Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Int J Obes (Lond) ; 46(5): 1009-1017, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35082385

RESUMO

BACKGROUND: Preliminary data suggested that fat embolism could explain the importance of visceral obesity as a critical determinant of coronavirus disease-2019 (COVID-19). METHODS: We performed a comprehensive histomorphologic analysis of autoptic visceral adipose tissue (VAT), lungs and livers of 19 subjects with COVID-19 (COVID-19+), and 23 people without COVID-19 (controls). Human adipocytes (hMADS) infected with SARS-CoV-2 were also studied. RESULTS: Although there were no between-group differences in body-mass-index and adipocytes size, a higher prevalence of CD68+ macrophages among COVID-19+ VAT was detected (p = 0.005) and accompanied by crown-like structures presence, signs of adipocytes stress and death. Consistently, human adipocytes were successfully infected by SARS-CoV-2 in vitro and displayed lower cell viability. Being VAT inflammation associated with lipids spill-over from dead adipocytes, we studied lipids distribution by ORO. Lipids were observed within lungs and livers interstitial spaces, macrophages, endothelial cells, and vessels lumen, features suggestive of fat embolism syndrome, more prevalent among COVID-19+ (p < 0.001). Notably, signs of fat embolism were more prevalent among people with obesity (p = 0.03) independently of COVID-19 diagnosis, suggesting that such condition may be an obesity complication exacerbated by SARS-CoV-2 infection. Importantly, all infected subjects' lungs presented lipids-rich (ORO+) hyaline membranes, formations associated with COVID-19-related pneumonia, present only in one control patient with non-COVID-19-related pneumonia. Importantly, transition aspects between embolic fat and hyaline membranes were also observed. CONCLUSIONS: This study confirms the lung fat embolism in COVID-19+ patients and describes for the first time novel COVID-19-related features possibly underlying the unfavorable prognosis in people with COVID-19 and obesity.


Assuntos
COVID-19 , Embolia Gordurosa , COVID-19/complicações , Teste para COVID-19 , Células Endoteliais/metabolismo , Humanos , Hialina/metabolismo , Inflamação/metabolismo , Gordura Intra-Abdominal/metabolismo , Lipídeos , Pulmão , Obesidade/metabolismo , SARS-CoV-2
2.
J Cell Physiol ; 235(2): 1184-1196, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31294462

RESUMO

Brown and brown-like adipocytes (BBAs) control thermogenesis and are detected in adult humans. They express UCP1, which transforms energy into heat. They appear as promising cells to fight obesity. Deciphering the molecular mechanisms leading to the browning of human white adipocytes or the whitening of BBAs represents a goal to properly and safely control the pathways involved in these processes. Here, we analyzed how drugs endowed with therapeutic potential affect the differentiation of human adipose progenitor-cells into BBAs and/or their phenotype. We showed that HIV-protease inhibitors (PI) reduced UCP1 expression in BBAs modifying their metabolic profile and the mitochondria functionality. Lopinavir (LPV) was more potent than darunavir (DRV), a last PI generation. PPARγ and PGC-1α were decreased in a PI or cell-specific manner, thus altering UCP1's constitutive expression. In addition, LPV altered p38 MAPK phosphorylation, blunting then the ß-adrenergic responses. In contrast, low doses of resveratrol stimulated the activatable expression of UCP1 in a p38 MAPK-dependent manner and counteracted the LPV induced loss of UCP1. This effect was independent of the resveratrol-induced sirtuin-1 expression. Altogether our results uncover how drugs impact crucial components of the networks regulating the expression of the thermogenic signature. They provide important information to control the relevant pathways involved in energy expenditure.


Assuntos
Adipócitos/efeitos dos fármacos , Darunavir/farmacologia , Resveratrol/farmacologia , Proteína Desacopladora 1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adipócitos/metabolismo , Antioxidantes/farmacologia , Linhagem Celular , Colforsina , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores da Protease de HIV/farmacologia , Humanos , Compostos Orgânicos/farmacologia , Fosforilação , Proteína Desacopladora 1/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
3.
BMC Cancer ; 20(1): 784, 2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32819314

RESUMO

BACKGROUND: Cancer cells cooperate with cells that compose their environment to promote tumor growth and invasion. Among them, adipocytes provide lipids used as a source of energy by cancer cells and adipokines that contribute to tumor expansion. Mechanisms supporting the dynamic interactions between cancer cells and stromal adipocytes, however, remain unclear. METHODS: We set-up a co-culture model with breast cancer cells grown in 3D as mammospheres and human adipocytes to accurately recapitulate intrinsic features of tumors, such as hypoxia and cancer cell-adipocytes interactions. RESULTS: Herein, we observed that the lipid droplets' size was reduced in adipocytes adjacent to the mammospheres, mimicking adipocyte morphology on histological sections. We showed that the uncoupling protein UCP1 was expressed in adipocytes close to tumor cells on breast cancer histological sections as well as in adipocytes in contact with the mammospheres. Mammospheres produced adrenomedullin (ADM), a multifactorial hypoxia-inducible peptide while ADM receptors were detected in adipocytes. Stimulation of adipocytes with ADM promoted UCP1 expression and increased HSL phosphorylation, which activated lipolysis. Invalidation of ADM in breast cancer cells dramatically reduced UCP1 expression in adipocytes. CONCLUSIONS: Breast tumor cells secreted ADM that modified cancer-associated adipocytes through paracrine signaling, leading to metabolic changes and delipidation. Hence, ADM appears to be crucial in controlling the interactions between cancer cells and adipocytes and represents an excellent target to hinder them.


Assuntos
Adipócitos/patologia , Adrenomedulina/metabolismo , Neoplasias da Mama/patologia , Comunicação Parácrina , Esferoides Celulares/metabolismo , Adipócitos/citologia , Mama/citologia , Mama/patologia , Hipóxia Celular , Técnicas de Cocultura , Feminino , Humanos , Gotículas Lipídicas/metabolismo , Lipólise , Células MCF-7 , Microambiente Tumoral , Proteína Desacopladora 1/metabolismo
4.
Stem Cells ; 33(8): 2564-73, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25827082

RESUMO

Adipose tissue expansion is well-orchestrated to fulfill the energy demand. It results from adipocyte hypertrophy and hyperplasia due to adipose progenitor cell (APC) expansion and differentiation. Chronic low grade inflammation and hypoxia take place in obese adipose tissue microenvironment. Both of these events were shown to impact the APC pool by promoting increased self-renewal along with a decrease in the APC differentiation potential. However, no common target has been identified so far. Here we show that the immediate early response 3 gene (IER3) is preferentially expressed in APCs and is essential for APC proliferation and self-renewal. Experiments based on RNA interference revealed that impairing IER3 expression altered cell proliferation through ERK1/2 phosphorylation and clonogenicity. IER3 expression was induced by Activin A, which plays a crucial role in adipocyte differentiation as well as by a decrease in oxygen tension through HIF1-induced transcriptional activation. Interestingly, high levels of IER3 were detected in native APCs (CD34+/CD31- cells) isolated from obese patients and conditioned media from obese adipose tissue-macrophages stimulated its expression. Overall, these results indicate that IER3 is a key player in expanding the pool of APC while highlighting the role of distinct effectors found in an obese microenvironment in this process.


Assuntos
Tecido Adiposo/metabolismo , Proteínas Reguladoras de Apoptose/biossíntese , Regulação da Expressão Gênica/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas de Membrana/biossíntese , Nicho de Células-Tronco/fisiologia , Células-Tronco/metabolismo , Tecido Adiposo/citologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células-Tronco/citologia
5.
Biomedicines ; 10(8)2022 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-36009475

RESUMO

Breast adipose tissue (AT) participates in the physiological evolution and remodeling of the mammary gland due to its high plasticity. It is also a favorable microenvironment for breast cancer progression. However, information on the properties of human breast adipose progenitor cells (APCs) involved in breast physiology or pathology is scant. We performed differential enzymatic dissociation of human breast AT lobules. We isolated and characterized two populations of APCs. Here we report that these distinct breast APC populations selectively expressed markers suitable for characterization. The population preferentially expressing ALPL (MSCA1) showed higher adipogenic potential. The population expressing higher levels of INHBA and CD142 acquired myofibroblast characteristics upon TGF-ß treatment and a myo-cancer-associated fibroblast profile in the presence of breast cancer cells. This population expressed the immune checkpoint CD274 (PD-L1) and facilitated the expansion of breast cancer mammospheres compared with the adipogenic population. Indeed, the breast, as with other fat depots, contains distinct types of APCs with differences in their ability to specialize. This indicates that they were differentially involved in breast remodeling. Their interactions with breast cancer cells revealed differences in the potential for tumor dissemination and estrogen receptor expression, and these differences might be relevant to improve therapies targeting the tumor microenvironment.

6.
Biomedicines ; 9(1)2021 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-33466493

RESUMO

Adipose tissue resides in specific depots scattered in peripheral or deeper locations all over the body and it enwraps most of the organs. This tissue is always in a dynamic evolution as it must adapt to the metabolic demand and constraints. It exhibits also endocrine functions important to regulate energy homeostasis. This complex organ is composed of depots able to produce opposite functions to monitor energy: the so called white adipose tissue acts to store energy as triglycerides preventing ectopic fat deposition while the brown adipose depots dissipate it. It is composed of many cell types. Different types of adipocytes constitute the mature cells specialized to store or burn energy. Immature adipose progenitors (AP) presenting stem cells properties contribute not only to the maintenance but also to the expansion of this tissue as observed in overweight or obese individuals. They display a high regeneration potential offering a great interest for cell therapy. In this review, we will depict the attributes of the distinct types of adipocytes and their contribution to the function and metabolic features of adipose tissue. We will examine the specific role and properties of distinct depots according to their location. We will consider their cellular heterogeneity to present an updated picture of this sophisticated tissue. We will also introduce new trends pointing out a rational targeting of adipose tissue for medical applications.

7.
Cells ; 9(10)2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-33049976

RESUMO

Cancer associated fibroblasts (CAFs) are central elements of the microenvironment that control tumor development. In breast cancer, CAFs can originate from adipose progenitors (APs). We, and others, have shown that the primary cilium, an antenna-shaped organelle, controls several aspects of APs' biology. We studied the conversion of human APs into CAFs by breast cancer cell lines (BCCs). Deletion of the cilium of APs by a pharmacological inhibitor, or by siRNA, allow us to demonstrate that the cilium is necessary for the differentiation of APs into CAFs. BCCs increase production of TGF-ß1 by APs, which is a known inducer of CAFs. Pharmacological inhibition of TGF-ß1 signaling in APs prevents their conversion into CAFs. Since we previously showed that deletion of the APs' cilium inhibits TGF-ß1 signaling, we propose that BCCs induce TGF-ß1 production in Aps, which binds to the primary cilium of Aps and leads to their differentiation into CAFs. Inhibition of APs conversion into CAFs induces a loss in some of the biological effects of CAFs since deletion of the cilium of APs decreases their effect on the migration of BCCs. This is the first observation of a function of the cilium of APs in their conversion into CAFs, and its consequences on BCCs.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Cílios/metabolismo , Células-Tronco Mesenquimais/metabolismo , Tecido Adiposo/metabolismo , Neoplasias da Mama/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Cílios/fisiologia , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Células-Tronco Mesenquimais/fisiologia , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Fator de Crescimento Transformador beta1/genética , Microambiente Tumoral/fisiologia
8.
Biochem Biophys Res Commun ; 386(1): 96-100, 2009 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-19501568

RESUMO

HIV-protease inhibitors (PIs) markedly decreased mortality of HIV-infected patients. However, their use has been associated with occurence of metabolic abnormalities the causes of which are not well understood. We report here that lopinavir, one of the most prescribed PI, dose-dependently co-induced insulin resistance and ER stress in human adipocytes obtained from differentiation of precursor cells. Insulin resistance was subsequent to IRS1 phosphorylation defects and resulted in a concentration-dependent decrease of glucose uptake. The major ER stress pathway involved was the phosphorylation of eIF2-alpha. Salubrinal, a selective eIF2-alpha dephosphorylation inhibitor, induced insulin resistance by targeting IRS1 phosphorylation at serine 312 and acted synergistically with LPV when both drugs were used in combination. This study points out the key role of eIF2-alpha phosphorylation in the development of PI-associated insulin resistance and ER stress. Thus, this protein represents a promising therapeutic target for development of new PIs devoid of adverse metabolic effects.


Assuntos
Adipócitos/efeitos dos fármacos , Fator de Iniciação 2 em Eucariotos/metabolismo , Inibidores da Protease de HIV/efeitos adversos , Resistência à Insulina , Pirimidinonas/efeitos adversos , Adipócitos/metabolismo , Linhagem Celular , Cinamatos/farmacologia , Retículo Endoplasmático/metabolismo , Inibidores da Protease de HIV/farmacologia , Humanos , Proteínas Substratos do Receptor de Insulina/metabolismo , Lopinavir , Fosforilação , Pirimidinonas/farmacologia , Tioureia/análogos & derivados , Tioureia/farmacologia
9.
Sci Rep ; 7(1): 2986, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28592814

RESUMO

Maintenance of the adipose tissue requires a proper balance between self-renewal and differentiation of adipose progenitors (AP). Any deregulation leads either to fat overexpansion and obesity or fat loss and consequent lipodystrophies. Depending on the fat pad location, APs and adipocytes are heterogeneous. However, information on the pharmacological sensitivity of distinct APs to drugs known to alter the function of adipose tissue, especially HIV protease inhibitors (PIs) is scant. Here we show that PIs decreased proliferation and clonal expansion of APs, modifying their self-renewal potential. Lopinavir was the most potent PI tested. Decrease in self-renewal was accompanied by a reduced expression of the immediate early response gene IER3, a gene associated with tissue expansion. It was more pronounced in chin-derived APs than in knee-derived APs. Furthermore, lopinavir lowered the activin A-induced ERK1/2 phosphorylation. Expressions of the transcription factor EGR1 and its targets, including INHBA were subsequently altered. Therefore, activin A secretion was reduced leading to a dramatic impairment of APs self-renewal sustained by the activin A autocrine loop. All together, these observations highlight the activin A autocrine loop as a crucial effector to maintain APs self-renewal. Targeting this pathway by HIV-PIs may participate in the induction of unwanted side effects.


Assuntos
Ativinas/antagonistas & inibidores , Tecido Adiposo/citologia , Proliferação de Células/efeitos dos fármacos , Inibidores da Protease de HIV/efeitos adversos , Lopinavir/efeitos adversos , Células-Tronco/fisiologia , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Mapas de Interação de Proteínas/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos
10.
Sci Rep ; 6: 32490, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27577850

RESUMO

Human induced pluripotent stem cells (hiPSCs) show great promise for obesity treatment as they represent an unlimited source of brown/brite adipose progenitors (BAPs). However, hiPSC-BAPs display a low adipogenic capacity compared to adult-BAPs when maintained in a traditional adipogenic cocktail. The reasons of this feature are unknown and hamper their use both in cell-based therapy and basic research. Here we show that treatment with TGFß pathway inhibitor SB431542 together with ascorbic acid and EGF were required to promote hiPSCs-BAP differentiation at a level similar to adult-BAP differentiation. hiPSC-BAPs expressed the molecular identity of adult-UCP1 expressing cells (PAX3, CIDEA, DIO2) with both brown (ZIC1) and brite (CD137) adipocyte markers. Altogether, these data highlighted the critical role of TGFß pathway in switching off hiPSC-brown adipogenesis and revealed novel factors to unlock their differentiation. As hiPSC-BAPs display similarities with adult-BAPs, it opens new opportunities to develop alternative strategies to counteract obesity.


Assuntos
Adipócitos Marrons/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adipócitos Marrons/citologia , Adipócitos Marrons/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Ácido Ascórbico/farmacologia , Benzamidas/farmacologia , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Dioxóis/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Iodotironina Desiodinase Tipo II
12.
Am J Stem Cells ; 1(1): 42-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23671796

RESUMO

Adipose tissue is an alternative source of mesenchymal stem cells and human adipose-derived stem cells (ASCs) display an attractive and substantial therapeutic potential when transplanted in animal models. To this end, an understanding of ASC biology is necessary and the knowledge of mechanisms that maintain ASCs in an undifferentiated state with no loss of differentiation potential during ex vivo expansion represents a crucial step. However, these mechanisms remain to be identified because appropriate human cellular models are scant. In this review we will describe a cellular model isolated from human adipose tissue displaying all the features of stem cells. Then, we will focus on the identification of intrinsic and extrinsic factors regulating the balance between human ASC proliferation and differentiation. We will point out the role of factors secreted by undifferentiated ASCs, such a FGF2, activin A, BMP4, Hedgehog molecules and secreted by adipose tissue macrophages. Finally, we will outline the role of miRNAs in these processes.

13.
Diabetes ; 59(10): 2513-21, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20530742

RESUMO

OBJECTIVE: Growth of white adipose tissue takes place in normal development and in obesity. A pool of adipose progenitors is responsible for the formation of new adipocytes and for the potential of this tissue to expand in response to chronic energy overload. However, factors controlling self-renewal of human adipose progenitors are largely unknown. We investigated the expression profile and the role of activin A in this process. RESEARCH DESIGN AND METHODS: Expression of INHBA/activin A was investigated in three types of human adipose progenitors. We then analyzed at the molecular level the function of activin A during human adipogenesis. We finally investigated the status of activin A in adipose tissues of lean and obese subjects and analyzed macrophage-induced regulation of its expression. RESULTS: INHBA/activin A is expressed by adipose progenitors from various fat depots, and its expression dramatically decreases as progenitors differentiate into adipocytes. Activin A regulates the number of undifferentiated progenitors. Sustained activation or inhibition of the activin A pathway impairs or promotes, respectively, adipocyte differentiation via the C/EBPß-LAP and Smad2 pathway in an autocrine/paracrine manner. Activin A is expressed at higher levels in adipose tissue of obese patients compared with the expression levels in lean subjects. Indeed, activin A levels in adipose progenitors are dramatically increased by factors secreted by macrophages derived from obese adipose tissue. CONCLUSIONS: Altogether, our data show that activin A plays a significant role in human adipogenesis. We propose a model in which macrophages that are located in adipose tissue regulate adipose progenitor self-renewal through activin A.


Assuntos
Ativinas/fisiologia , Tecido Adiposo/citologia , Glucosefosfato Desidrogenase/genética , Obesidade Mórbida/patologia , Células-Tronco/citologia , Magreza/patologia , Ativinas/genética , Ativinas/farmacologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/patologia , Adulto , Diferenciação Celular , Divisão Celular , RNA Polimerases Dirigidas por DNA/efeitos dos fármacos , RNA Polimerases Dirigidas por DNA/genética , Dexametasona/farmacologia , Regulação da Expressão Gênica , Glucosefosfato Desidrogenase/efeitos dos fármacos , Humanos , Obesidade Mórbida/genética , Obesidade Mórbida/prevenção & controle , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Proteína de Ligação a TATA-Box/efeitos dos fármacos , Proteína de Ligação a TATA-Box/genética
14.
J Biol Chem ; 279(29): 30316-25, 2004 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-15131110

RESUMO

The NAD(P)H cytochrome b5 oxidoreductase, Ncb5or (previously named b5+b5R), is widely expressed in human tissues and broadly distributed among the animal kingdom. NCB5OR is the first example of an animal flavohemoprotein containing cytochrome b5 and chrome b5 reductase cytodomains. We initially reported human NCB5OR to be a 487-residue soluble protein that reduces cytochrome c, methemoglobin, ferricyanide, and molecular oxygen in vitro. Bioinformatic analysis of genomic sequences suggested the presence of an upstream start codon. We confirm that endogenous NCB5OR indeed has additional NH2-terminal residues. By performing fractionation of subcellular organelles and confocal microscopy, we show that NCB5OR colocalizes with calreticulin, a marker for endoplasmic reticulum. Recombinant NCB5OR is soluble and has stoichiometric amounts of heme and flavin adenine dinucleotide. Resonance Raman spectroscopy of NCB5OR presents typical signatures of a six-coordinate low-spin heme similar to those found in other cytochrome b5 proteins. Kinetic measurements showed that full-length and truncated NCB5OR reduce cytochrome c actively in vitro. However, both full-length and truncated NCB5OR produce superoxide from oxygen with slow turnover rates: kcat = approximately 0.05 and approximately 1 s(-1), respectively. The redox potential at the heme center of NCB5OR is -108 mV, as determined by potentiometric titrations. Taken together, these data suggest that endogenous NCB5OR is a soluble NAD(P)H reductase preferentially reducing substrate(s) rather than transferring electrons to molecular oxygen and therefore not an NAD(P)H oxidase for superoxide production. The subcellular localization and redox properties of NCB5OR provide important insights into the biology of NCB5OR and the phenotype of the Ncb5or-null mouse.


Assuntos
Citocromo-B(5) Redutase/biossíntese , Citocromos b5/metabolismo , Retículo Endoplasmático/metabolismo , NADH NADPH Oxirredutases/metabolismo , Animais , Sequência de Bases , Western Blotting , Células COS , Calreticulina/metabolismo , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Biologia Computacional , Citocromo-B(5) Redutase/química , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Feminino , Ferricianetos/química , Heme/química , Humanos , Cinética , Fígado/metabolismo , Metemoglobina/química , Camundongos , Microscopia Confocal , Dados de Sequência Molecular , Oxirredução , Oxigênio/metabolismo , Fenótipo , Fótons , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Homologia de Sequência do Ácido Nucleico , Análise Espectral Raman , Frações Subcelulares/metabolismo , Superóxidos/química , Fatores de Tempo , Transfecção , Raios Ultravioleta
15.
Proc Natl Acad Sci U S A ; 101(29): 10750-5, 2004 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-15247412

RESUMO

NCB5OR is a highly conserved NAD(P)H reductase that contains a cytochrome b5-like domain at the N terminus and a cytochrome b5 reductase-like domain at the C terminus. The enzyme is located in the endoplasmic reticulum (ER) and is widely expressed in organs and tissues. Targeted inactivation of this gene in mice has no impact on embryonic or fetal viability. At 4 weeks of age, Ncb5or-/- mice have normal blood glucose levels but impaired glucose tolerance. Isolated Ncb5or-/- islets have markedly impaired glucose- or arginine-stimulated insulin secretion. By 7 weeks of age, these mice develop severe hyperglycemia with markedly decreased serum insulin levels and nearly normal insulin tolerance. As the animals age, there is a progressive loss of beta cells in pancreatic islets, but there is no loss of alpha, delta, or PP cells. Electron microscopy reveals degranulation of beta cells and hypertrophic and hyperplastic mitochondria, some of which contain electron dense inclusions. Four-week-old Ncb5or-/- mice have enhanced sensitivity to the diabetogenic agent streptozotocin. NCB5OR appears to play a critical role in protecting pancreatic beta cells against oxidant stress.


Assuntos
Citocromo-B(5) Redutase/metabolismo , Diabetes Mellitus/metabolismo , Insulina/metabolismo , Oxirredutases/metabolismo , Animais , Glicemia/metabolismo , Citocromo-B(5) Redutase/genética , Diabetes Mellitus/genética , Feminino , Glucagon/metabolismo , Teste de Tolerância a Glucose , Homeostase , Humanos , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Oxirredutases/genética , Estreptozocina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA