Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Immunol Res ; 12(6): 719-730, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38558120

RESUMO

Small-cell lung cancer (SCLC) is an aggressive cancer for which immune checkpoint inhibitors (ICI) have had only limited success. Bispecific T-cell engagers are promising therapeutic alternatives for ICI-resistant tumors, but not all patients with SCLC are responsive. Herein, to integrate CD137 costimulatory function into a T-cell engager format and thereby augment therapeutic efficacy, we generated a CD3/CD137 dual-specific Fab and engineered a DLL3-targeted trispecific antibody (DLL3 trispecific). The CD3/CD137 dual-specific Fab was generated to competitively bind to CD3 and CD137 to prevent DLL3-independent cross-linking of CD3 and CD137, which could lead to systemic T-cell activation. We demonstrated that DLL3 trispecific induced better tumor growth control and a marked increase in the number of intratumoral T cells compared with a conventional DLL3-targeted bispecific T-cell engager. These findings suggest that DLL3 trispecific can exert potent efficacy by inducing concurrent CD137 costimulation and provide a promising therapeutic option for SCLC.


Assuntos
Complexo CD3 , Peptídeos e Proteínas de Sinalização Intracelular , Neoplasias Pulmonares , Proteínas de Membrana , Carcinoma de Pequenas Células do Pulmão , Linfócitos T , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral , Carcinoma de Pequenas Células do Pulmão/imunologia , Carcinoma de Pequenas Células do Pulmão/patologia , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/terapia , Carcinoma de Pequenas Células do Pulmão/metabolismo , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/tratamento farmacológico , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Complexo CD3/imunologia , Animais , Camundongos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/imunologia , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Linhagem Celular Tumoral , Ativação Linfocitária/imunologia , Feminino , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Blood Adv ; 5(9): 2305-2318, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33929501

RESUMO

Acute myeloid leukemia (AML) is a heterogenous malignancy characterized by distinct lineage subtypes and various genetic/epigenetic alterations. As with other neoplasms, AML cells have well-known aerobic glycolysis, but metabolic variations depending on cellular lineages also exist. Lysine-specific demethylase-1 (LSD1) has been reported to be crucial for human leukemogenesis, which is currently one of the emerging therapeutic targets. However, metabolic roles of LSD1 and lineage-dependent factors remain to be elucidated in AML cells. Here, we show that LSD1 directs a hematopoietic lineage-specific metabolic program in AML subtypes. Erythroid leukemia (EL) cells particularly showed activated glycolysis and high expression of LSD1 in both AML cell lines and clinical samples. Transcriptome, chromatin immunoprecipitation-sequencing, and metabolomic analyses revealed that LSD1 was essential not only for glycolysis but also for heme synthesis, the most characteristic metabolic pathway of erythroid origin. Notably, LSD1 stabilized the erythroid transcription factor GATA1, which directly enhanced the expression of glycolysis and heme synthesis genes. In contrast, LSD1 epigenetically downregulated the granulo-monocytic transcription factor C/EBPα. Thus, the use of LSD1 knockdown or chemical inhibitor dominated C/EBPα instead of GATA1 in EL cells, resulting in metabolic shifts and growth arrest. Furthermore, GATA1 suppressed the gene encoding C/EBPα that then acted as a repressor of GATA1 target genes. Collectively, we conclude that LSD1 shapes metabolic phenotypes in EL cells by balancing these lineage-specific transcription factors and that LSD1 inhibitors pharmacologically cause lineage-dependent metabolic remodeling.


Assuntos
Leucemia Eritroblástica Aguda , Proteína alfa Estimuladora de Ligação a CCAAT , Fator de Transcrição GATA1/genética , Histona Desmetilases/genética , Humanos , Leucemia Eritroblástica Aguda/genética , Proteínas Proto-Oncogênicas , Fatores de Transcrição
3.
Cancer Discov ; 11(1): 158-175, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32847940

RESUMO

Agonistic antibodies targeting CD137 have been clinically unsuccessful due to systemic toxicity. Because conferring tumor selectivity through tumor-associated antigen limits its clinical use to cancers that highly express such antigens, we exploited extracellular adenosine triphosphate (exATP), which is a hallmark of the tumor microenvironment and highly elevated in solid tumors, as a broadly tumor-selective switch. We generated a novel anti-CD137 switch antibody, STA551, which exerts agonistic activity only in the presence of exATP. STA551 demonstrated potent and broad antitumor efficacy against all mouse and human tumors tested and a wide therapeutic window without systemic immune activation in mice. STA551 was well tolerated even at 150 mg/kg/week in cynomolgus monkeys. These results provide a strong rationale for the clinical testing of STA551 against a broad variety of cancers regardless of antigen expression, and for the further application of this novel platform to other targets in cancer therapy. SIGNIFICANCE: Reported CD137 agonists suffer from either systemic toxicity or limited efficacy against antigen-specific cancers. STA551, an antibody designed to agonize CD137 only in the presence of extracellular ATP, inhibited tumor growth in a broad variety of cancer models without any systemic toxicity or dependence on antigen expression.See related commentary by Keenan and Fong, p. 20.This article is highlighted in the In This Issue feature, p. 1.


Assuntos
Trifosfato de Adenosina , Neoplasias , Animais , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Microambiente Tumoral , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral
4.
Cancer Sci ; 111(10): 3902-3911, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32748499

RESUMO

Central nervous system tumors are classified based on an integrated diagnosis combining histology and molecular characteristics, including IDH1/2 and H3-K27M mutations, as well as 1p/19q codeletion. Here, we aimed to develop and assess the feasibility of a glioma-tailored 48-gene next-generation sequencing (NGS) panel for integrated glioma diagnosis. We designed a glioma-tailored 48-gene NGS panel for detecting 1p/19q codeletion and mutations in IDH1/2, TP53, PTEN, PDGFRA, NF1, RB1, CDKN2A/B, CDK4, and the TERT promoter (TERTp). We analyzed 106 glioma patients (grade II: 19 cases, grade III: 23 cases, grade IV: 64 cases) using this system. The 1p/19q codeletion was detected precisely in oligodendroglial tumors using our NGS panel. In a cohort of 64 grade Ⅳ gliomas, we identified 56 IDH-wildtype glioblastomas. Within these IDH-wildtype glioblastomas, 33 samples (58.9%) showed a mutation in TERTp. Notably, PDGFRA mutations and their amplification were more commonly seen in TERTp-wildtype glioblastomas (43%) than in TERTp-mutant glioblastomas (6%) (P = .001). Hierarchical molecular classification of IDH-wildtype glioblastomas revealed 3 distinct groups of IDH-wildtype glioblastomas. One major cluster was characterized by mutations in PDGFRA, amplification of CDK4 and PDGFRA, homozygous deletion of CDKN2A/B, and absence of TERTp mutations. This cluster was significantly associated with older age (P = .021), higher Ki-67 score (P = .007), poor prognosis (P = .012), and a periventricular tumor location. We report the development of a glioma-tailored NGS panel for detecting 1p/19q codeletion and driver gene mutations on a single platform. Our panel identified distinct subtypes of IDH- and TERTp-wildtype glioblastomas with frequent PDGFRA alterations.


Assuntos
Glioblastoma/genética , Isocitrato Desidrogenase/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Telomerase/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Glioblastoma/classificação , Glioblastoma/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas/genética
5.
Cell Rep ; 29(1): 89-103.e7, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31577958

RESUMO

Tolerance to severe tumor microenvironments, including hypoxia and nutrient starvation, is a common feature of aggressive cancer cells and can be targeted. However, metabolic alterations that support cancer cells upon nutrient starvation are not well understood. Here, by comprehensive metabolome analyses, we show that glutamine deprivation leads to phosphoethanolamine (PEtn) accumulation in cancer cells via the downregulation of PEtn cytidylyltransferase (PCYT2), a rate-limiting enzyme of phosphatidylethanolamine biosynthesis. PEtn accumulation correlated with tumor growth under nutrient starvation. PCYT2 suppression was partially mediated by downregulation of the transcription factor ELF3. Furthermore, PCYT2 overexpression reduced PEtn levels and tumor growth. In addition, PEtn accumulation and PCYT2 downregulation in human breast tumors correlated with poor prognosis. Thus, we show that glutamine deprivation leads to tumor progression by regulating PE biosynthesis via the ELF3-PCYT2 axis. Furthermore, manipulating glutamine-responsive genes could be a therapeutic approach to limit cancer progression.


Assuntos
Regulação para Baixo/genética , Etanolaminas/metabolismo , Glutamina/metabolismo , RNA Nucleotidiltransferases/genética , Inanição/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Progressão da Doença , Células HeLa , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Células MCF-7 , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-ets/genética , Transcrição Gênica/genética
6.
Nature ; 561(7722): 243-247, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30185909

RESUMO

Large cutaneous ulcers are, in severe cases, life threatening1,2. As the global population ages, non-healing ulcers are becoming increasingly common1,2. Treatment currently requires the transplantation of pre-existing epithelial components, such as skin grafts, or therapy using cultured cells2. Here we develop alternative supplies of epidermal coverage for the treatment of these kinds of wounds. We generated expandable epithelial tissues using in vivo reprogramming of wound-resident mesenchymal cells. Transduction of four transcription factors that specify the skin-cell lineage enabled efficient and rapid de novo epithelialization from the surface of cutaneous ulcers in mice. Our findings may provide a new therapeutic avenue for treating skin wounds and could be extended to other disease situations in which tissue homeostasis and repair are impaired.


Assuntos
Reprogramação Celular , Células Epiteliais/citologia , Úlcera Cutânea/patologia , Pele/citologia , Ferimentos e Lesões/patologia , Animais , Linhagem da Célula , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos , Medicina Regenerativa , Pele/patologia , Úlcera Cutânea/terapia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Cicatrização , Ferimentos e Lesões/terapia
7.
Nucleic Acids Res ; 46(11): 5441-5454, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29618057

RESUMO

The metabolic properties of cells are formed under the influence of environmental factors such as nutrients and hormones. Although such a metabolic program is likely initiated through epigenetic mechanisms, the direct links between metabolic cues and activities of chromatin modifiers remain largely unknown. In this study, we show that lysine-specific demethylase-1 (LSD1) controls the metabolic program in myogenic differentiation, under the action of catabolic hormone, glucocorticoids. By using transcriptomic and epigenomic approaches, we revealed that LSD1 bound to oxidative metabolism and slow-twitch myosin genes, and repressed their expression. Consistent with this, loss of LSD1 activity during differentiation enhanced the oxidative capacity of myotubes. By testing the effects of various hormones, we found that LSD1 levels were decreased by treatment with the glucocorticoid dexamethasone (Dex) in cultured myoblasts and in skeletal muscle from mice. Mechanistically, glucocorticoid signaling induced expression of a ubiquitin E3 ligase, JADE-2, which was responsible for proteasomal degradation of LSD1. Consequently, in differentiating myoblasts, chemical inhibition of LSD1, in combination with Dex treatment, synergistically de-repressed oxidative metabolism genes, concomitant with increased histone H3 lysine 4 methylation at these loci. These findings demonstrated that LSD1 serves as an epigenetic regulator linking glucocorticoid action to metabolic programming during myogenic differentiation.


Assuntos
Dexametasona/farmacologia , Glucocorticoides/farmacologia , Histona Desmetilases/metabolismo , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Animais , Diferenciação Celular/genética , Linhagem Celular Tumoral , Histona Desmetilases/antagonistas & inibidores , Histonas/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Oxirredução , Ubiquitina-Proteína Ligases/metabolismo
8.
Sci Transl Med ; 9(410)2017 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-28978751

RESUMO

Cancer care is being revolutionized by immunotherapies such as immune checkpoint inhibitors, engineered T cell transfer, and cell vaccines. The bispecific T cell-redirecting antibody (TRAB) is one such promising immunotherapy, which can redirect T cells to tumor cells by engaging CD3 on a T cell and an antigen on a tumor cell. Because T cells can be redirected to tumor cells regardless of the specificity of T cell receptors, TRAB is considered efficacious for less immunogenic tumors lacking enough neoantigens. Its clinical efficacy has been exemplified by blinatumomab, a bispecific T cell engager targeting CD19 and CD3, which has shown marked clinical responses against hematological malignancies. However, the success of TRAB in solid tumors has been hampered by the lack of a target molecule with sufficient tumor selectivity to avoid "on-target off-tumor" toxicity. Glypican 3 (GPC3) is a highly tumor-specific antigen that is expressed during fetal development but is strictly suppressed in normal adult tissues. We developed ERY974, a whole humanized immunoglobulin G-structured TRAB harboring a common light chain, which bispecifically binds to GPC3 and CD3. Using a mouse model with reconstituted human immune cells, we revealed that ERY974 is highly effective in killing various types of tumors that have GPC3 expression comparable to that in clinical tumors. ERY974 also induced a robust antitumor efficacy even against tumors with nonimmunogenic features, which are difficult to treat by inhibiting immune checkpoints such as PD-1 (programmed cell death protein-1) and CTLA-4 (cytotoxic T lymphocyte-associated protein-4). Immune monitoring revealed that ERY974 converted the poorly inflamed tumor microenvironment to a highly inflamed microenvironment. Toxicology studies in cynomolgus monkeys showed transient cytokine elevation, but this was manageable and reversible. No organ toxicity was evident. These data provide a rationale for clinical testing of ERY974 for the treatment of patients with GPC3-positive solid tumors.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Glipicanas/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T/imunologia , Animais , Anticorpos Biespecíficos/administração & dosagem , Anticorpos Biespecíficos/farmacocinética , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Complexo CD3/metabolismo , Citocinas/metabolismo , Humanos , Imunocompetência/efeitos dos fármacos , Injeções Intravenosas , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Macaca fascicularis , Camundongos Transgênicos , Esteroides/farmacologia , Esteroides/uso terapêutico , Linfócitos T/efeitos dos fármacos
9.
Cell Rep ; 18(9): 2148-2161, 2017 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28249161

RESUMO

Cellular senescence is an irreversible growth arrest that contributes to development, tumor suppression, and age-related conditions. Senescent cells show active metabolism compared with proliferating cells, but the underlying mechanisms remain unclear. Here we show that the SETD8/PR-Set7 methyltransferase, which catalyzes mono-methylation of histone H4 at lysine 20 (H4K20me1), suppresses nucleolar and mitochondrial activities to prevent cellular senescence. SETD8 protein was selectively downregulated in both oncogene-induced and replicative senescence. Inhibition of SETD8 alone was sufficient to trigger senescence. Under these states, the expression of genes encoding ribosomal proteins (RPs) and ribosomal RNAs as well as the cyclin-dependent kinase (CDK) inhibitor p16INK4A was increased, with a corresponding reduction of H4K20me1 at each locus. As a result, the loss of SETD8 concurrently stimulated nucleolar function and retinoblastoma protein-mediated mitochondrial metabolism. In conclusion, our data demonstrate that SETD8 acts as a barrier to prevent cellular senescence through chromatin-mediated regulation of senescence-associated metabolic remodeling.


Assuntos
Senescência Celular/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Metiltransferases/metabolismo , Linhagem Celular , Nucléolo Celular/metabolismo , Cromatina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Replicação do DNA/fisiologia , Regulação para Baixo/fisiologia , Regulação da Expressão Gênica/fisiologia , Histonas/metabolismo , Humanos , Lisina/metabolismo , Metilação , Mitocôndrias/metabolismo , RNA Ribossômico/metabolismo , Proteínas Ribossômicas/metabolismo
10.
Int J Cancer ; 138(2): 428-39, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26240060

RESUMO

Lysine-specific demethylase-1 (LSD1) removes the methyl groups from mono- and di-methylated lysine 4 of histone H3. Previous studies have linked LSD1 to malignancy in several human tumors, and LSD1 is considered to epigenetically regulate the energy metabolism genes in adipocytes and hepatocellular carcinoma. This study investigates the function of LSD1 in the invasive activity and the metabolism of esophageal cancer cells. We investigated whether LSD1 immunohistochemical expression levels are related to clinical and pathological features, including the maximum standard uptake value in fluorodeoxyglucose positron emission tomography assay. The influence of LSD1 on cell proliferation, invasion and glucose uptake was evaluated in vitro by using specific small interfering RNA for LSD1, and an LSD1 inhibitor. We also evaluated two major energy pathways (glycolytic pathway and mitochondrial respiration) by measuring the extracellular acidification rate (ECAR) and the oxygen consumption rate (OCR) with an extracellular flux analyzer. High LSD1 immunohistochemical expression was significantly associated with high tumor stage, lymphovascular invasion, poor prognosis, and high maximum standard uptake value in esophageal cancer patients. In the in vitro analysis, LSD1 knockdown significantly suppressed the invasive activity and glucose uptake of cancerous cells, reduced their ECAR and increased their OCR and OCR/ECAR. LSD1 may contribute to malignant behavior by regulating the invasive activity and metabolism, activating the glycolytic pathway and inhibiting the mitochondrial respiration of esophageal cancer cells. The results support LSD1 as a potential therapeutic target.


Assuntos
Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/patologia , Histona Desmetilases/metabolismo , Invasividade Neoplásica/patologia , Idoso , Western Blotting , Neoplasias Esofágicas/diagnóstico por imagem , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Reação em Cadeia da Polimerase em Tempo Real
11.
Aging Cell ; 14(4): 689-97, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26009982

RESUMO

Metabolism is closely linked with cellular state and biological processes, but the mechanisms controlling metabolic properties in different contexts remain unclear. Cellular senescence is an irreversible growth arrest induced by various stresses, which exhibits active secretory and metabolic phenotypes. Here, we show that retinoblastoma protein (RB) plays a critical role in promoting the metabolic flow by activating both glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) in cells that have undergone oncogene-induced senescence (OIS). A combination of real-time metabolic monitoring, and metabolome and gene expression analyses showed that OIS-induced fibroblasts developed an accelerated metabolic flow. The loss of RB downregulated a series of glycolytic genes and simultaneously reduced metabolites produced from the glycolytic pathway, indicating that RB upregulates glycolytic genes in OIS cells. Importantly, both mitochondrial OXPHOS and glycolytic activities were abolished in RB-depleted or downstream glycolytic enzyme-depleted OIS cells, suggesting that RB-mediated glycolytic activation induces a metabolic flux into the OXPHOS pathway. Collectively, our findings reveal that RB essentially functions in metabolic remodeling and the maintenance of the active energy production in OIS cells.


Assuntos
Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes ras , Glicólise/genética , Fosforilação Oxidativa , Proteína do Retinoblastoma/genética , Linhagem Celular Tumoral , Senescência Celular/genética , Ciclo do Ácido Cítrico/genética , Células Epiteliais/patologia , Perfilação da Expressão Gênica , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Proteína do Retinoblastoma/deficiência , Transdução de Sinais
12.
Cancer Res ; 75(7): 1445-56, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25649769

RESUMO

The hallmark of most cancer cells is the metabolic shift from mitochondrial to glycolytic metabolism for adapting to the surrounding environment. Although epigenetic modification is intimately linked to cancer, the molecular mechanism, by which epigenetic factors regulate cancer metabolism, is poorly understood. Here, we show that lysine-specific demethylase-1 (LSD1, KDM1A) has an essential role in maintaining the metabolic shift in human hepatocellular carcinoma cells. Inhibition of LSD1 reduced glucose uptake and glycolytic activity, with a concurrent activation of mitochondrial respiration. These metabolic changes coexisted with the inactivation of the hypoxia-inducible factor HIF1α, resulting in a decreased expression of GLUT1 and glycolytic enzymes. In contrast, during LSD1 inhibition, a set of mitochondrial metabolism genes was activated with the concomitant increase of methylated histone H3 at lysine 4 in the promoter regions. Consistently, both LSD1 and GLUT1 were significantly overexpressed in carcinoma tissues. These findings demonstrate the epigenetic plasticity of cancer cell metabolism, which involves an LSD1-mediated mechanism.


Assuntos
Carcinoma Hepatocelular/enzimologia , Histona Desmetilases/fisiologia , Neoplasias Hepáticas/enzimologia , Mitocôndrias Hepáticas/enzimologia , Animais , Carcinoma Hepatocelular/patologia , Expressão Gênica , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Glicólise , Células Hep G2 , Histonas/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Neoplasias Hepáticas/patologia , Metilação , Camundongos SCID , Transplante de Neoplasias , Processamento de Proteína Pós-Traducional , Carga Tumoral
13.
Mol Cell Biol ; 35(7): 1068-80, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25624347

RESUMO

Cells link environmental fluctuations, such as nutrition, to metabolic remodeling. Epigenetic factors are thought to be involved in such cellular processes, but the molecular basis remains unclear. Here we report that the lysine-specific demethylase 2 (LSD2) suppresses the flux and metabolism of lipids to maintain the energy balance in hepatic cells. Using transcriptome and chromatin immunoprecipitation-sequencing analyses, we revealed that LSD2 represses the genes involved in lipid influx and metabolism through demethylation of histone H3K4. Selective recruitment of LSD2 at lipid metabolism gene loci was mediated in part by a stress-responsive transcription factor, c-Jun. Intriguingly, LSD2 depletion increased the intracellular levels of many lipid metabolites, which was accompanied by an increased susceptibility to toxic cell damage in response to fatty acid exposure. Our data demonstrate that LSD2 maintains metabolic plasticity under fluctuating environment in hepatocytes by mediating the cross talk between the epigenome and metabolism.


Assuntos
Regulação para Baixo , Hepatócitos/metabolismo , Histona Desmetilases/metabolismo , Metabolismo dos Lipídeos , Lipídeos/genética , Animais , Células Hep G2 , Histonas/metabolismo , Humanos , Fígado/metabolismo , Lisina/metabolismo , Masculino , Metilação , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-jun/metabolismo
14.
Nat Commun ; 3: 758, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22453831

RESUMO

Environmental factors such as nutritional state may act on the epigenome that consequently contributes to the metabolic adaptation of cells and the organisms. The lysine-specific demethylase-1 (LSD1) is a unique nuclear protein that utilizes flavin adenosine dinucleotide (FAD) as a cofactor. Here we show that LSD1 epigenetically regulates energy-expenditure genes in adipocytes depending on the cellular FAD availability. We find that the loss of LSD1 function, either by short interfering RNA or by selective inhibitors in adipocytes, induces a number of regulators of energy expenditure and mitochondrial metabolism such as PPARγ coactivator-1α resulting in the activation of mitochondrial respiration. In the adipose tissues from mice on a high-fat diet, expression of LSD1-target genes is reduced, compared with that in tissues from mice on a normal diet, which can be reverted by suppressing LSD1 function. Our data suggest a novel mechanism where LSD1 regulates cellular energy balance through coupling with cellular FAD biosynthesis.


Assuntos
Metabolismo Energético , Flavina-Adenina Dinucleotídeo/metabolismo , Mitocôndrias/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Células 3T3 , Adipócitos/metabolismo , Animais , Linhagem Celular , Dieta Hiperlipídica , Ingestão de Alimentos , Perfilação da Expressão Gênica , Células HEK293 , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histona Desmetilases , Humanos , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Oxirredutases N-Desmetilantes/genética , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-raf/genética , Proteínas Proto-Oncogênicas c-raf/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA
15.
J Craniomaxillofac Surg ; 39(8): 624-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21211986

RESUMO

We report the case of a 49-year-old man who underwent a craniectomy for severe head trauma and subsequent cranioplasty with polymethylmethacrylate. He was discharged with moderate right-hand weakness. Five years after the cranioplasty he experienced double vision and slight right hemiparesis. Magnetic resonance imaging (MRI) revealed signs of tentorial herniation but no specific space-occupying lesions. The hand-moulded polymethylmethacrylate bone flap was found to be uneven and protrusions on the inner surface of the bone graft slightly compressed the brain below. His symptoms improved dramatically after a second cranioplasty using a ceramic implant. Although some complications including infection and cosmetic problems have been reported, tentorial herniation during late follow-up as a specific complication of cranioplasty has not been documented previously. We attribute his neurological improvement to the release of compression from the initial graft and to the consequent restoration of cerebrospinal fluid dynamics.


Assuntos
Substitutos Ósseos/efeitos adversos , Doenças Cerebelares/etiologia , Craniotomia/efeitos adversos , Encefalocele/etiologia , Procedimentos de Cirurgia Plástica/efeitos adversos , Polimetil Metacrilato/efeitos adversos , Materiais Biocompatíveis , Cerâmica , Descompressão Cirúrgica , Diplopia/etiologia , Seguimentos , Osso Frontal/lesões , Osso Frontal/cirurgia , Cefaleia/etiologia , Hematoma Subdural/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Complicações Pós-Operatórias , Reoperação , Fraturas Cranianas/cirurgia , Tomografia Computadorizada por Raios X
16.
Chem Pharm Bull (Tokyo) ; 58(1): 38-44, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20045964

RESUMO

Selective factor VIIa-tissue factor complex (FVIIa/TF) inhibition is regarded as a promising target for developing new anticoagulant drugs. In previous reports, we described a S3 subsite found in the X-ray crystal structure of compound 2 that bound to FVIIa/soluble tissue factor (sTF). Based on the X-ray crystal structure information and with the aim of improving the inhibition activity for FVIIa/TF and selectivity against other serine proteases, we synthesized derivatives by introducing substituents at position 5 of the indole ring of compound 2. Among them, compound 16 showed high selectivity against other serine proteases. Contrary to our expectations, compound 16 did not occupy the S3-subsite; X-ray structure analysis revealed that compound 16 improved selectivity by forming hydrogen bonds with Gln217, Thr99 and Asn100.


Assuntos
Fator VIIa/antagonistas & inibidores , Fator VIIa/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Biomimética , Cristalografia por Raios X , Fator VIIa/química , Modelos Moleculares , Ligação Proteica , Tromboplastina/antagonistas & inibidores , Tromboplastina/química , Tromboplastina/metabolismo
17.
Bioorg Med Chem Lett ; 18(16): 4533-7, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18674905

RESUMO

Selective factor VIIa-tissue factor complex (FVIIa/TF) inhibition is regarded as a promising target for developing new anticoagulant drugs. Compound 1 was discovered from focused screening of serine protease-directed compounds from our internal collection. Using parallel synthesis supported by structure-based drug design, we identified peptidemimetic FVIIa/TF inhibitors (compounds 4-11) containing L-Gln or L-Met as the P2 moiety. However, these compounds lacked the selectivity of other serine proteases in the coagulation cascade, especially thrombin. Further optimization of these compounds was carried out with a focus on the P4 moiety. Among the optimized compounds, 12b-f showed improved selectivity.


Assuntos
Química Farmacêutica/métodos , Fator VIIa/antagonistas & inibidores , Serina Endopeptidases/farmacologia , Inibidores de Serina Proteinase/síntese química , Tromboembolia/tratamento farmacológico , Coagulação Sanguínea/efeitos dos fármacos , Cristalografia por Raios X/métodos , Desenho de Fármacos , Humanos , Cinética , Modelos Químicos , Conformação Molecular , Peptídeos/química , Serina Endopeptidases/química , Inibidores de Serina Proteinase/química , Tromboembolia/enzimologia
18.
No Shinkei Geka ; 36(5): 423-8, 2008 May.
Artigo em Japonês | MEDLINE | ID: mdl-18516860

RESUMO

Revisible posterior leukoencephalopathy syndrome (RPLS) is a syndrome defined predominantly by a characteristic pattern of MR images but heterogeneity and reversibility of the abnormalities is unclear. Three patients with characteristic neuroimaging of RPLS are described. Case 1. A 29-year-old woman underwent cesarean section 3 days before developing headache, visual disturbance, mild conscious deterioration and increased blood pressure. MRI on admission showed predominantly white matter edema bilaterally affecting the occipital, temporal and posterior parietal lobes. Lesions were iso-, hypointense on T1 weighted and hyperintense on T2 weighted and fluid-attenuated inversion recovery (FLAIR) images. Diffusion-weighted image (DWI) did not demonstrate hyperintensity in the same regions. She responded well to initial treatment and showed complete recovery. Case 2. A 67-year-old man had headache and visual disturbance after thromboendarterectomy of the left femoral artery. Initial DWI demonstrated hyperintensity in the occipital lobe. The patient had visual abnormality at discharge. Case 3. A 40-year-old woman with early gastric cancer had headache immediately after the injection of contrast material during abdominal computed tomography (CT) examination followed by generalized convulsion. On the third day, she had severe headache and vomiting. Her consciousness deteriorated to lethargy. CT showed marked brain edema, intracerebral hemorrhage, thin subdural hematoma and midline shift. External decompression and hematoma evacuation was performed. She had left hemianopia and left hemiplegia 1 month later. This report has important implications concerning the accurate early diagnosis of RPLS using MRI including DWI and its utility in prompting initial treatment. It should be noted that some cases demonstrating the characteristic neuroimaging pattern of RPLS do not attain full recovery and that surgical intervention is required in some cases.


Assuntos
Imageamento por Ressonância Magnética , Síndrome da Leucoencefalopatia Posterior/diagnóstico , Adulto , Idoso , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Imagem de Difusão por Ressonância Magnética , Feminino , Humanos , Masculino , Síndrome da Leucoencefalopatia Posterior/diagnóstico por imagem , Síndrome da Leucoencefalopatia Posterior/patologia , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
19.
Biochem Biophys Res Commun ; 327(2): 589-96, 2005 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-15629154

RESUMO

Selective factor VIIa-tissue factor complex (FVIIa/TF) inhibition is seen as a promising target for developing new anticoagulant drugs. Structure-based designs of the P3 moiety in the peptide mimetic factor VIIa inhibitor successfully lead to novel inhibitors with selectivity for FVIIa/TF and extrinsic coagulation the same as or even higher than those of previously reported peptide mimetic factor VIIa inhibitors. X-ray crystal structure analysis reveals that one of the novel inhibitors shows improved selectivity by forming interactions between the inhibitor and FVIIa as expected. Another of the novel inhibitors achieves improved selectivity through an unexpected hydrogen bond with Gln217, with a unique bent conformation in FVIIa/TF accompanied by conformational changes of the inhibitor and the protein.


Assuntos
Materiais Biomiméticos/química , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Fator VIIa/antagonistas & inibidores , Peptídeos/síntese química , Peptídeos/farmacologia , Sequência de Aminoácidos , Sítios de Ligação , Materiais Biomiméticos/síntese química , Materiais Biomiméticos/farmacologia , Cristalografia por Raios X , Inibidores Enzimáticos/química , Fator VIIa/química , Fator VIIa/metabolismo , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Estrutura Terciária de Proteína , Alinhamento de Sequência , Relação Estrutura-Atividade
20.
Artigo em Inglês | MEDLINE | ID: mdl-16510984

RESUMO

The crystal structure of human factor VIIa/soluble tissue factor (FVIIa/sTF) in complex with a highly selective peptide-mimetic FVIIa inhibitor which shows 1670-fold selectivity against thrombin inhibition has been solved at 2.6 A resolution. The inhibitor is bound to FVIIa/sTF at the S1, S2 and S3 sites and at the additional S1 subsite. Two charged groups, the amidino group in P2 and the carboxylate group in P4, form ionic interactions with Asp60 and Lys192 of FVIIa, respectively. Structural comparisons between factor VIIa and thrombin show that thrombin has oppositely charged residues, Lys60F and Glu192, in the S2 site and the S1 subsites, respectively. These data suggest that the utilization of the differences of charge distribution in the S2 site and the S1 subsites between FVIIa and thrombin is critical for achieving high selectivity against thrombin inhibition. These results will provide valuable information for the structure-based drug design of specific inhibitors for FVIIa/TF.


Assuntos
Anticoagulantes/química , Fator VIIa/antagonistas & inibidores , Fator VIIa/química , Tromboplastina/química , Antitrombinas/química , Coagulação Sanguínea , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Substâncias Macromoleculares/química , Modelos Moleculares , Peptídeos/química , Estrutura Secundária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA