptibility of the retinal ganglion cell (RGC) in this disease remains uncertain. Here we identify a novel deletion of OPA1 gene in the GTPase domain in three patients affected by ADOA. Muscle biopsy of the patients showed neurogenic atrophy and abnormal morphology and distribution of mitochondria. Confocal microscopy revealed increased mitochondrial fragmentation in fibroblasts as well as in myotubes, where mitochondria were also unevenly distributed, with clustered organelles alternating with areas where mitochondria were sparse. These abnormalities were not associated with altered bioenergetics or increased susceptibility to pro-apoptotic stimuli. Therefore, changes in mitochondrial shape and distribution can be independent of other reported effects of OPA1 mutations, and therefore may be the primary cause of the disease. The arrangement of mitochondria in RGCs, which degenerate in ADOA, may be exquisitely sensitive to disturbance, and this may lead to bioenergetic crisis and/or induction of apoptosis. Our results highlight the importance of mitochondrial dynamics in the disease per se, and point to the loss of the fine positioning of mitochondria in the axons of RGCs as a possible explanation for their predominant degeneration in ADOA.


Asunto(s)
GTP Fosfohidrolasas/genética , Mitocondrias/metabolismo , Atrofia Óptica Autosómica Dominante/genética , Adolescente , Adulto , Apoptosis , Células Cultivadas , Niño , Metabolismo Energético , Femenino , GTP Fosfohidrolasas/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/patología , Músculo Esquelético/anomalías , Músculo Esquelético/enzimología , Atrofia Óptica Autosómica Dominante/fisiopatología , Linaje , Especies Reactivas de Oxígeno/metabolismo , Retina/patología , Eliminación de Secuencia , Adulto Joven
11.
Biochim Biophys Acta ; 1767(7): 913-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17568559

RESUMEN

Two point mutations (T>G and T>C) at the same 8993 nucleotide of mitochondrial DNA (at comparable mutant load), affecting the ATPase 6 subunit of the F1F0-ATPase, result in neurological phenotypes of variable severity in humans. We have investigated mitochondrial function in lymphocytes from individuals carrying the 8993T>C mutation: the results were compared with data from five 8993T>G NARP (Neuropathy, Ataxia and Retinitis Pigmentosa) patients. Both 8993T>G and 8993T>C mutations led to energy deprivation and ROS overproduction. However, the relative contribution of the two pathogenic components is different depending on the mutation considered. The 8993T>G change mainly induces an energy deficiency, whereas the 8993T>C favours an increased ROS production. These results possibly highlight the different pathogenic mechanism generated by the two mutations at position 8993 and provide useful information to better characterize the biochemical role of the highly conserved Leu-156 in ATPase 6 subunit of the mitochondrial ATP synthase complex.


Asunto(s)
Ataxia/genética , Genes Mitocondriales , Enfermedad de Leigh/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Enfermedades del Sistema Nervioso Periférico/genética , Retinitis Pigmentosa/genética , Adenosina Trifosfato/metabolismo , Adulto , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Femenino , Humanos , Leucina/química , Leucina/genética , Linfocitos/metabolismo , Potenciales de la Membrana , Persona de Mediana Edad , Mutación , Fenotipo , Especies Reactivas de Oxígeno/metabolismo
12.
J Clin Invest ; 128(10): 4397-4412, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30024860

RESUMEN

The paracaspase MALT1 plays an essential role in activated B cell-like diffuse large B cell lymphoma (ABC DLBCL) downstream of B cell and TLR pathway genes mutated in these tumors. Although MALT1 is considered a compelling therapeutic target, the development of tractable and specific MALT1 protease inhibitors has thus far been elusive. Here, we developed a target engagement assay that provides a quantitative readout for specific MALT1-inhibitory effects in living cells. This enabled a structure-guided medicinal chemistry effort culminating in the discovery of pharmacologically tractable, irreversible substrate-mimetic compounds that bind the MALT1 active site. We confirmed that MALT1 targeting with compound 3 is effective at suppressing ABC DLBCL cells in vitro and in vivo. We show that a reduction in serum IL-10 levels exquisitely correlates with the drug pharmacokinetics and degree of MALT1 inhibition in vitro and in vivo and could constitute a useful pharmacodynamic biomarker to evaluate these compounds in clinical trials. Compound 3 revealed insights into the biology of MALT1 in ABC DLBCL, such as the role of MALT1 in driving JAK/STAT signaling and suppressing the type I IFN response and MHC class II expression, suggesting that MALT1 inhibition could prime lymphomas for immune recognition by cytotoxic immune cells.


Asunto(s)
Inhibidores de Caspasas , Sistemas de Liberación de Medicamentos , Linfoma de Células B Grandes Difuso , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas , Proteínas de Neoplasias , Transducción de Señal , Animales , Inhibidores de Caspasas/química , Inhibidores de Caspasas/farmacología , Dominio Catalítico , Línea Celular Tumoral , Femenino , Humanos , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/enzimología , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Masculino , Ratones , Ratones Endogámicos NOD , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/antagonistas & inhibidores , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/química , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
13.
Diabetes ; 66(3): 763-778, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27899487

RESUMEN

The molecular signaling mechanisms between glomerular cell types during initiation/progression of diabetic kidney disease (DKD) remain poorly understood. We compared the early transcriptome profile between DKD-resistant C57BL/6J and DKD-susceptible DBA/2J (D2) glomeruli and demonstrated a significant downregulation of essential mitochondrial genes in glomeruli from diabetic D2 mice, but not in C57BL/6J, with comparable hyperglycemia. Diabetic D2 mice manifested increased mitochondrial DNA lesions (8-oxoguanine) exclusively localized to glomerular endothelial cells after 3 weeks of diabetes, and these accumulated over time in addition to increased urine secretion of 8-oxo-deoxyguanosine. Detailed assessment of glomerular capillaries from diabetic D2 mice demonstrated early signs of endothelial injury and loss of fenestrae. Glomerular endothelial mitochondrial dysfunction was associated with increased glomerular endothelin-1 receptor type A (Ednra) expression and increased circulating endothelin-1 (Edn1). Selective Ednra blockade or mitochondrial-targeted reactive oxygen species scavenging prevented mitochondrial oxidative stress of endothelial cells and ameliorated diabetes-induced endothelial injury, podocyte loss, albuminuria, and glomerulosclerosis. In human DKD, increased urine 8-oxo-deoxyguanosine was associated with rapid DKD progression, and biopsies from patients with DKD showed increased mitochondrial DNA damage associated with glomerular endothelial EDNRA expression. Our studies show that DKD susceptibility was linked to mitochondrial dysfunction, mediated largely by Edn1-Ednra in glomerular endothelial cells representing an early event in DKD progression, and suggest that cross talk between glomerular endothelial injury and podocytes leads to defects and depletion, albuminuria, and glomerulosclerosis.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Nefropatías Diabéticas/metabolismo , Endotelina-1/metabolismo , Endotelio/metabolismo , Glomérulos Renales/metabolismo , Mitocondrias/metabolismo , Receptor de Endotelina A/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Adulto , Anciano , Albuminuria , Animales , Antioxidantes/farmacología , Cromatografía Líquida de Alta Presión , ADN Mitocondrial/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/orina , Susceptibilidad a Enfermedades , Endotelio/efectos de los fármacos , Endotelio/ultraestructura , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Glomérulos Renales/ultraestructura , Masculino , Células Mesangiales/patología , Células Mesangiales/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Microscopía Electrónica de Rastreo , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Compuestos Organofosforados/farmacología , Consumo de Oxígeno , Piperidinas/farmacología , Podocitos/patología , Podocitos/ultraestructura , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Adulto Joven
14.
Nephron ; 131(4): 278-84, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26613578

RESUMEN

BACKGROUND/AIMS: In podocytes, the overexpression of TGFß ligands and receptors during glomerulosclerosis could be a causal factor for injury induction and perpetuation in glomerular tufts. Mitochondrial dysfunction and oxidative stress are emerging as potential therapeutic targets in glomerular injury, and TGFß has been shown to modulate mitochondrial metabolism in different cell types. This study aims at investigating the role of TGFß in podocyte energy metabolism and cytoskeleton dynamics. METHODS: Mitochondrial function and cytoskeleton dynamics were analyzed in TGFß-treated WT and Smad2/3 double KO podocytes. RESULTS: TGFß treatment in podocytes induced a significant Smad-dependent increase of mitochondrial oxygen consumption rate (OCR). ATP content was unchanged and increased respiration was not associated with increased mitochondrial mass. Increased cellular reactive oxygen species induced by Smad-mediated TGFß signaling were reverted by NADPH oxidase inhibitor apocynin. TGFß treatment did not induce mitochondrial oxidative stress, and Smad2/3-dependent TGFß signaling and increased mitochondrial OCR were found to be associated with actin cytoskeleton dynamics. The role of motor proteins myosin II and dynamin in TGFß-induced actin polymerization was demonstrated by specific inhibition, resulting in actin stabilization and normalization of mitochondrial OCR. CONCLUSION: TGFß-induced rearrangements of actin cytoskeleton are controlled by Smad2/3 signaling pathways and coupled with the activation of mitochondrial ATP synthesis as bioenergetic adaptation to ATP consumption by ATP- and GTP-dependent motor proteins, myosin II and dynamin.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Adaptación Fisiológica , Metabolismo Energético , Mitocondrias/metabolismo , Podocitos/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Citoesqueleto de Actina/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Células Cultivadas , Ratones , Podocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Proteínas Smad/fisiología
15.
J Clin Invest ; 124(4): 1608-21, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24590287

RESUMEN

Focal segmental glomerular sclerosis (FSGS) is a primary kidney disease that is commonly associated with proteinuria and progressive loss of glomerular function, leading to development of chronic kidney disease (CKD). FSGS is characterized by podocyte injury and depletion and collapse of glomerular capillary segments. Progression of FSGS is associated with TGF-ß activation in podocytes; however, it is not clear how TGF-ß signaling promotes disease. Here, we determined that podocyte-specific activation of TGF-ß signaling in transgenic mice and BALB/c mice with Adriamycin-induced glomerulosclerosis is associated with endothelin-1 (EDN1) release by podocytes, which mediates mitochondrial oxidative stress and dysfunction in adjacent endothelial cells via paracrine EDN1 receptor type A (EDNRA) activation. Endothelial dysfunction promoted podocyte apoptosis, and inhibition of EDNRA or scavenging of mitochondrial-targeted ROS prevented podocyte loss, albuminuria, glomerulosclerosis, and renal failure. We confirmed reciprocal crosstalk between podocytes and endothelial cells in a coculture system. Biopsies from patients with FSGS exhibited increased mitochondrial DNA damage, consistent with EDNRA-mediated glomerular endothelial mitochondrial oxidative stress. Our studies indicate that segmental glomerulosclerosis develops as a result of podocyte-endothelial crosstalk mediated by EDN1/EDNRA-dependent mitochondrial dysfunction and suggest that targeting the reciprocal interaction between podocytes and endothelia may provide opportunities for therapeutic intervention in FSGS.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Podocitos/metabolismo , Podocitos/patología , Animales , Línea Celular , Modelos Animales de Enfermedad , Endotelina-1/genética , Endotelina-1/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mitocondrias/metabolismo , Modelos Biológicos , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Endotelina A/genética , Receptor de Endotelina A/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
16.
Semin Nephrol ; 32(3): 295-303, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22835461

RESUMEN

The transforming growth factor-ß (TGF-ß) family comprises more than 30 family members that are structurally related secreted dimeric cytokines, including TGF-ß, activins, and bone morphogenetic proteins/growth and differentiation factors. TGF-ß are pluripotent regulators of cell proliferation, differentiation, apoptosis, migration, and adhesion of many different cell types. TGF-ß pathways are highly evolutionarily conserved and control embryogenesis, tissue repair, and tissue homeostasis in invertebrates and vertebrates. Aberrations in TGF-ß activity and signaling underlie a broad spectrum of developmental disorders and major pathologies in human beings, including cancer, fibrosis, and autoimmune diseases. Recent observations have indicated an emerging role for TGF-ß in the regulation of mitochondrial bioenergetics and oxidative stress responses characteristic of chronic degenerative diseases and aging. Conversely, energy and metabolic sensory pathways cross-regulate mediators of TGF-ß signaling. Here, we review TGF-ß and regulation of bioenergetic and mitochondrial functions, including energy and oxidant metabolism and apoptotic cell death, as well as their emerging relevance in renal biology and disease.


Asunto(s)
ADN Mitocondrial/metabolismo , Riñón/patología , Mitocondrias/metabolismo , Insuficiencia Renal Crónica/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis/fisiología , Metabolismo Energético/fisiología , Fibrosis , Humanos , Riñón/metabolismo , Ratones , Estrés Oxidativo/fisiología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Proteínas Smad/metabolismo
17.
Brain Res ; 1362: 1-12, 2010 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-20875800

RESUMEN

Although the role of STAT3 in cell physiology and tissue development has been largely investigated, its involvement in the development and maintenance of nervous tissue and in the mechanisms of neuroprotection is not yet known. The potentially wide range of STAT3 activities raises the question of tissue- and gender-specificity as putative mechanisms of regulation. To explore the function of STAT3 in the brain and the hypothesis of a gender-linked modulation of STAT3, we analyzed a neuron-specific STAT3 knockout mouse model investigating the influence of STAT3 activity in brain protein expression pattern in both males and females in the absence of neurological insult. We performed a proteomic study aimed to reveal the molecular pathways directly or indirectly controlled by STAT3 underscoring its role in brain development and maintenance. We identified several proteins, belonging to different neuronal pathways such as energy metabolism or synaptic transmission, controlled by STAT3 that confirm its crucial role in brain development and maintenance. Moreover, we investigated the different processes that could contribute to the sexual dimorphic behavior observed in the incidence of neurological and mental disease. Interestingly both STAT3 KO and gender factors influence the expression of several mitochondrial proteins conferring to mitochondrial activity high importance in the regulation of brain physiology and conceivable relevance as therapeutic target.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Proteínas del Tejido Nervioso/fisiología , Proteómica/métodos , Factor de Transcripción STAT3/fisiología , Caracteres Sexuales , Animales , Química Encefálica/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética
18.
Arch Neurol ; 66(8): 951-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19667215

RESUMEN

OBJECTIVE: To verify whether enhanced substrate-level phosphorylation increases viability and adenosine 5'-triphosphate (ATP) content of cells with neuropathy, ataxia, and retinitis pigmentosa/maternally inherited Leigh syndrome (NARP/MILS) mitochondrial DNA mutations and ATP synthase dysfunction. DESIGN: We used cell lines "poisoned" with oligomycin, the specific inhibitor of ATP synthase, and "natural" models, including transmitochondrial human cell lines (cybrids) harboring 2 different pathogenic mutations associated with the NARP/MILS phenotypes. MAIN OUTCOME MEASURES: Cell survival, morphology, and ATP content. RESULTS: When normal human fibroblasts cultured in glucose-free medium were forced to increase energy consumption by exposure to the ionophore gramicidin or were energy challenged by oligomycin inhibition, their survival at 72 hours was 5%, but this increased to 70% when the medium was supplemented with alpha-ketoglutarate/aspartate to boost mitochondrial substrate-level phosphorylation. Homoplasmic cybrids harboring the 8993T-->G NARP mutation were also protected from death (75% vs 15% survival at 72 hours) by the supplemented medium and their ATP content was similar to controls. CONCLUSIONS: These results show that ATP synthase-deficient cells can be rescued by increasing mitochondrial substrate-level phosphorylation and suggest potential dietary or pharmacological therapeutic approaches based on the supplementation of alpha-ketoglutarate/aspartate to patients with impaired ATP synthase activity.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ácido Aspártico/farmacología , Proteína C-Reactiva/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , ADN Mitocondrial/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Ácidos Cetoglutáricos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , ATPasas de Translocación de Protón Mitocondriales/genética , Mutación , Proteínas del Tejido Nervioso/genética , Oligomicinas/farmacología , Antiinfecciosos Locales/farmacología , Línea Celular , Gramicidina/farmacología , Humanos , Enfermedad de Leigh/genética , Fosforilación Oxidativa/efectos de los fármacos , Retinitis Pigmentosa/genética , Degeneraciones Espinocerebelosas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA