Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
PLoS One ; 15(6): e0232801, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32479500

RESUMEN

The myeloproliferative neoplasms, polycythemia vera, essential thrombocytosis and primary myelofibrosis are hematopoietic stem cell disorders and share driver mutations that either directly activate the thrombopoietin receptor, MPL, or activate it indirectly through gain-of-function mutations in the gene for JAK2, its cognate tyrosine kinase. Paradoxically, MPL surface expression in hematopoietic stem cells is also reduced in the myeloproliferative neoplasms due to abnormal post-translational glycosylation and premature destruction of JAK2, suggesting that the myeloproliferative neoplasms are disorders of MPL processing since MPL is the only hematopoietic growth factor receptor in hematopoietic stem cells. To examine this possibility, we genetically manipulated MPL expression and maturation in a JAK2V617F transgenic mouse model of polycythemia vera. Elimination of MPL expression completely abrogated the polycythemia vera phenotype in this JAK2V617F transgenic mouse model, which could only be partially restored by expression of one MPL allele. Most importantly, elimination of thrombopoietin gene expression abrogated the polycythemia vera phenotype in this JAK2V617F transgenic mouse model, which could be completely restored by expression of a single thrombopoietin allele. These data indicate that polycythemia vera is in part a thrombopoietin-dependent disorder and that targeting the MPL-thrombopoietin axis could be an effective, nonmyelotoxic therapeutic strategy in this disorder.


Asunto(s)
Janus Quinasa 2/genética , Policitemia Vera/genética , Policitemia Vera/metabolismo , Trombopoyetina/genética , Trombopoyetina/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Janus Quinasa 2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Trastornos Mieloproliferativos/genética , Fenotipo , Policitemia Vera/patología , Mielofibrosis Primaria/genética , Receptores de Trombopoyetina/genética , Trombocitemia Esencial/genética
4.
JCI Insight ; 5(1)2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31941838

RESUMEN

Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by cytopenia and extramedullary hematopoiesis, resulting in splenomegaly. Multiple pathological mechanisms (e.g., circulating cytokines and genetic alterations, such as JAKV617F mutation) have been implicated in the etiology of MF, but the molecular mechanism causing resistance to JAK2V617F inhibitor therapy remains unknown. Among MF patients who were treated with the JAK inhibitor ruxolitinib, we compared noncoding RNA profiles of ruxolitinib therapy responders versus nonresponders and found miR-543 was significantly upregulated in nonresponders. We validated these findings by reverse transcription-quantitative PCR. in this same cohort, in 2 additional independent MF patient cohorts from the United States and Romania, and in a JAK2V617F mouse model of MF. Both in vitro and in vivo models were used to determine the underlying molecular mechanism of miR-543 in MF. Here, we demonstrate that miR-543 targets the dioxygenases ten-eleven translocation 1 (TET1) and 2 (TET2) in patients and in vitro, causing increased levels of global 5-methylcytosine, while decreasing the acetylation of histone 3, STAT3, and tumor protein p53. Mechanistically, we found that activation of STAT3 by JAKs epigenetically controls miR-543 expression via binding the promoter region of miR-543. Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. Our findings suggest miR-543 as a potentially novel biomarker for the prognosis of MF patients with a high risk of treatment resistance and as a potentially new target for the development of new treatment options.


Asunto(s)
Proteínas de Unión al ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , MicroARNs/metabolismo , MicroARNs/farmacología , Mielofibrosis Primaria/tratamiento farmacológico , Proteínas Proto-Oncogénicas/efectos de los fármacos , Animales , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas , Modelos Animales de Enfermedad , Histonas , Humanos , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus/metabolismo , Ratones , MicroARNs/genética , Oxigenasas de Función Mixta , Mutación , Trastornos Mieloproliferativos , Nitrilos , Mielofibrosis Primaria/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/genética , Pirazoles/uso terapéutico , Pirimidinas , Factor de Transcripción STAT3 , Transcriptoma , Estados Unidos
5.
Science ; 359(6381): 1233-1239, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29590070

RESUMEN

Genetic association studies often examine features independently, potentially missing subpopulations with multiple phenotypes that share a single cause. We describe an approach that aggregates phenotypes on the basis of patterns described by Mendelian diseases. We mapped the clinical features of 1204 Mendelian diseases into phenotypes captured from the electronic health record (EHR) and summarized this evidence as phenotype risk scores (PheRSs). In an initial validation, PheRS distinguished cases and controls of five Mendelian diseases. Applying PheRS to 21,701 genotyped individuals uncovered 18 associations between rare variants and phenotypes consistent with Mendelian diseases. In 16 patients, the rare genetic variants were associated with severe outcomes such as organ transplants. PheRS can augment rare-variant interpretation and may identify subsets of patients with distinct genetic causes for common diseases.


Asunto(s)
Enfermedades Genéticas Congénitas/diagnóstico , Enfermedades Genéticas Congénitas/genética , Predisposición Genética a la Enfermedad , Análisis Mutacional de ADN , Bases de Datos Genéticas , Registros Electrónicos de Salud , Exoma , Estudios de Asociación Genética , Variación Genética , Humanos , Fenotipo , Factores de Riesgo
6.
Stem Cell Investig ; 4: 48, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28607922

RESUMEN

Acute myeloid leukemia (AML) is a cancer of the myeloid lineage of blood cells. Although significant progress has been made in treating many types of cancers during recent years, AML remains a deadly disease with survival rate lagging behind other blood cancers. A combination of toxic chemotherapies has been the standard AML treatment for more than 40 years. With intensive efforts to define the pathogenesis of AML, novel therapeutic drugs targeting key molecular defects in AML are being developed. Mutated in nearly 30% of AML, FMS-like tyrosine kinase 3 (FLT3) represents one of the most attractive targets. FLT3 mutants resulted from either internal tandem duplication (ITD) or point mutations possess enhanced kinase activity and cause constitutive activation of signaling. To date, several small molecule inhibitors of FLT3 have been developed but their clinical efficacy is limited due to a lack of potency and the generation of drug resistance. Therefore, next-generation FLT3 inhibitors overcoming these limitations are urgently in need. This review focuses on the pathological role of mutant FLT3 in the development of AML, the current status of FLT3 inhibitor development, and mechanisms underlining the development of resistance to existing FLT3 inhibitors.

7.
JCI Insight ; 2(7): e90932, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28405618

RESUMEN

Myelofibrosis (MF) is a bone marrow disorder characterized by clonal myeloproliferation, aberrant cytokine production, extramedullary hematopoiesis, and bone marrow fibrosis. Although somatic mutations in JAK2, MPL, and CALR have been identified in the pathogenesis of these diseases, inhibitors of the Jak2 pathway have not demonstrated efficacy in ameliorating MF in patients. TGF-ß family members are profibrotic cytokines and we observed significant TGF-ß1 isoform overexpression in a large cohort of primary MF patient samples. Significant overexpression of TGF-ß1 was also observed in murine clonal MPLW515L megakaryocytic cells. TGF-ß1 stimulated the deposition of excessive collagen by mesenchymal stromal cells (MSCs) by activating the TGF-ß receptor I kinase (ALK5)/Smad3 pathway. MSCs derived from MPLW515L mice demonstrated sustained overproduction of both collagen I and collagen III, effects that were abrogated by ALK5 inhibition in vitro and in vivo. Importantly, use of galunisertib, a clinically active ALK5 inhibitor, significantly improved MF in both MPLW515L and JAK2V617F mouse models. These data demonstrate the role of malignant hematopoietic stem cell (HSC)/TGF-ß/MSC axis in the pathogenesis of MF, and provide a preclinical rationale for ALK5 blockade as a therapeutic strategy in MF.


Asunto(s)
Janus Quinasa 2/metabolismo , Mielofibrosis Primaria/tratamiento farmacológico , Pirazoles/farmacología , Quinolinas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptores de Trombopoyetina/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Médula Ósea/patología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Células HEK293 , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mielofibrosis Primaria/metabolismo , Transducción de Señal , Proteína smad3/metabolismo
8.
Stem Cell Investig ; 2: 2, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27358870

RESUMEN

BACKGROUND: Myeloproliferative neoplasms (MPNs) are blood malignancies manifested in increased production of red blood cells, white blood cells, and/or platelets. Myelofibrosis is a subtype of MPNs characterized by the formation of scar-like tissues in the bone marrow due to abnormal hematopoiesis. It is considered a disease of both hematopoietic stem cells and stem cell niches. Patients with myelofibrosis have very poor prognosis, and there is no effective treatment so far. Myelofibrosis has routinely been detected by using histochemical staining methods which produce qualitative rather than quantitative results. In this study, we developed a quantitative assay of bone marrow myelofibrosis in JAK2V617F transgenic mice by determining hydroxyproline. METHODS: The JAK2V617F transgenic mice's tissue was collected to detect the bone marrow myelofibrosis. Statistical analyses were performed using the GraphPad Prism program. Differences of samples between two groups were accessed using t tests. P values less than 0.05 (2-tailed) were considered significantly different. RESULTS: We developed a quantitative method for detecting myelofibrosis by analyzing the content of hydroxyproline, a modified amino acid largely restricted to collagen which forms the fibrotic structure in bone marrow tissues. Our study also demonstrated age-dependent development of bone marrow myelofibrosis in JAK2V617F transgenic mice. CONCLUSIONS: In the present study, we have developed a new method for detecting bone marrow myelofibrosis by analyzing hydroxyproline contents. The method is highly sensitive and accurate. It provides more accurate, representative, and quantitative information than histochemical analyses. We believe that this method should find wide applications for analyzing the progression of myelofibrosis and efficacy of drug treatment.

9.
Stem Cell Investig ; 2: 22, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27358890

RESUMEN

Sickle-cell disease (SCD), also known as sickle-cell anemia, is a hereditary blood disorder characterized by the presence of abnormal hemoglobin, the oxygen-carrying protein found in red blood cells. This devastating hematologic disease affects millions of children worldwide. Currently the only available cure is an allogenic hematopoietic stem cell transplant (HSCT) which is limited by the scarcity of fully-matched donors. SCD is caused by a single nucleotide mutation in the beta-globin gene. Correction of this genetic defect would provide a cure for the disease. Two recent murine studies have provided proof of principle for such a strategy by correcting the mutation in hematopoietic stem cells (HSC) using genome editing techniques. With transformative advances being made in the genome editing field, effective and precise manipulation of cellular genomes is becoming highly feasible. Genome editing techniques in combination with stem cell therapy should provide a safe and curative treatment of various genetic diseases such as SCD.

10.
PLoS One ; 9(7): e99017, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25036984

RESUMEN

The JAK2V617F mutation is found in the majority of patients with myeloproliferative neoplasms (MPNs). Transgenic expression of the mutant gene causes MPN-like phenotypes in mice. We have produced JAK2V617F mice with p53 null background. Some of these mice developed acute erythroleukemia. From one of these mice, we derived a cell line designated J53Z1. J53Z1 cells were stained positive for surface markers CD71 and CD117 but negative for Sca-1, TER-119, CD11b, Gr-1, F4/80, CD11c, CD317, CD4, CD8a, CD3e, B220, CD19, CD41, CD42d, NK-1.1, and FceR1. Real time PCR analyses demonstrated expressions of erythropoietin receptor EpoR, GATA1, and GATA2 in these cells. J53Z1 cells grew rapidly in suspension culture containing fetal bovine serum with a doubling time of ∼18 hours. When transplanted into C57Bl/6 mice, J53Z1 cells induced acute erythroleukemia with massive infiltration of tumor cells in the spleen and liver. J53Z1 cells were responsive to stimulation with erythropoietin and stem cell factor and were selectively inhibited by JAK2 inhibitors which induced apoptosis of the cells. Together, J53Z1 cells belong to the erythroid lineage, and they may be useful for studying the role of JAK2V617F in proliferation and differentiation of erythroid cells and for identifying potential therapeutic drugs targeting JAK2.


Asunto(s)
Línea Celular Tumoral/enzimología , Janus Quinasa 2/genética , Leucemia Eritroblástica Aguda/patología , Mutación Missense , Proteínas de Neoplasias/genética , Mutación Puntual , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/trasplante , Cruzamientos Genéticos , Ensayos de Selección de Medicamentos Antitumorales , Eritropoyesis/efectos de los fármacos , Perfilación de la Expresión Génica , Genes p53 , Factores de Crecimiento de Célula Hematopoyética/farmacología , Humanos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Neoplasias/biosíntesis , Inhibidores de Proteínas Quinasas/farmacología , Bazo/patología
11.
J Immunol ; 193(2): 477-84, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24920845

RESUMEN

Pruritus occurs frequently in patients with polycythemia vera (PV), and the pathophysiology of PV-associated pruritus is unclear. We have previously demonstrated that transgenic mice expressing JAK2V617F displayed clear PV-like phenotypes. In the current study, we found frequent occurrence of pruritus with aged JAK2V617F transgenic mice and further investigated the underlying mechanisms by studying mast cells, key players in allergic reactions and anaphylaxis. Massive accumulations of mast cells were observed in the skin of pruritic JAK2V617F transgenic mice. In vitro culture yielded much higher mast cell counts from the bone marrow, spleen, peripheral blood, and peritoneal cavity of JAK2V617F transgenic mice than from controls. Cultured mast cells from JAK2V617F transgenic mice exhibited enhanced proliferative signals, relative resistance to cell death upon growth factor deprivation, and a growth advantage over control cells under suboptimal growth conditions. However, these mast cells displayed normal morphology and contained normal levels of mast cell proteases before and after degranulation. Finally, the JAK2 inhibitor G6 effectively reduced mast cell numbers and alleviated pruritus in JAK2V617F transgenic mice. Collectively, these data demonstrate that mast cells are involved in PV-associated pruritogenesis and that JAK2 inhibitors are potential antipruritus drugs.


Asunto(s)
Janus Quinasa 2/metabolismo , Mastocitos/metabolismo , Policitemia Vera/metabolismo , Prurito/metabolismo , Sustitución de Aminoácidos , Animales , Western Blotting , Células de la Médula Ósea/metabolismo , Células CHO , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Cricetulus , Femenino , Humanos , Interleucina-3/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Policitemia Vera/genética , Inhibidores de Proteínas Quinasas/farmacología , Prurito/genética , Prurito/prevención & control , Piel/metabolismo , Piel/patología , Factor de Células Madre/farmacología , Estilbenos/farmacología , Factores de Tiempo
12.
PLoS One ; 9(4): e93643, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24699465

RESUMEN

Although extremely rare, hematopoietic stem cells (HSCs) are divisible into subsets that differ with respect to differentiation potential and cell surface marker expression. For example, we recently found that CD86(-) CD150(+) CD48(-) HSCs have limited potential for lymphocyte production. This could be an important new tool for studying hematological abnormalities. Here, we analyzed HSC subsets with a series of stem cell markers in JAK2V617F transgenic (Tg) mice, where the mutation is sufficient to cause myeloproliferative neoplasia with lymphocyte deficiency. Total numbers of HSC were elevated 3 to 20 fold in bone marrow of JAK2V617F mice. Careful analysis suggested the accumulation involved multiple HSC subsets, but particularly those characterized as CD150(HI) CD86(-) CD18(L)°CD41(+) and excluding Hoechst dye. Real-Time PCR analysis of their HSC revealed that the erythropoiesis associated gene transcripts Gata1, Klf1 and Epor were particularly high. Flow cytometry analyses based on two differentiation schemes for multipotent progenitors (MPP) also suggested alteration by JAK2 signals. The low CD86 on HSC and multipotent progenitors paralleled the large reductions we found in lymphoid progenitors, but the few that were produced functioned normally when sorted and placed in culture. Either of two HSC subsets conferred disease when transplanted. Thus, flow cytometry can be used to observe the influence of abnormal JAK2 signaling on stem and progenitor subsets. Markers that similarly distinguish categories of human HSCs might be very valuable for monitoring such conditions. They could also serve as indicators of HSC fitness and suitability for transplantation.


Asunto(s)
Janus Quinasa 2/metabolismo , Transducción de Señal , Células Madre/citología , Animales , Citometría de Flujo , Ratones , Ratones Transgénicos
13.
J Hematol Oncol ; 7: 25, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24646493

RESUMEN

BACKGROUND: Myeloproliferative neoplasms (MPNs) are blood malignancies manifested in increased production of red blood cells, white blood cells, and/or platelets. A major molecular lesion associated with the diseases is JAK2V617F, an activation mutation form of tyrosine kinase JAK2. Cardiovascular events represent the leading cause of morbidity and mortality associated MPNs, but the underlying mechanism is not well understood. METHODS: Previously, we generated JAK2V617F transgenic mice which displayed MPN-like phenotypes. In the present study, we further characterized these mice by analyzing the time course of MPN phenotype development and associated cardiac abnormalities. We performed detailed histochemical staining of cardiac sections. RESULTS: JAK2V617F transgenic mice developed cardiomegaly as a subsequent event of increased blood cell production during the course of MPN phenotype development. The cardiomegaly is manifested in increased ventricular wall thickness and enlarged cardiomyocytes. Trichrome and reticulin staining revealed extensive collagen fibrosis in the heart of JAK2V617F transgenic mice. Thrombosis in the coronary artery and inflammatory cell infiltration into cardiac muscle were also observed in JAK2V617F transgenic mice, and the latter event was accompanied by fibrosis. CONCLUSION: JAK2V617F-induced blood disorders have a major impact on heart function and lead to cardiac hypertrophy. JAK2V617F transgenic mice represent an excellent model system to study both hematological malignancies and cardiovascular diseases.


Asunto(s)
Cardiomegalia/enzimología , Neoplasias Hematológicas/enzimología , Janus Quinasa 2/genética , Trastornos Mieloproliferativos/enzimología , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/genética , Cardiomegalia/patología , Femenino , Fibrosis/enzimología , Fibrosis/genética , Fibrosis/patología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología
14.
PLoS One ; 8(12): e84340, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24391943

RESUMEN

Mast cells are effector cells best known for their roles in IgE-associated allergy, but they also play a protective role in defense against pathogens. These cells express high levels of proteases including chymase, tryptase and carboxypeptidase. In the present study, we identified a congenic strain of C57BL/6 mice expressing an extraordinarily high level of chymases Mcp-2 and Mcp-4 in mast cells. The overexpression was associated with variant Mcp-2 and Mcp-4 genes originated from DBA/2 mice that also expressed high levels of the two enzymes. Real time PCR analysis revealed that Mcp-2 and Mcp-4 were selectively overexpressed as tryptases, Cpa3 and several other chymases were kept at normal levels. Reporter gene assays demonstrated that single-nucleotide polymorphisms (SNPs) in the promoter region of Mcp-2 gene may be partly responsible for the increased gene transcription. Our study provides a new model system to study the function of mast cell chymases. The data also suggest that expression of chymases differs considerably in different strains of mice and the increased chymase activity may be responsible for some unique phenotypes observed in DBA/2 mice.


Asunto(s)
Quimasas/metabolismo , Mastocitos/enzimología , Ratones Congénicos/metabolismo , Modelos Animales , Animales , Western Blotting , Clonación Molecular , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , Proteómica , Reacción en Cadena en Tiempo Real de la Polimerasa , Serina Endopeptidasas/metabolismo
15.
Exp Hematol Oncol ; 1(1): 15, 2012 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23210734

RESUMEN

BACKGROUND: JAK2V617F, a gain-of-function mutant form of tyrosine kinase JAK2, is found in the majority of patients with Ph- myeloproliferative neoplasms (MPNs), a group of chronic hematological diseases that often lead to acute leukemia. The current study is intended to find other gene mutations that collaborate with JAK2V617F to cause leukemic transformation. METHODS: Total RNA and genomic DNA were isolated from two JAK2V617F-positive cell lines, namely, erythroleukemic HEL and megakaryoblastic leukemic SET-2 cells. Candidate genes were amplified by PCR and further sequenced. RESULTS: Homozygous mutations of the TP53 gene which encodes tumor suppressor p53 were found in HEL and SET-2 cells. While HEL cells, which have homozygous JAK2V617F, contain a rare M133K p53 mutation, SET-2 cells, which have a heterozygous JAK2V617F mutation, contain a common R248W p53 alteration. Western blot analyses revealed high levels of p53 expression in both cells. M133K and R248W are located in the DNA binding domain of p53. Structural analyses revealed that they potentially disrupt the interaction of p53 with DNA, thereby causing loss of p53 function. CONCLUSIONS: JAK2V617F and p53 mutations coexist in leukemia cells. We believe that JAK2V617F is able to drive leukemic transformation when the function of tumor suppressor p53 is lost. The interplay of JAK2V617F with p53 may affect the progression of MPNs.

16.
BMC Cell Biol ; 13: 28, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23114011

RESUMEN

BACKGROUND: Autophagy is a ubiquitous cellular process responsible for the bulk degradation of cytoplasmic components through the autophagosomal-lysosomal pathway. In skeletal muscle, autophagy has been regarded as a key regulator for muscle mass maintenance, and its imbalance leads to sarcopenia. However, the underlying mechanism is poorly understood. RESULTS: In this study, we demonstrate that ceMTM3, a FYVE-domain containing myotubalarin family phosphatase, is required for the maintenance of muscle fibers by preventing excessive autophagy in Caenorhabditis elegans. Knockdown of ceMTM3 by using feeding-based RNA interference caused loss of muscle fibers accompanied by shortening of muscle cell and body size in aged C. elegans worms. This was preceded by the occurrence of excessive autophagy in the muscle and other tissues, which subsequently resulted in increased lysosomal activity and necrotic cell death. However, knockdown of ceMTM3 did not aggravate the abnormalities of muscle wasting in autophagy-deficient atg-18 mutant worms. CONCLUSIONS: Our data suggest an important role of ceMTM3 in regulating autophagy and maintaining muscle fibers. This study may have clinical implications for prevention and treatment of sarcopenia.


Asunto(s)
Autofagia , Proteínas de Caenorhabditis elegans/metabolismo , Músculos/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Animales , Tamaño Corporal , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación , Proteínas Tirosina Fosfatasas no Receptoras/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas no Receptoras/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA