Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 318(3): H671-H681, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32004074

RESUMEN

In the murine venous thrombosis model induced by ligation of the inferior vena cava (IVCL), genetic deficiency of heme oxygenase-1 (HO-1) increases clot size. This study examined whether induction of HO-1 or administration of its products reduces thrombosis. Venous HO-1 upregulation by gene delivery reduced clot size, as did products of HO activity, biliverdin, and carbon monoxide. Induction of HO-1 by hemin reduced clot formation, clot size, and upregulation of plasminogen activator inhibitor-1 (PAI-1) that occurs in the IVCL model, while leaving urokinase plasminogen activator (uPA) and tissue plasminogen activator (tPA) expression unaltered. The reductive effect of hemin on clot size required HO activity. The IVCL model exhibited relatively high concentrations of heme that peaked just before maximum clot size, then declined as clot size decreased. Administration of hemin decreased heme concentration in the IVCL model. HO-2 mRNA was induced twofold in the IVCL model (vs. 40-fold HO-1 induction), but clot size was not increased in HO-2-/- mice compared with HO-2+/+ mice. Hemopexin, the major heme-binding protein, was induced in the IVCL model, and clot size was increased in hemopexin-/- mice compared with hemopexin+/+ mice. We conclude that in the IVCL model, the heme-degrading protein HO-1 and HO products inhibit thrombus formation, as does the heme-binding protein, hemopexin. The reductive effects of hemin administration require HO activity and are mediated, in part, by reducing PAI-1 upregulation in the IVCL model. We speculate that HO-1, HO, and hemopexin reduce clot size by restraining the increase in clot concentration of heme (now recognized as a procoagulant) that otherwise occurs.NEW & NOTEWORTHY This study provides conclusive evidence that two proteins, one heme-degrading and the other heme-binding, inhibit clot formation. This may serve as a new therapeutic strategy in preventing and treating venous thromboembolic disease.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Proteínas de Unión al Hemo/metabolismo , Regulación hacia Arriba , Trombosis de la Vena/metabolismo , Animales , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/genética , Proteínas de Unión al Hemo/genética , Hemina/farmacología , Ratones , Ratones Noqueados , Trombosis de la Vena/genética
2.
Am J Physiol Renal Physiol ; 317(3): F695-F704, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31215802

RESUMEN

Heme oxygenase (HO) activity is exhibited by inducible (HO-1) and constitutive (HO-2) proteins. HO-1 protects against ischemic and nephrotoxic acute kidney injury (AKI). We have previously demonstrated that HO-2 protects against heme protein-induced AKI. The present study examined whether HO-2 is protective in ischemic AKI. Renal ischemia was imposed on young and aged HO-2+/+ and HO-2-/- mice. On days 1 and 2 after renal ischemia, there were no significant differences in renal function between young male HO-2+/+ and HO-2-/- mice, between young female HO-2+/+ and HO-2-/- mice, or between aged female HO-2+/+ and HO-2-/- mice. However, in aged male mice, HO-2 deficiency worsened renal function on days 1 and 2 after ischemic AKI, and, on day 2 after ischemia, such deficiency augmented upregulation of injury-related genes and worsened histological injury. Renal HO activity was markedly decreased in unstressed aged male HO-2-/- mice and remained so after ischemia, despite exaggerated HO-1 induction in HO-2-/- mice after ischemia. Such exacerbation of deficiency of HO-2 protein and HO activity may reflect phosphorylated STAT3, as activation of this proinflammatory transcription factor was accentuated early after ischemia in aged male HO-2-/- mice. This exacerbation may not reflect impaired induction of nephroprotectant genes, since the induction of HO-1, sirtuin 1, and ß-catenin was accentuated in aged male HO-2-/- mice after ischemia. We conclude that aged male mice are hypersensitive to ischemic AKI and that HO-2 mitigates such sensitivity. We speculate that this protective effect of HO-2 may be mediated, at least in part, by suppression of phosphorylated STAT3-dependent signaling.


Asunto(s)
Lesión Renal Aguda/prevención & control , Hemo Oxigenasa (Desciclizante)/metabolismo , Riñón/enzimología , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/enzimología , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Factores de Edad , Animales , Modelos Animales de Enfermedad , Femenino , Hemo Oxigenasa (Desciclizante)/deficiencia , Hemo Oxigenasa (Desciclizante)/genética , Riñón/patología , Riñón/fisiopatología , Masculino , Ratones Noqueados , Fosforilación , Daño por Reperfusión/enzimología , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Factor de Transcripción STAT3/metabolismo , Factores Sexuales , Transducción de Señal
3.
Am J Physiol Renal Physiol ; 314(5): F906-F914, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28978536

RESUMEN

Destabilized heme proteins release heme, and free heme is toxic. Heme is now recognized as an agonist for the Toll-like receptor-4 (TLR4) receptor. This study examined whether the TLR4 receptor mediates the nephrotoxicity of heme, specifically, the effects of heme on renal blood flow and inflammatory responses. We blocked TLR4 signaling by the specific antagonist TAK-242. Intravenous administration of heme to mice promptly reduced renal blood flow, an effect attenuated by TAK-242. In vitro, TAK-242 reduced heme-elicited activation of NF-κB and its downstream gene monocyte chemoattractant protein-1(MCP-1); in contrast, TAK-242 failed to reduce heme-induced activation of the anti-inflammatory transcription factor Nrf2 and its downstream gene heme oxygenase-1 (HO-1). TAK-242 did not reduce heme-induced renal MCP-1 upregulation in vivo. TAK-242 did not reduce dysfunction and histological injury in the glycerol model of heme protein-induced acute kidney injury (AKI), findings corroborated by studies in TLR4+/+ and TLR4-/- mice. We conclude that 1) acute heme-mediated renal vasoconstriction occurs through TLR4 signaling; 2) proinflammatory effects of heme in renal epithelial cells involve TLR4 signaling, whereas the anti-inflammatory effects of heme do not; 3) TLR4 signaling does not mediate the proinflammatory effects of heme in the kidney; and 4) major mechanisms underlying glycerol-induced, heme protein-mediated AKI do not involve TLR4 signaling. These findings in the glycerol model are in stark contrast with findings in virtually all other AKI models studied to date and emphasize the importance of TLR4-independent pathways of heme protein-mediated injury in this model. Finally, these studies urge caution when using observations derived in vitro to predict what occurs in vivo.


Asunto(s)
Lesión Renal Aguda/metabolismo , Células Epiteliales/metabolismo , Hemina , Riñón/irrigación sanguínea , Riñón/metabolismo , Circulación Renal , Transducción de Señal , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/metabolismo , Vasoconstricción , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Línea Celular , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Glicerol , Riñón/efectos de los fármacos , Riñón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Ratas , Circulación Renal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Vasoconstricción/efectos de los fármacos
4.
Am J Physiol Renal Physiol ; 315(5): F1493-F1499, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30019935

RESUMEN

There is no therapy that promotes maturation and functionality of a dialysis arteriovenous fistula (AVF). The search for such therapies largely relies on evaluation of vascular responses and putative therapies in experimental AVFs. We studied an AVF in mice with chronic kidney disease (CKD). We demonstrate numerous stressors in the vein of the AVF-CKD group, including pathological shear, mitogenic, inflammatory, and hypoxia-reoxygenation stress. Because stress promotes premature senescence, we examined whether senescence is induced in the vein of the AVF-CKD model. We demonstrate a senescence phenotype in the AVF-CKD model, as indicated by increased expression of p16Ink4a, p21Cip1, and p53 and expected changes for certain senescence-associated microRNAs. RNA-sequencing analysis demonstrated differential expression of ~10,000 genes, including upregulation of proinflammatory and proliferative genes, in the vein of the AVF-CKD group. The vein in the AVF-CKD group exhibited telomere erosion and increased senescence-associated ß-galactosidase activity and staining. Senescence was induced in the artery of the AVF-CKD group and in the vein of the AVF without CKD. Finally, given the rapidly rising clinical interest in senolytics, we provide proof of concept of senolytics as a therapeutic approach by demonstrating that senolytics decrease p16Ink4a expression in the AVF-CKD model. This study introduces a novel concept underlying the basis for maturational and functional failure in human dialysis AVFs and identifies a new target for senolytic therapy.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Senescencia Celular , Complicaciones Posoperatorias/patología , Diálisis Renal , Insuficiencia Renal Crónica/terapia , Cola (estructura animal)/irrigación sanguínea , Remodelación Vascular , Venas/cirugía , Animales , Derivación Arteriovenosa Quirúrgica/efectos adversos , Velocidad del Flujo Sanguíneo , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Dasatinib/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Fenotipo , Complicaciones Posoperatorias/tratamiento farmacológico , Complicaciones Posoperatorias/metabolismo , Complicaciones Posoperatorias/fisiopatología , Quercetina/farmacología , Flujo Sanguíneo Regional , Estrés Mecánico , Grado de Desobstrucción Vascular , Remodelación Vascular/efectos de los fármacos , Remodelación Vascular/genética , Venas/metabolismo , Venas/patología , Venas/fisiopatología
5.
Am J Physiol Renal Physiol ; 310(6): F466-76, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26672617

RESUMEN

The arteriovenous fistula (AVF) is the preferred hemodialysis vascular access, but it is complicated by high failure rates and attendant morbidity. This study provides the first description of a murine AVF model that recapitulates two salient features of hemodialysis AVFs, namely, anastomosis of end-vein to side-artery to create the AVF and the presence of chronic kidney disease (CKD). CKD reduced AVF blood flow, observed as early as 3 days after AVF creation, and increased neointimal hyperplasia, venous wall thickness, thrombus formation, and vasculopathic gene expression in the AVF. These adverse effects of CKD could not be ascribed to preexisting alterations in blood pressure or vascular reactivity in this CKD model. In addition to vasculopathic genes, CKD induced potentially vasoprotective genes in the AVF such as heme oxygenase-1 (HO-1) and HO-2. To determine whether prior HO-1 upregulation may protect in this model, we upregulated HO-1 by adeno-associated viral gene delivery, achieving marked venous induction of the HO-1 protein and HO activity. Such HO-1 upregulation improved AVF blood flow and decreased venous wall thickness in the AVF. Finally, we demonstrate that the administration of carbon monoxide, a product of HO, acutely increased AVF blood flow. This study thus demonstrates: 1) the feasibility of a clinically relevant murine AVF model created in the presence of CKD and involving an end-vein to side-artery anastomosis; 2) the exacerbatory effect of CKD on clinically relevant features of this model; and 3) the beneficial effects in this model conferred by HO-1 upregulation by adeno-associated viral gene delivery.


Asunto(s)
Anemia Hemolítica/complicaciones , Derivación Arteriovenosa Quirúrgica , Terapia Genética , Trastornos del Crecimiento/complicaciones , Hemo-Oxigenasa 1/deficiencia , Trastornos del Metabolismo del Hierro/complicaciones , Complicaciones Posoperatorias/etiología , Anemia Hemolítica/metabolismo , Anemia Hemolítica/terapia , Animales , Dependovirus , Estudios de Factibilidad , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/terapia , Hemo-Oxigenasa 1/metabolismo , Trastornos del Metabolismo del Hierro/metabolismo , Trastornos del Metabolismo del Hierro/terapia , Masculino , Ratones Endogámicos C57BL , Modelos Animales , Compuestos Organometálicos , Complicaciones Posoperatorias/metabolismo , Complicaciones Posoperatorias/terapia , Regulación hacia Arriba
6.
Am J Physiol Heart Circ Physiol ; 308(11): H1402-13, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25820397

RESUMEN

The present study examined the heme oxygenase (HO) system in an in vivo murine model of pathological shear stress induced by partial carotid artery ligation. In this model, along with upregulation of vasculopathic genes, HO-1 is induced in the endothelium and adventitia, whereas HO-2 is mainly upregulated in the endothelium. Within minutes of ligation, NF-κB, a transcription factor that upregulates vasculopathic genes and HO-1, is activated. Failure to express either HO-1 or HO-2 exaggerates the reduction in carotid blood flow and exacerbates vascular injury. After artery ligation, comparable induction of HO-2 occurred in HO-1(+/+) and HO-1(-/-) mice, whereas HO-1 induction was exaggerated in HO-2(-/-) mice compared with HO-2(+/+) mice. Upregulation of HO-1 by an adeno-associated viral vector increased vascular HO-1 expression and HO activity and augmented blood flow in both ligated and contralateral carotid arteries. Acute inhibition of HO activity decreased flow in the ligated carotid artery, whereas a product of HO, carbon monoxide (CO), delivered by CO-releasing molecule-3, increased carotid blood flow. In conclusion, in the partial carotid artery ligation model of pathological shear stress, this study provides the first demonstration of 1) upregulation and vasoprotective effects of HO-1 and HO-2 and the vasorelaxant effects of CO as well as 2) vascular upregulation of HO-1 in vivo by an adeno-associated viral vector that is attended by a salutary vascular response. Induction of HO-1 may reside in NF-κB activation, and, along with induced HO-2, such upregulation of HO-1 provides a countervailing vasoprotective response in pathological shear stress in vivo.


Asunto(s)
Enfermedades de las Arterias Carótidas/metabolismo , Endotelio Vascular/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/metabolismo , Mecanotransducción Celular , Proteínas de la Membrana/metabolismo , Adventicia/metabolismo , Adventicia/fisiopatología , Animales , Enfermedades de las Arterias Carótidas/fisiopatología , Endotelio Vascular/fisiopatología , Hemo Oxigenasa (Desciclizante)/genética , Hemo-Oxigenasa 1/genética , Hemodinámica , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Estrés Mecánico , Regulación hacia Arriba
7.
Am J Physiol Renal Physiol ; 305(4): F545-52, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23678042

RESUMEN

Heme oxygenase-2 (HO-2), the constitutive isoform of the heme-degrading enzyme heme oxygenase, may serve as an anti-inflammatory vasorelaxant, in part, by generating carbon monoxide. Arteriovenous fistulas (AVFs) are employed as hemodialysis vascular accesses because they provide an accessible, high-blood-flow vascular segment. We examined the role of vascular expression of HO-2 in AVF function. An AVF was created in mice by anastomosing the carotid artery to the jugular vein. HO-2 expression was detected by immunohistochemistry in the intact carotid artery, mainly in endothelial cells and smooth muscle cells; expression of HO-2 protein and mRNA was modestly increased in the artery of the AVF. Creating an AVF in HO-2(-/-) mice compared with an AVF in HO-2(+/+) mice led to markedly reduced AVF blood flow and increased numbers of nonfunctioning AVFs. The impairment of AVF function in the setting of HO-2 deficiency could not be ascribed to either preexisting intrinsic abnormalities in endothelium-dependent and endothelium-independent relaxation of the carotid artery in HO-2-deficient mice or to impaired vasorelaxant responses in the intact carotid artery in vivo. HO-1 mRNA was comparably induced in the AVF in HO-2(+/+) and HO-2(-/-) mice, whereas the AVF in HO-2(-/-) mice compared with that in HO-2(+/+) mice exhibited exaggerated induction of matrix metalloproteinase (MMP)-9 but similar induction of MMP-2. HO-2 deficiency also led to lower AVF blood flow when AVFs were created in uremia, the latter induced by subtotal nephrectomy. We conclude that HO-2 critically contributes to the adequacy of AVF blood flow and function.


Asunto(s)
Fístula Arteriovenosa/metabolismo , Arterias Carótidas/metabolismo , Hemo Oxigenasa (Desciclizante)/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Fístula Arteriovenosa/fisiopatología , Inmunohistoquímica , Venas Yugulares , Masculino , Ratones , Ratones Endogámicos C57BL
8.
Am J Physiol Renal Physiol ; 304(3): F317-25, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23195679

RESUMEN

Age increases the risk for ischemic acute kidney injury (AKI). We questioned whether a similar age-dependent injury occurs following exposure to hemoglobin, a known nephrotoxin. Old mice (~16 mo old), but not young mice (~6 mo old), when administered hemoglobin, exhibited marked elevation in blood urea nitrogen (BUN) and serum creatinine, and acute tubular necrosis with prominent tubular cast formation. The aged kidney exhibited induction of heme oxygenase-1 (HO-1) and other genes/proteins that may protect against heme-mediated renal injury, including ferritin, ferroportin, haptoglobin, and hemopexin. Old mice did not evince induction of HO-2 mRNA by hemoglobin, whereas a modest induction of HO-2 mRNA was observed in young mice. To determine the functional significance of HO-2 in heme protein-induced AKI, we administered hemoglobin to relatively young HO-2(+/+) and HO-2(-/-) mice: HO-2(-/-) mice, compared with HO-2(+/+) mice, exhibited greater renal dysfunction and histologic injury when administered hemoglobin. In addition to failing to elicit a protective system such as HO-2 in response to hemoglobin, old mice exhibited an exaggerated maladaptive response typified by markedly greater induction of the nephrotoxic cytokine IL-6 (130-fold increase vs. 10-fold increase in mRNA in young mice). We conclude that aged mice, unlike relatively younger mice, are exquisitely sensitive to the nephrotoxicity of hemoglobin, an effect attended by a failure to induce HO-2 mRNA and a fulminant upregulation of IL-6. Age thus markedly augments the sensitivity of the kidney to heme proteins, and HO-2 confers resistance to such insults.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Envejecimiento/fisiología , Hemoproteínas/efectos adversos , Hemoglobinas/efectos adversos , Necrosis Tubular Aguda/inducido químicamente , Riñón/fisiopatología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/fisiopatología , Animales , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Femenino , Hemo Oxigenasa (Desciclizante)/deficiencia , Hemo Oxigenasa (Desciclizante)/genética , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hemoproteínas/metabolismo , Hemoglobinas/metabolismo , Interleucina-6/metabolismo , Riñón/metabolismo , Necrosis Tubular Aguda/metabolismo , Necrosis Tubular Aguda/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , ARN Mensajero/metabolismo
9.
FASEB J ; 26(7): 2911-8, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22490782

RESUMEN

Underlying mechanisms of individual variation in severity of influenza infection and response to vaccination are poorly understood. We investigated the effect of reduced heme oxygenase-1 (HO-1) expression on vaccine response and outcome of influenza infection. HO-1-deficient and wild-type (WT) mice (kingdom, Animalia; phylum, Chordata; genus/species, Mus musculus) were infected with influenza virus A/PR/8/34 with or without prior vaccination with an adenoviral-based influenza vaccine. A genome-wide association study evaluated the expression of single-nucleotide polymorphisms (SNPs) in the HO-1 gene and the response to influenza vaccination in healthy humans. HO-1-deficient mice had decreased survival after influenza infection compared to WT mice (median survival 5.5 vs. 6.5 d, P=0.016). HO-1-deficient mice had impaired production of antibody following influenza vaccination compared to WT mice (mean antibody titer 869 vs. 1698, P=0.02). One SNP in HO-1 and one SNP in the constitutively expressed isoform HO-2 were independently associated with decreased antibody production after influenza vaccination in healthy human volunteers (P=0.017 and 0.014, respectively). HO-1 deficient mice were paired with sex- and age-matched WT controls. HO-1 affects the immune response to both influenza infection and vaccination, suggesting that therapeutic induction of HO-1 expression may represent a novel adjuvant to enhance influenza vaccine effectiveness.


Asunto(s)
Envejecimiento/inmunología , Envejecimiento/metabolismo , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Secuencia de Bases , Cartilla de ADN/genética , Femenino , Estudio de Asociación del Genoma Completo , Hemo-Oxigenasa 1/deficiencia , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Humanos , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/enzimología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Vacunación
10.
Am J Physiol Renal Physiol ; 303(12): F1601-7, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22993073

RESUMEN

Vascular access dysfunction causes morbidity in hemodialysis patients. This study examined the generation and pathobiological significance of superoxide anion in a rat femoral arteriovenous fistula (AVF). One week after AVF creation, there was increased production of superoxide anion accompanied by decreased total superoxide dismutase (SOD) and Cu/Zn SOD activities and induction of the redox-sensitive gene heme oxygenase-1. Immunohistochemical studies of nitrotyrosine formation demonstrated that peroxynitrite, a product of superoxide anion and nitric oxide, was present in increased amounts in endothelial and smooth muscle cells in the AVF. Because uncoupled NOS isoforms generate superoxide anion, and NOS coupling requires tetrahydrobiopterin (BH(4)) as a cofactor, we assessed NOS uncoupling by determining the ratio of BH(4) to dihydrobiopterin (BH(2)); the BH(4)-to-BH(2) ratio was markedly attenuated in the AVF. Because Src is a vasculopathic signaling species upstream and downstream of superoxide anion, such expression was evaluated; expression of Src and phosphorylated Src was both markedly increased in the AVF. Expression of NADPH oxidase (NOX) 1, NOX2, NOX4, cyclooxygenase (COX) 1, COX2, p47(phox), and p67(phox) was all unchanged, as assessed by Western analyses, thereby suggesting that these proteins may not be involved in increased production of superoxide anion. Finally, administration of tempol, a superoxide anion scavenger, decreased neointima formation in the juxta-anastomotic venous segment and improved AVF blood flow. We conclude that the AVF exhibits increased superoxide anion generation that may reflect the combined effects of decreased scavenging by SOD and increased generation by uncoupled NOS, and that enhanced superoxide anion production promotes juxta-anastomotic stenosis and impairs AVF function.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Vena Femoral/metabolismo , Fémur/irrigación sanguínea , Estrés Oxidativo/fisiología , Superóxidos/metabolismo , Animales , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Vena Femoral/patología , Vena Femoral/cirugía , Fémur/cirugía , Modelos Animales , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Familia-src Quinasas/metabolismo
11.
J Am Soc Nephrol ; 22(1): 43-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21115617

RESUMEN

Vascular access dysfunction compromises the care of patients on chronic hemodialysis. Elucidating the mechanisms of such dysfunction and devising strategies that may interrupt neointimal hyperplasia and relevant pathogenetic pathways are essential. Here, we show that, in the venous segment of a murine model of an arteriovenous fistula, monocyte chemoattractant protein-1 (MCP-1) mRNA and protein increase, accompanied by increased activity of the transcription factors NF-κB and AP-1. Genetic deficiency of MCP-1 proved markedly protective in this murine model, reflected by increased fistula patency 6 weeks after its formation, decreased venous wall thickness, and increased luminal area. An early effect of MCP-1 deficiency was the attenuation of the marked induction of CCL5 (RANTES) that occurred in this model, a chemokine recently recognized as a critical participant in vascular injury. Finally, in a rat model of an arteriovenous fistula, we localized expression of MCP-1 to the endothelium, proliferating smooth muscle cells and infiltrating leukocytes. In summary, marked upregulation of MCP-1 occurs in the venous segment of an arteriovenous fistula in rodents, and this vasculopathic chemokine contributes to failure of the fistula.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Quimiocina CCL2/metabolismo , Endotelio Vascular/metabolismo , Regulación hacia Arriba/fisiología , Animales , Proliferación Celular , Quimiocina CCL2/genética , Endotelio Vascular/citología , Leucocitos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Diálisis Renal/métodos , Factor de Transcripción AP-1/metabolismo
12.
Kidney360 ; 3(11): 1969-1979, 2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36514409

RESUMEN

Heme proteins, the stuff of life, represent an ingenious biologic strategy that capitalizes on the biochemical versatility of heme, and yet is one that avoids the inherent risks to cellular vitality posed by unfettered and promiscuously reactive heme. Heme proteins, however, may be a double-edged sword because they can damage the kidney in certain settings. Although such injury is often viewed mainly within the context of rhabdomyolysis and the nephrotoxicity of myoglobin, an increasing literature now attests to the fact that involvement of heme proteins in renal injury ranges well beyond the confines of this single disease (and its analog, hemolysis); indeed, through the release of the defining heme motif, destabilization of intracellular heme proteins may be a common pathway for acute kidney injury, in general, and irrespective of the underlying insult. This brief review outlines current understanding regarding processes underlying such heme protein-induced acute kidney injury (AKI) and chronic kidney disease (CKD). Topics covered include, among others, the basis for renal injury after the exposure of the kidney to and its incorporation of myoglobin and hemoglobin; auto-oxidation of myoglobin and hemoglobin; destabilization of heme proteins and the release of heme; heme/iron/oxidant pathways of renal injury; generation of reactive oxygen species and reactive nitrogen species by NOX, iNOS, and myeloperoxidase; and the role of circulating cell-free hemoglobin in AKI and CKD. Also covered are the characteristics of the kidney that render this organ uniquely vulnerable to injury after myolysis and hemolysis, and pathobiologic effects emanating from free, labile heme. Mechanisms that defend against the toxicity of heme proteins are discussed, and the review concludes by outlining the therapeutic strategies that have arisen from current understanding of mechanisms of renal injury caused by heme proteins and how such mechanisms may be interrupted.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Rabdomiólisis , Humanos , Mioglobina/efectos adversos , Hemólisis , Rabdomiólisis/inducido químicamente , Riñón/metabolismo , Lesión Renal Aguda/etiología , Hemo/efectos adversos , Hemoglobinas/efectos adversos , Insuficiencia Renal Crónica/complicaciones
13.
Kidney360 ; 3(8): 1417-1422, 2022 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-36176648

RESUMEN

Discovering new nephroprotectants may provide therapeutic strategies in AKI.This study provides the first evidence that KLF11, a member of the Krüppel-like factor (KLF) family of proteins, protects against AKI.In the absence of KLF11, exaggerated induction of endothelin-1 and IL-6 occurs after ischemic renal injury and may contribute to worse AKI.


Asunto(s)
Lesión Renal Aguda , Proteínas Reguladoras de la Apoptosis , Daño por Reperfusión , Proteínas Represoras , Lesión Renal Aguda/prevención & control , Proteínas Reguladoras de la Apoptosis/metabolismo , Endotelina-1/metabolismo , Humanos , Interleucina-6/metabolismo , Riñón/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Sustancias Protectoras/metabolismo , Daño por Reperfusión/prevención & control , Proteínas Represoras/metabolismo
14.
Kidney360 ; 3(10): 1672-1682, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36514726

RESUMEN

Background: Mitochondrial injury occurs in and underlies acute kidney injury (AKI) caused by ischemia-reperfusion and other forms of renal injury. However, to date, a comprehensive analysis of this issue has not been undertaken in heme protein-induced AKI (HP-AKI). We examined key aspects of mitochondrial function, expression of proteins relevant to mitochondrial quality control, and mitochondrial ultrastructure in HP-AKI, along with responses to heme in renal proximal tubule epithelial cells. Methods: The long-established murine glycerol model of HP-AKI was examined at 8 and 24 hours after HP-AKI. Indices of mitochondrial function (ATP and NAD+), expression of proteins relevant to mitochondrial dynamics, mitochondrial ultrastructure, and relevant gene/protein expression in heme-exposed renal proximal tubule epithelial cells in vitro were examined. Results: ATP and NAD+ content and the NAD+/NADH ratio were all reduced in HP-AKI. Expression of relevant proteins indicate that mitochondrial biogenesis (PGC-1α, NRF1, and TFAM) and fusion (MFN2) were impaired, as was expression of key proteins involved in the integrity of outer and inner mitochondrial membranes (VDAC, Tom20, and Tim23). Conversely, marked upregulation of proteins involved in mitochondrial fission (DRP1) occurred. Ultrastructural studies, including novel 3D imaging, indicate profound changes in mitochondrial structure, including mitochondrial fragmentation, mitochondrial swelling, and misshapen mitochondrial cristae; mitophagy was also observed. Exposure of renal proximal tubule epithelial cells to heme in vitro recapitulated suppression of PGC-1α (mitochondrial biogenesis) and upregulation of p-DRP1 (mitochondrial fission). Conclusions: Modern concepts pertaining to AKI apply to HP-AKI. This study validates the investigation of novel, clinically relevant therapies such as NAD+-boosting agents and mitoprotective agents in HP-AKI.


Asunto(s)
Lesión Renal Aguda , Hemoproteínas , Ratones , Animales , Hemoproteínas/metabolismo , NAD/metabolismo , Lesión Renal Aguda/etiología , Mitocondrias/metabolismo , Hemo/metabolismo , Adenosina Trifosfato/metabolismo
15.
Biochim Biophys Acta Mol Basis Dis ; 1868(3): 166322, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34920080

RESUMEN

BACKGROUND: Acute kidney injury (AKI) is both a consequence and determinant of outcomes in COVID-19. The kidney is one of the major organs infected by the causative virus, SARS-CoV-2. Viral entry into cells requires the viral spike protein, and both the virus and its spike protein appear in the urine of COVID-19 patients with AKI. We examined the effects of transfecting the viral spike protein of SARS-CoV-2 in kidney cell lines. METHODS: HEK293, HEK293-ACE2+ (stably overexpressing ACE2), and Vero E6 cells having endogenous ACE2 were transfected with SARS-CoV-2 spike or control plasmid. Assessment of gene and protein expression, and syncytia formation was performed, and the effects of quercetin on syncytia formation examined. FINDINGS: Spike transfection in HEK293-ACE2+ cells caused syncytia formation, cellular sloughing, and focal denudation of the cell monolayer; transfection in Vero E6 cells also caused syncytia formation. Spike expression upregulated potentially nephrotoxic genes (TNF-α, MCP-1, and ICAM1). Spike upregulated the cytoprotective gene HO-1 and relevant signaling pathways (p-Akt, p-STAT3, and p-p38). Quercetin, an HO-1 inducer, reduced syncytia formation and spike protein expression. INTERPRETATION: The major conclusions of the study are: 1) Spike protein expression in kidney cells provides a relevant model for the study of maladaptive and adaptive responses germane to AKI in COVID-19; 2) such spike protein expression upregulates HO-1; and 3) quercetin, an HO-1 inducer, may provide a clinically relevant/feasible protective strategy in AKI occurring in the setting of COVID-19. FUNDING: R01-DK119167 (KAN), R01-AI100911 (JPG), P30-DK079337; R01-DK059600 (AA).


Asunto(s)
COVID-19/metabolismo , Hemo-Oxigenasa 1/metabolismo , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , COVID-19/virología , Línea Celular , Chlorocebus aethiops , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/fisiología , Humanos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Quercetina/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Células Vero , Internalización del Virus/efectos de los fármacos
16.
Am J Physiol Renal Physiol ; 301(2): F436-42, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21525133

RESUMEN

TGF-ß1 contributes to chronic kidney disease, at least in part, via Smad3. TGF-ß1 is induced in the kidney following acute ischemia, and there is increasing evidence that TGF-ß1 may protect against acute kidney injury. As there is a paucity of information regarding the functional significance of Smad3 in acute kidney injury, the present study explored this issue in a murine model of ischemic acute kidney injury in Smad3(+/+) and Smad3(-/-) mice. We demonstrate that, at 24 h after ischemia, Smad3 is significantly induced in Smad3(+/+) mice, whereas Smad3(-/-) mice fail to express this protein in the kidney in either the sham or postischemic groups. Compared with Smad3(+/+) mice, and 24 h following ischemia, Smad3(-/-) mice exhibited greater preservation of renal function as measured by blood urea nitrogen (BUN) and serum creatinine; less histological injury assessed by both semiquantitative and qualitative analyses; markedly suppressed renal expression of IL-6 and endothelin-1 mRNA (but comparable expression of MCP-1, TNF-α, and heme oxygenase-1 mRNA); and no increase in plasma IL-6 levels, the latter increasing approximately sixfold in postischemic Smad3(+/+) mice. We conclude that genetic deficiency of Smad3 confers structural and functional protection against acute ischemic injury to the kidney. We speculate that these effects may be mediated through suppression of IL-6 production. Finally, we suggest that upregulation of Smad3 after an ischemic insult may contribute to the increased risk for chronic kidney disease that occurs after acute renal ischemia.


Asunto(s)
Lesión Renal Aguda/metabolismo , Daño por Reperfusión/metabolismo , Proteína smad3/metabolismo , Lesión Renal Aguda/patología , Animales , Femenino , Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Interleucina-6/sangre , Isquemia/metabolismo , Riñón/irrigación sanguínea , Riñón/metabolismo , Riñón/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Insuficiencia Renal Crónica/metabolismo , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/metabolismo
17.
Am J Physiol Renal Physiol ; 301(4): F845-51, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21697243

RESUMEN

The study of hemodynamic alterations following the creation of an arteriovenous fistula (AVF) is relevant to vascular adaptive responses and hemodialysis access dysfunction. This study examined such alterations in a murine AVF created by anastomosing the carotid artery to the jugular vein. AVF blood flow was markedly increased due to reduced AVF vascular resistance. Despite such markedly increased basal blood flow, AVF blood flow further increased in response to acetylcholine. This AVF model exhibited increased cardiac output and decreased systemic vascular resistance; the kidney, in contrast, exhibited decreased blood flow and increased vascular resistance. Augmentation in AVF blood flow was attended by increased arterial heme oxygenase-1 (HO-1) mRNA and protein expression, the latter localized to smooth muscle cells of the AVF artery; AVF blood flow was substantially reduced in HO-1(-/-) mice compared with HO-1(+/+) mice. Finally, in a murine model of a representative disease known to exhibit impaired hemodynamic responses (sickle cell disease), the creation of an AVF was attended by decreased AVF flow and impaired AVF function. We conclude that this AVF model exhibits markedly increased AVF blood flow, a vasodilatory reserve capacity, increased cardiac output, decreased renal blood flow, and a dependency on intact hemodynamic responses, in general, and HO-1 expression, in particular, in achieving and maintaining AVF blood flow. We suggest that these findings support the utility of this model in investigating the basis for and the consequences of hemodynamic stress, including shear stress, and the pathobiology of hemodialysis AVF dysfunction.


Asunto(s)
Fístula Arteriovenosa/fisiopatología , Hemodinámica/fisiología , Acetilcolina/farmacología , Anemia de Células Falciformes/fisiopatología , Animales , Fístula Arteriovenosa/enzimología , Gasto Cardíaco/efectos de los fármacos , Gasto Cardíaco/fisiología , Arterias Carótidas , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/biosíntesis , Hemodinámica/efectos de los fármacos , Venas Yugulares , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/fisiopatología , Flujo Sanguíneo Regional , Resistencia Vascular/efectos de los fármacos , Resistencia Vascular/fisiología
18.
Am J Pathol ; 176(5): 2530-41, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20363917

RESUMEN

Vascular access dysfunction contributes to the mortality of patients undergoing chronic hemodialysis. The present study analyzed the changes that evolve in a femoral arteriovenous fistula in the rat. The venous segment of this model exhibited, at 1 week, activation of pro-inflammatory transcription factors and up-regulation of pro-inflammatory, proliferative, procoagulant, and profibrotic genes; and at 4 weeks, the venous segment displayed neointimal hyperplasia, smooth muscle proliferation, and thrombus formation. These changes were accompanied by endothelial (e) nitric oxide synthase (NOS) and inducible (i) NOS up-regulation. The administration of NG-nitro-L-arginine methyl ester, an inhibitor of NOS activity, increased venous neointimal hyperplasia and pro-inflammatory gene expression (monocyte chemoattractant protein-1 and cytokine-induced neutrophil chemoattractant-1), increased systolic blood pressure, and decreased blood flow through the fistula. In another hypertensive model, the rat subtotal nephrectomy model, venous neointimal hyperplasia in the arteriovenous fistula was also exacerbated. We conclude that this arteriovenous fistula model recapitulates the salient features observed in dysfunctional, hemodialysis arteriovenous fistulas, and that venous neointimal hyperplasia is exacerbated when this model is superimposed in two different models of systemic hypertension. Since the uremic milieu contains increased amounts of asymmetric dimethylarginine, we speculate that such accumulation of this endogenous inhibitor of NOS, by virtue of its pressor or nitric oxide-depleting effects, or a combination thereof, may contribute to the limited longevity of arteriovenous fistulas used for hemodialysis.


Asunto(s)
Fístula Arteriovenosa/tratamiento farmacológico , Fístula Arteriovenosa/metabolismo , Regulación de la Expresión Génica , NG-Nitroarginina Metil Éster/farmacología , Animales , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Inflamación , Modelos Biológicos , Miocitos del Músculo Liso/citología , Óxido Nítrico/metabolismo , Ratas , Ratas Sprague-Dawley , Diálisis Renal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trombosis/patología
19.
Kidney360 ; 2(7): 1095-1106, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35368365

RESUMEN

Background: The actions of angiotensin-converting enzyme 2 (ACE2) oppose those of the renin-angiotensin-aldosterone system. ACE2 may be a cytoprotectant in some tissues. This study examined ACE2 expression in models of AKI. Methods: ACE2 mRNA and protein expression and ACE2 activity were assessed in murine ischemic AKI. Renal ACE2 mRNA expression was evaluated in LPS-induced AKI in wild-type (C57BL/6J) mice, in heme oxygenase-1+/+ and heme oxygenase-1-/- mice, and after unilateral ureteral obstruction (UUO) in wild-type mice. The effect of sex and age on renal ACE2 protein expression was also assessed. Results: In ischemic AKI, ACE2 mRNA and protein expression and ACE2 activity were reduced as compared with such indices in the intact kidney. In ischemic AKI, ACE2, which, in health, is prominently expressed in the tubular epithelium, especially proximal tubules, is decreased in expression in these segments. Decreased ACE2 expression in AKI did not reflect reduced GFR, because ACE2 mRNA expression was unaltered after UUO. LPS induced renal ACE2 mRNA expression in wild-type mice, but this effect did not occur in heme oxygenase-1-deficient mice. In ischemic and LPS-induced AKI, renal expression of the Mas receptor was increased. In the intact kidney, renal ACE2 protein expression decreased in female mice as compared with male mice, but was unaltered with age. Conclusion: We conclude that renal ACE2 expression is decreased in ischemic AKI, characterized by decreased GFR and abundant cell death, but is upregulated in LPS-induced AKI, an effect requiring heme oxygenase-1. Determining the significance of ACE2 expression in experimental AKI merits further study. We suggest that understanding the mechanism underlying ACE2 downregulation in AKI may offer insights relevant to COVID-19: ACE2 expression is downregulated after ACE2 mediates SARS-CoV-2 cellular entry; such downregulation is proinflammatory; and AKI commonly occurs and determines outcomes in COVID-19.


Asunto(s)
Lesión Renal Aguda , Enzima Convertidora de Angiotensina 2 , Lesión Renal Aguda/genética , Enzima Convertidora de Angiotensina 2/genética , Animales , Femenino , Riñón , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Am J Physiol Renal Physiol ; 299(6): F1270-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20881035

RESUMEN

Neointimal hyperplasia contributes to failure of hemodialysis arteriovenous fistulas (AVFs). Increased expression of matrix metalloproteinase (MMP)-9 occurs in AVFs, and MMP-9 is implicated in neointimal hyperplasia and vascular injury. Recent studies demonstrate that MMP-9, by degrading N-cadherin, leads to increased expression of ß-catenin and ß-catenin-dependent proliferation of smooth muscle cells. The present study examined this pathway in the venous limb of a murine AVF model. Western analyses demonstrate that, in this model, there is diminished expression of N-cadherin accompanied by increased expression of ß-catenin, c-Myc, and proliferating cell nuclear antigen (PCNA). By immunohistochemistry, ß-catenin and c-Myc localized to proliferating smooth muscle cells in the venous limb of the AVF. Increased expression of ß-catenin was accompanied by augmented expression of phosphorylated (p)-glycogen synthase kinase (GSK)-3ß, GSK-3ß, and integrin-linked kinase. The administration of doxycycline suppressed MMP-9 expression but did not reduce venous histological injury in the AVF, or increase AVF patency assessed 6 wk after its creation. Doxycycline did not influence expression of ß-catenin, c-Myc, GSK-3ß, or integrin-linked kinase. Thus, in this vascular injury model, the upregulation of ß-catenin cannot be readily attributed to MMP-9 upregulation; increased ß-catenin expression may reflect either the upregulation of p-GSK-3ß, GSK-3ß, or integrin-linked kinase. This study provides the first exploration of ß-catenin in an AVF, demonstrating substantial upregulation of this mitogenic signaling molecule and uncovering possible mechanisms that may account for such upregulation.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Doxiciclina/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Neointima/patología , beta Catenina/biosíntesis , Animales , Cadherinas/biosíntesis , Arterias Carótidas/cirugía , Glucógeno Sintasa Quinasa 3/biosíntesis , Glucógeno Sintasa Quinasa 3 beta , Hiperplasia/prevención & control , Venas Yugulares/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA