Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Cell ; 184(5): 1214-1231.e16, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33636133

RESUMO

Although enteric helminth infections modulate immunity to mucosal pathogens, their effects on systemic microbes remain less established. Here, we observe increased mortality in mice coinfected with the enteric helminth Heligmosomoides polygyrus bakeri (Hpb) and West Nile virus (WNV). This enhanced susceptibility is associated with altered gut morphology and transit, translocation of commensal bacteria, impaired WNV-specific T cell responses, and increased virus infection in the gastrointestinal tract and central nervous system. These outcomes were due to type 2 immune skewing, because coinfection in Stat6-/- mice rescues mortality, treatment of helminth-free WNV-infected mice with interleukin (IL)-4 mirrors coinfection, and IL-4 receptor signaling in intestinal epithelial cells mediates the susceptibility phenotypes. Moreover, tuft cell-deficient mice show improved outcomes with coinfection, whereas treatment of helminth-free mice with tuft cell-derived cytokine IL-25 or ligand succinate worsens WNV disease. Thus, helminth activation of tuft cell-IL-4-receptor circuits in the gut exacerbates infection and disease of a neurotropic flavivirus.


Assuntos
Coinfecção , Nematospiroides dubius/fisiologia , Transdução de Sinais , Infecções por Strongylida/patologia , Vírus do Nilo Ocidental/fisiologia , Animais , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Mucosa Intestinal/parasitologia , Mucosa Intestinal/virologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/parasitologia , Neurônios/virologia , Receptores de Interleucina-4/metabolismo , Fator de Transcrição STAT6/genética , Índice de Gravidade de Doença , Infecções por Strongylida/parasitologia
2.
Cell ; 175(5): 1198-1212.e12, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30293866

RESUMO

Although chronic gastrointestinal dysmotility syndromes are a common worldwide health problem, underlying causes for these disorders are poorly understood. We show that flavivirus infection of enteric neurons leads to acute neuronal injury and cell death, inflammation, bowel dilation, and slowing of intestinal transit in mice. Flavivirus-primed CD8+ T cells promote these phenotypes, as their absence diminished enteric neuron injury and intestinal transit delays, and their adoptive transfer reestablished dysmotility after flavivirus infection. Remarkably, mice surviving acute flavivirus infection developed chronic gastrointestinal dysmotility that was exacerbated by immunization with an unrelated alphavirus vaccine or exposure to a non-infectious inflammatory stimulus. This model of chronic post-infectious gastrointestinal dysmotility in mice suggests that viral infections with tropism for enteric neurons and the ensuing immune response might contribute to the development of bowel motility disorders in humans. These results suggest an opportunity for unique approaches to diagnosis and therapy of gastrointestinal dysmotility syndromes.


Assuntos
Infecções por Flavivirus/patologia , Flavivirus/patogenicidade , Motilidade Gastrointestinal , Intestinos/patologia , Animais , Linfócitos T CD8-Positivos/imunologia , Flavivirus/genética , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/virologia , Intestinos/virologia , Leucócitos/citologia , Leucócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/patologia , Neurônios/ultraestrutura , RNA Viral/isolamento & purificação , RNA Viral/metabolismo , Síndrome
3.
Cell ; 157(6): 1279-1291, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24906147

RESUMO

Exercise training benefits many organ systems and offers protection against metabolic disorders such as obesity and diabetes. Using the recently identified isoform of PGC1-α (PGC1-α4) as a discovery tool, we report the identification of meteorin-like (Metrnl), a circulating factor that is induced in muscle after exercise and in adipose tissue upon cold exposure. Increasing circulating levels of Metrnl stimulates energy expenditure and improves glucose tolerance and the expression of genes associated with beige fat thermogenesis and anti-inflammatory cytokines. Metrnl stimulates an eosinophil-dependent increase in IL-4 expression and promotes alternative activation of adipose tissue macrophages, which are required for the increased expression of the thermogenic and anti-inflammatory gene programs in fat. Importantly, blocking Metrnl actions in vivo significantly attenuates chronic cold-exposure-induced alternative macrophage activation and thermogenic gene responses. Thus, Metrnl links host-adaptive responses to the regulation of energy homeostasis and tissue inflammation and has therapeutic potential for metabolic and inflammatory diseases.


Assuntos
Tecido Adiposo Marrom/metabolismo , Fatores de Crescimento Neural/metabolismo , Animais , Glucose/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Fígado/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Fatores de Crescimento Neural/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Termogênese , Fatores de Transcrição/genética
4.
Cell ; 155(7): 1624-38, 2013 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-24360282

RESUMO

Ever since eukaryotes subsumed the bacterial ancestor of mitochondria, the nuclear and mitochondrial genomes have had to closely coordinate their activities, as each encode different subunits of the oxidative phosphorylation (OXPHOS) system. Mitochondrial dysfunction is a hallmark of aging, but its causes are debated. We show that, during aging, there is a specific loss of mitochondrial, but not nuclear, encoded OXPHOS subunits. We trace the cause to an alternate PGC-1α/ß-independent pathway of nuclear-mitochondrial communication that is induced by a decline in nuclear NAD(+) and the accumulation of HIF-1α under normoxic conditions, with parallels to Warburg reprogramming. Deleting SIRT1 accelerates this process, whereas raising NAD(+) levels in old mice restores mitochondrial function to that of a young mouse in a SIRT1-dependent manner. Thus, a pseudohypoxic state that disrupts PGC-1α/ß-independent nuclear-mitochondrial communication contributes to the decline in mitochondrial function with age, a process that is apparently reversible.


Assuntos
Envelhecimento/patologia , Núcleo Celular/metabolismo , Mitocôndrias/metabolismo , NAD/metabolismo , Fosforilação Oxidativa , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 1/metabolismo , Fatores de Transcrição/metabolismo
5.
Immunity ; 48(3): 487-499.e5, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29525521

RESUMO

Although interferon-induced proteins with tetratricopeptide repeats (IFIT proteins) inhibit infection of many viruses by recognizing their RNA, the regulatory mechanisms involved remain unclear. Here we report a crystal structure of cap 0 (m7GpppN) RNA bound to human IFIT1 in complex with the C-terminal domain of human IFIT3. Structural, biochemical, and genetic studies suggest that IFIT3 binding to IFIT1 has dual regulatory functions: (1) extending the half-life of IFIT1 and thereby increasing its steady-state amounts in cells; and (2) allosterically regulating the IFIT1 RNA-binding channel, thereby enhancing the specificity of recognition for cap 0 but not cap 1 (m7GpppNm) or 5'-ppp RNA. Mouse Ifit3 lacks this key C-terminal domain and does not bind mouse Ifit1. The IFIT3 interaction with IFIT1 is important for restricting infection of viruses lacking 2'-O methylation in their RNA cap structures. Our experiments establish differences in the regulation of IFIT1 orthologs and define targets for modulation of human IFIT protein activity.


Assuntos
Proteínas de Transporte/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Metilação , Camundongos , Modelos Moleculares , Conformação Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , RNA/química , RNA/genética , RNA/metabolismo , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Especificidade da Espécie , Relação Estrutura-Atividade
6.
Cell ; 151(6): 1319-31, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23217713

RESUMO

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.


Assuntos
Proteínas de Choque Térmico/metabolismo , Músculo Esquelético/metabolismo , Condicionamento Físico Animal , Treinamento Resistido , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Adiposidade , Animais , Glucose/metabolismo , Humanos , Hipertrofia , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/metabolismo , Miostatina/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Isoformas de Proteínas/metabolismo
7.
Nature ; 535(7610): 164-8, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27383988

RESUMO

Flaviviruses infect hundreds of millions of people annually, and no antiviral therapy is available. We performed a genome-wide CRISPR/Cas9-based screen to identify host genes that, when edited, resulted in reduced flavivirus infection. Here, we validated nine human genes required for flavivirus infectivity, and these were associated with endoplasmic reticulum functions including translocation, protein degradation, and N-linked glycosylation. In particular, a subset of endoplasmic reticulum-associated signal peptidase complex (SPCS) proteins was necessary for proper cleavage of the flavivirus structural proteins (prM and E) and secretion of viral particles. Loss of SPCS1 expression resulted in markedly reduced yield of all Flaviviridae family members tested (West Nile, Dengue, Zika, yellow fever, Japanese encephalitis, and hepatitis C viruses), but had little impact on alphavirus, bunyavirus, or rhabdovirus infection or the surface expression or secretion of diverse host proteins. We found that SPCS1 dependence could be bypassed by replacing the native prM protein leader sequences with a class I major histocompatibility complex (MHC) antigen leader sequence. Thus, SPCS1, either directly or indirectly via its interactions with unknown host proteins, preferentially promotes the processing of specific protein cargo, and Flaviviridae have a unique dependence on this signal peptide processing pathway. SPCS1 and other signal processing pathway members could represent pharmacological targets for inhibiting infection by the expanding number of flaviviruses of medical concern.


Assuntos
Sistemas CRISPR-Cas/genética , Flavivirus/fisiologia , Genoma Humano/genética , Fatores Celulares Derivados do Hospedeiro/genética , Sinais Direcionadores de Proteínas/fisiologia , Animais , Linhagem Celular , Drosophila/citologia , Drosophila/genética , Drosophila/virologia , Descoberta de Drogas , Retículo Endoplasmático/metabolismo , Feminino , Flavivirus/metabolismo , Infecções por Flavivirus/genética , Infecções por Flavivirus/virologia , Glicosilação , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas de Membrana/genética , Terapia de Alvo Molecular , Transporte Proteico/genética , Proteólise , Reprodutibilidade dos Testes , Serina Endopeptidases/genética , Especificidade da Espécie , Proteínas Virais/química , Proteínas Virais/metabolismo , Proteínas Estruturais Virais/metabolismo
8.
Proc Natl Acad Sci U S A ; 115(3): 561-566, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29295932

RESUMO

The peroxisome-proliferator receptor-γ (PPARγ) is expressed in multiple cancer types. Recently, our group has shown that PPARγ is phosphorylated on serine 273 (S273), which selectively modulates the transcriptional program controlled by this protein. PPARγ ligands, including thiazolidinediones (TZDs), block S273 phosphorylation. This activity is chemically separable from the canonical activation of the receptor by agonist ligands and, importantly, these noncanonical agonist ligands do not cause some of the known side effects of TZDs. Here, we show that phosphorylation of S273 of PPARγ occurs in cancer cells on exposure to DNA damaging agents. Blocking this phosphorylation genetically or pharmacologically increases accumulation of DNA damage, resulting in apoptotic cell death. A genetic signature of PPARγ phosphorylation is associated with worse outcomes in response to chemotherapy in human patients. Noncanonical agonist ligands sensitize lung cancer xenografts and genetically induced lung tumors to carboplatin therapy. Moreover, inhibition of this phosphorylation results in deregulation of p53 signaling, and biochemical studies show that PPARγ physically interacts with p53 in a manner dependent on S273 phosphorylation. These data implicate a role for PPARγ in modifying the p53 response to cytotoxic therapy, which can be modulated for therapeutic gain using these compounds.


Assuntos
Antineoplásicos/administração & dosagem , Dano ao DNA , Neoplasias Pulmonares/tratamento farmacológico , PPAR gama/metabolismo , Tiazolidinedionas/administração & dosagem , Motivos de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Carboplatina/administração & dosagem , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Humanos , Ligantes , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Nus , PPAR gama/agonistas , PPAR gama/química , PPAR gama/genética , Fosforilação , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
J Infect Dis ; 221(9): 1506-1517, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-31616920

RESUMO

BACKGROUND: Sexual transmission and persistence of Zika virus (ZIKV) in the male reproductive tract has raised concerned for potential damaging effects on function. Animal studies have demonstrated that ZIKV virus can infect and damage the testis and epididymis, and these results has been correlated to lower sperm counts in ZIKV-infected humans. The prostate plays a vital role in the male reproductive tract, with acute and chronic prostatitis linked to male infertility. METHODS: In this study, we evaluated the effects of ZIKV virus on the prostate in mice and nonhuman primates. RESULTS: In mice, ZIKV infected the prostate and triggered inflammation that persisted even after virus clearance. Evidence of chronic prostatitis associated with ZIKV infection remained for several months. Similar histological findings were observed in the prostate of ZIKV-infected rhesus macaques. CONCLUSIONS: These studies establish that ZIKV replicates in the prostate and can cause acute and chronic inflammatory and proliferative changes in mouse and nonhuman primate models.


Assuntos
Prostatite/virologia , Testículo/virologia , Infecção por Zika virus/complicações , Doença Aguda , Animais , Doença Crônica , Modelos Animais de Doenças , Epididimo , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Prostatite/patologia , Sêmen/virologia , Testículo/patologia , Zika virus , Infecção por Zika virus/transmissão
10.
Nature ; 513(7516): 100-4, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25043053

RESUMO

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.


Assuntos
Tecido Adiposo Marrom/metabolismo , Caquexia/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/patologia , Animais , Caquexia/patologia , Carcinoma Pulmonar de Lewis/genética , Meios de Cultivo Condicionados/farmacologia , Metabolismo Energético/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Tamanho do Órgão/efeitos dos fármacos , Proteína Relacionada ao Hormônio Paratireóideo/antagonistas & inibidores , Termogênese/efeitos dos fármacos , Termogênese/genética
11.
J Virol ; 90(23): 10499-10512, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27630236

RESUMO

While natural hepatitis C virus (HCV) infection results in highly diverse quasispecies of related viruses over time, mutations accumulate more slowly in tissue culture, in part because of the inefficiency of replication in cells. To create a highly diverse population of HCV particles in cell culture and identify novel growth-enhancing mutations, we engineered a library of infectious HCV with all codons represented at most positions in the ectodomain of the E2 gene. We identified many putative growth-adaptive mutations and selected nine highly represented E2 mutants for further study: Q412R, T416R, S449P, T563V, A579R, L619T, V626S, K632T, and L644I. We evaluated these mutants for changes in particle-to-infectious-unit ratio, sensitivity to neutralizing antibody or CD81 large extracellular loop (CD81-LEL) inhibition, entry factor usage, and buoyant density profiles. Q412R, T416R, S449P, T563V, and L619T were neutralized more efficiently by anti-E2 antibodies and T416R, T563V, and L619T by CD81-LEL. Remarkably, all nine variants showed reduced dependence on scavenger receptor class B type I (SR-BI) for infection. This shift from SR-BI usage did not correlate with a change in the buoyant density profiles of the variants, suggesting an altered E2-SR-BI interaction rather than changes in the virus-associated lipoprotein-E2 interaction. Our results demonstrate that residues influencing SR-BI usage are distributed across E2 and support the development of large-scale mutagenesis studies to identify viral variants with unique functional properties. IMPORTANCE: Characterizing variant viruses can reveal new information about the life cycle of HCV and the roles played by different viral genes. However, it is difficult to recapitulate high levels of diversity in the laboratory because of limitations in the HCV culture system. To overcome this limitation, we engineered a library of mutations into the E2 gene in the context of an infectious clone of the virus. We used this library of viruses to identify nine mutations that enhance the growth rate of HCV. These growth-enhancing mutations reduced the dependence on a key entry receptor, SR-BI. By generating a highly diverse library of infectious HCV, we mapped regions of the E2 protein that influence a key virus-host interaction and provide proof of principle for the generation of large-scale mutant libraries for the study of pathogens with great sequence variability.


Assuntos
Hepacivirus/genética , Mutação , Receptores Depuradores Classe B/fisiologia , Proteínas do Envelope Viral/genética , Adaptação Fisiológica/genética , Substituição de Aminoácidos , Anticorpos Antivirais , Sistemas CRISPR-Cas , Linhagem Celular , Biblioteca Gênica , Engenharia Genética , Genoma Viral , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/fisiologia , Hepatite C/imunologia , Hepatite C/prevenção & controle , Hepatite C/virologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Tetraspanina 28 , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia
12.
PLoS Pathog ; 11(4): e1004863, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25927359

RESUMO

Alphaviruses are a group of widely distributed human and animal pathogens. It is well established that their replication is sensitive to type I IFN treatment, but the mechanism of IFN inhibitory function remains poorly understood. Using a new experimental system, we demonstrate that in the presence of IFN-ß, activation of interferon-stimulated genes (ISGs) does not interfere with either attachment of alphavirus virions to the cells, or their entry and nucleocapsid disassembly. However, it strongly affects translation of the virion-delivered virus-specific RNAs. One of the ISG products, IFIT1 protein, plays a major role in this translation block, although an IFIT1-independent mechanism is also involved. The 5'UTRs of the alphavirus genomes were found to differ significantly in their ability to drive translation in the presence of increased concentration of IFIT1. Prior studies have shown that adaptation of naturally circulating alphaviruses to replication in tissue culture results in accumulation of mutations in the 5'UTR, which increase the efficiency of the promoter located in the 5'end of the genome. Here, we show that these mutations also decrease resistance of viral RNA to IFIT1-induced translation inhibition. In the presence of higher levels of IFIT1, alphaviruses with wt 5'UTRs became potent inducers of type I IFN, suggesting a new mechanism of type I IFN induction. We applied this knowledge of IFIT1 interaction with alphaviruses to develop new attenuated variants of Venezuelan equine encephalitis and chikungunya viruses that are more sensitive to the antiviral effects of IFIT1, and thus could serve as novel vaccine candidates.


Assuntos
Alphavirus/fisiologia , Proteínas de Transporte/metabolismo , Regulação Viral da Expressão Gênica , Genoma Viral , Interações Hospedeiro-Patógeno , Interferon Tipo I/agonistas , Replicação Viral , Regiões 5' não Traduzidas , Proteínas Adaptadoras de Transdução de Sinal , Aedes , Alphavirus/genética , Alphavirus/imunologia , Animais , Linhagem Celular , Células Cultivadas , Vírus Chikungunya/genética , Vírus Chikungunya/imunologia , Vírus Chikungunya/fisiologia , Regulação para Baixo , Vírus da Encefalite Equina Venezuelana/genética , Vírus da Encefalite Equina Venezuelana/imunologia , Vírus da Encefalite Equina Venezuelana/fisiologia , Vacinas Fúngicas/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/imunologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/virologia , Mutação , Células NIH 3T3 , RNA/metabolismo , Proteínas de Ligação a RNA , Tropismo Viral
13.
Proc Natl Acad Sci U S A ; 111(44): 15756-61, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25336758

RESUMO

Peroxisome proliferator-activated receptor gamma coactivator 1-alpha 4 (PGC-1α4) is a protein isoform derived by alternative splicing of the PGC1α mRNA and has been shown to promote muscle hypertrophy. We show here that G protein-coupled receptor 56 (GPR56) is a transcriptional target of PGC-1α4 and is induced in humans by resistance exercise. Furthermore, the anabolic effects of PGC-1α4 in cultured murine muscle cells are dependent on GPR56 signaling, because knockdown of GPR56 attenuates PGC-1α4-induced muscle hypertrophy in vitro. Forced expression of GPR56 results in myotube hypertrophy through the expression of insulin-like growth factor 1, which is dependent on Gα12/13 signaling. A murine model of overload-induced muscle hypertrophy is associated with increased expression of both GPR56 and its ligand collagen type III, whereas genetic ablation of GPR56 expression attenuates overload-induced muscle hypertrophy and associated anabolic signaling. These data illustrate a signaling pathway through GPR56 which regulates muscle hypertrophy associated with resistance/loading-type exercise.


Assuntos
Regulação da Expressão Gênica/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Condicionamento Físico Animal , Receptores Acoplados a Proteínas G/biossíntese , Transdução de Sinais/fisiologia , Animais , Colágeno Tipo III/biossíntese , Hipertrofia/metabolismo , Fator de Crescimento Insulin-Like I/biossíntese , Masculino , Camundongos , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/metabolismo
14.
J Virol ; 89(18): 9465-76, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26157117

RESUMO

UNLABELLED: Interferon-induced protein with tetratricopeptide repeats 1 (IFIT1) is a host protein with reported cell-intrinsic antiviral activity against several RNA viruses. The proposed basis for the activity against negative-sense RNA viruses is the binding to exposed 5'-triphosphates (5'-ppp) on the genome of viral RNA. However, recent studies reported relatively low binding affinities of IFIT1 for 5'-ppp RNA, suggesting that IFIT1 may not interact efficiently with this moiety under physiological conditions. To evaluate the ability of IFIT1 to have an impact on negative-sense RNA viruses, we infected Ifit1(-/-) and wild-type control mice and primary cells with four negative-sense RNA viruses (influenza A virus [IAV], La Crosse virus [LACV], Oropouche virus [OROV], and Ebola virus) corresponding to three distinct families. Unexpectedly, a lack of Ifit1 gene expression did not result in increased infection by any of these viruses in cell culture. Analogously, morbidity, mortality, and viral burdens in tissues were identical between Ifit1(-/-) and control mice after infection with IAV, LACV, or OROV. Finally, deletion of the human IFIT1 protein in A549 cells did not affect IAV replication or infection, and reciprocally, ectopic expression of IFIT1 in HEK293T cells did not inhibit IAV infection. To explain the lack of antiviral activity against IAV, we measured the binding affinity of IFIT1 for RNA oligonucleotides resembling the 5' ends of IAV gene segments. The affinity for 5'-ppp RNA was approximately 10-fold lower than that for non-2'-O-methylated (cap 0) RNA oligonucleotides. Based on this analysis, we conclude that IFIT1 is not a dominant restriction factor against negative-sense RNA viruses. IMPORTANCE: Negative-sense RNA viruses, including influenza virus and Ebola virus, have been responsible for some of the most deadly outbreaks in recent history. The host interferon response and induction of antiviral genes contribute to the control of infections by these viruses. IFIT1 is highly induced after virus infection and reportedly has antiviral activity against several RNA and DNA viruses. However, its role in restricting infection by negative-sense RNA viruses remains unclear. In this study, we evaluated the ability of IFIT1 to inhibit negative-sense RNA virus replication and pathogenesis both in vitro and in vivo. Detailed cell culture and animal studies demonstrated that IFIT1 is not a dominant restriction factor against three different families of negative-sense RNA viruses.


Assuntos
Proteínas de Transporte/metabolismo , Ebolavirus/metabolismo , Vírus da Influenza A/metabolismo , Vírus La Crosse/metabolismo , Infecções por Vírus de RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Linhagem Celular , Ebolavirus/genética , Humanos , Vírus da Influenza A/genética , Vírus La Crosse/genética , Camundongos , Camundongos Knockout , Infecções por Vírus de RNA/genética , Proteínas de Ligação a RNA
15.
Handb Exp Pharmacol ; 234: 299-308, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27832493

RESUMO

Skeletal muscle homeostasis is regulated by a constant influx of chemicals and exposure to mechanical stimuli. A number of key signaling pathways that translate these stimuli into changes in muscle physiology have been established. The GPCR family known as adhesion GPCRs (aGPCRs) has largely elusive roles in skeletal muscle biology; however, their unique capacity to activate adhesion and G protein signaling pathways makes them an attractive point of investigation. The skeletal muscle myofiber contains a highly organized cytoarchitecture to ensure contractile function. This requires intricate interactions with components of the extracellular matrix (ECM) surrounding each fiber. aGPCRs possess extended N-termini known to interact with ECM proteins and complexes suggesting a compatible role in skeletal muscle biology. Furthermore, recent work demonstrated the involvement of certain aGPCRs in whole muscle hypertrophy and differentiation of muscle progenitor cells. Signaling pathways downstream of aGPCRs are still incompletely understood; however, initial findings show involvement of the Gα12/13 subunit signaling to the pro-anabolic Akt/mTOR pathway. Together, this chapter will review the emerging role of aGPCRs in skeletal muscle biology and putative mechanism(s) employed to regulate skeletal muscle growth.


Assuntos
Adesão Celular , Membrana Celular/metabolismo , Músculo Esquelético/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Sítios de Ligação , Matriz Extracelular/metabolismo , Humanos , Mecanotransdução Celular , Modelos Moleculares , Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores Acoplados a Proteínas G/química , Estresse Mecânico , Relação Estrutura-Atividade
16.
Cell Rep ; 43(3): 113881, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38442019

RESUMO

An intriguing effect of short-term caloric restriction (CR) is the expansion of certain stem cell populations, including muscle stem cells (satellite cells), which facilitate an accelerated regenerative program after injury. Here, we utilized the MetRSL274G (MetRS) transgenic mouse to identify liver-secreted plasminogen as a candidate for regulating satellite cell expansion during short-term CR. Knockdown of circulating plasminogen prevents satellite cell expansion during short-term CR. Furthermore, loss of the plasminogen receptor KT (Plg-RKT) is also sufficient to prevent CR-related satellite cell expansion, consistent with direct signaling of plasminogen through the plasminogen receptor Plg-RKT/ERK kinase to promote proliferation of satellite cells. Importantly, we are able to replicate many of these findings in human participants from the CALERIE trial. Our results demonstrate that CR enhances liver protein secretion of plasminogen, which signals directly to the muscle satellite cell through Plg-RKT to promote proliferation and subsequent muscle resilience during CR.


Assuntos
Plasminogênio , Receptores de Superfície Celular , Camundongos , Animais , Humanos , Plasminogênio/metabolismo , Receptores de Superfície Celular/metabolismo , Restrição Calórica , Fígado/metabolismo , Camundongos Transgênicos , Serina Proteases , Proliferação de Células , Músculos/metabolismo
17.
Am J Physiol Endocrinol Metab ; 304(10): E1042-52, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23531613

RESUMO

Although catabolic signaling has a well-established role in muscle wasting during cancer cachexia, the suppression of anabolic signaling also warrants further investigation. In cachectic tumor-bearing mice, circulating IL-6 levels are associated with suppressed muscle protein synthesis and mTORC1 signaling. We have found AMPK and IGF-I/insulin signaling, two well-known regulators of the mammalian target of rapamycin (mTOR), are altered with the progression of cachexia. How IL-6 can induce suppression of mTORC1 signaling remains to be established. The purpose of this study was to examine mTOR complex 1 (mTORC1) activation and regulation by IL-6 during cancer cachexia. IL-6 effects on mTOR activation were examined in Apc(Min/+) mouse skeletal muscle and C2C12 myotubes. Systemic IL-6 overexpression in Apc(Min/+) mice produced a dose-dependent suppression of mTOR signaling that corresponded to induction of STAT3 and AMPK phosphorylation. This result was also evident in IL-6-treated myotubes. Basal mTOR activation and mTOR responsiveness to glucose administration were suppressed in cachectic skeletal muscle. However, insulin induction of mTOR activity was maintained in IL-6-treated myotubes. Whereas IL-6 suppression of myotube mTOR activity was rescued by AMPK inhibition, inhibition of STAT3 signaling was not sufficient to rescue IL-6 suppression of mTOR activity. Last, treadmill exercise training was able to prevent IL-6-induced inhibition of mTOR signaling in Apc(Min/+) mice independently of activated STAT. In conclusion, we report dose-dependent suppression of mTOR activity by IL-6 and suppressed mTOR responsiveness to glucose administration in Apc(Min/+) mice. IL-6 suppression of mTOR activity was dependent on AMPK activation and independent of STAT signaling in myotubes.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Caquexia/metabolismo , Interleucina-6/metabolismo , Músculo Esquelético/metabolismo , Neoplasias Experimentais/metabolismo , Proteínas/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/genética , Aminoimidazol Carboxamida/farmacologia , Animais , Western Blotting , Caquexia/enzimologia , Interleucina-6/sangue , Interleucina-6/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/enzimologia , Neoplasias Experimentais/enzimologia , Fosforilação , Condicionamento Físico Animal/fisiologia , Proteínas/antagonistas & inibidores , Proteínas/genética , Pirazóis/farmacologia , Pirimidinas/farmacologia , RNA Mensageiro/química , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Recombinantes/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR
18.
Nat Aging ; 3(8): 948-964, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37500973

RESUMO

Heterochronic parabiosis (HPB) is known for its functional rejuvenation effects across several mouse tissues. However, its impact on biological age and long-term health is unknown. Here we performed extended (3-month) HPB, followed by a 2-month detachment period of anastomosed pairs. Old detached mice exhibited improved physiological parameters and lived longer than control isochronic mice. HPB drastically reduced the epigenetic age of blood and liver based on several clock models using two independent platforms. Remarkably, this rejuvenation effect persisted even after 2 months of detachment. Transcriptomic and epigenomic profiles of anastomosed mice showed an intermediate phenotype between old and young, suggesting a global multi-omic rejuvenation effect. In addition, old HPB mice showed gene expression changes opposite to aging but akin to several life span-extending interventions. Altogether, we reveal that long-term HPB results in lasting epigenetic and transcriptome remodeling, culminating in the extension of life span and health span.


Assuntos
Longevidade , Rejuvenescimento , Camundongos , Animais , Longevidade/genética , Multiômica , Envelhecimento/genética
19.
Cell Metab ; 35(5): 807-820.e5, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-37086720

RESUMO

Aging is classically conceptualized as an ever-increasing trajectory of damage accumulation and loss of function, leading to increases in morbidity and mortality. However, recent in vitro studies have raised the possibility of age reversal. Here, we report that biological age is fluid and exhibits rapid changes in both directions. At epigenetic, transcriptomic, and metabolomic levels, we find that the biological age of young mice is increased by heterochronic parabiosis and restored following surgical detachment. We also identify transient changes in biological age during major surgery, pregnancy, and severe COVID-19 in humans and/or mice. Together, these data show that biological age undergoes a rapid increase in response to diverse forms of stress, which is reversed following recovery from stress. Our study uncovers a new layer of aging dynamics that should be considered in future studies. The elevation of biological age by stress may be a quantifiable and actionable target for future interventions.


Assuntos
COVID-19 , Humanos , Animais , Camundongos , Envelhecimento/fisiologia , Parabiose
20.
Biochim Biophys Acta ; 1812(12): 1601-6, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21914473

RESUMO

BACKGROUND: The Apc(Min/+) mouse, an animal model of colorectal cancer and cachexia, has a heterologous mutation in the Apc tumor suppressor gene, predisposing the mouse to intestinal and colon tumor development. This mouse develops intestinal polyps by ~4 weeks of age, and loses body weight gradually between ~14 and ~20 weeks of age. The strengths of this cachexia model derive from several features that mimic human cancer, including a gradual increase in tumor burden, chronic inflammation, and anemia. Little is known about the role of gut barrier dysfunction and endotoxemia in the development of cancer cachexia. We sought to determine how gut permeability and resultant endotoxemia change with the progression of cachexia. METHODS: Intestinal gut barrier integrity was assessed by permeability to FITC-dextran (MW(av)=4000kDa; FD4). Plasma glucose and triglycerides were measured by enzymatic assays, IL-6 by enzyme-linked immunosorbent assay, and endotoxin by the limulus amoebocyte assay. Body temperature was measured using a rectal probe. RESULTS: Progression of cachexia was accompanied by development of gut barrier dysfunction (permeability to FD4), hypertrophy of mesenteric lymph nodes, and an increase in plasma endotoxin concentration. Changes in blood glucose and glucose tolerance, plasma IL-6, triglycerides, and body temperature were characteristic of endotoxemia. CONCLUSION: We propose a role for gut barrier dysfunction (GBD) and subsequent endotoxemia in the development of inflammation and progression of cachexia in the Apc(Min/+) mouse.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Caquexia/etiologia , Neoplasias do Colo/complicações , Trato Gastrointestinal/metabolismo , Animais , Caquexia/metabolismo , Caquexia/fisiopatologia , Modelos Animais de Doenças , Endotoxemia/etiologia , Endotoxinas/sangue , Humanos , Hiperlipidemias/etiologia , Hipotermia/etiologia , Resistência à Insulina , Interleucina-6/sangue , Linfonodos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Permeabilidade , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA