Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Dis Model Mech ; 12(8)2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31383794

RESUMO

Amyotrophic lateral sclerosis (ALS) presents a poorly understood pathogenesis. Evidence from patients and mutant SOD1 mouse models suggests vascular damage may precede or aggravate motor dysfunction in ALS. We have previously shown angiogenin (ANG) treatment enhances motor neuron survival, delays motor dysfunction and prevents vascular regression in the SOD1G93A ALS model. However, the existence of vascular defects at different stages of disease progression remains to be established in other ALS models. Here, we assessed vascular integrity in vivo throughout different disease stages, and investigated whether ANG treatment reverses vascular regression and prolongs motor neuron survival in the FUS (1-359) mouse model of ALS. Lumbar spinal cord tissue was collected from FUS (1-359) and non-transgenic control mice at postnatal day (P)50, P90 and P120. We found a significant decrease in vascular network density in lumbar spinal cords from FUS (1-359) mice by day 90, at which point motor neuron numbers were unaffected. ANG treatment did not affect survival or counter vascular regression. Endogenous Ang1 and Vegf expression were unchanged at P50 and P90; however, we found a significant decrease in miRNA 126 at P50, indicating vascular integrity in FUS mice may be compromised via an alternative pathway. Our study demonstrates that vascular regression occurs before motor neuron degeneration in FUS (1-359) mice, and highlights that heterogeneity in responses to novel ALS therapeutics can already be detected in preclinical mouse models of ALS.This article has an associated First Person interview with the joint first authors of the paper.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Vasos Sanguíneos/patologia , Neurônios Motores/patologia , Proteína FUS de Ligação a RNA/genética , Animais , Contagem de Células , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/metabolismo , Neurônios Motores/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Ribonuclease Pancreático/farmacologia , Sialoglicoproteínas/metabolismo , Análise de Sobrevida
3.
Neuropharmacology ; 133: 503-511, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29486168

RESUMO

Loss-of-function mutations in the angiogenin (ANG) gene have been identified in familial and sporadic ALS patients. Previous work from our group identified human ANG (huANG) to protect motoneurons in vitro, and provided proof-of-concept that daily intraperitoneal (i.p.) huANG injections post-symptom onset increased lifespan and delayed disease progression in SOD1G93A mice. huANG's mechanism of action remains less well understood. Here, we implemented a preclinical in vivo design to validate our previous results, provide pharmacokinetic and protein distribution data after systemic administration, and explore potential pleiotropic activities of huANG in vivo. SOD1G93A mice (n = 45) and non-transgenic controls (n = 31) were sex- age- and litter-matched according to the 2010 European ALS/MND group guidelines, and treated with huANG (1 µg, i.p., 3 times/week) or vehicle from 90 days on. huANG treatment increased survival and delayed motor dysfunction as assessed by rotarod in SOD1G93A mice. Increased huANG serum levels were detectable 2 and 24 h after i.p. injection equally in transgenic and non-transgenic mice. Exogenous huANG localized to spinal cord astrocytes, supporting a glia-mediated, paracrine mechanism of action; uptake into endothelial cells was also observed. 1 µg huANG or vehicle were administered from 90 to 115 days of age for histological analysis. Vehicle-treated SOD1G93A mice showed decreased motoneuron numbers and vascular length per ventral horn area, while huANG treatment resulted in improved vascular network maintenance and motoneuron survival. Our data suggest huANG represents a new class of pleiotropic ALS therapeutic that acts on the spinal cord vasculature and glia to delay motoneuron degeneration and disease progression.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Indutores da Angiogênese/uso terapêutico , Ribonuclease Pancreático/uso terapêutico , Fatores Etários , Esclerose Lateral Amiotrófica/sangue , Esclerose Lateral Amiotrófica/complicações , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Transtornos dos Movimentos/tratamento farmacológico , Transtornos dos Movimentos/etiologia , Ribonuclease Pancreático/sangue , Teste de Desempenho do Rota-Rod , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Análise de Sobrevida , Fatores de Tempo
4.
Artigo em Inglês | MEDLINE | ID: mdl-29221425

RESUMO

BACKGROUND: Riluzole is the most widespread therapeutic for treatment of the progressive degenerative disease amyotrophic lateral sclerosis (ALS). Riluzole gained FDA approval in 1995 before the development of ALS mouse models. We assessed riluzole in three transgenic ALS mouse models: the SOD1G93A model, the TDP-43A315T model, and the recently developed FUS (1-359) model. METHODS: Age, sex and litter-matched mice were treated with riluzole (22 mg/kg) in drinking water or vehicle (DMSO) from symptom onset. Lifespan was assessed and motor function tests were carried out twice weekly to determine whether riluzole slowed disease progression. RESULTS: Riluzole treatment had no significant benefit on lifespan in any of the ALS mouse models tested. Riluzole had no significant impact on decline in motor performance in the FUS (1-359) and SOD1G93A transgenic mice as assessed by Rotarod and stride length analysis. CONCLUSIONS: Riluzole is widely prescribed for ALS patients despite questions surrounding its efficacy. Our data suggest that if riluzole was identified as a therapeutic candidate today it would not progress past pre-clinical assessment. This raises questions about the standards used in pre-clinical assessment of therapeutic candidates for the treatment of ALS.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/mortalidade , Longevidade/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Riluzol/uso terapêutico , Esclerose Lateral Amiotrófica/genética , Animais , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Progressão da Doença , Estimativa de Kaplan-Meier , Longevidade/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína FUS de Ligação a RNA/genética , Superóxido Dismutase/genética
5.
Dis Model Mech ; 9(9): 1029-37, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27491077

RESUMO

Transgenic transactivation response DNA-binding protein 43 (TDP-43) mice expressing the A315T mutation under control of the murine prion promoter progressively develop motor function deficits and are considered a new model for the study of amyotrophic lateral sclerosis (ALS); however, premature sudden death resulting from intestinal obstruction halts disease phenotype progression in 100% of C57BL6/J congenic TDP-43(A315T) mice. Similar to our recent results in SOD1(G93A) mice, TDP-43(A315T) mice fed a standard pellet diet showed increased 5' adenosine monophosphate-activated protein kinase (AMPK) activation at postnatal day (P)80, indicating elevated energetic stress during disease progression. We therefore investigated the effects of a high-fat jelly diet on bioenergetic status and lifespan in TDP-43(A315T) mice. In contrast to standard pellet-fed mice, mice fed high-fat jelly showed no difference in AMPK activation up to P120 and decreased phosphorylation of acetly-CoA carboxylase (ACC) at early-stage time points. Exposure to a high-fat jelly diet prevented sudden death and extended survival, allowing development of a motor neuron disease phenotype with significantly decreased body weight from P80 onward that was characterised by deficits in Rotarod abilities and stride length measurements. Development of this phenotype was associated with a significant motor neuron loss as assessed by Nissl staining in the lumbar spinal cord. Our work suggests that a high-fat jelly diet improves the pre-clinical utility of the TDP-43(A315T) model by extending lifespan and allowing the motor neuron disease phenotype to progress, and indicates the potential benefit of this diet in TDP-43-associated ALS.


Assuntos
Proteínas de Ligação a DNA/genética , Dieta Hiperlipídica , Metabolismo Energético , Longevidade , Vértebras Lombares/fisiopatologia , Neurônios Motores/patologia , Medula Espinal/fisiopatologia , Adenilato Quinase/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/fisiopatologia , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Progressão da Doença , Ativação Enzimática , Ácidos Graxos/metabolismo , Intestinos/patologia , Vértebras Lombares/metabolismo , Vértebras Lombares/patologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora , Neurônios Motores/metabolismo , Oxirredução , Fenótipo , Medula Espinal/metabolismo , Medula Espinal/patologia
6.
Neurobiol Aging ; 36(2): 1140-50, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25443289

RESUMO

Adenosine 5'-monophosphate-activated protein kinase (AMPK) is a master regulator of energy balance. As energy imbalance is documented as a key pathologic feature of amyotrophic lateral sclerosis (ALS), we investigated AMPK as a pharmacologic target in SOD1(G93A) mice. We noted a strong activation of AMPK in lumbar spinal cords of SOD1(G93A) mice. Pharmacologic activation of AMPK has shown protective effects in neuronal "preconditioning" models. We tested the hypothesis that "preconditioning" with a small molecule activator of AMPK, latrepirdine, exerts beneficial effects on disease progression. SOD1(G93A) mice (n = 24 animals per group; sex and litter matched) were treated with latrepirdine (1 µg/kg, intraperitoneal) or vehicle from postnatal day 70 to 120. Treatment with latrepirdine increased AMPK activity in primary mouse motor neuron cultures and in SOD1(G93A) lumbar spinal cords. Mice "preconditioned" with latrepirdine showed a delayed symptom onset and a significant increase in life span (p < 0.01). Our study suggests that "preconditioning" with latrepirdine may represent a possible therapeutic strategy for individuals harboring ALS-associated gene mutations who are at risk for developing ALS.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/prevenção & controle , Indóis/administração & dosagem , Superóxido Dismutase/genética , Proteínas Quinases Ativadas por AMP/fisiologia , Esclerose Lateral Amiotrófica/enzimologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Progressão da Doença , Metabolismo Energético , Ativação Enzimática/efeitos dos fármacos , Feminino , Injeções Intraperitoneais , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios Motores/enzimologia , Neurônios Motores/patologia , Mutação , Risco , Medula Espinal/enzimologia , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
7.
Sci Rep ; 4: 6307, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25200751

RESUMO

Xanthine oxidase (XOD) is an enzyme which plays a central role in purine catabolism by converting hypoxanthine into xanthine and then further into uric acid. Here we report that XOD is activated in THP-1 human myeloid cells in response to pro-inflammatory and growth factor stimulation. This effect occurred following stimulation of THP-1 cells with ligands of plasma membrane associated TLRs 2 and 4, endosomal TLRs 7 and 8 as well as stem cell growth factor (SCF). Hypoxia-inducible factor 1 (HIF-1) and activator protein 1 (AP-1) transcription complexes were found to be responsible for XOD upregulation. Importantly, the mammalian target of rapamycin (mTOR), a major myeloid cell translation regulator, was also found to be essential for XOD activation. Specific inhibition of XOD by allopurinol and sodium tungstate led to an increase in intracellular AMP levels triggering downregulation of mTOR activation by phosphorylation of its T2446 residue. Taken together, our results demonstrate for the first time that XOD is not only activated by pro-inflammatory stimuli or SCF but also plays an important role in maintaining mTOR-dependent translational control during the biological responses of human myeloid cells.


Assuntos
Inflamação/imunologia , Células Mieloides/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Xantina Oxidase/metabolismo , Alopurinol/farmacologia , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Ativação Enzimática , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lipopolissacarídeos , Fígado/metabolismo , Células MCF-7 , Masculino , Camundongos , Peptidoglicano , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Interferência de RNA , RNA Interferente Pequeno , Fator de Células-Tronco/metabolismo , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/metabolismo , Compostos de Tungstênio/farmacologia , Ácido Úrico/análise , Xantina Oxidase/antagonistas & inibidores , Xantina Oxidase/biossíntese
8.
Neurobiol Dis ; 70: 99-107, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24956542

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of motoneurons in the spinal cord, brainstem and motor cortex. Mutations in the superoxide dismutase 1 (SOD1) gene represent a frequent genetic determinant and recapitulate a disease phenotype similar to ALS when expressed in mice. Previous studies using SOD1(G93A) transgenic mice have suggested a paracrine mechanism of neuronal loss, in which cytokines and other toxic factors released from astroglia or microglia trigger motoneuron degeneration. Several pro-inflammatory cytokines activate death receptors and may downstream from this activate the Bcl-2 family protein, Bid. We here sought to investigate the role of Bid in astrocyte activation and non-cell autonomous motoneuron degeneration. We found that spinal cord Bid protein levels increased significantly during disease progression in SOD1(G93A) mice. Subsequent experiments in vitro indicated that Bid was expressed at relatively low levels in motoneurons, but was enriched in astrocytes and microglia. Bid was strongly induced in astrocytes in response to pro-inflammatory cytokines or exposure to lipopolysaccharide. Experiments in bid-deficient astrocytes or astrocytes treated with a small molecule Bid inhibitor demonstrated that Bid was required for the efficient activation of transcription factor nuclear factor-κB in response to these pro-inflammatory stimuli. Finally, we found that conditioned medium from wild-type astrocytes, but not from bid-deficient astrocytes, was toxic when applied to primary motoneuron cultures. Collectively, our data demonstrate a new role for the Bcl-2 family protein Bid as a mediator of astrocyte activation during neuroinflammation, and suggest that Bid activation may contribute to non-cell autonomous motoneuron degeneration in ALS.


Assuntos
Astrócitos/imunologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Superóxido Dismutase/metabolismo , Esclerose Lateral Amiotrófica , Animais , Células do Corno Anterior/fisiologia , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/antagonistas & inibidores , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/genética , Morte Celular/fisiologia , Células Cultivadas , Humanos , Lipopolissacarídeos , Camundongos Knockout , Camundongos Transgênicos , Microglia/imunologia , Neurônios Motores/fisiologia , NF-kappa B/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Neuroimunomodulação/fisiologia , Superóxido Dismutase/genética , Superóxido Dismutase-1
9.
J Neurosci ; 32(15): 5024-38, 2012 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22496549

RESUMO

Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disorder affecting motoneurons. Mutations in angiogenin, encoding a member of the pancreatic RNase A superfamily, segregate with ALS. We previously demonstrated that angiogenin administration shows promise as a neuroprotective therapeutic in studies using transgenic ALS mice and primary motoneuron cultures. Its mechanism of action and target cells in the spinal cord, however, are largely unknown. Using mixed motoneuron cultures, motoneuron-like NSC34 cells, and primary astroglia cultures as model systems, we here demonstrate that angiogenin is a neuronally secreted factor that is endocytosed by astroglia and mediates neuroprotection in paracrine. We show that wild-type angiogenin acts unidirectionally to induce RNA cleavage in astroglia, while the ALS-associated K40I mutant is also secreted and endocytosed, but fails to induce RNA cleavage. Angiogenin uptake into astroglia requires heparan sulfate proteoglycans, and engages clathrin-mediated endocytosis. We show that this uptake mechanism exists for mouse and human angiogenin, and delivers a functional RNase output. Moreover, we identify syndecan 4 as the angiogenin receptor mediating the selective uptake of angiogenin into astroglia. Our data provide new insights into the paracrine activities of angiogenin in the nervous system, and further highlight the critical role of non-neuronal cells in the pathogenesis of ALS.


Assuntos
Astrócitos/metabolismo , Astrócitos/fisiologia , Neurônios Motores/metabolismo , Clivagem do RNA/fisiologia , Ribonuclease Pancreático/metabolismo , Ribonuclease Pancreático/fisiologia , Animais , Western Blotting , Sobrevivência Celular/fisiologia , Células Cultivadas , Clatrina/fisiologia , Meios de Cultivo Condicionados , Endocitose/fisiologia , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores , Comunicação Parácrina/fisiologia , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real , Sindecana-4/metabolismo , Transfecção
10.
Int J Biochem Cell Biol ; 43(4): 674-81, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21262374

RESUMO

Mitochondria, known to share many common features with prokaryotic cells, accumulate several endogenous ligands of the pattern-recognition Toll-like receptor 4 (TLR4), such as the heat shock proteins (Hsp) 70 and 60. TLR4 specifically recognises and responds to LPS of Gram-negative bacteria and participates in both autoimmune reactions and tissue regeneration due to its ability to recognise endogenous ligands. In the present study we show that mitochondria extracts obtained from hydrogen peroxide-dysfunctionalised cells induce a pro-inflammatory response in human THP-1 myeloid leukaemia cells. This inflammatory response was similar to that caused by LPS and much stronger than that induced by the extracts of normal mitochondria. Such reactions include activation of stress-adaptation hypoxia-inducible factor 1 alpha (HIF-1α) and expression/release of the pro-inflammatory cytokines IL-6 and TNF-α. Pre-treatment of THP-1 myeloid macrophages with TLR4-neutralising antibody before exposure to mitochondria extracts or LPS attenuated the inflammatory responses. Signalling pathways recruited by TLR4 in response to LPS and mitochondria-derived ligands were found to be the same. An in vitro ELISA-based TLR4-ligand binding assay, in which the ligand-binding domain of human TLR4 was immobilised, showed that mitochondria extracts contain endogenous TLR4 ligands. These results were verified in surface plasmon resonance experiments in which the affinity of the ligands derived from dysfunctional mitochondria was comparable with that of LPS and was much higher than that observed for normal mitochondria.


Assuntos
Mitocôndrias/metabolismo , Receptor 4 Toll-Like/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Ligantes , Lipopolissacarídeos/farmacologia
11.
Immunol Cell Biol ; 89(2): 268-74, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20661259

RESUMO

Toll-like receptors (TLRs) lie in the core of resistance to infectious diseases allowing host immune cells to specifically detect pathogens by recognising their specific molecular patterns. Cell membrane-associated TLR4 (recognises lipopolysaccharide (LPS) of Gram-negative bacteria) and endosomal TLR7/8 (recognise viral single-stranded RNA) are known to activate hypoxia inducible factor-1α (HIF-1α) protein (necessary for cellular adaptation to the inflammatory stress) via redox-dependent mechanism. TLR4 triggers the cross talk between HIF-1α and apoptosis signal-regulating kinase 1 (ASK1), whereas TLR7/8 activates HIF-1α in the ASK1-independent manner. Here, we report that in THP-1 and RAW264.7 macrophages, ligand-induced activation of the TLR4 but not TLR7/8 induces activation and transcriptional upregulation of sphingosine kinase 1 (SphK1) in extracellular signal-regulating kinase and phospholipase C-1γ/PI3 kinase-dependent manner. TLR4-mediated SphK1 activation was found to be critical for the redox-dependent activation of HIF-1α and ASK1, as well as for the prevention of LPS-induced activation of caspase 3 and the expression of pro-inflammatory cytokine interleukin-6.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-6/biossíntese , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase Quinase 5/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Caspase 3/metabolismo , Ativação Enzimática/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Camundongos , Modelos Biológicos
12.
Apoptosis ; 13(12): 1410-6, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18841476

RESUMO

Nitric oxide (NO) is a reactive secondary mediator, which has been found to participate in cell cycle regulation and apoptosis in myeloid macrophages, the key effectors of inflammatory and innate immune responses. However, the molecular mechanisms of nitric oxide-induced death of myeloid macrophages are not well understood. In this study we have found that NO derived from S-nitrosoglutathione (GSNO) activates ASK1 in THP-1 human myeloid macrophages in a concentration and time-dependent manner. It also induces accumulation of HIF-1alpha protein in a concentration-dependent manner, which peaks at 4 h of exposure to 1 mM GSNO. GSNO does not affect the level of HIF-1alpha mRNA as detected by the RT-PCR. In addition, GSNO was found to induce accumulation of p53 in normal but not HIF-1alpha knockdown THP-1 cells, where expression of this protein was silenced by specific siRNA. It has also been found that GSNO-mediated accumulation of p53 depends on activation of ASK1 since no GSNO-induced p53 stabilisation was observed in THP-1 cells transfected with dominant-negative form of this kinase. However, in both HIF-1alpha knockdown THP-1 cells and those transfected with the dominant-negative form of ASK1, GSNO was able to induce cell death as detected by the MTS cell viability assay leading to an increase in release of LDH.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Estabilidade Enzimática , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , MAP Quinase Quinase Quinase 5/genética , Macrófagos/citologia , S-Nitrosoglutationa/metabolismo , Proteína Supressora de Tumor p53/genética
13.
Mol Immunol ; 45(11): 3045-9, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18462799

RESUMO

Sepsis is the leading cause of death in intensive care units, which reflects detrimental host response to infection where lipopolysaccharide (LPS) shared by Gram-negative bacteria acts as a potent activator of immune cells via Toll-like receptor 4 (TLR4). Recently it was found that TLR4 downstream signalling leads to the accumulation of hypoxia-inducible factor 1 alpha (HIF-1alpha), which is important for TLR4-dependent expression of pro-inflammatory cytokines, however, basic biochemical mechanisms of involvement of this protein in TLR4 downstream signalling remains unclear. Here we found that knockdown of the expression of HIF-1alpha protein by siRNA led to the depletion of ATP, which corresponded to the constant increase in the activity of apoptosis signal-regulating kinase 1 (ASK1) and therefore apoptosis as estimated based on the increase in the activity of caspase 3. On the other hand, LPS-dependent production of IL-6 was attenuated. Treatment of HIF-1alpha knockdown cells with extracellular ATP in combination with LPS preserved the IL-6 expression but not the activity of ASK1 on the level observed in LPS-stimulated control cells. We therefore suggested that HIF-1alpha protein supports LPS-dependent expression of IL-6 by preventing depletion of ATP. On the other hand HIF-1alpha protein is selectively required for down-regulation of ASK1 activated during LPS-induced TLR4 downstream signalling.


Assuntos
Trifosfato de Adenosina/deficiência , Apoptose/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-6/biossíntese , Lipopolissacarídeos/farmacologia , Células Mieloides/citologia , Células Mieloides/metabolismo , Receptor 4 Toll-Like/imunologia , Linhagem Celular Tumoral , Cobalto/farmacologia , Regulação para Baixo/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Humanos , MAP Quinase Quinase Quinase 5/genética , Células Mieloides/efeitos dos fármacos , Células Mieloides/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA