Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(29): e2221249120, 2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37410813

RESUMEN

Asynchronous skeletal muscle degeneration/regeneration is a hallmark feature of Duchenne muscular dystrophy (DMD); however, traditional -omics technologies that lack spatial context make it difficult to study the biological mechanisms of how asynchronous regeneration contributes to disease progression. Here, using the severely dystrophic D2-mdx mouse model, we generated a high-resolution cellular and molecular spatial atlas of dystrophic muscle by integrating spatial transcriptomics and single-cell RNAseq datasets. Unbiased clustering revealed nonuniform distribution of unique cell populations throughout D2-mdx muscle that were associated with multiple regenerative timepoints, demonstrating that this model faithfully recapitulates the asynchronous regeneration observed in human DMD muscle. By probing spatiotemporal gene expression signatures, we found that propagation of inflammatory and fibrotic signals from locally damaged areas contributes to widespread pathology and that querying expression signatures within discrete microenvironments can identify targetable pathways for DMD therapy. Overall, this spatial atlas of dystrophic muscle provides a valuable resource for studying DMD disease biology and therapeutic target discovery.


Asunto(s)
Músculo Esquelético , Distrofia Muscular de Duchenne , Animales , Ratones , Humanos , Músculo Esquelético/metabolismo , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/metabolismo , Progresión de la Enfermedad , Modelos Animales de Enfermedad
2.
JBMR Plus ; 7(7): e10753, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37457877

RESUMEN

Mutations in the COL1A1 and COL1A2 genes, which encode type I collagen, are present in around 85%-90% of osteogenesis imperfecta (OI) patients. Because type I collagen is the principal protein composition of bones, any changes in its gene sequences or synthesis can severely affect bone structure. As a result, skeletal deformity and bone frailty are defining characteristics of OI. Homozygous oim/oim mice are utilized as models of severe progressive type III OI. Bone adapts to external forces by altering its mass and architecture. Previous attempts to leverage the relationship between muscle and bone involved using a soluble activin receptor type IIB-mFc (sActRIIB-mFc) fusion protein to lower circulating concentrations of activin A and myostatin. These two proteins are part of the TGF-ß superfamily that regulate muscle and bone function. While this approach resulted in increased muscle masses and enhanced bone properties, adverse effects emerged due to ligand promiscuity, limiting clinical efficacy and obscuring the precise contributions of myostatin and activin A. In this study, we investigated the musculoskeletal and whole-body metabolism effect of treating 5-week-old wildtype (Wt) and oim/oim mice for 11 weeks with either control antibody (Ctrl-Ab) or monoclonal anti-activin A antibody (ActA-Ab), anti-myostatin antibody (Mstn-Ab), or a combination of ActA-Ab and Mstn-Ab (Combo). We demonstrated that ActA-Ab treatment minimally impacts muscle mass in oim/oim mice, whereas Mstn-Ab and Combo treatments substantially increased muscle mass and overall lean mass regardless of genotype and sex. Further, while no improvements in cortical bone microarchitecture were observed with all treatments, minimal improvements in trabecular bone microarchitecture were observed with the Combo treatment in oim/oim mice. Our findings suggest that individual or combinatorial inhibition of myostatin and activin A alone is insufficient to robustly improve femoral biomechanical and microarchitectural properties in severely affected OI mice. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

3.
Cells ; 12(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36980244

RESUMEN

The rare SLC30A8 mutation encoding a truncating p.Arg138* variant (R138X) in zinc transporter 8 (ZnT8) is associated with a 65% reduced risk for type 2 diabetes. To determine whether ZnT8 is required for beta cell development and function, we derived human pluripotent stem cells carrying the R138X mutation and differentiated them into insulin-producing cells. We found that human pluripotent stem cells with homozygous or heterozygous R138X mutation and the null (KO) mutation have normal efficiency of differentiation towards insulin-producing cells, but these cells show diffuse granules that lack crystalline zinc-containing insulin granules. Insulin secretion is not compromised in vitro by KO or R138X mutations in human embryonic stem cell-derived beta cells (sc-beta cells). Likewise, the ability of sc-beta cells to secrete insulin and maintain glucose homeostasis after transplantation into mice was comparable across different genotypes. Interestingly, sc-beta cells with the SLC30A8 KO mutation showed increased cytoplasmic zinc, and cells with either KO or R138X mutation were resistant to apoptosis when extracellular zinc was limiting. These findings are consistent with a protective role of zinc in cell death and with the protective role of zinc in T2D.


Asunto(s)
Proteínas de Transporte de Catión , Diabetes Mellitus Tipo 2 , Células Madre Embrionarias Humanas , Transportador 8 de Zinc , Zinc , Animales , Humanos , Ratones , Apoptosis/genética , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/fisiología , Insulina/metabolismo , Mutación con Pérdida de Función , Mutación/genética , Zinc/metabolismo , Transportador 8 de Zinc/genética , Transportador 8 de Zinc/metabolismo
4.
J Bone Miner Res ; 37(5): 938-953, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35195284

RESUMEN

Osteogenesis imperfecta (OI) is a collagen-related bone disorder characterized by fragile osteopenic bone and muscle weakness. We have previously shown that the soluble activin receptor type IIB decoy (sActRIIB) molecule increases muscle mass and improves bone strength in the mild to moderate G610C mouse model of OI. The sActRIIB molecule binds multiple transforming growth factor-ß (TGF-ß) ligands, including myostatin and activin A. Here, we investigate the musculoskeletal effects of inhibiting activin A alone, myostatin alone, or both myostatin and activin A in wild-type (Wt) and heterozygous G610C (+/G610C) mice using specific monoclonal antibodies. Male and female Wt and +/G610C mice were treated twice weekly with intraperitoneal injections of monoclonal control antibody (Ctrl-Ab, Regn1945), anti-activin A antibody (ActA-Ab, Regn2476), anti-myostatin antibody (Mstn-Ab, Regn647), or both ActA-Ab and Mstn-Ab (Combo, Regn2476, and Regn647) from 5 to 16 weeks of age. Prior to euthanasia, whole body composition, metabolism and muscle force generation assessments were performed. Post euthanasia, hindlimb muscles were evaluated for mass, and femurs were evaluated for changes in microarchitecture and biomechanical strength using micro-computed tomography (µCT) and three-point bend analyses. ActA-Ab treatment minimally impacted the +/G610C musculoskeleton, and was detrimental to bone strength in male +/G610C mice. Mstn-Ab treatment, as previously reported, resulted in substantial increases in hindlimb muscle weights and overall body weights in Wt and male +/G610C mice, but had minimal skeletal impact in +/G610C mice. Conversely, the Combo treatment outperformed ActA-Ab alone or Mstn-Ab alone, consistently increasing hindlimb muscle and body weights regardless of sex or genotype and improving bone microarchitecture and strength in both male and female +/G610C and Wt mice. Combinatorial inhibition of activin A and myostatin more potently increased muscle mass and bone microarchitecture and strength than either antibody alone, recapturing most of the observed benefits of sActRIIB treatment in +/G610C mice. © 2022 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Osteogénesis Imperfecta , Activinas , Animales , Peso Corporal , Modelos Animales de Enfermedad , Femenino , Fémur/diagnóstico por imagen , Fémur/metabolismo , Masculino , Ratones , Miostatina/genética , Osteogénesis Imperfecta/diagnóstico por imagen , Osteogénesis Imperfecta/tratamiento farmacológico , Osteogénesis Imperfecta/genética , Microtomografía por Rayos X
5.
Cell Metab ; 33(11): 2215-2230.e8, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34592133

RESUMEN

Endurance exercise promotes skeletal muscle vascularization, oxidative metabolism, fiber-type switching, and neuromuscular junction integrity. Importantly, the metabolic and contractile properties of the muscle fiber must be coupled to the identity of the innervating motor neuron (MN). Here, we show that muscle-derived neurturin (NRTN) acts on muscle fibers and MNs to couple their characteristics. Using a muscle-specific NRTN transgenic mouse (HSA-NRTN) and RNA sequencing of MN somas, we observed that retrograde NRTN signaling promotes a shift toward a slow MN identity. In muscle, NRTN increased capillary density and oxidative capacity and induced a transcriptional reprograming favoring fatty acid metabolism over glycolysis. This combination of effects on muscle and MNs makes HSA-NRTN mice lean with remarkable exercise performance and motor coordination. Interestingly, HSA-NRTN mice largely recapitulate the phenotype of mice with muscle-specific expression of its upstream regulator PGC-1ɑ1. This work identifies NRTN as a myokine that couples muscle oxidative capacity to slow MN identity.


Asunto(s)
Neuronas Motoras , Neurturina , Animales , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Músculo Esquelético/metabolismo , Neurturina/genética , Neurturina/metabolismo , Neurturina/farmacología , Estrés Oxidativo
6.
JCI Insight ; 6(5)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33529174

RESUMEN

Limitations in cell proliferation are important for normal function of differentiated tissues and essential for the safety of cell replacement products made from pluripotent stem cells, which have unlimited proliferative potential. To evaluate whether these limitations can be established pharmacologically, we exposed pancreatic progenitors differentiating from human pluripotent stem cells to small molecules that interfere with cell cycle progression either by inducing G1 arrest or by impairing S phase entry or S phase completion and determined growth potential, differentiation, and function of insulin-producing endocrine cells. We found that the combination of G1 arrest with a compromised ability to complete DNA replication promoted the differentiation of pancreatic progenitor cells toward insulin-producing cells and could substitute for endocrine differentiation factors. Reduced replication fork speed during differentiation improved the stability of insulin expression, and the resulting cells protected mice from diabetes without the formation of cystic growths. The proliferative potential of grafts was proportional to the reduction of replication fork speed during pancreatic differentiation. Therefore, a compromised ability to enter and complete S phase is a functionally important property of pancreatic endocrine differentiation, can be achieved by reducing replication fork speed, and is an important determinant of cell-intrinsic limitations of growth.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Replicación del ADN , Diabetes Mellitus , Células Madre Pluripotentes Inducidas , Células Secretoras de Insulina , Trasplante de Células Madre , Animales , Afidicolina , Proliferación Celular , Diabetes Mellitus/terapia , Humanos , Insulina/metabolismo , Islotes Pancreáticos , Ratones , Páncreas , Células Madre Pluripotentes , Trasplantes
7.
J Bone Miner Res ; 36(4): 739-756, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33249643

RESUMEN

Osteogenesis imperfecta (OI) is a genetic connective tissue disorder characterized by compromised skeletal integrity, altered microarchitecture, and bone fragility. Current OI treatment strategies focus on bone antiresorptives and surgical intervention with limited effectiveness, and thus identifying alternative therapeutic options remains critical. Muscle is an important stimulus for bone formation. Myostatin, a TGF-ß superfamily myokine, acts through ActRIIB to negatively regulate muscle growth. Recent studies demonstrated the potential benefit of myostatin inhibition with the soluble ActRIIB fusion protein on skeletal properties, although various OI mouse models exhibited variable skeletal responses. The genetic and clinical heterogeneity associated with OI, the lack of specificity of the ActRIIB decoy molecule for myostatin alone, and adverse events in human clinical trials further the need to clarify myostatin's therapeutic potential and role in skeletal integrity. In this study, we determined musculoskeletal outcomes of genetic myostatin deficiency and postnatal pharmacological myostatin inhibition by a monoclonal anti-myostatin antibody (Regn647) in the G610C mouse, a model of mild-moderate type I/IV human OI. In the postnatal study, 5-week-old wild-type and +/G610C male and female littermates were treated with Regn647 or a control antibody for 11 weeks or for 7 weeks followed by a 4-week treatment holiday. Inhibition of myostatin, whether genetically or pharmacologically, increased muscle mass regardless of OI genotype, although to varying degrees. Genetic myostatin deficiency increased hindlimb muscle weights by 6.9% to 34.4%, whereas pharmacological inhibition increased them by 13.5% to 29.6%. Female +/mstn +/G610C (Dbl.Het) mice tended to have similar trabecular and cortical bone parameters as Wt showing reversal of +/G610C characteristics but with minimal effect of +/mstn occurring in male mice. Pharmacologic myostatin inhibition failed to improve skeletal bone properties of male or female +/G610C mice, although skeletal microarchitectural and biomechanical improvements were observed in male wild-type mice. Four-week treatment holiday did not alter skeletal outcomes. © 2020 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Osteogénesis Imperfecta , Animales , Huesos , Colágeno Tipo I , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Miostatina/genética , Osteogénesis , Osteogénesis Imperfecta/tratamiento farmacológico , Osteogénesis Imperfecta/genética
8.
Nat Genet ; 51(11): 1596-1606, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31676859

RESUMEN

A rare loss-of-function allele p.Arg138* in SLC30A8 encoding the zinc transporter 8 (ZnT8), which is enriched in Western Finland, protects against type 2 diabetes (T2D). We recruited relatives of the identified carriers and showed that protection was associated with better insulin secretion due to enhanced glucose responsiveness and proinsulin conversion, particularly when compared with individuals matched for the genotype of a common T2D-risk allele in SLC30A8, p.Arg325. In genome-edited human induced pluripotent stem cell (iPSC)-derived ß-like cells, we establish that the p.Arg138* allele results in reduced SLC30A8 expression due to haploinsufficiency. In human ß cells, loss of SLC30A8 leads to increased glucose responsiveness and reduced KATP channel function similar to isolated islets from carriers of the T2D-protective allele p.Trp325. These data position ZnT8 as an appealing target for treatment aimed at maintaining insulin secretion capacity in T2D.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/prevención & control , Glucosa/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Transportador 8 de Zinc/metabolismo , Adolescente , Adulto , Anciano , Diabetes Mellitus Tipo 2/patología , Femenino , Genotipo , Humanos , Células Madre Pluripotentes Inducidas/patología , Islotes Pancreáticos/patología , Masculino , Persona de Mediana Edad , Adulto Joven , Transportador 8 de Zinc/genética
9.
Proc Natl Acad Sci U S A ; 115(32): E7642-E7649, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30038024

RESUMEN

SLC30A8 encodes a zinc transporter that is primarily expressed in the pancreatic islets of Langerhans. In ß-cells it transports zinc into insulin-containing secretory granules. Loss-of-function (LOF) mutations in SLC30A8 protect against type 2 diabetes in humans. In this study, we generated a knockin mouse model carrying one of the most common human LOF mutations for SLC30A8, R138X. The R138X mice had normal body weight, glucose tolerance, and pancreatic ß-cell mass. Interestingly, in hyperglycemic conditions induced by the insulin receptor antagonist S961, the R138X mice showed a 50% increase in insulin secretion. This effect was not associated with enhanced ß-cell proliferation or mass. Our data suggest that the SLC30A8 R138X LOF mutation may exert beneficial effects on glucose metabolism by increasing the capacity of ß-cells to secrete insulin under hyperglycemic conditions.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Transportador 8 de Zinc/genética , Alelos , Animales , Glucemia , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Humanos , Hiperglucemia/sangre , Hiperglucemia/inducido químicamente , Hiperglucemia/metabolismo , Secreción de Insulina , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/farmacología , Receptor de Insulina/antagonistas & inhibidores , Receptor de Insulina/metabolismo , Transportador 8 de Zinc/metabolismo
10.
J Lipid Res ; 58(6): 1166-1173, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28413163

RESUMEN

Angiopoietin-like (ANGPTL)3 and ANGPTL8 are secreted proteins and inhibitors of LPL-mediated plasma triglyceride (TG) clearance. It is unclear how these two ANGPTL proteins interact to regulate LPL activity. ANGPTL3 inhibits LPL activity and increases serum TG independent of ANGPTL8. These effects are reversed with an ANGPTL3 blocking antibody. Here, we show that ANGPTL8, although it possesses a functional inhibitory motif, is inactive by itself and requires ANGPTL3 expression to inhibit LPL and increase plasma TG. Using a mutated form of ANGPTL3 that lacks LPL inhibitory activity, we demonstrate that ANGPTL3 activity is not required for its ability to activate ANGPTL8. Moreover, coexpression of ANGPTL3 and ANGPTL8 leads to a far more efficacious increase in TG in mice than ANGPTL3 alone, suggesting the major inhibitory activity of this complex derives from ANGPTL8. An antibody to the C terminus of ANGPTL8 reversed LPL inhibition by ANGPTL8 in the presence of ANGPTL3. The antibody did not disrupt the ANGPTL8:ANGPTL3 complex, but came in close proximity to the LPL inhibitory motif in the N terminus of ANGPTL8. Collectively, these data show that ANGPTL8 has a functional LPL inhibitory motif, but only inhibits LPL and increases plasma TG levels in mice in the presence of ANGPTL3.


Asunto(s)
Proteínas Similares a la Angiopoyetina/metabolismo , Lipoproteína Lipasa/antagonistas & inhibidores , Hormonas Peptídicas/metabolismo , Triglicéridos/sangre , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteína 3 Similar a la Angiopoyetina , Proteína 8 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina/química , Proteínas Similares a la Angiopoyetina/deficiencia , Animales , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Hipertrigliceridemia/metabolismo , Lipoproteína Lipasa/metabolismo , Ratones , Hormonas Peptídicas/química , Hormonas Peptídicas/deficiencia
11.
Cell ; 158(1): 69-83, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24995979

RESUMEN

Brown fat can reduce obesity through the dissipation of calories as heat. Control of thermogenic gene expression occurs via the induction of various coactivators, most notably PGC-1α. In contrast, the transcription factor partner(s) of these cofactors are poorly described. Here, we identify interferon regulatory factor 4 (IRF4) as a dominant transcriptional effector of thermogenesis. IRF4 is induced by cold and cAMP in adipocytes and is sufficient to promote increased thermogenic gene expression, energy expenditure, and cold tolerance. Conversely, knockout of IRF4 in UCP1(+) cells causes reduced thermogenic gene expression and energy expenditure, obesity, and cold intolerance. IRF4 also induces the expression of PGC-1α and PRDM16 and interacts with PGC-1α, driving Ucp1 expression. Finally, cold, ß-agonists, or forced expression of PGC-1α are unable to cause thermogenic gene expression in the absence of IRF4. These studies establish IRF4 as a transcriptional driver of a program of thermogenic gene expression and energy expenditure.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Factores Reguladores del Interferón/metabolismo , Termogénesis , Factores de Transcripción/metabolismo , Activación Transcripcional , Adipocitos/metabolismo , Tejido Adiposo Pardo/citología , Agonistas de Receptores Adrenérgicos beta 3/farmacología , Animales , Frío , AMP Cíclico/metabolismo , Metabolismo Energético , Humanos , Canales Iónicos/genética , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Obesidad/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Delgadez/metabolismo , Activación Transcripcional/efectos de los fármacos , Proteína Desacopladora 1
12.
Nature ; 513(7516): 100-4, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-25043053

RESUMEN

Cachexia is a wasting disorder of adipose and skeletal muscle tissues that leads to profound weight loss and frailty. About half of all cancer patients suffer from cachexia, which impairs quality of life, limits cancer therapy and decreases survival. One key characteristic of cachexia is higher resting energy expenditure levels than in healthy individuals, which has been linked to greater thermogenesis by brown fat. How tumours induce brown fat activity is unknown. Here, using a Lewis lung carcinoma model of cancer cachexia, we show that tumour-derived parathyroid-hormone-related protein (PTHrP) has an important role in wasting, through driving the expression of genes involved in thermogenesis in adipose tissues. Neutralization of PTHrP in tumour-bearing mice blocked adipose tissue browning and the loss of muscle mass and strength. Our results demonstrate that PTHrP mediates energy wasting in fat tissues and contributes to the broader aspects of cancer cachexia. Thus, neutralization of PTHrP might hold promise for ameliorating cancer cachexia and improving patient survival.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Caquexia/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/patología , Animales , Caquexia/patología , Carcinoma Pulmonar de Lewis/genética , Medios de Cultivo Condicionados/farmacología , Metabolismo Energético/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Tamaño de los Órganos/efectos de los fármacos , Proteína Relacionada con la Hormona Paratiroidea/antagonistas & inhibidores , Termogénesis/efectos de los fármacos , Termogénesis/genética
13.
Am J Physiol Endocrinol Metab ; 306(2): E157-67, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24280126

RESUMEN

Diabetes risk increases significantly with age and correlates with lower oxidative capacity in muscle. Decreased expression of peroxisome proliferator-activated receptor-γ coactivator-1α (Pgc-1α) and target gene pathways involved in mitochondrial oxidative phosphorylation are associated with muscle insulin resistance, but a causative role has not been established. We sought to determine whether a decline in Pgc-1α and oxidative gene expression occurs during aging and potentiates the development of age-associated insulin resistance. Muscle-specific Pgc-1α knockout (MKO) mice and wild-type littermate controls were aged for 2 yr. Genetic signatures of skeletal muscle (microarray and mRNA expression) and metabolic profiles (glucose homeostasis, mitochondrial metabolism, body composition, lipids, and indirect calorimetry) of mice were compared at 3, 12, and 24 mo of age. Microarray and gene set enrichment analysis highlighted decreased function of the electron transport chain as characteristic of both aging muscle and loss of Pgc-1α expression. Despite significant reductions in oxidative gene expression and succinate dehydrogenase activity, young mice lacking Pgc-1α in muscle had lower fasting glucose and insulin. Consistent with loss of oxidative capacity during aging, Pgc-1α and Pgc-1ß expression were reduced in aged wild-type mouse muscle. Interestingly, the combination of age and loss of muscle Pgc-1α expression impaired glucose tolerance and led to increased fat mass, insulin resistance, and inflammatory markers in white adipose and liver tissues. Therefore, loss of Pgc-1α expression and decreased mitochondrial oxidative capacity contribute to worsening glucose tolerance and chronic systemic inflammation associated with aging.


Asunto(s)
Envejecimiento/fisiología , Intolerancia a la Glucosa/genética , Inflamación/genética , Músculo Esquelético/metabolismo , Factores de Transcripción/genética , Animales , Eliminación de Gen , Perfilación de la Expresión Génica , Intolerancia a la Glucosa/metabolismo , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Mitocondrias Musculares/metabolismo , Músculo Esquelético/patología , Fosforilación Oxidativa , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma
14.
Cell ; 151(6): 1319-31, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23217713

RESUMEN

PGC-1α is a transcriptional coactivator induced by exercise that gives muscle many of the best known adaptations to endurance-type exercise but has no effects on muscle strength or hypertrophy. We have identified a form of PGC-1α (PGC-1α4) that results from alternative promoter usage and splicing of the primary transcript. PGC-1α4 is highly expressed in exercised muscle but does not regulate most known PGC-1α targets such as the mitochondrial OXPHOS genes. Rather, it specifically induces IGF1 and represses myostatin, and expression of PGC-1α4 in vitro and in vivo induces robust skeletal muscle hypertrophy. Importantly, mice with skeletal muscle-specific transgenic expression of PGC-1α4 show increased muscle mass and strength and dramatic resistance to the muscle wasting of cancer cachexia. Expression of PGC-1α4 is preferentially induced in mouse and human muscle during resistance exercise. These studies identify a PGC-1α protein that regulates and coordinates factors involved in skeletal muscle hypertrophy.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal , Entrenamiento de Fuerza , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Adiposidad , Animales , Glucosa/metabolismo , Humanos , Hipertrofia , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/metabolismo , Miostatina/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Isoformas de Proteínas/metabolismo
15.
Cell ; 151(1): 96-110, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23021218

RESUMEN

PGC1α is a key transcriptional coregulator of oxidative metabolism and thermogenesis. Through a high-throughput chemical screen, we found that molecules antagonizing the TRPVs (transient receptor potential vanilloid), a family of ion channels, induced PGC1α expression in adipocytes. In particular, TRPV4 negatively regulated the expression of PGC1α, UCP1, and cellular respiration. Additionally, it potently controlled the expression of multiple proinflammatory genes involved in the development of insulin resistance. Mice with a null mutation for TRPV4 or wild-type mice treated with a TRPV4 antagonist showed elevated thermogenesis in adipose tissues and were protected from diet-induced obesity, adipose inflammation, and insulin resistance. This role of TRPV4 as a cell-autonomous mediator for both the thermogenic and proinflammatory programs in adipocytes could offer a target for treating obesity and related metabolic diseases.


Asunto(s)
Metabolismo Energético , Canales Catiónicos TRPV/metabolismo , Termogénesis , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Femenino , Técnicas de Silenciamiento del Gen , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Obesidad/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Transactivadores/metabolismo , Factores de Transcripción , Proteína Desacopladora 1
16.
Proc Natl Acad Sci U S A ; 109(24): 9635-40, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22645355

RESUMEN

Reduced peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) expression and mitochondrial dysfunction in adipose tissue have been associated with obesity and insulin resistance. Whether this association is causally involved in the development of insulin resistance or is only a consequence of this condition has not been clearly determined. Here we studied the effects of adipose-specific deficiency of PGC-1α on systemic glucose homeostasis. Loss of PGC-1α in white fat resulted in reduced expression of the thermogenic and mitochondrial genes in mice housed at ambient temperature, whereas gene expression patterns in brown fat were not altered. When challenged with a high-fat diet, insulin resistance was observed in the mutant mice, characterized by reduced suppression of hepatic glucose output. Resistance to insulin was also associated with an increase in circulating lipids, along with a decrease in the expression of genes regulating lipid metabolism and fatty acid uptake in adipose tissues. Taken together, these data demonstrate a critical role for adipose PGC-1α in the regulation of glucose homeostasis and a potentially causal involvement in the development of insulin resistance.


Asunto(s)
Tejido Adiposo/metabolismo , Resistencia a la Insulina , Transactivadores/fisiología , Animales , Grasas de la Dieta/administración & dosificación , Prueba de Tolerancia a la Glucosa , Homeostasis , Ratones , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Transactivadores/genética , Factores de Transcripción
17.
Genes Dev ; 26(3): 271-81, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22302939

RESUMEN

Certain white adipose tissue (WAT) depots are readily able to convert to a "brown-like" state with prolonged cold exposure or exposure to ß-adrenergic compounds. This process is characterized by the appearance of pockets of uncoupling protein 1 (UCP1)-positive, multilocular adipocytes and serves to increase the thermogenic capacity of the organism. We show here that fibroblast growth factor 21 (FGF21) plays a physiologic role in this thermogenic recruitment of WATs. In fact, mice deficient in FGF21 display an impaired ability to adapt to chronic cold exposure, with diminished browning of WAT. Adipose-derived FGF21 acts in an autocrine/paracrine manner to increase expression of UCP1 and other thermogenic genes in fat tissues. FGF21 regulates this process, at least in part, by enhancing adipose tissue PGC-1α protein levels independently of mRNA expression. We conclude that FGF21 acts to activate and expand the thermogenic machinery in vivo to provide a robust defense against hypothermia.


Asunto(s)
Adaptación Fisiológica/fisiología , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Factores de Crecimiento de Fibroblastos/metabolismo , Termogénesis/fisiología , Transactivadores/metabolismo , Adaptación Fisiológica/genética , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Diferenciación Celular , Células Cultivadas , Frío , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Procesamiento Postranscripcional del ARN , Transactivadores/genética , Factores de Transcripción
18.
Cell Metab ; 15(2): 230-9, 2012 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-22326224

RESUMEN

Progress has been made in elucidating the cell-surface phenotype of primary adipose progenitors; however, specific functional markers and distinct molecular signatures of fat depot-specific preadipocytes have remained elusive. In this study, we label committed murine adipose progenitors through expression of GFP from the genetic locus for Zfp423, a gene controlling preadipocyte determination. Selection of GFP-expressing fibroblasts from either subcutaneous or visceral adipose-derived stromal vascular cultures isolates stably committed preadipocytes that undergo robust adipogenesis. Immunohistochemistry for Zfp423-driven GFP expression in vivo confirms a perivascular origin of preadipocytes within both white and brown adipose tissues. Interestingly, a small subset of capillary endothelial cells within white and brown fat also express this marker, suggesting a contribution of specialized endothelial cells to the adipose lineage. Zfp423(GFP) mice represent a simple tool for the specific localization and isolation of molecularly defined preadipocytes from distinct adipose tissue depots.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/metabolismo , Pericitos/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Proteínas de Unión al ADN/genética , Citometría de Flujo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Ratones , Ratones Transgénicos , Células Madre/citología , Factores de Transcripción/genética
19.
J Biol Chem ; 284(28): 18624-33, 2009 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-19435887

RESUMEN

PPARdelta (peroxisome proliferator-activated receptor delta) is a regulator of lipid metabolism and has been shown to induce fatty acid oxidation (FAO). PPARdelta transgenic and knock-out mice indicate an involvement of PPARdelta in regulating mitochondrial biogenesis and oxidative capacity; however, the precise mechanisms by which PPARdelta regulates these pathways in skeletal muscle remain unclear. In this study, we determined the effect of selective PPARdelta agonism with the synthetic ligand, GW501516, on FAO and mitochondrial gene expression in vitro and in vivo. Our results show that activation of PPARdelta by GW501516 led to a robust increase in mRNA levels of key lipid metabolism genes. Mitochondrial gene expression and function were not induced under the same conditions. Additionally, the activation of Pdk4 transcription by PPARdelta was coactivated by PGC-1alpha. PGC-1alpha, but not PGC-1beta, was essential for full activation of Cpt-1b and Pdk4 gene expression via PPARdelta agonism. Furthermore, the induction of FAO by PPARdelta agonism was completely abolished in the absence of both PGC-1alpha and PGC-1beta. Conversely, PGC-1alpha-driven FAO was independent of PPARdelta. Neither GW501516 treatment nor knockdown of PPARdelta affects PGC-1alpha-induced mitochondrial gene expression in primary myotubes. These results demonstrate that pharmacological activation of PPARdelta induces FAO via PGC-1alpha. However, PPARdelta agonism does not induce mitochondrial gene expression and function. PGC-1alpha-induced FAO and mitochondrial biogenesis appear to be independent of PPARdelta.


Asunto(s)
Ácidos Grasos/metabolismo , Regulación de la Expresión Génica , Mitocondrias/metabolismo , Oxígeno/química , PPAR delta/metabolismo , Transactivadores/metabolismo , Animales , Células HeLa , Humanos , Lípidos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción , Transcripción Genética , Activación Transcripcional
20.
FEBS J ; 274(1): 227-40, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17222183

RESUMEN

Loss of E-cadherin-mediated cell-cell adhesion and expression of proteolytic enzymes characterize the transition from benign lesions to invasive, metastatic tumor, a rate-limiting step in the progression from adenoma to carcinoma in vivo. A soluble E-cadherin fragment found recently in the serum and urine of cancer patients has been shown to disrupt cell-cell adhesion and to drive cell invasion in a dominant-interfering manner. Physical disruption of cell-cell adhesion can be mimicked by the function-blocking antibody Decma. We have shown previously in MCF7 and T47D cells that urokinase-type plasminogen activator (uPA) activity is up-regulated upon disruption of E-cadherin-dependent cell-cell adhesion. We explored the underlying molecular mechanisms and found that blockage of E-cadherin by Decma elicits a signaling pathway downstream of E-cadherin that leads to Src-dependent Shc and extracellular regulated kinase (Erk) activation and results in uPAgene activation. siRNA-mediated knockdown of endogenous Src-homology collagen protein (Shc) and subsequent expression of single Shc isoforms revealed that p46(Shc) and p52(Shc) but not p66(Shc) were able to mediate Erk activation. A parallel pathway involving PI3K contributed partially to Decma-induced Erk activation. This report describes that disruption of E-cadherin-dependent cell-cell adhesion induces intracellular signaling with the potential to enhance tumorigenesis and, thus, offers new insights into the pathophysiological mechanisms of tumor development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cadherinas/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Activador de Plasminógeno de Tipo Uroquinasa/genética , Familia-src Quinasas/metabolismo , Animales , Anticuerpos/farmacología , Adhesión Celular , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , ARN Interferente Pequeño/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...