Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 312
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Breast Cancer Res Treat ; 195(3): 223-236, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35918499

RESUMO

PURPOSE: Arylamine N-acetyltransferase 1 (NAT1) deficiency has been associated with drug resistance and poor outcomes in breast cancer patients. The current study aimed to investigate drug resistance in vitro using normal breast cancer cell lines and NAT1-deficient cell lines to understand the changes induced by the lack of NAT1 that resulted in poor drug response. METHODS: The response to seven chemotherapeutic agents was quantified following NAT1 deletion using CRISPR-Cas 9 in MDA-MB-231 and T-47D cells. Apoptosis was monitored by annexin V staining and caspase 3/7 activity. Cytochrome C release and caspase 8 and 9 activities were measured by Western blots. Caspase 8 was inhibited using Z-IETD-FMK and necroptosis was inhibited using necrostatin and necrosulfonamide. RESULTS: Compared to parental cells, NAT1 depleted cells were resistant to drug treatment. This could be reversed following NAT1 rescue of the NAT1 deleted cells. Release of cytochrome C in response to treatment was decreased in the NAT1 depleted cells, suggesting suppression of the intrinsic apoptotic pathway. In addition, NAT1 knockout resulted in a decrease in caspase 8 activation. Treatment with necrosulfonamide showed that NAT1 deficient cells switched from intrinsic apoptosis to necroptosis when treated with the anti-cancer drug cisplatin. CONCLUSIONS: NAT1 deficiency can switch cell death from apoptosis to necroptosis resulting in decreased response to cytotoxic drugs. The absence of NAT1 in patient tumours may be a useful biomarker for selecting alternative treatments in a subset of breast cancer patients.


Assuntos
Antineoplásicos , Arilamina N-Acetiltransferase , Neoplasias da Mama , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Arilamina N-Acetiltransferase/deficiência , Arilamina N-Acetiltransferase/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Caspase 8/uso terapêutico , Morte Celular , Citocromos c/metabolismo , Citocromos c/uso terapêutico , Feminino , Humanos , Isoenzimas/deficiência , Isoenzimas/genética , Necroptose
2.
Neurosci Lett ; 735: 135206, 2020 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-32593773

RESUMO

Inositol-1,4,5-trisphosphate 3-kinase-A (ITPKA) is the neuronal isoform of ITPKs and exhibits both actin bundling and InsP3kinase activity. In addition to neurons, ITPKA is ectopically expressed in tumor cells, where its oncogenic activity increases tumor cell malignancy. In order to analyze the physiological relevance of ITPKA, here we performed a broad phenotypic screening of itpka deficient mice. Our data show that among the neurobehavioral tests analyzed, itpka deficient mice reacted faster to a hotplate, prepulse inhibition was impaired and the accelerating rotarod test showed decreased latency of itpka deficient mice to fall. These data indicate that ITPKA is involved in the regulation of nociceptive pathways, sensorimotor gating and motor learning. Analysis of extracerebral functions in control and itpka deficient mice revealed significantly reduced glucose, lactate, and triglyceride plasma concentrations in itpka deficient mice. Based on this finding, expression of ITPKA was analyzed in extracerebral tissues and the highest level was found in the small intestine. However, functional studies on CaCo-2 control and ITPKA depleted cells showed that glucose, as well as triglyceride uptake, were not significantly different between the cell lines. Altogether, these data show that ITPKA exhibits distinct functions in the central nervous system and reveal an involvement of ITPKA in energy metabolism.


Assuntos
Neurônios/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Inibição Pré-Pulso/fisiologia , Animais , Células CACO-2 , Feminino , Humanos , Isoenzimas/deficiência , Isoenzimas/genética , Masculino , Camundongos , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/genética
3.
Cancer Cell ; 36(2): 156-167.e7, 2019 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31378680

RESUMO

We report that mouse LSL-KrasG12D;Trp53fl/fl (KP)-mediated lung adenocarcinoma (LADC) tumorigenesis can proceed through both PKCι-dependent and PKCι-independent pathways. The predominant pathway involves PKCι-dependent transformation of bronchoalveolar stem cells (BASCs). However, KP mice harboring conditional knock out Prkci alleles (KPI mice) develop LADC tumors through PKCι-independent transformation of Axin2+ alveolar type 2 (AT2) stem cells. Transformed growth of KPI, but not KP, tumors is blocked by Wnt pathway inhibition in vitro and in vivo. Furthermore, a KPI-derived genomic signature predicts sensitivity of human LADC cells to Wnt inhibition, and identifies a distinct subset of primary LADC tumors exhibiting a KPI-like genotype. Thus, LADC can develop through both PKCι-dependent and PKCι-independent pathways, resulting in tumors exhibiting distinct oncogenic signaling and pharmacologic vulnerabilities.


Assuntos
Adenocarcinoma de Pulmão/enzimologia , Transformação Celular Neoplásica/metabolismo , Genes ras , Isoenzimas/metabolismo , Neoplasias Pulmonares/enzimologia , Proteína Quinase C/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Via de Sinalização Wnt , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/patologia , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Isoenzimas/deficiência , Isoenzimas/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Inibidores de Proteínas Quinases/farmacologia , Carga Tumoral , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , beta Catenina/genética , beta Catenina/metabolismo
4.
Mol Pharmacol ; 96(5): 573-579, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31444237

RESUMO

Arylamine N-acetyltransferase 1 (NAT1) is a drug-metabolizing enzyme that influences cancer cell proliferation and survival. However, the mechanism for these effects is unknown. Because of previous observations that NAT1 inhibition decreases invasiveness, we investigated the expression of the metalloproteinase matrix metalloproteinase 9 (MMP9) in human breast cancer samples and in cancer cells. We found a negative correlation between the expression of NAT1 and MMP9 in 1904 breast cancer samples. Moreover, when NAT1 was deleted in highly invasive breast cancer cells, MMP9 mRNA and protein significantly increased, both of which were reversed by reintroducing NAT1 into the knockout cells. After NAT1 deletion, there was an increased association of acetylated histone H3 with the SET and MYND-domain containing 3 (SMYD3) element in the MMP9 promoter, consistent with an increase in MMP9 transcription. NAT1 deletion also up-regulated hypoxia-inducible factor 1-α (HIF1-α). Treatment of the NAT1 knockout cells with small interfering RNA directed toward HIF1-α mRNA inhibited the increased expression of MMP9. Taken together, these results show a direct inverse relationship between NAT1 and MMP9 and suggest that HIF1-α may be essential for the regulation of MMP9 expression by NAT1. SIGNIFICANCE STATEMENT: The expression of the enzyme NAT1 was found to be negatively correlated with MMP9 expression in tumor tissue from breast cancer patients. In cells, NAT1 regulated MMP9 expression at a transcriptional level via HIF1-α. This finding is important as it may explain some of the pathological features associated with changes in NAT1 expression in cancer.


Assuntos
Arilamina N-Acetiltransferase/deficiência , Neoplasias da Mama/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Isoenzimas/deficiência , Metaloproteinase 9 da Matriz/biossíntese , Arilamina N-Acetiltransferase/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Técnicas de Inativação de Genes , Células HT29 , Células HeLa , Humanos , Isoenzimas/genética , Metaloproteinase 9 da Matriz/deficiência
5.
FASEB J ; 33(10): 10668-10679, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31268747

RESUMO

PLC-ß exerts biologic influences through GPCR. GPCRs are involved in regulating glucose-stimulated insulin secretion (GSIS). Previous studies have suggested that PLC-ßs might play an important role in pancreatic ß cells. However, because of a lack of the specific inhibitors of PLC-ß isozymes and appropriate genetic models, the in vivo function of specific PLC-ß isozymes in pancreatic ß cells and their physiologic relevance in the regulation of insulin secretion have not been studied so far. The present study showed that PLC-ß1 was crucial for ß-cell function by generation of each PLC-ß conditional knockout mouse. Mice lacking PLC-ß1 in ß cells exhibited a marked defect in GSIS, leading to glucose intolerance. In ex vivo studies, the secreted insulin level and Ca2+ response in Plcb1f/f; pancreas/duodenum homeobox protein 1 (Pdx1)-Cre recombinase-estrogen receptor T2 (CreERt2) islets was lower than those in the Plcb1f/f islets under the high-glucose condition. PLC-ß1 led to potentiate insulin secretion via stimulation of particular Gq-protein-coupled receptors. Plcb1f/f; Pdx1-CreERt2 mice fed a high-fat diet developed more severe glucose intolerance because of a defect in insulin secretion. The present study identified PLC-ß1 as an important molecule that regulates ß cell insulin secretion and can be considered a candidate for therapeutic intervention in diabetes mellitus.-Hwang, H.-J., Yang, Y. R., Kim, H. Y., Choi, Y., Park, K.-S., Lee, H., Ma, J. S., Yamamoto, M., Kim, J., Chae, Y. C., Choi, J. H., Cocco, L., Berggren, P.-O., Jang, H.-J., Suh, P.-G. Phospholipase Cß1 potentiates glucose-stimulated insulin secretion.


Assuntos
Glucose/metabolismo , Secreção de Insulina/fisiologia , Fosfolipase C beta/metabolismo , Animais , Linhagem Celular , Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Técnicas In Vitro , Secreção de Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfolipase C beta/deficiência , Fosfolipase C beta/genética , Receptores Acoplados a Proteínas G/metabolismo , Transativadores/genética , Transativadores/metabolismo
7.
Cancer Cell ; 35(3): 385-400.e9, 2019 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-30827887

RESUMO

Increasingly effective therapies targeting the androgen receptor have paradoxically promoted the incidence of neuroendocrine prostate cancer (NEPC), the most lethal subtype of castration-resistant prostate cancer (PCa), for which there is no effective therapy. Here we report that protein kinase C (PKC)λ/ι is downregulated in de novo and during therapy-induced NEPC, which results in the upregulation of serine biosynthesis through an mTORC1/ATF4-driven pathway. This metabolic reprogramming supports cell proliferation and increases intracellular S-adenosyl methionine (SAM) levels to feed epigenetic changes that favor the development of NEPC characteristics. Altogether, we have uncovered a metabolic vulnerability triggered by PKCλ/ι deficiency in NEPC, which offers potentially actionable targets to prevent therapy resistance in PCa.


Assuntos
Carcinoma Neuroendócrino/patologia , Regulação para Baixo , Isoenzimas/deficiência , Neoplasias da Próstata/patologia , Proteína Quinase C/deficiência , Serina/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Vias Biossintéticas , Carcinoma Neuroendócrino/genética , Carcinoma Neuroendócrino/metabolismo , Linhagem Celular Tumoral , Metilação de DNA , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , S-Adenosilmetionina/metabolismo
8.
Int J Biochem Cell Biol ; 110: 84-90, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30836144

RESUMO

Human arylamine N-acetyltransferase 1 (NAT1) has been widely reported to affect cancer cell growth and survival and recent studies suggest it may alter cell metabolism. In this study, the effects of NAT1 deletion on mitochondrial function was examined in 2 human cell lines, breast carcinoma MDA-MB-231 and colon carcinoma HT-29 cells. Using a Seahorse XFe96 Flux Analyzer, NAT1 deletion was shown to decrease oxidative phosphorylation with a significant loss in respiratory reserve capacity in both cell lines. There also was a decrease in glycolysis without a change in glucose uptake. The changes in mitochondrial function was due to a decrease in pyruvate dehydrogenase activity, which could be reversed with the pyruvate dehydrogenase kinase inhibitor dichloroacetate. In the MDA-MB-231 and HT-29 cells, pyruvate dehydrogenase activity was attenuated either by an increase in phosphorylation or a decrease in total protein expression. These results may help explain some of the cellular events that have been reported recently in cell and animal models of NAT1 deficiency.


Assuntos
Arilamina N-Acetiltransferase/deficiência , Arilamina N-Acetiltransferase/genética , Deleção de Genes , Isoenzimas/deficiência , Isoenzimas/genética , Mitocôndrias/metabolismo , Complexo Piruvato Desidrogenase/metabolismo , Transporte Biológico/genética , Regulação Neoplásica da Expressão Gênica , Glucose/metabolismo , Células HT29 , Humanos , Complexo Piruvato Desidrogenase/antagonistas & inibidores
9.
Eur J Pediatr Surg ; 29(3): 290-297, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29723880

RESUMO

INTRODUCTION: Prematurity, formula feeding, and early weaning strongly influence enterocyte differentiation. Intestinal alkaline phosphatase (IAP), an endogenous protein expressed in the intestines, is one enzyme that is affected by these factors. IAP supplementation decreases the severity of necrotizing enterocolitis (NEC) injury. We, therefore, hypothesized that prematurity predisposes this population to NEC due to IAP deficiency and investigated IAP expression and function in a neonatal rat model. MATERIALS AND METHODS: Pre- and full-term newborn Sprague-Dawley rat pups were sacrificed on consecutive days of life both after vaginal or cesarean birth and following either breast or formula feeding. RESULTS: Compared with controls, cesarean delivery and formula feeding are associated with lower levels of IAP. The formula-fed pups continued to have low baseline IAP activity. Neither prematurity nor formula feeding led to differences of intestinal injury. CONCLUSION: Prematurity and formula feeding are associated with inhibition of IAP expression and activity. Both may increase the risk of NEC and early enteral supplementation of IAP to newborns at risk of NEC may be of therapeutic benefit.


Assuntos
Fosfatase Alcalina/deficiência , Enterocolite Necrosante/etiologia , Doenças do Prematuro/etiologia , Isoenzimas/deficiência , Fosfatase Alcalina/metabolismo , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Cesárea/efeitos adversos , Enterocolite Necrosante/metabolismo , Humanos , Fórmulas Infantis/efeitos adversos , Recém-Nascido , Doenças do Prematuro/metabolismo , Isoenzimas/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Risco
10.
Nucleus ; 9(1): 431-441, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30059280

RESUMO

Aberrant levels of histone modifications lead to chromatin malfunctioning and consequently to various developmental defects and human diseases. Therefore, the proteins bearing the ability to modify histones have been extensively studied and the molecular mechanisms of their action are now fairly well understood. However, little attention has been paid to naturally occurring alternative isoforms of chromatin modifying proteins and to their biological roles. In this review, we focus on mammalian KDM2A and KDM2B, the only two lysine demethylases whose genes have been described to produce also an alternative isoform lacking the N-terminal demethylase domain. These short KDM2A/B-SF isoforms arise through alternative promoter usage and seem to play important roles in development and disease. We hypothesise about the biological significance of these alternative isoforms, which might represent a more common evolutionarily conserved regulatory mechanism.


Assuntos
Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias/enzimologia , Animais , Humanos , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias/metabolismo
11.
Elife ; 72018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29916805

RESUMO

Hematopoietic stem cells require MLL1, which is one of six Set1/Trithorax-type histone 3 lysine 4 (H3K4) methyltransferases in mammals and clinically the most important leukemia gene. Here, we add to emerging evidence that all six H3K4 methyltransferases play essential roles in the hematopoietic system by showing that conditional mutagenesis of Setd1b in adult mice provoked aberrant homeostasis of hematopoietic stem and progenitor cells (HSPCs). Using both ubiquitous and hematopoietic-specific deletion strategies, the loss of Setd1b resulted in peripheral thrombo- and lymphocytopenia, multilineage dysplasia, myeloid-biased extramedullary hematopoiesis in the spleen, and lethality. By transplantation experiments and expression profiling, we determined that Setd1b is autonomously required in the hematopoietic lineages where it regulates key lineage specification components, including Cebpa, Gata1, and Klf1. Altogether, these data imply that the Set1/Trithorax-type epigenetic machinery sustains different aspects of hematopoiesis and constitutes a second framework additional to the transcription factor hierarchy of hematopoietic homeostasis.


Assuntos
Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Histona-Lisina N-Metiltransferase/genética , Homeostase/genética , Linfopenia/genética , Proteína de Leucina Linfoide-Mieloide/genética , Trombocitopenia/genética , Animais , Transplante de Medula Óssea , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem da Célula/genética , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genes Letais , Células-Tronco Hematopoéticas/citologia , Histona-Lisina N-Metiltransferase/deficiência , Isoenzimas/deficiência , Isoenzimas/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Linfopenia/metabolismo , Linfopenia/patologia , Camundongos , Camundongos Knockout , Proteína de Leucina Linfoide-Mieloide/deficiência , Baço/metabolismo , Baço/patologia , Trombocitopenia/metabolismo , Trombocitopenia/patologia , Irradiação Corporal Total
12.
Nat Cell Biol ; 19(9): 1017-1026, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28812580

RESUMO

Although normally dormant, hair follicle stem cells (HFSCs) quickly become activated to divide during a new hair cycle. The quiescence of HFSCs is known to be regulated by a number of intrinsic and extrinsic mechanisms. Here we provide several lines of evidence to demonstrate that HFSCs utilize glycolytic metabolism and produce significantly more lactate than other cells in the epidermis. Furthermore, lactate generation appears to be critical for the activation of HFSCs as deletion of lactate dehydrogenase (Ldha) prevented their activation. Conversely, genetically promoting lactate production in HFSCs through mitochondrial pyruvate carrier 1 (Mpc1) deletion accelerated their activation and the hair cycle. Finally, we identify small molecules that increase lactate production by stimulating Myc levels or inhibiting Mpc1 carrier activity and can topically induce the hair cycle. These data suggest that HFSCs maintain a metabolic state that allows them to remain dormant and yet quickly respond to appropriate proliferative stimuli.


Assuntos
Proliferação de Células , Senescência Celular , Glicólise , Folículo Piloso/enzimologia , L-Lactato Desidrogenase/metabolismo , Ácido Láctico/metabolismo , Células-Tronco/enzimologia , Acrilatos/farmacologia , Animais , Proteínas de Transporte de Ânions/antagonistas & inibidores , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Feminino , Genótipo , Glicólise/efeitos dos fármacos , Folículo Piloso/citologia , Folículo Piloso/efeitos dos fármacos , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Desidrogenase/deficiência , L-Lactato Desidrogenase/genética , Lactato Desidrogenase 5 , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Transporte da Membrana Mitocondrial/antagonistas & inibidores , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos , Fenótipo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transdução de Sinais , Células-Tronco/efeitos dos fármacos , Fatores de Tempo
13.
Cell Commun Signal ; 15(1): 28, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28724384

RESUMO

BACKGROUND: Phosphoinositide 3-kinase γ (PI3Kγ) and PI3Kδ are second messenger-generating enzymes with key roles in proliferation, differentiation, survival, and function of leukocytes. Deficiency of the catalytic subunits p110γ and p110δ of PI3Kγ and PI3Kδ in p110γ/δ-/- mice leads to defective B- and T-cell homeostasis. Here we examined the role of p110γ and p110δ in the homeostasis of neutrophils by analyzing p110γ-/-, p110δ-/- and p110γ/δ-/- mice. METHODS: Neutrophils and T cells in leukocyte suspensions from the bone marrow (BM), blood, spleen and lung were analyzed by flow cytometry. Serum concentrations of IL-17, of the neutrophilic growth factor G-CSF, and of the neutrophil mobilizing CXC chemokines CXCL1/KC and CXCL2/MIP-2 were measured by Bio-Plex assay. Production of G-CSF and CXCL1/KC by IL-17-stimulated primary lung tissue cells were determined by ELISA, whereas IL-17-dependent signaling in lung tissue cells was analyzed by measuring Akt phosphorylation using immunoblot. RESULTS: We found that in contrast to single knock-out mice, p110γ/δ-/- mice exhibited significantly elevated neutrophil counts in blood, spleen, and lung. Increased granulocytic differentiation stages in the bone marrow of p110γ/δ-/- mice were paralleled by increased serum concentrations of G-CSF, CXCL1/KC, and CXCL2/MIP-2. As IL-17 induces neutrophilia via the induction of G-CSF and CXC chemokines, we measured IL-17 and IL-17-producing T cells. IL-17 serum concentrations and frequencies of IL-17+ splenic T cells were significantly increased in p110γ/δ-/- mice. Moreover, IFN-γ+, IL-4+, and IL-5+ T cell subsets were drastically increased in p110γ/δ-/- mice, suggesting that IL-17+ T cells were up-regulated in the context of a general percentage increase of other cytokine producing T cell subsets. CONCLUSIONS: We found that p110γ/δ deficiency in mice induces complex immunological changes, which might in concert contribute to neutrophilia. These findings emphasize a crucial but indirect role of both p110γ and p110δ in the regulation of neutrophil homeostasis.


Assuntos
Transtornos Leucocíticos/genética , Neutrófilos/metabolismo , Fosfatidilinositol 3-Quinases/deficiência , Animais , Células Cultivadas , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Homeostase , Interleucina-17/metabolismo , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Transtornos Leucocíticos/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Baço/metabolismo , Linfócitos T/metabolismo
14.
Cell Signal ; 38: 223-229, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28739484

RESUMO

Platelet activation at sites of vascular injury is crucial for hemostasis, but it may also cause myocardial infarction or ischemic stroke. Upon platelet activation, cytoskeletal reorganization is essential for platelet secretion and thrombus formation. Members of the protein kinase C family, which includes 12 isoforms, are involved in most platelet responses required for thrombus formation. The atypical protein kinase Cι/λ (PKCι/λ) has been implicated as an important mediator of cell polarity, carcinogenesis and immune cell responses. PKCι/λ is known to be associated with the small GTPase Cdc42, an important mediator of multiple platelet functions; however, its exact function in platelets is not known. To study the role of PKCι/λ, we generated platelet- and megakaryocyte-specific PKCι/λ knockout mice (Prkcifl/fl, Pf4-Cre) and used them to investigate the function of PKCι/λ in platelet activation and aggregation in vitro and in vivo. Surprisingly, lack of PKCι/λ had no detectable effect on platelet spreading and function in vitro and in vivo under all tested conditions. These results indicate that PKCι/λ is dispensable for Cdc42-triggered processes and for thrombosis and hemostasis in mice.


Assuntos
Plaquetas/metabolismo , Hemostasia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Trombose/enzimologia , Trombose/patologia , Animais , Colágeno/metabolismo , Feminino , Fibrinogênio/metabolismo , Humanos , Integrinas/metabolismo , Isoenzimas/deficiência , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Adesividade Plaquetária , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Proteína Quinase C/deficiência , Transdução de Sinais
15.
Cancer Res ; 77(13): 3632-3643, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28446465

RESUMO

Immunometabolism is emerging as a critical determinant of cancer pathophysiology. In this study, we explored the contributions of macrophage-expressed lactate dehydrogenase-A (LDH-A) to tumor formation in a K-Ras murine model of lung carcinoma. Myeloid-specific deletion of LDH-A promoted accumulation of macrophages with a CD86high and MCP-1high M1-like phenotype that suppressed tumor growth. This phenotypic effect was accompanied by reduced VEGF expression and angiogenesis, diminished numbers of PD-L1+ cancer cells, increased numbers of CD3+ T cells, and activation status of CD8+ T cells. Furthermore, it was associated with more pronounced antitumor T-cell immunity via induction of IL17 and IFNγ-producing CD8+ T (Tc17 and Tc1) cells, likely via suppression of lactate-driven PD-L1 expression. Our results suggest that expressions of LDH-A and lactate by macrophage in the tumor microenvironment are major drivers of T-cell immunosuppression, strongly supporting the concept of targeting stromal LDH-A as an effective strategy to blunt tumoral immune escape. Cancer Res; 77(13); 3632-43. ©2017 AACR.


Assuntos
L-Lactato Desidrogenase/deficiência , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/imunologia , Células Mieloides/imunologia , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Humanos , Isoenzimas/deficiência , Isoenzimas/imunologia , Isoenzimas/metabolismo , L-Lactato Desidrogenase/imunologia , L-Lactato Desidrogenase/metabolismo , Lactato Desidrogenase 5 , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/enzimologia , Células Mieloides/patologia , Microambiente Tumoral/imunologia
16.
Artigo em Russo | MEDLINE | ID: mdl-27735906

RESUMO

Fabry disease (Anderson-Fabry disease) is an X-linked recessive lysosomal storage disorder resulting from deficient activity of lysosomal hydrolase, alpha-galactosidase A (alpha-Gal A), which leads to progressive accumulation of globotriaosylceramide (Gb3) in various cells, predominantly endothelial and vascular smooth muscle cells, with clinical manifestations affecting major organs including the central nervous system. Manifestations of Fabry disease include progressive renal and cardiac insufficiency, neuropathic pain, stroke and cerebral disease, skin and gastrointestinal symptoms. Clinical onset usually occurs in childhood, but many severe patients are diagnosed in adulthood. Females may be severely affected as males and both may die prematurely due to stroke, heart disease and renal failure. Early recognition of symptoms, enzyme activity levels, concentration of Gb3 levels in the blood, urine and skin biopsies, as well as genetic testing (GLA gene) enable establishment of early diagnosis and therapeutic intervention with enzyme replacement therapy. Enzyme replacement therapy can stabilize or reduce the progression of the disease. Early therapy may prevent complications of the disease.


Assuntos
Doença de Fabry/diagnóstico , Doença de Fabry/fisiopatologia , Progressão da Doença , Diagnóstico Precoce , Terapia de Reposição de Enzimas , Doença de Fabry/complicações , Doença de Fabry/tratamento farmacológico , Feminino , Humanos , Isoenzimas/deficiência , Masculino , Neuralgia/diagnóstico , Neuralgia/etiologia , Acidente Vascular Cerebral/etiologia , Triexosilceramidas/metabolismo , alfa-Galactosidase
17.
Cell Rep ; 17(2): 527-540, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27705799

RESUMO

We recently identified human N-acetyltransferase 2 (NAT2) as an insulin resistance (IR) gene. Here, we examine the cellular mechanism linking NAT2 to IR and find that Nat1 (mouse ortholog of NAT2) is co-regulated with key mitochondrial genes. RNAi-mediated silencing of Nat1 led to mitochondrial dysfunction characterized by increased intracellular reactive oxygen species and mitochondrial fragmentation as well as decreased mitochondrial membrane potential, biogenesis, mass, cellular respiration, and ATP generation. These effects were consistent in 3T3-L1 adipocytes, C2C12 myoblasts, and in tissues from Nat1-deficient mice, including white adipose tissue, heart, and skeletal muscle. Nat1-deficient mice had changes in plasma metabolites and lipids consistent with a decreased ability to utilize fats for energy and a decrease in basal metabolic rate and exercise capacity without altered thermogenesis. Collectively, our results suggest that Nat1 deficiency results in mitochondrial dysfunction, which may constitute a mechanistic link between this gene and IR.


Assuntos
Arilamina N-Acetiltransferase/genética , Resistência à Insulina/genética , Isoenzimas/genética , Mitocôndrias/metabolismo , Condicionamento Físico Animal , Células 3T3-L1 , Trifosfato de Adenosina/biossíntese , Adipócitos/metabolismo , Animais , Arilamina N-Acetiltransferase/deficiência , Arilamina N-Acetiltransferase/metabolismo , Isoenzimas/deficiência , Potencial da Membrana Mitocondrial/genética , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Camundongos , Mitocôndrias/patologia , Miocárdio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
18.
Antimicrob Agents Chemother ; 60(11): 6460-6470, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27527086

RESUMO

Mycobacterium tuberculosis remains a global health threat largely due to the lengthy duration of curative antibiotic treatment, contributing to medical nonadherence and the emergence of drug resistance. This prolonged therapy is likely due to the presence of M. tuberculosis persisters, which exhibit antibiotic tolerance. Inorganic polyphosphate [poly(P)] is a key regulatory molecule in the M. tuberculosis stringent response mediating antibiotic tolerance. The polyphosphate kinase PPK1 is responsible for poly(P) synthesis in M. tuberculosis, while the exopolyphosphatases PPX1 and PPX2 and the GTP synthase PPK2 are responsible for poly(P) hydrolysis. In the present study, we show by liquid chromatography-tandem mass spectrometry that poly(P)-accumulating M. tuberculosis mutant strains deficient in ppx1 or ppk2 had significantly lower intracellular levels of glycerol-3-phosphate (G3P) and 1-deoxy-xylulose-5-phosphate. Real-time PCR revealed decreased expression of genes in the G3P synthesis pathway in each mutant. The ppx1-deficient mutant also showed a significant accumulation of metabolites in the tricarboxylic acid cycle, as well as altered arginine and NADH metabolism. Each poly(P)-accumulating strain showed defective biofilm formation, while deficiency of ppk2 was associated with increased sensitivity to plumbagin and meropenem and deficiency of ppx1 led to enhanced susceptibility to clofazimine. A DNA vaccine expressing ppx1 and ppk2, together with two other members of the M. tuberculosis stringent response, M. tuberculosis rel and sigE, did not show protective activity against aerosol challenge with M. tuberculosis, but vaccine-induced immunity enhanced the killing activity of isoniazid in a murine model of chronic tuberculosis. In summary, poly(P)-regulating factors of the M. tuberculosis stringent response play an important role in M. tuberculosis metabolism, biofilm formation, and antibiotic sensitivity in vivo.


Assuntos
Hidrolases Anidrido Ácido/genética , Antituberculosos/farmacologia , Farmacorresistência Bacteriana/genética , Isoniazida/farmacologia , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Vacinas contra a Tuberculose/administração & dosagem , Tuberculose Resistente a Múltiplos Medicamentos/tratamento farmacológico , Hidrolases Anidrido Ácido/deficiência , Animais , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Ciclo do Ácido Cítrico/genética , Clofazimina/farmacologia , Modelos Animais de Doenças , Expressão Gênica , Glicerofosfatos/metabolismo , Isoenzimas/deficiência , Isoenzimas/genética , Meropeném , Camundongos , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Naftoquinonas/farmacologia , Fosfotransferases (Aceptor do Grupo Fosfato)/deficiência , Polifosfatos/metabolismo , Tienamicinas/farmacologia , Tuberculose Resistente a Múltiplos Medicamentos/imunologia , Tuberculose Resistente a Múltiplos Medicamentos/microbiologia , Tuberculose Resistente a Múltiplos Medicamentos/prevenção & controle , Vacinas de DNA/administração & dosagem , Xilose/análogos & derivados , Xilose/metabolismo
19.
J Dermatol ; 43(12): 1429-1432, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27450766

RESUMO

Hereditary lactate dehydrogenase (LDH) M-subunit deficiency is very rare and we have found reports of close to a dozen cases in the published work, two of which were associated with pustular psoriasis-like lesions. We report a third case of pustular psoriasis-like eruptions associated with LDH M-subunit deficiency, which occurred 24 years after the diagnosis of LDH M-subunit deficiency. These cases indicate that abnormal activity of LDH can induce pustular psoriatic lesions in the long term. Some patients with symptoms of hereditary LDH M-subunit deficiency have antecedent annular scaly plaque lesions, that resemble psoriatic lesions. We discuss a hypothesis to explain this scenario.


Assuntos
L-Lactato Desidrogenase/deficiência , L-Lactato Desidrogenase/genética , Psoríase/genética , Doenças Raras/genética , Deleção de Sequência , Adulto , Sequência de Aminoácidos/genética , Biópsia , Éxons/genética , Feminino , Glucocorticoides/uso terapêutico , Humanos , Imuno-Histoquímica , Isoenzimas/deficiência , Isoenzimas/genética , Lactato Desidrogenase 5 , Prednisolona/uso terapêutico , Psoríase/tratamento farmacológico , RNA Mensageiro/genética , Análise de Sequência de DNA , Análise de Sequência de RNA , Pele/patologia , Fatores de Tempo , Valina/genética
20.
Mol Biol Cell ; 27(14): 2186-97, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27226486

RESUMO

The conserved proteins of the polarity complex made up of atypical PKC (aPKC, isoforms ι and ζ), Par6, and Par3 determine asymmetry in several cell types, from Caenorhabditis elegans oocytes to vertebrate epithelia and neurons. We previously showed that aPKC is down-regulated in intestinal epithelia under inflammatory stimulation. Further, expression of constitutively active PKCι decreases NF-κB activity in an epithelial cell line, the opposite of the effect reported in other cells. Here we tested the hypothesis that aPKC has a dual function in epithelia, inhibiting the NF-κB pathway in addition to having a role in apicobasal polarity. We achieved full aPKC down-regulation in small intestine villi and colon surface epithelium using a conditional epithelium-specific knockout mouse. The results show that aPKC is dispensable for polarity after cell differentiation, except for known targets, including ROCK and ezrin, claudin-4 expression, and barrier permeability. The aPKC defect resulted in increased NF-κB activity, which could be rescued by IKK and ROCK inhibitors. It also increased expression of proinflammatory cytokines. In contrast, expression of anti-inflammatory IL-10 decreased. We conclude that epithelial aPKC acts upstream of multiple mechanisms that participate in the inflammatory response in the intestine, including, but not restricted to, NF-κB.


Assuntos
Isoenzimas/metabolismo , NF-kappa B/metabolismo , Proteína Quinase C/metabolismo , Animais , Diferenciação Celular/fisiologia , Polaridade Celular/fisiologia , Regulação para Baixo , Células Epiteliais/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Isoenzimas/deficiência , Isoenzimas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA