Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Bone Miner Metab ; 41(4): 470-480, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37036533

RESUMO

INTRODUCTION: The conditional manipulation of genes using the Cre recombinase-locus of crossover in P1 (Cre/loxP) system is an important tool for revealing gene functions and cell lineages in vivo. The outcome of this method is dependent on the performance of Cre-driver mouse strains. In most cases, Cre knock-in mice show better specificity than randomly inserted Cre transgenic mice. However, following knock-in, the expression of the original gene replaced by Cre is lost. MATERIALS AND METHODS: We generated a new differentiated osteoblast- and osteocyte-specific Cre knock-in mouse line that carries the viral T2A sequence encoding a 2A self-cleaving peptide at the end of the coding region of the dentin matrix protein 1 (Dmp1) gene accompanied by the Cre gene. RESULTS: We confirmed that Dmp1-T2A-Cre mice showed high Cre expression in osteoblasts, osteocytes, odontoblasts, and periodontal ligament cells and that the 2A self-cleaving peptide efficiently produced both Dmp1 and Cre proteins. Furthermore, unlike the Dmp1 knockout mice, homozygous Dmp1-T2A-Cre mice showed no skeletal abnormalities. Analysis using the Cre reporter strain confirmed differentiated osteoblast- and osteocyte-specific Cre-mediated recombination in the skeleton. Furthermore, recombination was also detected in some nuclei of skeletal muscle cells, spermatocytes, and intestinal cells. CONCLUSION: 2A-Cre functions effectively in vivo, and Dmp1-T2A-Cre knock-in mice are a useful tool for studying the functioning of various genes in hard tissues.


Assuntos
Integrases , Peptídeos , Masculino , Camundongos , Animais , Integrases/genética , Integrases/metabolismo , Camundongos Transgênicos , Peptídeos/genética , Diferenciação Celular/genética , Camundongos Knockout , Proteínas da Matriz Extracelular/genética
2.
Cytokine ; 157: 155936, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35738051

RESUMO

Brown/beige adipocytes, which are derived from skeletal muscle/smooth muscle-lineage cells, consume excess energy as heat through the expression of mitochondrial uncoupling protein 1 (UCP1). Previous studies have shown that forced expression of PR/SET domain (PRDM)-16 or early B-cell factor (EBF)-2 induced UCP1-positive adipocytes in C2C12 myogenic cells. Here, we explored the culture conditions to induce Ucp1 expression in C2C12 cells without introducing exogenous genes. Treatment with rosiglitazone (a peroxisome proliferator-activated receptor (PPAR)-γ agonist), GW501516 (a PPARδ agonist), and bone morphogenetic protein (BMP)-7 for 8 days efficiently increased Ucp1 expression in response to treatment with forskolin, an activator of the protein kinase A pathway. BMP7 dose-dependently increased forskolin-induced Ucp1 expression in the presence of rosiglitazone and GW501516; however, GW501516 was not required for Ucp1 induction. Additionally, the structurally related proteins, BMP6 and BMP9, efficiently increased forskolin-induced Ucp1 expression in rosiglitazone-treated cells. UCP1 protein was localized in cells with lipid droplets, but adipocytes were not always positive for UCP1. Continuous treatment with BMP7 was needed for the efficient induction of Ucp1 by forskolin treatment. Significant expression of Prdm16 was not detected, irrespective of the treatment, and treatment with rosiglitazone, GW501516, and BMP7 did not affect the expression levels of Ebf2. Fibroblast growth factor receptor (Fgfr)-3 expression levels were increased by BMP9 in rosiglitazone-treated cells, and molecules that upregulate Fgfr3 transcription partly overlapped with those that stimulate Ucp1 transcription. The present results provide basic information on the practical differentiation of myogenic cells to brown adipocytes.


Assuntos
Canais Iônicos , Proteínas Mitocondriais , Adipócitos Marrons , Tecido Adiposo Marrom/metabolismo , Colforsina/metabolismo , Colforsina/farmacologia , Canais Iônicos/genética , Canais Iônicos/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , PPAR gama/metabolismo , Rosiglitazona/farmacologia , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
3.
Development ; 144(1): 63-73, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28049690

RESUMO

Adapting the energy metabolism state to changing bioenergetic demands is essential for mammalian development accompanying massive cell proliferation and cell differentiation. However, it remains unclear how developing embryos meet the changing bioenergetic demands during the chorioallantoic branching (CB) stage, when the maternal-fetal exchange of gases and nutrients is promoted. In this study, using metabolome analysis with mass-labeled glucose, we found that developing embryos redirected glucose carbon flow into the pentose phosphate pathway via suppression of the key glycolytic enzymes PFK-1 and aldolase during CB. Concomitantly, embryos exhibited an increase in lactate pool size and in the fractional contribution of glycolysis to lactate biosynthesis. Imaging mass spectrometry visualized lactate-rich tissues, such as the dorsal or posterior neural tube, somites and head mesenchyme. Furthermore, we found that the heterochronic gene Lin28a could act as a regulator of the metabolic changes observed during CB. Perturbation of glucose metabolism rewiring by suppressing Lin28a downregulation resulted in perinatal lethality. Thus, our work demonstrates that developing embryos rewire glucose metabolism following CB for normal development.


Assuntos
Membrana Corioalantoide/embriologia , Membrana Corioalantoide/metabolismo , Metabolismo Energético/genética , Frutose-Bifosfato Aldolase/genética , Glucose/metabolismo , Fosfofrutoquinase-1/genética , Animais , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Feminino , Frutose-Bifosfato Aldolase/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Glicólise/genética , Troca Materno-Fetal/genética , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Fosfofrutoquinase-1/metabolismo , Gravidez , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia
4.
J Immunol ; 195(4): 1883-90, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26170385

RESUMO

Immunodeficient hosts exhibit high acceptance of xenogeneic or neoplastic cells mainly due to lack of adaptive immunity, although it still remains to be elucidated how innate response affects the engraftment. IL-2R common γ-chain (IL-2Rγc) signaling is required for development of NK cells and a subset of dendritic cells producing IFN-γ. To better understand innate response in the absence of adaptive immunity, we examined amounts of metastatic foci in the livers after intrasplenic transfer of human colon cancer HCT116 cells into NOD/SCID versus NOD/SCID/IL-2Rγc (null) (NOG) hosts. The intravital microscopic imaging of livers in the hosts depleted of NK cells and/or macrophages revealed that IL-2Rγc function critically contributes to elimination of cancer cells without the need for NK cells and macrophages. In the absence of IL-2Rγc, macrophages play a role in the defense against tumors despite the NOD Sirpa allele, which allows human CD47 to bind to the encoded signal regulatory protein α to inhibit macrophage phagocytosis of human cells. Analogous experiments using human pancreas cancer MIA PaCa-2 cells provided findings roughly similar to those from the experiments using HCT116 cells except for lack of suppression of metastases by macrophages in NOG hosts. Administration of mouse IFN-γ to NOG hosts appeared to partially compensate lack of IL-2Rγc-dependent elimination of transferred HCT116 cells. These results provide insights into the nature of innate response in the absence of adaptive immunity, aiding in developing tumor xenograft models in experimental oncology.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Subunidade gama Comum de Receptores de Interleucina/genética , Neoplasias/genética , Neoplasias/imunologia , Receptores de Interleucina-2/genética , Animais , Pontos de Checagem do Ciclo Celular , Modelos Animais de Doenças , Células HCT116 , Humanos , Interferon gama/administração & dosagem , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Neoplasias Hepáticas/secundário , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/patologia
5.
Microcirculation ; 23(3): 183-90, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26537442

RESUMO

CO is a gaseous mediator generated by HO. Our previous studies revealed that CO generated from inducible HO-1 or from constitutive HO-2 modulates function of different heme proteins or enzymes through binding to their prosthetic ferrous heme to alter their structures, regulating biological function of cells and organs. Such CO-directed target macromolecules include sGC and CBS. In the liver, CO serves as a sinusoidal dilator through its action on sGC in hepatic stellate cells, while the same gas accounts for vasoconstrictor that inhibits H2S generated by CO-sensitive CBS in astrocytes. Since molecular O2 is a substrate for HO, the latter mechanism contributes to hypoxic vasodilation in neurovascular units. We have recently uncovered that stress-inducible CO in and around cancer cells suppresses CBS to result in decreased methylation of PFKFB3, the enzyme regulating PFK-1, leading to a shift of glucose biotransformation from glycolysis toward pentose phosphate pathway; such a metabolic remodeling causes chemoresistance through increasing NADPH and reduced glutathione under stress conditions for cancer cells. This article reviews the intriguing networks of CO-sensitive metabolic regulatory mechanisms in microcirculation and cancer.


Assuntos
Monóxido de Carbono/metabolismo , Cistationina beta-Sintase/metabolismo , Sulfeto de Hidrogênio/metabolismo , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Capilares/metabolismo , Capilares/patologia , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1/metabolismo , Humanos , Células de Kupffer/metabolismo , Células de Kupffer/patologia , Fígado/patologia , Neoplasias Hepáticas/patologia , Transmissão Sináptica
6.
Nitric Oxide ; 46: 102-13, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25461272

RESUMO

CD44 expressed in cancer cells was shown to stabilize cystine transporter (xCT) that uptakes cystine and excretes glutamate to supply cysteine as a substrate for reduced glutathione (GSH) for survival. While targeting CD44 serves as a potentially therapeutic stratagem to attack cancer growth and chemoresistance, the impact of CD44 targeting in cancer cells on metabolic systems of tumors and host tissues in vivo remains to be fully determined. This study aimed to reveal effects of CD44 silencing on alterations in energy metabolism and sulfur-containing metabolites in vitro and in vivo using capillary electrophoresis-mass spectrometry and quantitative imaging mass spectrometry (Q-IMS), respectively. In an experimental model of xenograft transplantation of human colon cancer HCT116 cells in superimmunodeficient NOG mice, snap-frozen liver tissues containing metastatic tumors were examined by Q-IMS. As reported previously, short hairpin CD44 RNA interference (shCD44) in cancer cells caused significant regression of tumor growth in the host liver. Under these circumstances, the CD44 knockdown suppressed polyamines, GSH and energy charges not only in metastatic tumors but also in the host liver. In culture, HCT116 cells treated with shCD44 decreased total amounts of methionine-pool metabolites including spermidine and spermine, and reactive cysteine persulfides, suggesting roles of these metabolites for cancer growth. Collectively, these results suggest that CD44 expressed in cancer accounts for a key regulator of metabolic interplay between tumor and the host tissue.


Assuntos
Cisteína/metabolismo , Receptores de Hialuronatos/metabolismo , Metaboloma/fisiologia , Neoplasias/metabolismo , Animais , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , Receptores de Hialuronatos/genética , Fígado/química , Fígado/metabolismo , Masculino , Espectrometria de Massas , Metaboloma/efeitos dos fármacos , Metaboloma/genética , Metabolômica , Camundongos , Neoplasias/genética , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Transplante Heterólogo
7.
Shinrigaku Kenkyu ; 85(2): 121-9, 2014 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-25016832

RESUMO

The present study examined the negative evaluations and discrimination against smokers among the Japanese. In Study 1, 52 students rated one of four target-persons differentially depicted in terms of gender and smoking habit using scales to measure coolness, sociability, intellectuality, and earnestness. The results showed that participants rated smokers more negatively than nonsmokers except for sociability. Those who perceived smoking as controllable rated smokers' earnestness even more negatively, suggesting that the negative evaluations are partially moderated by the perceived controllability of smoking. To examine a hypothesis that negative evaluations of smokers would mediate discrimination, in Study 2 we measured how participants (96 students) responded to target persons asking for a loan or a job, as well as their ratings of the targets on the Big Five personality dimensions. The results support the hypothesis of mediation.


Assuntos
Fumar , Discriminação Social , Feminino , Humanos , Estilo de Vida , Masculino , Estereotipagem , Adulto Jovem
8.
Gene ; 893: 147907, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-37858745

RESUMO

Long noncoding RNAs (lncRNAs) have recently been proved to be functional in the testis. Tesra, a testis-specific lncRNA, was suggested to activate the transcription of Prss42/Tessp-2, a gene that is involved in meiotic progression, in mouse spermatocytes. To reveal the molecular mechanism underlying the activation, we searched for Tesra-binding proteins by a Ribotrap assay followed by LC-MS/MS analysis and identified polypyrimidine tract binding protein 2 (PTBP2) as a candidate. Analysis of public RNA-seq data and our qRT-PCR results indicated that Ptbp2 mRNA showed an expression pattern similar to the expression patterns of Tesra and Prss42/Tessp-2 during testis development. Moreover, PTBP2 was found to be associated with Tesra in testicular germ cells by RNA immunoprecipitation. To evaluate the effect of PTBP2 on the Prss42/Tessp-2 promoter, we established an in vitro reporter gene assay system in which Tesra expression could be induced by the Tet-on system and thereby Prss42/Tessp-2 promoter activity could be increased. In this system, the Prss42/Tessp-2 promoter activity was significantly decreased by the knockdown of PTBP2. These results suggest that PTBP2 contributes to Prss42/Tessp-2 transcriptional activation by Tesra in spermatocytes. The finding provides a precious example of a molecular mechanism of testis lncRNA functioning in spermatogenesis.


Assuntos
RNA Longo não Codificante , Testículo , Masculino , Camundongos , Animais , Testículo/metabolismo , RNA Longo não Codificante/metabolismo , Cromatografia Líquida , Espectrometria de Massas em Tandem , Espermatogênese/fisiologia , Espermatócitos/metabolismo
9.
Elife ; 122024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38573813

RESUMO

Metabolic pathways are plastic and rapidly change in response to stress or perturbation. Current metabolic profiling techniques require lysis of many cells, complicating the tracking of metabolic changes over time after stress in rare cells such as hematopoietic stem cells (HSCs). Here, we aimed to identify the key metabolic enzymes that define differences in glycolytic metabolism between steady-state and stress conditions in murine HSCs and elucidate their regulatory mechanisms. Through quantitative 13C metabolic flux analysis of glucose metabolism using high-sensitivity glucose tracing and mathematical modeling, we found that HSCs activate the glycolytic rate-limiting enzyme phosphofructokinase (PFK) during proliferation and oxidative phosphorylation (OXPHOS) inhibition. Real-time measurement of ATP levels in single HSCs demonstrated that proliferative stress or OXPHOS inhibition led to accelerated glycolysis via increased activity of PFKFB3, the enzyme regulating an allosteric PFK activator, within seconds to meet ATP requirements. Furthermore, varying stresses differentially activated PFKFB3 via PRMT1-dependent methylation during proliferative stress and via AMPK-dependent phosphorylation during OXPHOS inhibition. Overexpression of Pfkfb3 induced HSC proliferation and promoted differentiated cell production, whereas inhibition or loss of Pfkfb3 suppressed them. This study reveals the flexible and multilayered regulation of HSC glycolytic metabolism to sustain hematopoiesis under stress and provides techniques to better understand the physiological metabolism of rare hematopoietic cells.


Assuntos
Glicólise , Fosfofrutoquinase-2 , Animais , Camundongos , Trifosfato de Adenosina/metabolismo , Anaerobiose , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Fosforilação Oxidativa , Fosfofrutoquinase-2/genética , Fosfofrutoquinase-2/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo
10.
Cancer Res ; 84(7): 1065-1083, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38383964

RESUMO

Triple-negative breast cancer (TNBC) chemoresistance hampers the ability to effectively treat patients. Identification of mechanisms driving chemoresistance can lead to strategies to improve treatment. Here, we revealed that protein arginine methyltransferase-1 (PRMT1) simultaneously methylates D-3-phosphoglycerate dehydrogenase (PHGDH), a critical enzyme in serine synthesis, and the glycolytic enzymes PFKFB3 and PKM2 in TNBC cells. 13C metabolic flux analyses showed that PRMT1-dependent methylation of these three enzymes diverts glucose toward intermediates in the serine-synthesizing and serine/glycine cleavage pathways, thereby accelerating the production of methyl donors in TNBC cells. Mechanistically, PRMT1-dependent methylation of PHGDH at R54 or R20 activated its enzymatic activity by stabilizing 3-phosphoglycerate binding and suppressing polyubiquitination. PRMT1-mediated PHGDH methylation drove chemoresistance independently of glutathione synthesis. Rather, activation of the serine synthesis pathway supplied α-ketoglutarate and citrate to increase palmitate levels through activation of fatty acid synthase (FASN). Increased palmitate induced protein S-palmitoylation of PHGDH and FASN to further enhance fatty acid synthesis in a PRMT1-dependent manner. Loss of PRMT1 or pharmacologic inhibition of FASN or protein S-palmitoyltransferase reversed chemoresistance in TNBC. Furthermore, IHC coupled with imaging MS in clinical TNBC specimens substantiated that PRMT1-mediated methylation of PHGDH, PFKFB3, and PKM2 correlates with chemoresistance and that metabolites required for methylation and fatty acid synthesis are enriched in TNBC. Together, these results suggest that enhanced de novo fatty acid synthesis mediated by coordinated protein arginine methylation and protein S-palmitoylation is a therapeutic target for overcoming chemoresistance in TNBC. SIGNIFICANCE: PRMT1 promotes chemoresistance in TNBC by methylating metabolic enzymes PFKFB3, PKM2, and PHGDH to augment de novo fatty acid synthesis, indicating that targeting this axis is a potential treatment strategy.


Assuntos
Fosfoglicerato Desidrogenase , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Resistencia a Medicamentos Antineoplásicos , Serina/metabolismo , Palmitatos , Ácidos Graxos , Linhagem Celular Tumoral , Proteína-Arginina N-Metiltransferases/genética , Proteínas Repressoras
11.
Sci Adv ; 9(47): eadg6532, 2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-38000026

RESUMO

Eggs accumulate thousands of translationally repressed mRNAs that are translated into proteins after fertilization to direct diverse developmental processes. However, molecular mechanisms underlying the translation of stored mRNAs after fertilization remain unclear. Here, we report a previously unknown RNA processing of 3' end sequences of mature mRNAs that activates the translation of stored mRNAs. Specifically, 9 to 72 nucleotides at the 3' ends of zebrafish pou5f3 and mouse Pou5f1 mRNAs were deleted in the early stages of development. Reporter assays illustrated the effective translation of the truncated forms of mRNAs. Moreover, promotion and inhibition of the shortening of 3' ends accelerated and attenuated Pou5f3 accumulation, respectively, resulting in defective development. Identification of proteins binding to unprocessed and/or processed mRNAs revealed that mRNA shortening acts as molecular switches. Comprehensive analysis revealed that >250 mRNAs underwent this processing. Therefore, our results provide a molecular principle that triggers the translational activation and directs development.


Assuntos
Biossíntese de Proteínas , Peixe-Zebra , Animais , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Desenvolvimento Embrionário/genética , Processamento Pós-Transcricional do RNA
13.
Redox Biol ; 41: 101926, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33752108

RESUMO

Chemosensitivity to cisplatin derivatives varies among individual patients with intractable malignancies including ovarian cancer, while how to unlock the resistance remain unknown. Ovarian cancer tissues were collected the debulking surgery in discovery- (n = 135) and validation- (n = 47) cohorts, to be analyzed with high-throughput automated immunohistochemistry which identified cystathionine γ-lyase (CSE) as an independent marker distinguishing non-responders from responders to post-operative platinum-based chemotherapy. We aimed to identify CSE-derived metabolites responsible for chemoresistant mechanisms: gold-nanoparticle (AuN)-based surface-enhanced Raman spectroscopy (SERS) was used to enhance electromagnetic fields which enabled to visualize multiple sulfur-containing metabolites through detecting scattering light from Au-S vibration two-dimensionally. Clear cell carcinoma (CCC) who turned out less sensitive to cisplatin than serous adenocarcinoma was classified into two groups by the intensities of SERS intensities at 480 cm-1; patients with greater intensities displayed the shorter overall survival after the debulking surgery. The SERS signals were eliminated by topically applied monobromobimane that breaks sulfane-sulfur bonds of polysulfides to result in formation of sulfodibimane which was detected at 580 cm-1, manifesting the presence of polysulfides in cancer tissues. CCC-derived cancer cell lines in culture were resistant against cisplatin, but treatment with ambroxol, an expectorant degrading polysulfides, renders the cells CDDP-susceptible. Co-administration of ambroxol with cisplatin significantly suppressed growth of cancer xenografts in nude mice. Furthermore, polysulfides, but neither glutathione nor hypotaurine, attenuated cisplatin-induced disturbance of DNA supercoiling. Polysulfide detection by on-tissue SERS thus enables to predict prognosis of cisplatin-based chemotherapy. The current findings suggest polysulfide degradation as a stratagem unlocking cisplatin chemoresistance.


Assuntos
Antineoplásicos , Neoplasias Ovarianas , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Ovarianas/tratamento farmacológico , Análise Espectral Raman , Sulfetos
14.
Hepatology ; 49(1): 141-50, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19085910

RESUMO

UNLABELLED: Carbon monoxide (CO) is a stress-inducible gas generated by heme oxygenase (HO) eliciting adaptive responses against toxicants; however, mechanisms for its reception remain unknown. Serendipitous observation in metabolome analysis in CO-overproducing livers suggested roles of cystathionine beta-synthase (CBS) that rate-limits transsulfuration pathway and H(2)S generation, for the gas-responsive receptor. Studies using recombinant CBS indicated that CO binds to the prosthetic heme, stabilizing 6-coordinated CO-Fe(II)-histidine complex to block the activity, whereas nitric oxide (NO) forms 5-coordinated structure without inhibiting it. The CO-overproducing livers down-regulated H(2)S to stimulate HCO(3) (-)-dependent choleresis: these responses were attenuated by blocking HO or by donating H(2)S. Livers of heterozygous CBS knockout mice neither down-regulated H(2)S nor exhibited the choleresis while overproducing CO. In the mouse model of estradiol-induced cholestasis, CO overproduction by inducing HO-1 significantly improved the bile output through stimulating HCO(3) (-) excretion; such a choleretic response did not occur in the knockout mice. CONCLUSION: Results collected from metabolome analyses suggested that CBS serves as a CO-sensitive modulator of H(2)S to support biliary excretion, shedding light on a putative role of the enzyme for stress-elicited adaptive response against bile-dependent detoxification processes.


Assuntos
Bile/metabolismo , Monóxido de Carbono/fisiologia , Cistationina beta-Sintase/metabolismo , Animais , Masculino , Metaboloma/fisiologia , Camundongos
15.
Adv Exp Med Biol ; 662: 101-7, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20204778

RESUMO

Carbon monoxide (CO) is the stress-inducible gas generated by heme oxygenase (HO). Although the HO/CO system appears to contribute to cell protection and tissue repair under stress conditions, its mode of actions remains largely unknown. We hypothesized that CO might alter the cellular energetic conditions and thereby modulate oxygen metabolism. To examine this hypothesis, we attempted to establish a method to follow the global flux of (13)C-glucose in the cells using metabolomic approaches with liquid chromatography-mass spectrometry (LC-MS/MS). The human monoblastic leukemia cell line U937 was exposed to the CO-releasing molecule (CORM). The CO exposure attenuated the conversion of the mass-labeled glucose to its downstream metabolites, while significantly stimulating its conversion to those for pentose phosphate pathway, suggesting roles of stress-inducible CO in a shift of glucose biotransformation.


Assuntos
Monóxido de Carbono/farmacologia , Cromatografia Líquida/métodos , Glucose/metabolismo , Espectrometria de Massas/métodos , Metaboloma/efeitos dos fármacos , Biotransformação/efeitos dos fármacos , Isótopos de Carbono , Linhagem Celular Tumoral , Humanos , Marcação por Isótopo
16.
Surg Case Rep ; 6(1): 242, 2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-32997223

RESUMO

BACKGROUND: The thoracic cavity is the most frequent site of extrapelvic endometriosis. It exhibits a wide variety of clinical manifestations, such as chest pain, cough, and respiratory distress, and is frequently associated with pelvic endometriosis. Although histological confirmation is the gold standard for a definitive diagnosis, endoscopic identification of the affected area is often difficult. Narrow band imaging (NBI) is an imaging technique that emphasizes vascular structures and is reported to be useful in the diagnosis of pelvic endometriosis. CASE PRESENTATIONS: A 31-year-old woman and 39-year-old woman developed a recurrent right pneumothorax during their menstruation cycles. They both had no medical history suggesting pelvic endometriosis. We planned an elective video-assisted thoracoscopic surgery for the suspicion of thoracic endometriosis. In addition to white light alone, an NBI observation enhanced the microvasculature of the suspected lesions and allowed us to identify the affected area more clearly. Partial resections of the diaphragm were performed. Histopathological and immunohistochemical studies of each specimen confirmed the diagnosis of extrapelvic endometriosis. CONCLUSIONS: NBI may improve the diagnostic accuracy for thoracic endometriosis, especially in clinically suspected patients but without a history of pelvic endometriosis.

17.
Commun Biol ; 3(1): 479, 2020 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-32887925

RESUMO

Progesterone receptor membrane associated component 1 (PGRMC1) exhibits haem-dependent dimerization on cell membrane and binds to EGF receptor and cytochromes P450 to regulate cancer proliferation and chemoresistance. However, its physiological functions remain unknown. Herein, we demonstrate that PGRMC1 is required for adipogenesis, and its expression is significantly enhanced by insulin or thiazolidine, an agonist for PPARγ. The haem-dimerized PGRMC1 interacts with low-density lipoprotein receptors (VLDL-R and LDL-R) or GLUT4 to regulate their translocation to the plasma membrane, facilitating lipid uptake and accumulation, and de-novo fatty acid synthesis in adipocytes. These events are cancelled by CO through interfering with PGRMC1 dimerization. PGRMC1 expression in mouse adipose tissues is enhanced during obesity induced by a high fat diet. Furthermore, adipose tissue-specific PGRMC1 knockout in mice dramatically suppressed high-fat-diet induced adipocyte hypertrophy. Our results indicate a pivotal role of PGRMC1 in developing obesity through its metabolic regulation of lipids and carbohydrates in adipocytes.


Assuntos
Adipócitos/metabolismo , Progressão da Doença , Metabolismo dos Lipídeos , Proteínas de Membrana/metabolismo , Obesidade/patologia , Receptores de Progesterona/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Animais , Monóxido de Carbono/farmacologia , Diferenciação Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Hipertrofia , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/metabolismo , Camundongos , Modelos Biológicos , Obesidade/sangue , Transporte Proteico/efeitos dos fármacos , Receptores de LDL/metabolismo
18.
Clin Cancer Res ; 13(18 Pt 1): 5436-45, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17875773

RESUMO

PURPOSE: We reported previously that reactive oxygen species (ROS) are key mediators of apoptosis induced by a polyphenol, (-)-epigallocatechin-3-gallate (EGCG), in myeloid leukemic cells. This study aimed to further examine the mechanism of ROS-mediated apoptosis induced by EGCG and its relationship to the heme enzyme myeloperoxidase (MPO). EXPERIMENTAL DESIGN: We established stably transfected K562 cells expressing wild-type and mutant MPO. Then, sensitivity against EGCG and other ROS-inducing agent was examined and further investigated the detailed molecular mechanism of ROS-inducing apoptosis in MPO-positive leukemic cells. RESULTS: EGCG rapidly induced apoptosis in MPO-positive leukemia cells. Preincubation of myeloid leukemic cells with the MPO-specific inhibitor, 4-aminobenzoic acid hydrazide, and the heme biosynthesis inhibitor, succinylacetone, resulted in inhibition of the intracellular MPO activity, ROS production, and induction of apoptosis following addition of EGCG. Overexpression of MPO sensitized EGCG-resistant K562 cells to apoptosis induced by EGCG. In contrast, an enzymatically inactive MPO mutant-expressing K562 cell could not respond to EGCG, suggesting that MPO is important for determining the sensitivity to EGCG-induced oxidative stress. Hypochlorous acid scavengers and the hydroxyl radical (.OH) scavenger inhibited EGCG-induced apoptosis in myeloid leukemic cells. The fluorescence intensity of both aminophenyl fluorescein- and hydroxyphenyl fluorescein-loaded myeloid leukemic cells significantly increased on stimulation with EGCG, indicating that EGCG generated highly toxic ROS in myeloid leukemic cells. CONCLUSIONS: These results indicated that highly toxic ROS such as .OH generated via the hydrogen peroxide/MPO/halide system induce apoptosis and that ROS may be the direct mediators of EGCG-induced apoptosis in MPO-positive leukemic cells.


Assuntos
Antioxidantes/farmacologia , Apoptose , Catequina/análogos & derivados , Leucemia Mieloide Aguda/enzimologia , Peroxidase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Compostos de Anilina/farmacologia , Apoptose/efeitos dos fármacos , Catequina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Heme/antagonistas & inibidores , Humanos , Estresse Oxidativo , Peroxidase/antagonistas & inibidores , Peroxidase/genética , Superóxidos/metabolismo
19.
Front Microbiol ; 9: 2902, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555442

RESUMO

Coenzyme A (CoA) is an essential cofactor for numerous cellular reactions in all living organisms. In the protozoan parasite Entamoeba histolytica, CoA is synthesized in a pathway consisting of four enzymes with dephospho-CoA kinase (DPCK) catalyzing the last step. However, the metabolic and physiological roles of E. histolytica DPCK remain elusive. In this study, we took biochemical, reverse genetic, and metabolomic approaches to elucidate role of DPCK in E. histolytica. The E. histolytica genome encodes two DPCK isotypes (EhDPCK1 and EhDPCK2). Epigenetic gene silencing of Ehdpck1 and Ehdpck2 caused significant reduction of DPCK activity, intracellular CoA concentrations, and also led to growth retardation in vitro, suggesting importance of DPCK for CoA synthesis and proliferation. Furthermore, metabolomic analysis showed that suppression of Ehdpck gene expression also caused decrease in the level of acetyl-CoA, and metabolites involved in amino acid, glycogen, hexosamine, nucleic acid metabolisms, chitin, and polyamine biosynthesis. The kinetic properties of E. histolytica and human DPCK showed remarkable differences, e.g., the Km values of E. histolytica and human DPCK were 58-114 and 5.2 µM toward dephospho-CoA and 15-20 and 192 µM for ATP, respectively. Phylogenetic analysis also supported the uniqueness of the amebic enzyme compared to the human counterpart. These biochemical, evolutionary features, and physiological importance of EhDPCKs indicate that EhDPCK represents the rational target for the development of anti-amebic agents.

20.
Int J Parasitol Drugs Drug Resist ; 8(1): 125-136, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29518650

RESUMO

The Coenzyme A (CoA), as a cofactor involved in >100 metabolic reactions, is essential to the basic biochemistry of life. Here, we investigated the CoA biosynthetic pathway of Entamoeba histolytica (E. histolytica), an enteric protozoan parasite responsible for human amebiasis. We identified four key enzymes involved in the CoA pathway: pantothenate kinase (PanK, EC 2.7.1.33), bifunctional phosphopantothenate-cysteine ligase/decarboxylase (PPCS-PPCDC), phosphopantetheine adenylyltransferase (PPAT) and dephospho-CoA kinase (DPCK). Cytosolic enzyme PanK, was selected for further biochemical, genetic, and phylogenetic characterization. Since E. histolytica PanK (EhPanK) is physiologically important and sufficiently divergent from its human orthologs, this enzyme represents an attractive target for the development of novel anti-amebic chemotherapies. Epigenetic gene silencing of PanK resulted in a significant reduction of PanK activity, intracellular CoA concentrations, and growth retardation in vitro, reinforcing the importance of this gene in E. histolytica. Furthermore, we screened the Kitasato Natural Products Library for inhibitors of recombinant EhPanK, and identified 14 such compounds. One compound demonstrated moderate inhibition of PanK activity and cell growth at a low concentration, as well as differential toxicity towards E. histolytica and human cells.


Assuntos
Antiprotozoários/isolamento & purificação , Vias Biossintéticas/efeitos dos fármacos , Entamoeba histolytica/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/isolamento & purificação , Amebíase/tratamento farmacológico , Produtos Biológicos , Vias Biossintéticas/genética , Coenzima A/análise , Coenzima A/biossíntese , Coenzima A/genética , Sistemas de Liberação de Medicamentos , Descoberta de Drogas , Entamoeba histolytica/efeitos dos fármacos , Entamoeba histolytica/genética , Entamoeba histolytica/crescimento & desenvolvimento , Epigenômica , Inativação Gênica , Humanos , Nucleotidiltransferases/genética , Nucleotidiltransferases/isolamento & purificação , Peptídeo Sintases/genética , Peptídeo Sintases/isolamento & purificação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Filogenia , Bibliotecas de Moléculas Pequenas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA