Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(1): e2214874120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36574710

RESUMEN

Adequate mass and function of adipose tissues (ATs) play essential roles in preventing metabolic perturbations. The pathological reduction of ATs in lipodystrophy leads to an array of metabolic diseases. Understanding the underlying mechanisms may benefit the development of effective therapies. Several cellular processes, including autophagy and vesicle trafficking, function collectively to maintain AT homeostasis. Here, we investigated the impact of adipocyte-specific deletion of the lipid kinase phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3) on AT homeostasis and systemic metabolism in mice. We report that PIK3C3 functions in all ATs and that its absence disturbs adipocyte autophagy and hinders adipocyte differentiation, survival, and function with differential effects on brown and white ATs. These abnormalities cause loss of white ATs, whitening followed by loss of brown ATs, and impaired "browning" of white ATs. Consequently, mice exhibit compromised thermogenic capacity and develop dyslipidemia, hepatic steatosis, insulin resistance, and type 2 diabetes. While these effects of PIK3C3 largely contrast previous findings with the autophagy-related (ATG) protein ATG7 in adipocytes, mice with a combined deficiency in both factors reveal a dominant role of the PIK3C3-deficient phenotype. We have also found that dietary lipid excess exacerbates AT pathologies caused by PIK3C3 deficiency. Surprisingly, glucose tolerance is spared in adipocyte-specific PIK3C3-deficient mice, a phenotype that is more evident during dietary lipid excess. These findings reveal a crucial yet complex role for PIK3C3 in ATs, with potential therapeutic implications.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Animales , Ratones , Fosfatidilinositol 3-Quinasas Clase III/genética , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Adipocitos/metabolismo , Lípidos , Tejido Adiposo Pardo/metabolismo , Adipocitos Marrones/metabolismo
2.
Am J Physiol Renal Physiol ; 323(1): F81-F91, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35499237

RESUMEN

Disruption of the blood-urine barrier can result in acute or chronic inflammatory bladder injury. Activation of the oxygen-regulated hypoxia-inducible factor (HIF) pathway has been shown to protect mucosal membranes by increasing the expression of cytoprotective genes and by suppressing inflammation. The activity of HIF is controlled by prolyl hydroxylase domain (PHD) dioxygenases, which have been exploited as therapeutic targets for the treatment of anemia of chronic kidney disease. Here, we established a mouse model of acute cyclophosphamide (CYP)-induced blood-urine barrier disruption associated with inflammation and severe urinary dysfunction to investigate the HIF-PHD axis in inflammatory bladder injury. We found that systemic administration of dimethyloxalylglycine or molidustat, two small-molecule inhibitors of HIF-prolyl hydroxylases, profoundly mitigated CYP-induced bladder injury and inflammation as assessed by morphological analysis of transmural edema and urothelial integrity and by measuring tissue cytokine expression. Void spot analysis to examine bladder function quantitatively demonstrated that HIF-prolyl hydroxylase inhibitor administration normalized micturition patterns and protected against CYP-induced alteration of urinary frequency and micturition patterns. Our study highlights the therapeutic potential of HIF-activating small-molecule compounds for the prevention or therapy of bladder injury and urinary dysfunction due to blood-urine barrier disruption.NEW & NOTEWORTHY Disruption of the blood-urine barrier can result in acute or chronic inflammatory bladder injury. Here, we demonstrate that pharmacological inhibition of hypoxia-inducible factor (HIF)-prolyl hydroxylation prevented bladder injury and protected from urinary dysfunction in a mouse model of cyclophosphamide-induced disruption of the blood-urine barrier. Our study highlights a potential role for HIF-activating small-molecule compounds in the prevention or therapy of bladder injury and urinary dysfunction and provides a rationale for future clinical studies.


Asunto(s)
Prolina Dioxigenasas del Factor Inducible por Hipoxia , Vejiga Urinaria , Animales , Ciclofosfamida/toxicidad , Modelos Animales de Enfermedad , Hidroxilación , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Inflamación/metabolismo , Ratones , Procolágeno-Prolina Dioxigenasa/metabolismo , Prolil Hidroxilasas/metabolismo , Vejiga Urinaria/metabolismo
3.
Acta Physiol (Oxf) ; 235(4): e13826, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35491502

RESUMEN

AIM: Erythropoietin (EPO) is regulated by hypoxia-inducible factor (HIF)-2. In the kidney, it is produced by cortico-medullary perivascular interstitial cells, which transdifferentiate into collagen-producing myofibroblasts in response to injury. Inhibitors of prolyl hydroxylase domain (PHD) dioxygenases (HIF-PHIs) activate HIF-2 and stimulate kidney and liver EPO synthesis in patients with anemia of chronic kidney disease (CKD). We examined whether HIF-PHIs can reactivate EPO synthesis in interstitial cells that have undergone myofibroblast transdifferentiation in established kidney fibrosis. METHODS: We investigated Epo transcription in myofibroblasts and characterized the histological distribution of kidney Epo transcripts by RNA in situ hybridization combined with immunofluorescence in mice with adenine nephropathy (AN) treated with HIF-PHI molidustat.  Lectin absorption chromatography was used to assess liver-derived EPO.  In addition, we examined kidney Epo transcription in Phd2 knockout mice with obstructive nephropathy. RESULTS: In AN, molidustat-induced Epo transcripts were not found in areas of fibrosis and did not colocalize with interstitial cells that expressed α-smooth muscle actin, a marker of myofibroblast transdifferentiation. Epo transcription was associated with megalin-expressing, kidney injury molecule 1-negative nephron segments and contingent on residual renal function. Liver-derived EPO did not contribute to serum EPO in molidustat-treated mice. Epo transcription was not associated with myofibroblasts in Phd2 knockout mice with obstructive nephropathy. CONCLUSIONS: Our studies suggest that HIF-PHIs do not reactivate Epo transcription in interstitial myofibroblasts and that their efficacy in inducing kidney EPO in CKD is dependent on the degree of myofibroblast formation, the preservation of renal parenchyma and the level of residual renal function.


Asunto(s)
Eritropoyetina , Insuficiencia Renal Crónica , Animales , Transdiferenciación Celular , Eritropoyetina/farmacología , Fibrosis , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Riñón , Ratones , Ratones Noqueados , Miofibroblastos , Nefronas , Prolil Hidroxilasas , Insuficiencia Renal Crónica/complicaciones
4.
Kidney Int ; 102(1): 108-120, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35341793

RESUMEN

Oxidative metabolism in mitochondria regulates cellular differentiation and gene expression through intermediary metabolites and reactive oxygen species. Its role in kidney development and pathogenesis is not completely understood. Here we inactivated ubiquinone-binding protein QPC, a subunit of mitochondrial complex III, in two types of kidney progenitor cells to investigate the role of mitochondrial electron transport in kidney homeostasis. Inactivation of QPC in sine oculis-related homeobox 2 (SIX2)-expressing cap mesenchyme progenitors, which give rise to podocytes and all nephron segments except collecting ducts, resulted in perinatal death from severe kidney dysplasia. This was characterized by decreased proliferation of SIX2 progenitors and their failure to differentiate into kidney epithelium. QPC inactivation in cap mesenchyme progenitors induced activating transcription factor 4-mediated nutritional stress responses and was associated with a reduction in kidney tricarboxylic acid cycle metabolites and amino acid levels, which negatively impacted purine and pyrimidine synthesis. In contrast, QPC inactivation in ureteric tree epithelial cells, which give rise to the kidney collecting system, did not inhibit ureteric differentiation, and resulted in the development of functional kidneys that were smaller in size. Thus, our data demonstrate that mitochondrial oxidative metabolism is critical for the formation of cap mesenchyme-derived nephron segments but dispensable for formation of the kidney collecting system. Hence, our studies reveal compartment-specific needs for metabolic reprogramming during kidney development.


Asunto(s)
Complejo III de Transporte de Electrones , Riñón , Nefronas , Organogénesis , Podocitos , Aminoácidos/deficiencia , Diferenciación Celular , Complejo III de Transporte de Electrones/metabolismo , Femenino , Humanos , Riñón/embriología , Riñón/metabolismo , Mesodermo/metabolismo , Nefronas/metabolismo , Organogénesis/genética , Podocitos/metabolismo , Embarazo , Uréter/embriología
5.
Acta Physiol (Oxf) ; 233(1): e13668, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33900001

RESUMEN

AIM: Hypoxia-inducible factors (HIFs) are O2 -sensitive transcription factors that regulate multiple biological processes which are essential for cellular adaptation to hypoxia. Small molecule inhibitors of HIF-prolyl hydroxylase domain (PHD) dioxygenases (HIF-PHIs) activate HIF-dependent transcriptional programs and have broad clinical potential. HIF-PHIs are currently in global late-stage clinical development for the treatment of anaemia associated with chronic kidney disease. Although the effects of hypoxia on renal haemodynamics and function have been studied in animal models and in humans living at high altitude, the effects of pharmacological HIF activation on renal haemodynamics, O2 metabolism and metabolic efficiency are not well understood. METHODS: Using a cross-sectional study design, we investigated renal haemodynamics, O2 metabolism, gene expression and NO production in healthy rats treated with different doses of HIF-PHIs roxadustat or molidustat compared to vehicle control. RESULTS: Systemic administration of roxadustat or molidustat resulted in a dose-dependent reduction in renovascular resistance (RVR). This was associated with increased glomerular filtration rate (GFR), urine flow and tubular sodium transport rate (TNa ). Although both total O2 delivery and TNa were increased, more O2 was extracted per transported sodium in rats treated with high-doses of HIF-PHIs, suggesting a reduction in metabolic efficiency. Changes in RVR and GFR were associated with increased nitric oxide (NO) generation and substantially suppressed by pharmacological inhibition of NO synthesis. CONCLUSIONS: Our data provide mechanistic insights into dose-dependent effects of short-term pharmacological HIF activation on renal haemodynamics, glomerular filtration and O2 metabolism and identify NO as a major mediator of these effects.


Asunto(s)
Fenómenos Biológicos , Insuficiencia Renal Crónica , Animales , Estudios Transversales , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Óxido Nítrico , Prolil Hidroxilasas , Ratas
6.
Kidney Int ; 99(3): 657-670, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33159962

RESUMEN

Abnormal mitochondrial function is a well-recognized feature of acute and chronic kidney diseases. To gain insight into the role of mitochondria in kidney homeostasis and pathogenesis, we targeted mitochondrial transcription factor A (TFAM), a protein required for mitochondrial DNA replication and transcription that plays a critical part in the maintenance of mitochondrial mass and function. To examine the consequences of disrupted mitochondrial function in kidney epithelial cells, we inactivated TFAM in sine oculis-related homeobox 2-expressing kidney progenitor cells. TFAM deficiency resulted in significantly decreased mitochondrial gene expression, mitochondrial depletion, inhibition of nephron maturation and the development of severe postnatal cystic disease, which resulted in premature death. This was associated with abnormal mitochondrial morphology, a reduction in oxygen consumption and increased glycolytic flux. Furthermore, we found that TFAM expression was reduced in murine and human polycystic kidneys, which was accompanied by mitochondrial depletion. Thus, our data suggest that dysregulation of TFAM expression and mitochondrial depletion are molecular features of kidney cystic disease that may contribute to its pathogenesis.


Asunto(s)
Proteínas de Unión al ADN , Factores de Transcripción , Animales , Proteínas del Grupo de Alta Movilidad , Humanos , Riñón , Ratones , Proteínas Mitocondriales/genética , Factores de Transcripción/genética
7.
Kidney Int ; 92(6): 1370-1383, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28847650

RESUMEN

Hypoxia in the embryo is a frequent cause of intra-uterine growth retardation, low birth weight, and multiple organ defects. In the kidney, this can lead to low nephron endowment, predisposing to chronic kidney disease and arterial hypertension. A key component in cellular adaptation to hypoxia is the hypoxia-inducible factor pathway, which is regulated by prolyl-4-hydroxylase domain (PHD) dioxygenases PHD1, PHD2, and PHD3. In the adult kidney, PHD oxygen sensors are differentially expressed in a cell type-dependent manner and control the production of erythropoietin in interstitial cells. However, the role of interstitial cell PHDs in renal development has not been examined. Here we used a genetic approach in mice to interrogate PHD function in FOXD1-expressing stroma during nephrogenesis. We demonstrate that PHD2 and PHD3 are essential for normal kidney development as the combined inactivation of stromal PHD2 and PHD3 resulted in renal failure that was associated with reduced kidney size, decreased numbers of glomeruli, and abnormal postnatal nephron formation. In contrast, nephrogenesis was normal in animals with individual PHD inactivation. We furthermore demonstrate that the defect in nephron formation in PHD2/PHD3 double mutants required intact hypoxia-inducible factor-2 signaling and was dependent on the extent of stromal hypoxia-inducible factor activation. Thus, hypoxia-inducible factor prolyl-4-hydroxylation in renal interstitial cells is critical for normal nephron formation.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/fisiología , Riñón/crecimiento & desarrollo , Procolágeno-Prolina Dioxigenasa/fisiología , Insuficiencia Renal/genética , Anemia/sangre , Anemia/tratamiento farmacológico , Anemia/etiología , Animales , Hipoxia de la Célula/fisiología , Ensayos Clínicos Fase III como Asunto , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Hidroxilación/fisiología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Riñón/citología , Riñón/metabolismo , Enfermedades Renales/complicaciones , Enfermedades Renales/tratamiento farmacológico , Ratones , Terapia Molecular Dirigida/métodos , Mutación , Tamaño de los Órganos/fisiología , Procolágeno-Prolina Dioxigenasa/antagonistas & inhibidores , Procolágeno-Prolina Dioxigenasa/genética , Insuficiencia Renal/mortalidad , Insuficiencia Renal/patología , Células del Estroma/metabolismo
8.
J Clin Invest ; 126(5): 1926-38, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-27088801

RESUMEN

Renal peritubular interstitial fibroblast-like cells are critical for adult erythropoiesis, as they are the main source of erythropoietin (EPO). Hypoxia-inducible factor 2 (HIF-2) controls EPO synthesis in the kidney and liver and is regulated by prolyl-4-hydroxylase domain (PHD) dioxygenases PHD1, PHD2, and PHD3, which function as cellular oxygen sensors. Renal interstitial cells with EPO-producing capacity are poorly characterized, and the role of the PHD/HIF-2 axis in renal EPO-producing cell (REPC) plasticity is unclear. Here we targeted the PHD/HIF-2/EPO axis in FOXD1 stroma-derived renal interstitial cells and examined the role of individual PHDs in REPC pool size regulation and renal EPO output. Renal interstitial cells with EPO-producing capacity were entirely derived from FOXD1-expressing stroma, and Phd2 inactivation alone induced renal Epo in a limited number of renal interstitial cells. EPO induction was submaximal, as hypoxia or pharmacologic PHD inhibition further increased the REPC fraction among Phd2-/- renal interstitial cells. Moreover, Phd1 and Phd3 were differentially expressed in renal interstitium, and heterozygous deficiency for Phd1 and Phd3 increased REPC numbers in Phd2-/- mice. We propose that FOXD1 lineage renal interstitial cells consist of distinct subpopulations that differ in their responsiveness to Phd2 inactivation and thus regulation of HIF-2 activity and EPO production under hypoxia or conditions of pharmacologic or genetic PHD inactivation.


Asunto(s)
Eritropoyetina/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Hipoxia/metabolismo , Riñón/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Eritropoyetina/genética , Factores de Transcripción Forkhead/genética , Hipoxia/genética , Hipoxia/patología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Riñón/irrigación sanguínea , Riñón/patología , Ratones , Ratones Noqueados , Procolágeno-Prolina Dioxigenasa/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología
9.
J Clin Invest ; 126(4): 1425-37, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26927670

RESUMEN

The adult kidney plays a central role in erythropoiesis and is the main source of erythropoietin (EPO), an oxygen-sensitive glycoprotein that is essential for red blood cell production. Decreases of renal pO2 promote hypoxia-inducible factor 2-mediated (HIF-2-mediated) induction of EPO in peritubular interstitial fibroblast-like cells, which serve as the cellular site of EPO synthesis in the kidney. It is not clear whether HIF signaling in other renal cell types also contributes to the regulation of EPO production. Here, we used a genetic approach in mice to investigate the role of renal epithelial HIF in erythropoiesis. Specifically, we found that HIF activation in the proximal nephron via induced inactivation of the von Hippel-Lindau tumor suppressor, which targets the HIF-α subunit for proteasomal degradation, led to rapid development of hypoproliferative anemia that was associated with a reduction in the number of EPO-producing renal interstitial cells. Moreover, suppression of renal EPO production was associated with increased glucose uptake, enhanced glycolysis, reduced mitochondrial mass, diminished O2 consumption, and elevated renal tissue pO2. Our genetic analysis suggests that tubulointerstitial cellular crosstalk modulates renal EPO production under conditions of epithelial HIF activation in the kidney.


Asunto(s)
Eritropoyesis , Eritropoyetina/biosíntesis , Túbulos Renales Proximales/metabolismo , Factores de Transcripción/metabolismo , Animales , Epitelio/metabolismo , Eritropoyetina/genética , Glucosa/genética , Glucosa/metabolismo , Ratones , Ratones Transgénicos , Consumo de Oxígeno , Factores de Transcripción/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
10.
Am J Physiol Renal Physiol ; 308(12): F1452-62, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25925251

RESUMEN

Ischemia-reperfusion injury (IRI) due to hypotension is a common cause of human acute kidney injury (AKI). Hypoxia-inducible transcription factors (HIFs) orchestrate a protective response in renal endothelial and epithelial cells in AKI models. As human mucin 1 (MUC1) is induced by hypoxia and enhances HIF-1 activity in cultured epithelial cells, we asked whether mouse mucin 1 (Muc1) regulates HIF-1 activity in kidney tissue during IRI. Whereas Muc1 was localized on the apical surface of the thick ascending limb, distal convoluted tubule, and collecting duct in the kidneys of sham-treated mice, Muc1 appeared in the cytoplasm and nucleus of all tubular epithelia during IRI. Muc1 was induced during IRI, and Muc1 transcripts and protein were also present in recovering proximal tubule cells. Kidney damage was worse and recovery was blocked during IRI in Muc1 knockout mice compared with congenic control mice. Muc1 knockout mice had reduced levels of HIF-1α, reduced or aberrant induction of HIF-1 target genes involved in the shift of glucose metabolism to glycolysis, and prolonged activation of AMP-activated protein kinase, indicating metabolic stress. Muc1 clearly plays a significant role in enhancing the HIF protective pathway during ischemic insult and recovery in kidney epithelia, providing a new target for developing therapies to treat AKI. Moreover, our data support a role specifically for HIF-1 in epithelial protection of the kidney during IRI as Muc1 is present only in tubule epithelial cells.


Asunto(s)
Mucina-1/metabolismo , Daño por Reperfusión/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Riñón/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/fisiopatología
11.
J Clin Invest ; 124(6): 2396-409, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24789906

RESUMEN

The hypoxia-inducible transcription factors HIF-1 and HIF-2 mediate key cellular adaptions to hypoxia and contribute to renal homeostasis and pathophysiology; however, little is known about the cell type-specific functions of HIF-1 and HIF-2 in response to ischemic kidney injury. Here, we used a genetic approach to specifically dissect the roles of endothelial HIF-1 and HIF-2 in murine models of hypoxic kidney injury induced by ischemia reperfusion or ureteral obstruction. In both models, inactivation of endothelial HIF increased injury-associated renal inflammation and fibrosis. Specifically, inactivation of endothelial HIF-2α, but not endothelial HIF-1α, resulted in increased expression of renal injury markers and inflammatory cell infiltration in the postischemic kidney, which was reversed by blockade of vascular cell adhesion molecule-1 (VCAM1) and very late antigen-4 (VLA4) using monoclonal antibodies. In contrast, pharmacologic or genetic activation of HIF via HIF prolyl-hydroxylase inhibition protected wild-type animals from ischemic kidney injury and inflammation; however, these same protective effects were not observed in HIF prolyl-hydroxylase inhibitor-treated animals lacking endothelial HIF-2. Taken together, our data indicate that endothelial HIF-2 protects from hypoxia-induced renal damage and represents a potential therapeutic target for renoprotection and prevention of fibrosis following acute ischemic injury.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Isquemia/fisiopatología , Riñón/lesiones , Riñón/fisiopatología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Fibrosis , Células Endoteliales de la Vena Umbilical Humana , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Integrina alfa4beta1/antagonistas & inhibidores , Integrina alfa4beta1/fisiología , Isquemia/patología , Isquemia/prevención & control , Riñón/irrigación sanguínea , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/prevención & control , Obstrucción Ureteral/complicaciones , Molécula 1 de Adhesión Celular Vascular/fisiología
12.
J Immunol ; 188(10): 5106-15, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22490864

RESUMEN

Renal fibrosis and inflammation are associated with hypoxia, and tissue pO(2) plays a central role in modulating the progression of chronic kidney disease. Key mediators of cellular adaptation to hypoxia are hypoxia-inducible factor (HIF)-1 and -2. In the kidney, they are expressed in a cell type-specific manner; to what degree activation of each homolog modulates renal fibrogenesis and inflammation has not been established. To address this issue, we used Cre-loxP recombination to activate or to delete both Hif-1 and Hif-2 either globally or cell type specifically in myeloid cells. Global activation of Hif suppressed inflammation and fibrogenesis in mice subjected to unilateral ureteral obstruction, whereas activation of Hif in myeloid cells suppressed inflammation only. Suppression of inflammatory cell infiltration was associated with downregulation of CC chemokine receptors in renal macrophages. Conversely, global deletion or myeloid-specific inactivation of Hif promoted inflammation. Furthermore, prolonged hypoxia suppressed the expression of multiple inflammatory molecules in noninjured kidneys. Collectively, we provide experimental evidence that hypoxia and/or myeloid cell-specific HIF activation attenuates renal inflammation associated with chronic kidney injury.


Asunto(s)
Lesión Renal Aguda/inmunología , Lesión Renal Aguda/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Células Mieloides/inmunología , Células Mieloides/patología , Obstrucción Ureteral/inmunología , Obstrucción Ureteral/patología , Lesión Renal Aguda/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Modelos Animales de Enfermedad , Fibrosis/inmunología , Fibrosis/prevención & control , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Inflamación/inmunología , Inflamación/patología , Inflamación/prevención & control , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Cultivo Primario de Células , Obstrucción Ureteral/genética
13.
PLoS One ; 5(3): e9458, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20221440

RESUMEN

BACKGROUND: Inflammation is associated with most diseases, which makes understanding the mechanisms of inflammation vitally important. METHODOLOGY/PRINCIPAL FINDINGS: Here, we demonstrate a critical function of interleukin-32beta (IL-32beta) in vascular inflammation. IL-32beta is present in tissues from humans, but is absent in rodents. We found that the gene is highly expressed in endothelial cells. Three isoforms of IL-32, named IL-32alpha, beta, and epsilon, were cloned from human endothelial cells, with IL-32beta being the major isoform. Pro-inflammatory cytokines (TNFalpha and IL-1beta) induced IL-32beta expression through NF-kappaB. Conversely, IL-32beta propagated vascular inflammation via induction of vascular cell adhesion molecules and inflammatory cytokines. Accordingly, IL-32beta increased adhesion of inflammatory cells to activated endothelial cells, a paramount process in inflammation. These results illustrate a positive feedback regulation that intensifies and prolongs inflammation. Importantly, endothelial/hematopoietic expression of IL-32beta in transgenic mice elevated inflammation and worsened sepsis. This was demonstrated by significant elevation of leukocyte infiltration and serum levels of TNFalpha and IL-1beta, increased vascular permeability and lung damage, and accelerated animal death. Together, our results reveal an important function of IL-32 in vascular inflammation and sepsis development. CONCLUSIONS/SIGNIFICANCE: Our results reveal an important function of IL-32 in vascular inflammation and sepsis development.


Asunto(s)
Vasos Sanguíneos/patología , Interleucinas/metabolismo , Sepsis/metabolismo , Animales , Clonación Molecular , Endotelio Vascular/metabolismo , Humanos , Inflamación , Interleucina-1beta/metabolismo , Pulmón/patología , Ratones , Ratones Transgénicos , Isoformas de Proteínas , Sepsis/patología , Factor de Necrosis Tumoral alfa/metabolismo
14.
Int J Radiat Oncol Biol Phys ; 74(5): 1573-9, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19616744

RESUMEN

PURPOSE: To study the role of interleukin-32 (IL-32), a novel protein only detected in human tissues, in ionizing radiation (IR)-induced vascular inflammation. METHODS AND MATERIALS: Irradiated (0-6 Gy) human umbilical vein endothelial cells treated with or without various agents--a cytosolic phospholipase A2 (cPLA2) inhibitor, a cyclooxygenase-2 (Cox-2) inhibitor, or lysophosphatidylcholines (LPCs)--were used to assess IL-32 expression by Northern blot analysis and quantitative reverse transcriptase-polymerase chain reaction. Expression of cell adhesion molecules and leukocyte adhesion to endothelial cells using human acute monocytic leukemia cell line (THP-1) cells was also analyzed. RESULTS: Ionizing radiation dramatically increased IL-32 expression in vascular endothelial cells through multiple pathways. Ionizing radiation induced IL-32 expression through nuclear factor kappaB activation, through induction of cPLA2 and LPC, as well as induction of Cox-2 and subsequent conversion of arachidonic acid to prostacyclin. Conversely, blocking nuclear factor kappaB, cPLA2, and Cox-2 activity impaired IR-induced IL-32 expression. Importantly, IL-32 significantly enhanced IR-induced expression of vascular cell adhesion molecules and leukocyte adhesion on endothelial cells. CONCLUSION: This study identifies IL-32 as a positive regulator in IR-induced vascular inflammation, and neutralization of IL-32 may be beneficial in protecting from IR-induced inflammation.


Asunto(s)
Endotelio Vascular/efectos de la radiación , Interleucinas/fisiología , Vasculitis/metabolismo , Moléculas de Adhesión Celular/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Endotelio Vascular/metabolismo , Fosfolipasas A2 Grupo IV/antagonistas & inhibidores , Fosfolipasas A2 Grupo IV/metabolismo , Fosfolipasas A2 Grupo IV/fisiología , Humanos , Interleucinas/antagonistas & inhibidores , Interleucinas/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , FN-kappa B/fisiología , Subtilisinas/antagonistas & inhibidores , Subtilisinas/metabolismo , Venas Umbilicales/metabolismo , Venas Umbilicales/efectos de la radiación , Molécula 1 de Adhesión Celular Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vasculitis/etiología
15.
Methods Mol Biol ; 511: 185-91, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19347298

RESUMEN

Angiogenesis, the formation of new blood vessels from existing vessels, is tightly linked to chronic inflammation and cancer. Angiogenesis is one of the molecular events bridging the gap between inflammation and cancer. One of the events linking inflammation and cancer is an increase in cellular adhesion molecules that are expressed on the luminal surface of endothelium upon inflammation. Cellular adhesion molecules are involved in leukocyte recruitment and subsequently lead to extravasation of leukocytes to the injury site. These adhesion molecules are known to be shared by some cancer cells and have the ability to contribute to metastasis. Thus, an elevation of these molecules in chronic inflammation may be a risk factor for metastasis. In this chapter, we discuss the method used to determine the adhesion molecules expressed on endothelium, and leukocyte adhesion to endothelium.


Asunto(s)
Bioensayo , Inflamación/patología , Neoplasias/patología , Neovascularización Patológica , Bioensayo/instrumentación , Bioensayo/métodos , Adhesión Celular , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Enfermedad Crónica , Humanos , Leucocitos/metabolismo
16.
Cytokine ; 46(3): 351-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19364659

RESUMEN

IL-32 is a newly discovered protein found in human and certain primates, but absent in rodent. Various reports suggest its role as a proinflammatory mediator. Since vascular endothelium is critical in inflammation, we investigate IL-32 in endothelial cells. We found that the gene is expressed in human endothelial cells and Akt strongly induces its expression. Sequence analysis indicates IL-32 beta as the major isoform in endothelial cells. Surprisingly, we did not detect any secretion of IL-32 beta in human endothelial cells; instead we observed co-localization of IL-32 beta with endoplasmic reticulum, suggesting IL-32 beta is an intracellular protein in these cells. Promoter analysis identified a minimum required region for IL-32 transcription at -0.1 to +0.5 kb around the initially identified transcription start site. We also defined a transcriptional suppressor-binding site at -2.0 to -1.5 kb. Importantly, RNA ligase mediated rapid amplification of cDNA ends in endothelial cells determined the transcription start site at the 328 bp downstream from the original identified site. Finally, we found a positive correlation of IL-32 levels with human breast cancer and glioblastoma multiforme (GBM). These findings improve our understanding of IL-32 in vascular endothelium. IL-32 expression might be valuable as a biomarker for cancer.


Asunto(s)
Células Endoteliales/inmunología , Interleucinas/inmunología , Isoformas de Proteínas/inmunología , Animales , Secuencia de Bases , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Bovinos , Células Cultivadas , Células Endoteliales/citología , Femenino , Regulación de la Expresión Génica , Humanos , Interleucinas/genética , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Isoformas de Proteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sitio de Iniciación de la Transcripción
17.
Curr Med Chem ; 14(4): 377-86, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17305540

RESUMEN

The role of cell adhesion molecules (CAMs), such as intercellular cell adhesion molecule-1 (ICAM-1), vascular endothelial cell adhesion molecule-1 (VCAM-1), E-selectin, and P-selectin, has been studied extensively in the process of inflammation. These molecules are responsible for recruiting leukocytes onto the vascular endothelium before extravasation to the injured tissues. Some circulating cancer cells have been shown to extravasate to a secondary site using a process similar to inflammatory cells. The most studied ligands for CAMs expressed on cancer cells, sialyl Lewis (a/x) antigens, are shown to be involved in adhesion to endothelial cells by binding to E-selectin. This process, shared by inflammatory cells and cancer cells, may partially explain the link between inflammation and tumorigenesis. Furthermore, this process may elucidate the therapeutic benefit of anti-inflammatory drugs in cancer treatment. The complexity of the tumor microenvironment has been revealed in the past decade. Currently, intense investigation is aimed at various aspects of the tumor microenvironment in addition to the tumor cells themselves. Here, we review the role of CAMs in extravasation of circulating cancer cells, a key step in metastasis.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Neoplasias/patología , Moléculas de Adhesión Celular/metabolismo , Progresión de la Enfermedad , Selectina E/metabolismo , Humanos , Metástasis de la Neoplasia/patología , Neoplasias/etiología
18.
Blood ; 108(4): 1260-6, 2006 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16638932

RESUMEN

Communication between endothelial cells (ECs) and mural cells is critical in vascular maturation. Genetic studies suggest that angiopoietin/Tie2 signaling may play a role in the recruitment of pericytes or smooth muscle cells (SMCs) during vascular maturation. However, the molecular mechanism is unclear. We used microarray technology to analyze genes regulated by angiopoietin-1 (Ang1), an agonist ligand for Tie2, in endothelial cells (ECs). We observed that hepatocyte growth factor (HGF), a mediator of mural cell motility, was up-regulated by Ang1 stimulation. We confirmed this finding by Northern blot and Western blot analyses in cultured vascular endothelial cells. Furthermore, stimulation of ECs with Ang1 increased SMC migration toward endothelial cells in a coculture assay. Addition of a neutralizing anti-HGF antibody inhibited Ang1-induced SMC recruitment, indicating that the induction of SMC migration by Ang1 was caused by the increase of HGF. Interestingly, Ang2, an antagonist ligand of Tie2, inhibited Ang1-induced HGF production and Ang1-induced SMC migration. Finally, we showed that deletion of Tie2 in transgenic mouse reduced HGF production. Collectively, our data reveal a novel mechanism of Ang/Tie2 signaling in regulating vascular maturation and suggest that a delicate balance between Ang1 and Ang2 is critical in this process.


Asunto(s)
Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Movimiento Celular/fisiología , Factor de Crecimiento de Hepatocito/biosíntesis , Miocitos del Músculo Liso/fisiología , Transducción de Señal/fisiología , Angiopoyetina 1/farmacología , Angiopoyetina 2/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/fisiología , Perfilación de la Expresión Génica , Humanos , Miocitos del Músculo Liso/citología , Análisis de Secuencia por Matrices de Oligonucleótidos , Pericitos/citología , Pericitos/fisiología , Receptor TIE-2/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
19.
Nanomedicine (Lond) ; 1(1): 17-22, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17716205

RESUMEN

Nanotechnology has advanced greatly in recent years and antiangiogenic therapy is becoming a promising approach for cancer treatment. Therefore, a combination of nanotechnology with antiangiogenic therapy should significantly enhance our ability to treat this deadly disease. alpha(v)beta(3)-integrin has often been used to target 'angiogenically active' tumor endothelium and many nanoparticles have been developed by combining this molecule with other agents for imaging and/or therapy. Development of improved and multifunctional nanoparticles is in progress for the enhancement of imaging, targeting, delivery and other processes. Modification for clinical use has been challenging. Concerns remain regarding safety, stability and tissue targeting of the particles, yet significant progress has been made. In this review, studies targeting 'angiogenically active' tumor endothelium using nanoparticles are discussed. The number of studies using multifunctional nanotechnology in tumor angiogenesis is still limited; however, significant advancement in this area is expected to come in the near future.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Nanotecnología/métodos , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/química , Animales , Portadores de Fármacos , Humanos , Nanopartículas/química , Nanotecnología/tendencias , Neoplasias/irrigación sanguínea , Neoplasias/patología
20.
Arthritis Rheum ; 52(5): 1585-94, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15880817

RESUMEN

OBJECTIVE: In a previous study, we demonstrated that Tie2 regulates angiogenesis in arthritis. The current study was performed to determine whether systemic delivery of a soluble Tie2 receptor (ExTek) using an adenoviral vector (AdExTek) as a Tie2 inhibitor affects arthritis development and progression in an animal model. METHODS: We used a collagen-induced arthritis (CIA) mouse model to study the outcome of treatment with either AdExTek or a control vector. The onset, incidence, and severity of arthritis were quantified. Immunohistologic analysis of endothelium obtained from the paws was performed. Bone destruction in paws was analyzed using phase-contrast radiography. RESULTS: The data showed that systemic delivery of ExTek before disease development significantly inhibited the onset, incidence, and severity of arthritis. When AdExTek was given after disease onset, the severity of disease in mice treated with AdExTek was significantly lower than that in the control group at 35 days postimmunization, which correlated with significantly diminished angiogenesis in mouse paws. Strikingly, AdExTek treatment protected bone from erosion in the CIA model and reduced levels of RANKL. No differences in collagen-specific antibodies were detected between these 2 groups. CONCLUSION: We demonstrated that blocking Tie2 receptor activation inhibits angiogenesis and arthritis development and protects against bone destruction in a CIA mouse model. These findings identify Tie2 as a therapeutic target for arthritis treatment and imply that interventions designed to target the Tie2 pathway could be clinically beneficial.


Asunto(s)
Artritis/terapia , Terapia Genética , Receptor TIE-2/genética , Adenoviridae , Animales , Artritis/inducido químicamente , Enfermedades Óseas/prevención & control , Colágeno/administración & dosificación , Progresión de la Enfermedad , Ratones , Ratones Endogámicos DBA , Neovascularización Patológica/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...