Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Cell Biol ; 44(4): 138-148, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38644795

RESUMO

Pharmacologic inhibitors of cellular hydroxylase oxygen sensors are protective in multiple preclinical in vivo models of inflammation. However, the molecular mechanisms underlying this regulation are only partly understood, preventing clinical translation. We previously proposed a new mechanism for cellular oxygen sensing: oxygen-dependent, (likely) covalent protein oligomer (oxomer) formation. Here, we report that the oxygen sensor factor inhibiting HIF (FIH) forms an oxomer with the NF-κB inhibitor ß (IκBß). The formation of this protein complex required FIH enzymatic activity and was prevented by pharmacologic inhibitors. Oxomer formation was highly hypoxia-sensitive and very stable. No other member of the IκB protein family formed an oxomer with FIH, demonstrating that FIH-IκBß oxomer formation was highly selective. In contrast to the known FIH-dependent oxomer formation with the deubiquitinase OTUB1, FIH-IκBß oxomer formation did not occur via an IκBß asparagine residue, but depended on the amino acid sequence VAERR contained within a loop between IκBß ankyrin repeat domains 2 and 3. Oxomer formation prevented IκBß from binding to its primary interaction partners p65 and c-Rel, subunits of NF-κB, the master regulator of the cellular transcriptional response to pro-inflammatory stimuli. We therefore propose that FIH-mediated oxomer formation with IκBß contributes to the hypoxia-dependent regulation of inflammation.


Assuntos
NF-kappa B , Humanos , NF-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Ligação Proteica , Hipóxia Celular , Oxigênio/metabolismo , Células HEK293 , Oxigenases de Função Mista/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Hipóxia/metabolismo , Proteínas Repressoras
2.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35163456

RESUMO

Dysregulated energy metabolism is a major contributor to a multitude of pathologies, including obesity and diabetes. Understanding the regulation of metabolic homeostasis is of utmost importance for the identification of therapeutic targets for the treatment of metabolically driven diseases. We previously identified the deubiquitinase OTUB1 as substrate for the cellular oxygen sensor factor-inhibiting HIF (FIH) with regulatory effects on cellular energy metabolism, but the physiological relevance of OTUB1 is unclear. Here, we report that the induced global deletion of OTUB1 in adult mice (Otub1 iKO) elevated energy expenditure, reduced age-dependent body weight gain, facilitated blood glucose clearance and lowered basal plasma insulin levels. The respiratory exchange ratio was maintained, indicating an unaltered nutrient oxidation. In addition, Otub1 deletion in cells enhanced AKT activity, leading to a larger cell size, higher ATP levels and reduced AMPK phosphorylation. AKT is an integral part of insulin-mediated signaling and Otub1 iKO mice presented with increased AKT phosphorylation following acute insulin administration combined with insulin hypersensitivity. We conclude that OTUB1 is an important regulator of metabolic homeostasis.


Assuntos
Trifosfato de Adenosina/metabolismo , Cisteína Endopeptidases/genética , Deleção de Genes , Resistência à Insulina/genética , Insulina/administração & dosagem , Oxigenases de Função Mista/metabolismo , Adenilato Quinase/metabolismo , Animais , Glicemia , Peso Corporal , Tamanho Celular , Células Cultivadas , Cisteína Endopeptidases/metabolismo , Metabolismo Energético , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Insulina/efeitos adversos , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo
3.
FASEB J ; 35(12): e22039, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34793600

RESUMO

OTUB1 is one of the most highly expressed deubiquitinases, counter-regulating the two most abundant ubiquitin chain types. OTUB1 expression is linked to the development and progression of lung cancer and idiopathic pulmonary fibrosis in humans. However, the physiological function of OTUB1 is unknown. Here, we show that constitutive whole-body Otub1 deletion in mice leads to perinatal lethality by asphyxiation. Analysis of (single-cell) RNA sequencing and proteome data demonstrated that OTUB1 is expressed in all lung cell types with a particularly high expression during late-stage lung development (E16.5, E18.5). At E18.5, the lungs of animals with Otub1 deletion presented with increased cell proliferation that decreased saccular air space and prevented inhalation. Flow cytometry-based analysis of E18.5 lung tissue revealed that Otub1 deletion increased proliferation of major lung parenchymal and mesenchymal/other non-hematopoietic cell types. Adult mice with conditional whole-body Otub1 deletion (wbOtub1del/del ) also displayed increased lung cell proliferation in addition to hyperventilation and failure to adapt the respiratory pattern to hypoxia. On the molecular level, Otub1 deletion enhanced mTOR signaling in embryonic and adult lung tissues. Based on these results, we propose that OTUB1 is a negative regulator of mTOR signaling with essential functions for lung cell proliferation, lung development, adult lung tissue homeostasis, and respiratory regulation.


Assuntos
Proliferação de Células , Cisteína Endopeptidases/fisiologia , Homeostase , Hiperventilação/patologia , Pneumopatias/patologia , Insuficiência Respiratória/patologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Feminino , Hiperventilação/etiologia , Pneumopatias/etiologia , Pneumopatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Insuficiência Respiratória/etiologia , Serina-Treonina Quinases TOR/genética
4.
Redox Biol ; 26: 101265, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31299612

RESUMO

Protein:protein interactions are the basis of molecular communication and are usually of transient non-covalent nature, while covalent interactions other than ubiquitination are rare. For cellular adaptations, the cellular oxygen and peroxide sensor factor inhibiting HIF (FIH) confers oxygen and oxidant stress sensitivity to the hypoxia inducible factor (HIF) by asparagine hydroxylation. We investigated whether FIH contributes to hypoxia adaptation also through other mechanisms and identified a hypoxia sensitive, likely covalent, bond formation by FIH with several client proteins, including the deubiquitinase ovarian tumor domain containing ubiquitin aldehyde binding protein 1 (OTUB1). Biochemical analyses were consistent with a co-translational amide bond formation between FIH and OTUB1, occurring within mammalian and bacterial cells but not between separately purified proteins. Bond formation is catalysed by FIH and highly dependent on oxygen availability in the cellular microenvironment. Within cells, a heterotrimeric complex is formed, consisting of two FIH and one covalently linked OTUB1. Complexation of OTUB1 by FIH regulates OTUB1 deubiquitinase activity. Our findings reveal an alternative mechanism for hypoxia adaptation with remarkably high oxygen sensitivity, mediated through covalent protein-protein interactions catalysed by an asparagine modifying dioxygenase.


Assuntos
Cisteína Endopeptidases/genética , Fator 1 Induzível por Hipóxia/metabolismo , Oxigênio/metabolismo , Linhagem Celular Tumoral , Cisteína Endopeptidases/metabolismo , Enzimas Desubiquitinantes , Humanos , Espectrometria de Massas , Oxirredução , Oxigênio/química
5.
Kidney Int ; 95(2): 375-387, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30502050

RESUMO

Erythropoietin (Epo) is essential for erythropoiesis and is mainly produced by the fetal liver and the adult kidney following hypoxic stimulation. Epo regulation is commonly studied in hepatoma cell lines, but differences in Epo regulation between kidney and liver limit the understanding of Epo dysregulation in polycythaemia and anaemia. To overcome this limitation, we have generated a novel transgenic mouse model expressing Cre recombinase specifically in the active fraction of renal Epo-producing (REP) cells. Crossing with reporter mice confirmed the inducible and highly specific tagging of REP cells, located in the corticomedullary border region where there is a steep drop in oxygen bioavailability. A novel method was developed to selectively grow primary REP cells in culture and to generate immortalized clonal cell lines, called fibroblastoid atypical interstitial kidney (FAIK) cells. FAIK cells show very early hypoxia-inducible factor (HIF)-2α induction, which precedes Epo transcription. Epo induction in FAIK cells reverses rapidly despite ongoing hypoxia, suggesting a cell autonomous feedback mechanism. In contrast, HIF stabilizing drugs resulted in chronic Epo induction in FAIK cells. RNA sequencing of three FAIK cell lines derived from independent kidneys revealed a high degree of overlap and suggests that REP cells represent a unique cell type with properties of pericytes, fibroblasts, and neurons, known as telocytes. These novel cell lines may be helpful to investigate myofibroblast differentiation in chronic kidney disease and to elucidate the molecular mechanisms of HIF stabilizing drugs currently in phase III studies to treat anemia in end-stage kidney disease.


Assuntos
Eritropoetina/metabolismo , Telócitos/patologia , Fatores de Transcrição/metabolismo , Anemia/etiologia , Anemia/patologia , Animais , Hipóxia Celular , Linhagem Celular , Eritropoetina/genética , Retroalimentação Fisiológica , Rim/citologia , Rim/patologia , Camundongos , Camundongos Transgênicos , Cultura Primária de Células , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/patologia , Telócitos/metabolismo
6.
J Biol Chem ; 292(27): 11561-11571, 2017 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-28507099

RESUMO

CO2 is a physiological gas normally produced in the body during aerobic respiration. Hypercapnia (elevated blood pCO2 >≈50 mm Hg) is a feature of several lung pathologies, e.g. chronic obstructive pulmonary disease. Hypercapnia is associated with increased susceptibility to bacterial infections and suppression of inflammatory signaling. The NF-κB pathway has been implicated in these effects; however, the molecular mechanisms underpinning cellular sensitivity of the NF-κB pathway to CO2 are not fully elucidated. Here, we identify several novel CO2-dependent changes in the NF-κB pathway. NF-κB family members p100 and RelB translocate to the nucleus in response to CO2 A cohort of RelB protein-protein interactions (e.g. with Raf-1 and IκBα) are altered by CO2 exposure, although others are maintained (e.g. with p100). RelB is processed by CO2 in a manner dependent on a key C-terminal domain located in its transactivation domain. Loss of the RelB transactivation domain alters NF-κB-dependent transcriptional activity, and loss of p100 alters sensitivity of RelB to CO2 Thus, we provide molecular insight into the CO2 sensitivity of the NF-κB pathway and implicate altered RelB/p100-dependent signaling in the CO2-dependent regulation of inflammatory signaling.


Assuntos
Dióxido de Carbono/imunologia , Hipercapnia/imunologia , Subunidade p52 de NF-kappa B/imunologia , Transdução de Sinais/imunologia , Fator de Transcrição RelB/imunologia , Células A549 , Animais , Humanos , Hipercapnia/genética , Hipercapnia/patologia , Camundongos , Subunidade p52 de NF-kappa B/genética , Domínios Proteicos , Transdução de Sinais/genética , Fator de Transcrição RelB/genética , Transcrição Gênica/genética , Transcrição Gênica/imunologia
7.
Exp Cell Res ; 356(2): 152-159, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28315321

RESUMO

The hypoxia inducible factor (HIF) pathway and the ubiquitin system represent major cellular processes that are involved in the regulation of a plethora of cellular signaling pathways and tissue functions. The ubiquitin system controls the ubiquitination of proteins, which is the covalent linkage of one or several ubiquitin molecules to specific targets. This ubiquitination is catalyzed by approximately 1000 different E3 ubiquitin ligases and can lead to different effects, depending on the type of internal ubiquitin chain linkage. The best-studied function is the targeting of proteins for proteasomal degradation. The activity of E3 ligases is antagonized by proteins called deubiquitinases (or deubiquitinating enzymes), which negatively regulate ubiquitin chains. This is performed in most cases by the catalytic removal of these chains from the targeted protein. The HIF pathway is regulated in an oxygen-dependent manner by oxygen-sensing hydroxylases. Covalent modification of HIFα subunits leads to the recruitment of an E3 ligase complex via the von Hippel-Lindau (VHL) protein and the subsequent polyubiquitination and proteasomal degradation of HIFα subunits, demonstrating the regulation of the HIF pathway by the ubiquitin system. This unidirectional effect of an E3 ligase on the HIF pathway is the best-studied example for the interplay between these two important cellular processes. However, additional regulatory mechanisms of the HIF pathway through the ubiquitin system are emerging and, more recently, also the reciprocal regulation of the ubiquitin system through components of the HIF pathway. Understanding these mechanisms and their relevance for the activity of each other is of major importance for the comprehensive elucidation of the oxygen-dependent regulation of cellular processes. This review describes the current knowledge of the functional bidirectional interplay between the HIF pathway and the ubiquitin system on the protein level.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Oxigênio/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina/metabolismo , Animais , Humanos , Proteínas Supressoras de Tumor/metabolismo
8.
Free Radic Biol Med ; 101: 129-142, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27717868

RESUMO

Inflammatory breast cancer (IBC) is the most aggressive form of breast cancer. Treatment options are limited and the mechanisms underlying its aggressiveness are poorly understood. Intermittent hypoxia (IH) causes oxidative stress and is emerging as important regulator of tumor metastasis. Vessels in IBC tumors have been shown to be immature, which is a primary cause of IH. We therefore investigated the relevance of IH for the modulation of gene expression in IBC cells in order to assess IH as potential regulator of IBC aggressiveness. Gene array analysis of IBC cells following chronic IH (45-60 days) demonstrated increased expression of pro-metastatic genes of the extracellular matrix, such as tenascin-C (TNC; an essential factor of the metastatic niche) and matrix metalloproteinase 9 (MMP9), and of pro-inflammatory processes, such as cyclooxygenase-2 (COX-2). Investigating the oxidative stress-dependent regulation of TNC, we found a gradual sensitivity on mRNA and protein levels. Oxidative stress activated NF-E2-related factor 2 (Nrf2), c-Jun N-terminal kinase (JNK), c-Jun and nuclear factor κB (NF-κB), but TNC upregulation was only dependent on NF-κB activation. Pharmacological inhibition of inhibitor of NF-κB α (IκBα) phosphorylation as well as overexpression of IκBα prevented TNC, MMP9 and COX-2 induction, whereas the pro-inflammatory cytokine interleukin-1ß (IL-1ß) increased their expression levels. Analysis of the gene array data showed NF-κB binding sites for 64% of all upregulated genes, linking NF-κB with IH-dependent regulation of pro-metastatic gene expression in IBC cells. Our results provide a first link between intermittent hypoxia and pro-metastatic gene expression in IBC cells, revealing a putative novel mechanism for the high metastatic potential of IBC.


Assuntos
Células Epiteliais/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Metaloproteinase 9 da Matriz/genética , NF-kappa B/genética , Oxigênio/farmacologia , Tenascina/genética , Acetilcisteína/farmacologia , Antracenos/farmacologia , Hipóxia Celular , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Interleucina-1beta/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Metaloproteinase 9 da Matriz/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Metástase Neoplásica , Nitrilas/farmacologia , Estresse Oxidativo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-jun/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais , Sulfonas/farmacologia , Tenascina/metabolismo
9.
PLoS Biol ; 14(1): e1002347, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26752685

RESUMO

The asparagine hydroxylase, factor inhibiting HIF (FIH), confers oxygen-dependence upon the hypoxia-inducible factor (HIF), a master regulator of the cellular adaptive response to hypoxia. Studies investigating whether asparagine hydroxylation is a general regulatory oxygen-dependent modification have identified multiple non-HIF targets for FIH. However, the functional consequences of this outside of the HIF pathway remain unclear. Here, we demonstrate that the deubiquitinase ovarian tumor domain containing ubiquitin aldehyde binding protein 1 (OTUB1) is a substrate for hydroxylation by FIH on N22. Mutation of N22 leads to a profound change in the interaction of OTUB1 with proteins important in cellular metabolism. Furthermore, in cultured cells, overexpression of N22A mutant OTUB1 impairs cellular metabolic processes when compared to wild type. Based on these data, we hypothesize that OTUB1 is a target for functional hydroxylation by FIH. Additionally, we propose that our results provide new insight into the regulation of cellular energy metabolism during hypoxic stress and the potential for targeting hydroxylases for therapeutic benefit.


Assuntos
Cisteína Endopeptidases/metabolismo , Oxigenases de Função Mista/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Cisteína Endopeptidases/genética , Enzimas Desubiquitinantes , Metabolismo Energético , Células HEK293 , Humanos , Hidroxilação , Mutagênese Sítio-Dirigida , Estabilidade Proteica
10.
Cell Host Microbe ; 17(5): 662-71, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25865369

RESUMO

Interactions between the microbiota and distal gut are fundamental determinants of human health. Such interactions are concentrated at the colonic mucosa and provide energy for the host epithelium through the production of the short-chain fatty acid butyrate. We sought to determine the role of epithelial butyrate metabolism in establishing the austere oxygenation profile of the distal gut. Bacteria-derived butyrate affects epithelial O2 consumption and results in stabilization of hypoxia-inducible factor (HIF), a transcription factor coordinating barrier protection. Antibiotic-mediated depletion of the microbiota reduces colonic butyrate and HIF expression, both of which are restored by butyrate supplementation. Additionally, germ-free mice exhibit diminished retention of O2-sensitive dyes and decreased stabilized HIF. Furthermore, the influences of butyrate are lost in cells lacking HIF, thus linking butyrate metabolism to stabilized HIF and barrier function. This work highlights a mechanism where host-microbe interactions augment barrier function in the distal gut.


Assuntos
Bactérias/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Ácidos Graxos Voláteis/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 1 Induzível por Hipóxia/biossíntese , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Humanos , Camundongos , Consumo de Oxigênio
11.
Exp Cell Res ; 330(2): 371-381, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25107382

RESUMO

Epithelial injury and tubulointerstitial fibrosis (TIF) within a hypoxic microenvironment are associated with progressive loss of renal function in chronic kidney disease [CKD]. Transforming growth factor beta-1 (TGF-ß1) is an important mediator of renal fibrosis. Growing evidence suggests that Vitamin D [1,25-(OH)2D] and its analogues may have a renoprotective effect in CKD. Here we examined the protective effect of the vitamin D analogue paricalcitol [PC; 19-nor-1α,3ß,25-trihydroxy-9,10-secoergosta-5(Z),7(E) 22(E)-triene] on the responses of human renal epithelial cells to TGF-ß1. PC attenuated TGF-ß1-induced Smad 2 phosphorylation and upregulation of the Notch ligand Jagged-1, α-smooth muscle actin and thrombospondin-1 and prevented the TGF-ß1-mediated loss of E-Cadherin. To mimic the hypoxic milieu of CKD we cultured renal epithelial cells in hypoxia [1% O2] and observed similar attenuation by PC of TGF-ß1-induced fibrotic responses. Furthermore, in cells cultured in normoxia [21% O2], PC induced an accumulation of hypoxia-inducible transcription factors (HIF) 1α and HIF-2α in a time and concentration [1 µM-2 µM] dependent manner. Here, PC-induced HIF stabilisation was dependent on activation of the PI-3Kinase pathway. This is the first study to demonstrate regulation of the HIF pathway by PC which may have importance in the mechanism underlying renoprotection by PC.


Assuntos
Células Epiteliais/efeitos dos fármacos , Ergocalciferóis/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Actinas/biossíntese , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Caderinas/metabolismo , Proteínas de Ligação ao Cálcio/biossíntese , Hipóxia Celular , Linhagem Celular Transformada , Células Epiteliais/patologia , Fibrose , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Proteína Jagged-1 , Proteínas de Membrana/biossíntese , Nefrite Intersticial/patologia , Fosforilação , Estabilidade Proteica , Interferência de RNA , Proteínas Serrate-Jagged , Proteína Smad2/metabolismo , Trombospondina 1/biossíntese , Fator de Crescimento Transformador beta1/metabolismo
13.
Proc Natl Acad Sci U S A ; 110(46): 18490-5, 2013 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-24145445

RESUMO

Hypoxia is a prominent feature of chronically inflamed tissues. Oxygen-sensing hydroxylases control transcriptional adaptation to hypoxia through the regulation of hypoxia-inducible factor (HIF) and nuclear factor κB (NF-κB), both of which can regulate the inflammatory response. Furthermore, pharmacologic hydroxylase inhibitors reduce inflammation in multiple animal models. However, the underlying mechanism(s) linking hydroxylase activity to inflammatory signaling remains unclear. IL-1ß, a major proinflammatory cytokine that regulates NF-κB, is associated with multiple inflammatory pathologies. We demonstrate that a combination of prolyl hydroxylase 1 and factor inhibiting HIF hydroxylase isoforms regulates IL-1ß-induced NF-κB at the level of (or downstream of) the tumor necrosis factor receptor-associated factor 6 complex. Multiple proteins of the distal IL-1ß-signaling pathway are subject to hydroxylation and form complexes with either prolyl hydroxylase 1 or factor inhibiting HIF. Thus, we hypothesize that hydroxylases regulate IL-1ß signaling and subsequent inflammatory gene expression. Furthermore, hydroxylase inhibition represents a unique approach to the inhibition of IL-1ß-dependent inflammatory signaling.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hipóxia/fisiopatologia , Inflamação/fisiopatologia , Oxigenases de Função Mista/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Western Blotting , Células HeLa , Humanos , Hidroxilação , Hipóxia/metabolismo , Imunoprecipitação , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Luciferases , Espectrometria de Massas , Prolil Hidroxilases/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo
14.
Curr Opin Pharmacol ; 13(4): 646-53, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23660374

RESUMO

Oxygen deprivation (hypoxia) is a frequently encountered condition in both health and disease. Metazoans have evolved an elegant and direct cellular mechanism by which to sense local oxygen levels and mount an adaptive transcriptional response to hypoxia which is mediated by a transcription factor termed the hypoxia-inducible factor (HIF). In normoxia, HIF is repressed primarily through the action of a family of hydroxylases, which target HIFα subunits for degradation in an oxygen-dependent manner. In hypoxia, HIF is rapidly stabilized in cells thus allowing it to regulate the expression of hundreds of genes which promote an adaptive response including genes expressing regulators of angiogenesis, metabolism, growth and survival. Initial studies into the HIF pathway focused mainly on its role in supporting tumor adaptation through enhancing processes such as angiogenesis, glycolytic metabolism and cell survival. More recently however, it has become clear that the HIF pathway also plays a key role in the regulation of immunity and inflammation. In fact, conditional knockout of the HIF-1α subunit has identified key immune roles in T-cells, dendritic cells, macrophages, neutrophils and epithelial cells. In this review, we will consider the role for HIF in the regulation of the immune response and its possible contribution to inflammation. Furthermore, we will consider potential therapeutic strategies, which target the HIF pathway in chronic inflammatory and infectious disease.


Assuntos
Fator 1 Induzível por Hipóxia/imunologia , Hipóxia/imunologia , Animais , Doenças Transmissíveis/imunologia , Humanos , Imunidade , Inflamação/imunologia
15.
Biol Chem ; 394(4): 479-93, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23362195

RESUMO

Hypoxia is associated with a diverse range of physiological and pathophysiological processes, including development, wound healing, inflammation, vascular disease and cancer. The requirement that eukaryotic cells have for molecular oxygen as the terminal electron acceptor for the electron transport chain means that the maintenance of oxygen delivery is key for bioenergetic homeostasis. Metazoans have evolved an effective way to adapt to hypoxic stress at the molecular level through a transcription factor termed the hypoxia inducible factor. A family of oxygen-sensing hydroxylases utilizes molecular oxygen as a co-substrate for the hydroxylation of hypoxia inducible factor α subunits, thereby reducing its expression and transcriptional activity when oxygen is available. Recent studies have indicated that other hypoxia-responsive transcriptional pathways may also be hydroxylase-dependent. In this review, we will discuss the role of hydroxylases in the regulation of NF-κB, a key regulator of immunity and inflammation. Developing our understanding of the role of hydroxylases in hypoxic inflammation may identify novel therapeutic approaches in chronic inflammatory disease.


Assuntos
Oxigenases de Função Mista/metabolismo , NF-kappa B/metabolismo , Animais , Humanos , Oxigenases de Função Mista/genética , Modelos Biológicos , Oxigênio/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
16.
J Biol Chem ; 286(6): 4718-26, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21123177

RESUMO

Under conditions of hypoxia, most eukaryotic cells undergo a shift in metabolic strategy, which involves increased flux through the glycolytic pathway. Although this is critical for bioenergetic homeostasis, the underlying mechanisms have remained incompletely understood. Here, we report that the induction of hypoxia-induced glycolysis is retained in cells when gene transcription or protein synthesis are inhibited suggesting the involvement of additional post-translational mechanisms. Post-translational protein modification by the small ubiquitin related modifier-1 (SUMO-1) is induced in hypoxia and mass spectrometric analysis using yeast cells expressing tap-tagged Smt3 (the yeast homolog of mammalian SUMO) revealed hypoxia-dependent modification of a number of key glycolytic enzymes. Overexpression of SUMO-1 in mammalian cancer cells resulted in increased hypoxia-induced glycolysis and resistance to hypoxia-dependent ATP depletion. Supporting this, non-transformed cells also demonstrated increased glucose uptake upon SUMO-1 overexpression. Conversely, cells overexpressing the de-SUMOylating enzyme SENP-2 failed to demonstrate hypoxia-induced glycolysis. SUMO-1 overexpressing cells demonstrated focal clustering of glycolytic enzymes in response to hypoxia leading us to hypothesize a role for SUMOylation in promoting spatial re-organization of the glycolytic pathway. In summary, we hypothesize that SUMO modification of key metabolic enzymes plays an important role in shifting cellular metabolic strategies toward increased flux through the glycolytic pathway during periods of hypoxic stress.


Assuntos
Glicólise/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Proteína SUMO-1/metabolismo , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Hipóxia Celular/fisiologia , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Células HeLa , Humanos , Proteína SUMO-1/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo
17.
J Immunol ; 185(7): 4439-45, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20817876

RESUMO

Molecular O(2) and CO(2) are the primary substrate and product of aerobic metabolism, respectively. Levels of these physiologic gases in the cell microenvironment vary dramatically both in health and in diseases, such as chronic inflammation, ischemia, and cancer, in which metabolism is significantly altered. The identification of the hypoxia-inducible factor led to the discovery of an ancient and direct link between tissue O(2) and gene transcription. In this study, we demonstrate that mammalian cells (mouse embryonic fibroblasts and others) also sense changes in local CO(2) levels, leading to altered gene expression via the NF-κB pathway. IKKα, a central regulatory component of NF-κB, rapidly and reversibly translocates to the nucleus in response to elevated CO(2). This response is independent of hypoxia-inducible factor hydroxylases, extracellular and intracellular pH, and pathways that mediate acute CO(2)-sensing in nematodes and flies and leads to attenuation of bacterial LPS-induced gene expression. These results suggest the existence of a molecular CO(2) sensor in mammalian cells that is linked to the regulation of genes involved in innate immunity and inflammation.


Assuntos
Dióxido de Carbono/metabolismo , Regulação da Expressão Gênica/imunologia , Imunidade Inata/fisiologia , Inflamação/metabolismo , NF-kappa B/imunologia , Animais , Western Blotting , Células Cultivadas , Expressão Gênica , Humanos , Quinase I-kappa B/metabolismo , Inflamação/imunologia , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Transporte Proteico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia
18.
Gastroenterology ; 139(6): 2093-101, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20600011

RESUMO

BACKGROUND & AIMS: Hypoxia inducible factor (HIF) prolyl hydroxylase inhibitors are protective in mouse models of inflammatory bowel disease (IBD). Here, we investigated the therapeutic target(s) and mechanism(s) involved. METHODS: The effect of genetic deletion of individual HIF-prolyl hydroxylase (PHD) enzymes on the development of dextran sulphate sodium (DSS)-induced colitis was examined in mice. RESULTS: PHD1(-/-), but not PHD2(+/-) or PHD3(-/-), mice were less susceptible to the development of colitis than wild-type controls as determined by weight loss, disease activity, colon histology, neutrophil infiltration, and cytokine expression. Reduced susceptibility of PHD1(-/-) mice to colitis was associated with increased density of colonic epithelial cells relative to wild-type controls, which was because of decreased levels of apoptosis that resulted in enhanced epithelial barrier function. Furthermore, with the use of cultured epithelial cells it was confirmed that hydroxylase inhibition reversed DSS-induced apoptosis and barrier dysfunction. Finally, PHD1 levels were increased with disease severity in intestinal tissue from patients with IBD and in colonic tissues from DSS-treated mice. CONCLUSIONS: These results imply a role for PHD1 as a positive regulator of intestinal epithelial cell apoptosis in the inflamed colon. Genetic loss of PHD1 is protective against colitis through decreased epithelial cell apoptosis and consequent enhancement of intestinal epithelial barrier function. Thus, targeted PHD1 inhibition may represent a new therapeutic approach in IBD.


Assuntos
Apoptose/fisiologia , Colite/metabolismo , Dioxigenases/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Proteínas Nucleares/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Animais , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana/toxicidade , Dioxigenases/genética , Modelos Animais de Doenças , Enterócitos/metabolismo , Enterócitos/patologia , Células Epiteliais/patologia , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Prolina Dioxigenases do Fator Induzível por Hipóxia , Absorção Intestinal/fisiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Proteínas Nucleares/genética , Pró-Colágeno-Prolina Dioxigenase/genética , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA