Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Am Soc Nephrol ; 27(1): 172-88, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25999406

RESUMO

Necrotizing and crescentic rapidly progressive GN (RPGN) is a life-threatening syndrome characterized by a rapid loss of renal function. Evidence suggests that podocyte expression of the transcription factor peroxisome proliferator-activated receptor γ (PPARγ) may prevent podocyte injury, but the function of glomerular PPARγ in acute, severe inflammatory GN is unknown. Here, we observed marked loss of PPARγ abundance and transcriptional activity in glomerular podocytes in experimental RPGN. Blunted expression of PPARγ in podocyte nuclei was also found in kidneys from patients diagnosed with crescentic GN. Podocyte-specific Pparγ gene targeting accentuated glomerular damage, with increased urinary loss of albumin and severe kidney failure. Furthermore, a PPARγ gain-of-function approach achieved by systemic administration of thiazolidinedione (TZD) failed to prevent severe RPGN in mice with podocyte-specific Pparγ gene deficiency. In nuclear factor erythroid 2-related factor 2 (NRF2)-deficient mice, loss of podocyte PPARγ was observed at baseline. NRF2 deficiency markedly aggravated the course of RPGN, an effect that was partially prevented by TZD administration. Furthermore, delayed administration of TZD, initiated after the onset of RPGN, still alleviated the severity of experimental RPGN. These findings establish a requirement for the NRF2-PPARγ cascade in podocytes, and we suggest that these transcription factors have a role in augmenting the tolerance of glomeruli to severe immune-complex mediated injury. The NRF2-PPARγ pathway may be a therapeutic target for RPGN.


Assuntos
Glomerulonefrite/etiologia , Fator 2 Relacionado a NF-E2/fisiologia , PPAR gama/biossíntese , Podócitos/metabolismo , Animais , Masculino , Camundongos
2.
Am J Respir Crit Care Med ; 188(6): 684-92, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23924269

RESUMO

RATIONALE: Purpura fulminans in adults is a rare but devastating disease. Its pathophysiology is not well known. OBJECTIVES: To understand the pathophysiology of skin lesions in purpura fulminans, the interplay between circulating blood and vascular alterations was assessed. METHODS: Prospective multicenter study in four intensive care units. Patients with severe sepsis without skin lesions were recruited as control subjects. MEASUREMENTS AND MAIN RESULTS: Twenty patients with severe sepsis and purpura fulminans were recruited for blood sampling, and skin biopsy was performed in deceased patients. High severity of disease and mortality rates (80%) was observed. Skin biopsies in purpura fulminans lesions revealed thrombosis and extensive vascular damage: vascular congestion and dilation, endothelial necrosis, alteration of markers of endothelial integrity (CD31) and of the protein C pathway receptors (endothelial protein C receptor, thrombomodulin). Elevated plasminogen activating inhibitor-1 mRNA was also observed. Comparison with control patients showed that these lesions were specific to purpura fulminans. By contrast, no difference was observed for blood hemostasis parameters, including soluble thrombomodulin, activated protein C, and disseminated intravascular coagulation markers. Bacterial presence at the vascular wall was observed specifically in areas of vascular damage in eight of nine patients tested (including patients with Streptococcus pneumoniae, Neisseria meningitidis, Escherichia coli, and Pseudomonas aeruginosa infection). CONCLUSIONS: Thrombi and extensive vascular damage with multifaceted prothrombotic local imbalance are characteristics of purpura fulminans. A "vascular wall infection" hypothesis, responsible for endothelial damage and subsequent skin lesions, can be put forward.


Assuntos
Endotélio Vascular/patologia , Púrpura Fulminante/patologia , Trombose/complicações , Malformações Vasculares/complicações , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Biópsia , Endotélio Vascular/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Estudos Prospectivos , Púrpura Fulminante/complicações , Púrpura Fulminante/metabolismo , Sepse/metabolismo , Pele/irrigação sanguínea , Trombomodulina/metabolismo , Trombose/patologia , Malformações Vasculares/metabolismo , Malformações Vasculares/patologia
3.
Kidney Int ; 79(6): 635-642, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21160460

RESUMO

IgA nephropathy (IgAN) often shows lesions morphologically identical with those of focal segmental glomerulosclerosis (FSGS). In order to determine the possible role of FSGS in IgAN lesions, we measured glomerular capsular adhesions, often the first step toward FSGS, in biopsies from 127 patients with IgAN, 100 with lupus nephritis, and 26 with primary FSGS. Capsular adhesions with no lesions in the underlying tuft, consistent with podocyte abnormality or loss, were found regularly in FSGS and IgAN, but infrequently in lupus. Fifteen biopsies of patients with IgAN were studied immunohistochemically using markers for podocytes, Bowman's parietal epithelial cells, proliferating cells, and macrophages. Cytokeratins CK-8 and C2562 differentiated normal podocytes (negative) from parietal epithelial cells (variably positive). There was focal loss of the podocyte markers synaptopodin, glomerular epithelial protein 1 (GLEPP-1), nephrin, and vascular endothelial growth factor (VEGF), particularly at sites of capsular adhesions in otherwise histologically normal glomeruli. Cells displaying the parietal epithelial cell markers PAX2 (paired box gene 2) and the cytokeratins were also positive for the proliferating cell marker, proliferating cell nuclear antigen. These cells gathered at sites of adhesion, and in response to active lesions in the tuft, grew inward along the adhesion onto the tuft, forming a monolayer positive for parietal markers and the podocyte marker Wilms tumor protein-1 (WT-1). These cells deposited a layer of collagen over the sclerosing tuft. Thus, all biopsies of patients with IgAN had changes basically identical to those classically described in FSGS. Hence, our study strongly suggests that podocytopathy of a type similar to that in primary FSGS occurs frequently in IgAN.


Assuntos
Glomerulonefrite por IGA/metabolismo , Glomerulosclerose Segmentar e Focal/metabolismo , Imuno-Histoquímica , Glomérulos Renais/química , Nefrite Lúpica/metabolismo , Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Biomarcadores/análise , Biópsia , Distribuição de Qui-Quadrado , Progressão da Doença , Células Epiteliais/química , Glomerulonefrite por IGA/imunologia , Glomerulonefrite por IGA/patologia , Glomerulosclerose Segmentar e Focal/imunologia , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Queratina-8/análise , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Fator de Transcrição PAX2/análise , Paris , Podócitos/química , Prognóstico , Antígeno Nuclear de Célula em Proliferação/análise , Proteínas WT1/análise
4.
FASEB J ; 24(12): 4691-700, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20667972

RESUMO

Angiotensin I-converting enzyme (ACE; kininase II) levels in humans are genetically determined. ACE levels have been linked to risk of myocardial infarction, but the association has been inconsistent, and the causality underlying it remains undocumented. We tested the hypothesis that genetic variation in ACE levels influences myocardial tolerance to ischemia. We studied ischemia-reperfusion injury in mice bearing 1 (ACE1c), 2 (ACE2c, wild type), or 3 (ACE3c) functional copies of the ACE gene and displaying an ACE level range similar to humans. Infarct size in ACE1c was 29% lower than in ACE2c (P<0.05). Pretreatment with a kinin B2 receptor antagonist suppressed this reduction. In ACE3c, infarct size was the same as in ACE2c. But ischemic preconditioning, which reduced infarct size in ACE2c (-63%, P<0.001) and ACE1c (-52%, P<0.05), was not efficient in ACE3c (-2%, NS, P<0.01 vs. ACE2c). In ACE3c, ischemic preconditioning did not decrease myocardial inflammation or cardiomyocyte apoptosis. Pretreatment with a renin inhibitor had no cardioprotective effect in ACE2c, but in ACE3c partially restored (38%) the cardioprotection of ischemic preconditioning. Thus, a modest genetic increase in ACE impairs myocardial tolerance to ischemia. ACE level plays a critical role in cardiac ischemia, through both kinin and angiotensin mediated mechanisms.


Assuntos
Coração/efeitos dos fármacos , Infarto do Miocárdio/enzimologia , Isquemia Miocárdica/enzimologia , Miocárdio/enzimologia , Peptidil Dipeptidase A/metabolismo , Traumatismo por Reperfusão/genética , Amidas/farmacologia , Angiotensina I/farmacologia , Angiotensina II/farmacologia , Animais , Apoptose/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Bradicinina/análogos & derivados , Bradicinina/farmacologia , Antagonistas dos Receptores da Bradicinina , Fumaratos/farmacologia , Cininas/farmacologia , Pulmão/enzimologia , Camundongos , Camundongos Mutantes , Infarto do Miocárdio/genética , Isquemia Miocárdica/genética , Peptidil Dipeptidase A/genética , Renina/antagonistas & inibidores
5.
Eur Heart J ; 31(12): 1529-37, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20028694

RESUMO

AIMS: To investigate the adaptations of left ventricular function and calcium handling to chronic heart rate reduction with ivabradine in the reperfused heart. METHODS AND RESULTS: Rabbits underwent 20 min coronary artery occlusion followed by 3 weeks of reperfusion. Throughout reperfusion, rabbits received ivabradine (10 mg/kg/day) or vehicle (control). Ivabradine reduced heart rate by about 20% and improved both ejection fraction (+35%) and systolic displacement (+26%) after 3 weeks of treatment. Interestingly, this was associated with a two-fold increase expression of FKBP12/12.6. There was no difference in the expressions of phospholamban, SERCA2a, calsequestrin, ryanodine, phospho-ryanodine, and Na(2+)/Ca(2+) exchanger in the two groups. Infarct scar and vascular density were similar in both groups. Administration of a single intravenous bolus of ivabradine (1 mg/kg) in control rabbits at 3 weeks of reperfusion also significantly improved acutely ejection fraction and systolic displacement. CONCLUSION: Chronic heart rate reduction protects the myocardium against ventricular dysfunction induced by myocardial ischaemia followed by 3 weeks of reperfusion. Beyond pure heart rate reduction, ivabradine improves global and regional systolic function of the reperfused heart through a dual mechanism involving a direct mechanical effect and a long-term adaptation in calcium handling, as supported by the increase in FKBP12/12.6 expression.


Assuntos
Antiarrítmicos/farmacologia , Benzazepinas/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Proteína 1A de Ligação a Tacrolimo/metabolismo , Animais , Western Blotting , Proteínas de Ligação ao Cálcio/metabolismo , Oclusão Coronária/fisiopatologia , Ivabradina , Infarto do Miocárdio/fisiopatologia , Reperfusão Miocárdica/métodos , Coelhos , Volume Sistólico/efeitos dos fármacos , Disfunção Ventricular Esquerda/tratamento farmacológico , Disfunção Ventricular Esquerda/fisiopatologia
6.
J Mol Cell Cardiol ; 46(5): 713-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19233193

RESUMO

The purpose of the present study was to investigate the potential cardioprotective effects of an original approach based on the properties of the X chromosome-linked Inhibitor of Apoptosis (XIAP), the most effective endogenous inhibitor of apoptosis. For this purpose, the C-terminal part of XIAP (BIR3 and RING domains) was fused to the protein transduction domain (PTD) of the HIV1 transactivator of transcription, which confers to fused protein the ability to cross cell membranes. This protein, so-called PTD-BIR3/RING, was administered intravenously in C57BL/6J mice subjected to 30 min coronary artery occlusion and 24 h of reperfusion. Administration of PTD-BIR3/RING at 5 min before and 30 min after the onset of reperfusion reduced infarct size vs control (23+/-2% vs 41+/-4% and 27+/-4% vs 41+/-3%, respectively, p<0.05). Similar reduction in infarct size was observed when PTD-BIR3/RING was administered prior to ischemia (28+/-1% vs 44+/-3%). In addition to inhibition of caspase-3 and -9 activities, PTD-BIR3/RING induced an inhibition of caspase-8 and several other actors of the apoptotic pathways. In conclusion, this study demonstrates that the administration of PTD-BIR3/RING reduces myocardial infarct size even when injected during reperfusion through interruption of caspase activation by pharmacologically mimicking endogenous XIAP.


Assuntos
Mimetismo Molecular , Infarto do Miocárdio/prevenção & controle , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Animais , Western Blotting , Peso Corporal , Caspases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Estrutura Terciária de Proteína , Ratos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/química
7.
Nephrol Dial Transplant ; 24(2): 630-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18927120

RESUMO

BACKGROUND: The specific mTor inhibitor sirolimus has been implicated in the pathogenesis of renal glomerular lesions and nephrotic syndrome appearance after transplantation. Podocyte injury and focal segmental glomerulosclerosis have been related to sirolimus therapy in some patients but the pathways underlying these lesions remain hypothetical. METHODS: To go further in the comprehension of these mechanisms, primary cultures of human podocytes were exposed to therapeutic-range concentrations of sirolimus. RESULTS: Cell viability was not affected after 2 days' exposure to the drug but changes in cell phenotype and cytoskeleton reorganization were observed. We also evidenced that vascular endothelial growth factor (VEGF) synthesis and Akt phosphorylation were decreased by sirolimus addition. We did not observe any loss of podocyte differentiation markers with the notable exception of WT1, a transcription factor essential for maintaining podocyte integrity. WT1 gene and protein expression in podocytes were decreased in a dose-dependent manner after incubation with sirolimus. CONCLUSION: Taken together, these data suggest that sirolimus could impair pathways essential for podocyte integrity and therefore predisposes to glomerular injury.


Assuntos
Podócitos/efeitos dos fármacos , Podócitos/metabolismo , Sirolimo/efeitos adversos , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Primers do DNA/genética , Genes do Tumor de Wilms/efeitos dos fármacos , Humanos , Imunossupressores/efeitos adversos , Modelos Biológicos , Fenótipo , Podócitos/patologia , Reação em Cadeia da Polimerase , Proteínas Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR , Fator A de Crescimento do Endotélio Vascular/biossíntese , Proteínas WT1/genética , Proteínas WT1/metabolismo
8.
Nat Commun ; 10(1): 3303, 2019 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-31341160

RESUMO

The mechanisms driving the development of extracapillary lesions in focal segmental glomerulosclerosis (FSGS) and crescentic glomerulonephritis (CGN) remain poorly understood. A key question is how parietal epithelial cells (PECs) invade glomerular capillaries, thereby promoting injury and kidney failure. Here we show that expression of the tetraspanin CD9 increases markedly in PECs in mouse models of CGN and FSGS, and in kidneys from individuals diagnosed with these diseases. Cd9 gene targeting in PECs prevents glomerular damage in CGN and FSGS mouse models. Mechanistically, CD9 deficiency prevents the oriented migration of PECs into the glomerular tuft and their acquisition of CD44 and ß1 integrin expression. These findings highlight a critical role for de novo expression of CD9 as a common pathogenic switch driving the PEC phenotype in CGN and FSGS, while offering a potential therapeutic avenue to treat these conditions.


Assuntos
Nefropatias/patologia , Tetraspanina 29/fisiologia , Animais , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Feminino , Glomerulonefrite/genética , Glomerulonefrite/metabolismo , Glomerulonefrite/patologia , Glomerulosclerose Segmentar e Focal/genética , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Humanos , Nefropatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tetraspanina 29/genética , Tetraspanina 29/metabolismo
9.
Transplantation ; 85(2): 281-9, 2008 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-18212634

RESUMO

BACKGROUND: Lumen loss in graft arteriosclerosis is the consequence of the development of a thick neointima and constrictive arterial remodeling. The latter is due to adventitial chronic inflammation and excessive perivascular collagen deposition. We reasoned that blockade of the portal of entry of inflammatory effectors may constitute a strategy to prevent constrictive arterial remodeling. METHODS AND RESULTS: We found that an anti-angiogenic therapy (ABT-510 nonapeptide), devoid of direct immunomodulatory properties, dramatically reduced adventitial angiogenesis by 66% (P<0.0001) in the rat aortic interposition model of graft arteriosclerosis. The associated decreased entry of inflammatory cells (44%; P<0.00001) resulted in drastic reduction of collagen deposition (57%; P<0.0001) thereby preventing subsequent adventitial constrictive remodeling and reduction of lumen surface area (5.26+/-0.74 vs. 8.58+/-2.48 microm2; Control vs. ABT-510-treated rats; P<0.0001). ABT-510 had no effect on the development of the neointima. CONCLUSION: This work supports the idea that targeting angiogenesis may act synergistically with conventional immunosuppressive therapy in preventing graft arteriosclerosis, a crucial feature of chronic graft rejection.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Aorta/transplante , Arteriosclerose/prevenção & controle , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Aorta/ultraestrutura , Colágeno/análise , Citometria de Fluxo , Teste de Cultura Mista de Linfócitos , Masculino , Modelos Animais , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos Lew , Transplante de Pele/patologia , Transplante de Pele/fisiologia , Transplante Homólogo , Transplante Isogênico
10.
Transplantation ; 80(5): 660-5, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16177642

RESUMO

OBJECTIVE: Cell death remains a major limitation of skeletal myoblast (SM) transplantation but the patterns of cell survival and proliferation in heart and their potential modulation by thermic stresses like heat shock (HS) and cryopreservation (Cryo) are still incompletely characterized. METHODS: To track SMs in situ, we developed a dual-marker system based on the semiconservative expression of the foreign soluble protein, beta-Galactosidase (beta-Gal) and the constitutive expression of the Y chromosome in a myocardial infarction model. Control medium or Lewis male rat SMs (fresh or subjected to Cryo or HS) were injected in Lewis female rats. RESULTS: There was a massive cell loss early after transplantation in the fresh group, which was only partially compensated for by a subsequent proliferation. Conversely, both Cryo and HS significantly improved early cell survival but blunted subsequent proliferation so that, at 15 days posttransplantation, the total number of engrafted donor-derived Y-positive cells did not differ significantly between the three groups. Most of them expressed a skeletal muscle phenotype. CONCLUSIONS: These data confirm the high death rate of in-scar transplanted myoblasts, demonstrate the ability of those that survive to proliferate and differentiate along the myogenic pathway but do not support the efficacy of either Cryo or HS for increasing the ultimate magnitude of myoblast engraftment.


Assuntos
Criopreservação , Sobrevivência de Enxerto , Resposta ao Choque Térmico , Mioblastos Esqueléticos/transplante , Infarto do Miocárdio/terapia , Adenoviridae/genética , Animais , Diferenciação Celular/genética , Divisão Celular/genética , Temperatura Baixa , Feminino , Marcadores Genéticos , Proteínas de Choque Térmico HSP70/metabolismo , Temperatura Alta , Masculino , Músculo Esquelético/citologia , Mioblastos Esqueléticos/citologia , Fenótipo , Ratos , Ratos Endogâmicos Lew , Cromossomo Y , beta-Galactosidase/genética
11.
J Thorac Cardiovasc Surg ; 130(1): 173-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15999059

RESUMO

OBJECTIVE: Early cell death remains a major limitation of skeletal myoblast transplantation. Because the poor vascularization of the target scars contributes to cell loss, we assessed the effects of combining skeletal myoblast transplantation with administration of hypoxia-inducible factor 1alpha, a master gene that controls the expression of a wide array of angiogenic factors. METHODS: A myocardial infarction was created in 56 rats by means of coronary artery ligation. Eight days later, rats were randomly allocated to receive in-scar injections of culture medium (control animals, n = 11), skeletal myoblasts (5 x 10(6) , n = 13), adenovirus-encoded hypoxia-inducible factor 1alpha (1.0 x 10(10) pfu/mL, n = 7), or skeletal myoblasts (5 x 10(6)) in combination with an empty vector (n = 3) or active hypoxia-inducible factor 1alpha (1.0 x 10(10) pfu/mL, n = 13). A fifth group (n = 9) underwent a staged approach in which hypoxia-inducible factor 1alpha (1.0 x 10(10) pfu/mL) was injected at the time of infarction, followed 8 days later by skeletal myoblasts (5 x 10(6)). Left ventricular function was assessed echocardiographically before transplantation and 1 month thereafter. Explanted hearts were then processed for the immunohistochemical detection of myotubes, quantification of angiogenesis, myoblast engraftment, and cell survival. RESULTS: Baseline ejection fractions were not significantly different among groups (35%-40%). One month later, ejection fraction had decreased from baseline in control hearts and in those injected with hypoxia-inducible factor 1alpha. In contrast, it did not deteriorate after injections of skeletal myoblasts alone or combined with either the empty vector or active hypoxia-inducible factor 1alpha administered sequentially. The most striking change occurred in the skeletal myoblast plus hypoxia-inducible factor 1alpha combined group in which ejection fraction increased dramatically (by 27%) above baseline levels and was thus markedly higher than in all other groups ( P = .0001 and P = .001 vs control animals and animals receiving hypoxia-inducible factor 1alpha, respectively). Compared with skeletal myoblasts alone, the coadministration of hypoxia-inducible factor 1alpha resulted in a significantly greater degree of angiogenesis, cell engraftment, and cell survival. CONCLUSION: Induction of angiogenesis is an effective means of potentiating the functional benefits of myoblast transplantation, and hypoxia-inducible factor 1alpha can successfully achieve this goal.


Assuntos
Mioblastos Esqueléticos/transplante , Neovascularização Fisiológica/efeitos dos fármacos , Fatores de Transcrição/farmacologia , Adenoviridae , Animais , Morte Celular/fisiologia , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia , Masculino , Mioblastos Esqueléticos/fisiologia , Infarto do Miocárdio/cirurgia , Distribuição Aleatória , Ratos , Ratos Wistar , Fatores de Transcrição/sangue , Fatores de Transcrição/uso terapêutico , Função Ventricular Esquerda
12.
J Heart Lung Transplant ; 34(9): 1198-207, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25534019

RESUMO

BACKGROUND: Cardiac-committed cells and biomimetic scaffolds independently improve the therapeutic efficacy of stem cells. In this study we tested the long-term effects of their combination. METHODS: Eighty immune-deficient rats underwent permanent coronary artery ligation. Five to 7 weeks later, those with an echocardiographically measured ejection fraction (EF) ≤55% were re-operated on and randomly allocated to receive a cell-free fibrin patch (n = 25), a fibrin patch loaded with 700,000 human embryonic stem cells (ESC) pre-treated to promote early cardiac differentiation (SSEA-1(+) progenitors [n = 30]), or to serve as sham-operated animals (n = 25). Left ventricular function was assessed by echocardiography at baseline and every month thereafter until 4 months. Hearts were then processed for assessment of fibrosis and angiogenesis and a 5-component heart failure score was constructed by integrating the absolute change in left ventricular end-systolic volume (LVESV) between 4 months and baseline, and the quantitative polymerase chain reaction (qPCR)-based expression of natriuretic peptides A and B, myosin heavy chain 7 and periostin. All data were recorded and analyzed in a blinded manner. RESULTS: The cell-treated group consistently yielded better functional outcomes than the sham-operated group (p = 0.002 for EF; p = 0.01 for LVESV). Angiogenesis in the border zone was also significantly greater in the cell-fibrin group (p = 0.006), which yielded the lowest heart failure score (p = 0.04 vs sham). Engrafted progenitors were only detected shortly after transplantation; no grafted cells were identified after 4 months. There was no teratoma identified. CONCLUSIONS: A fibrin scaffold loaded with ESC-derived cardiac progenitors resulted in sustained improvement in contractility and attenuation of remodeling without sustained donor cell engraftment. A paracrine effect, possibly on innate reparative responses, is a possible mechanism for this enduring effect.


Assuntos
Células-Tronco Embrionárias , Fibrina , Miocárdio/citologia , Células-Tronco , Alicerces Teciduais , Indutores da Angiogênese , Animais , Moléculas de Adesão Celular/análise , Vasos Coronários , Ecocardiografia , Feminino , Insuficiência Cardíaca , Humanos , Imuno-Histoquímica , Ligadura , Camundongos Nus , Cadeias Pesadas de Miosina , Peptídeos Natriuréticos/análise , Reação em Cadeia da Polimerase , Ratos , Volume Sistólico , Função Ventricular Esquerda
13.
Eur J Heart Fail ; 5(4): 435-42, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12921804

RESUMO

BACKGROUND: Pulmonary endothelium is an early upstream hemodynamic target of left ventricular dysfunction. Interleukin 6 (IL-6) is a pro-inflammatory cytokine reported to increase in congestive heart failure (CHF) patients. AIMS: We sought to determine the origin of IL-6, IL-6 receptor (IL-6R) and gp130 in experimental CHF. METHODS: We used rats with coronary artery ligation as an experimental model of either compensated or decompensated heart failure. Lung and aorta samples were analysed by RT-PCR, ELISA and immunohistochemistry for IL-6 and its receptors. RESULTS: IL-6 mRNA expression increased in the lung of rats with decompensated heart failure and was positively correlated with infarct severity whereas IL-6R mRNA decreased in the lung of myocardial infarction rats and gp130 mRNA remained unchanged. In contrast, there were no changes in IL-6 mRNA expression in the aorta and left ventricular myocardium. IL-6 peptide content as determined by ELISA and Western Blot in lung tissue was 2-fold higher in decompensated heart failure as compared to control rats. These data were confirmed by immunohistochemistry showing a preferential endothelial localization of IL-6 in the CHF lung. IL-6 peptide was also present in the pleural effusion of decompensated heart failure and was positively correlated with IL-6 mRNA expression in the lungs of decompensated HF rats. Pulmonary IL-6 overexpression was associated with nuclear translocation of NF-kappaB and cytosolic degradation of IkappaB. CONCLUSION: Dysfunctional pulmonary endothelium is a source of synthesis and storage of IL-6 in an experimental model of CHF.


Assuntos
Endotélio Vascular/metabolismo , Insuficiência Cardíaca/fisiopatologia , Interleucina-6/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Animais , Antígenos CD/metabolismo , Western Blotting , Receptor gp130 de Citocina , Regulação para Baixo/fisiologia , Insuficiência Cardíaca/metabolismo , Humanos , Masculino , Glicoproteínas de Membrana/metabolismo , NF-kappa B/fisiologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/fisiologia , Disfunção Ventricular Esquerda/metabolismo
14.
Fundam Clin Pharmacol ; 18(4): 437-47, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15312150

RESUMO

Some combinations of antihypertensive agents were shown to reduce proteinuria in patients with renal failure. However, preventive effects of such combinations on renal structure and function are presently unknown when treatment is administered before the onset of renal abnormalities. We thus investigated the long-term effects of an angiotensin-converting enzyme (ACE) inhibitor (perindopril)/diuretic (indapamide) combination (per/ind) in the Zucker rat, a classical model of chronic renal failure associated with obesity, hyperlipidemia, and insulin resistance. Two-month-old lean and obese Zucker rats, presenting normal renal structure and function at this young age, received per/ind (0.76 + 0.24 mg/kg of body weight/day) or the vehicle of this combination by daily gavage. After 8.5 consecutive months of treatment, those 10.5-month-old rats were used for determination of renal structural and functional parameters which were examined using standard renal clearance experiments and kidney tissue analysis. Per/ind prevented focal and segmental glomerular hyalinosis and tubulo-interstitial damage in obese rats. Treatment was also associated with a significant reduction in several staining markers of glomerular and interstitial fibrosis. The hypertrophy of superficial glomeruli and the mesangial expansion of deep glomeruli observed in control rats were reduced in per/ind-treated obese rats. The severe proteinuria observed in 10.5-month-old control obese rats was prevented by per/ind, while glomerular filtration and renal hemodynamic parameters reached similar values to those obtained in lean animals. These results show that long-term treatment with this ACE inhibitor/diuretic combination protects renal structure and function in the obese Zucker rat, emphasizing the potential efficiency of such therapy in renal failure prevention.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Diuréticos/uso terapêutico , Indapamida/uso terapêutico , Perindopril/uso terapêutico , Proteinúria/prevenção & controle , Insuficiência Renal/prevenção & controle , Inibidores da Enzima Conversora de Angiotensina/administração & dosagem , Inibidores da Enzima Conversora de Angiotensina/farmacocinética , Animais , Diuréticos/administração & dosagem , Diuréticos/farmacocinética , Combinação de Medicamentos , Fibrose/prevenção & controle , Indapamida/administração & dosagem , Indapamida/farmacocinética , Testes de Função Renal , Masculino , Taxa de Depuração Metabólica , Obesidade/complicações , Perindopril/administração & dosagem , Perindopril/farmacocinética , Proteinúria/etiologia , Ratos , Ratos Zucker , Insuficiência Renal/etiologia , Insuficiência Renal/patologia
15.
Cell Transplant ; 23(1): 87-96, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23127387

RESUMO

Both enzymatic dissociation of cells prior to needle-based injections and poor vascularization of myocardial infarct areas are two important contributors to cell death and impede the efficacy of cardiac cell therapy. Because these limitations could be overcome by scaffolds ensuring cell cohesiveness and codelivery of angiogenic cells, we used a chronic rat model of myocardial infarction to assess the long-term (6 months) effects of the epicardial delivery of a composite collagen-based patch harboring both cardiomyogenesis-targeted human embryonic SSEA-1(+) (stem cell-derived stage-specific embryonic antigen-1 positive) cardiovascular progenitors and autologous (rat) adipose tissue-derived angiogenesis-targeted stromal cells (n = 27). Cell-free patches served as controls (n = 28). Serial follow-up echocardiographic measurements of left ventricular ejection fraction (LVEF) showed that the composite patch group yielded a significantly better preservation of left ventricular function that was sustained over time as compared with controls, and this pattern persisted when the assessment was restricted to the subgroup of rats with initial LVEFs below 50%. The composite patch group was also associated with significantly less fibrosis and more vessels in the infarct area. However, although human progenitors expressing cardiac markers were present in the patches before implantation, none of them could be subsequently identified in the grafted tissue. These data confirm the efficacy of epicardial scaffolds as cell carriers for ensuring long-term functional benefits and suggest that these effects are likely related to paracrine effects and call for optimizing cross-talks between codelivered cell populations to achieve the ultimate goal of myocardial regeneration.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Infarto do Miocárdio/terapia , Pericárdio/citologia , Tecido Adiposo/citologia , Animais , Colágeno/administração & dosagem , Desfibriladores Implantáveis , Modelos Animais de Doenças , Feminino , Humanos , Miocárdio/citologia , Ratos , Ratos Wistar , Alicerces Teciduais
16.
Toxicol Sci ; 131(1): 259-70, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22977165

RESUMO

Nephrogenic systemic fibrosis (NSF) is a scleroderma-like disease associated with prior administration of certain gadolinium chelates (GCs). NSF occurs in patients with severe renal failure. The purpose of this study was to set up a rat model of GC-associated NSF to elucidate the mechanism of this devastating disease. Firstly, after characterization of the model, male Wistar rats received a 0.75% adenine-enriched diet for 8, 14, or 16 days to obtain various degrees of renal failure. Rats received five consecutive daily iv injections of saline or gadodiamide (2.5 mmol/kg/day). Secondly, the safety profile and in vivo propensity to dissociate of all categories of marketed GCs (gadoterate, gadobutrol, gadobenate, gadopentetate, and gadodiamide) were compared in rats receiving adenine-enriched diet for 16 days. Serial skin biopsies were performed for blinded histopathological study. Total Gd concentration in tissues was measured by Inductively Coupled Plasma Mass Spectrometry. Relaxometry was used to evaluate the presence of dissociated Gd in skin and bone. Gadodiamide-induced high mortality and skin lesions (dermal fibrosis, calcification, and inflammation) were related to adenine diet duration. No skin lesions were observed with other molecules. Unlike macrocyclic GCs, gadodiamide, gadopentetate, and gadobenate gradually increased the r(1) relaxivity value, consistent with in vivo dissociation and release of soluble Gd (or, in the case of gadobenate, the consequence of protein binding). Gadodiamide-induced cutaneous and systemic toxicity depended on baseline renal function. We demonstrate in vivo dissociation of linear GCs, gadodiamide, and gadopentetate, whereas macrocyclic agents remained stable over the study period.


Assuntos
Adenina/administração & dosagem , Meios de Contraste/toxicidade , Gadolínio/toxicidade , Dermopatia Fibrosante Nefrogênica/induzido quimicamente , Compostos Organometálicos/toxicidade , Insuficiência Renal/complicações , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Meios de Contraste/química , Meios de Contraste/farmacocinética , Dieta , Modelos Animais de Doenças , Gadolínio/química , Gadolínio/farmacocinética , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Testes de Função Renal , Masculino , Dermopatia Fibrosante Nefrogênica/etiologia , Dermopatia Fibrosante Nefrogênica/metabolismo , Dermopatia Fibrosante Nefrogênica/patologia , Compostos Organometálicos/química , Compostos Organometálicos/farmacocinética , Ratos , Ratos Wistar , Insuficiência Renal/induzido quimicamente , Insuficiência Renal/metabolismo , Pele/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Espectrofotometria Atômica , Distribuição Tecidual
17.
J Clin Invest ; 122(9): 3355-67, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22886306

RESUMO

Tight regulation of calcium levels is required for many critical biological functions. The Ca2+-sensing receptor (CaSR) expressed by parathyroid cells controls blood calcium concentration by regulating parathyroid hormone (PTH) secretion. However, CaSR is also expressed in other organs, such as the kidney, but the importance of extraparathyroid CaSR in calcium metabolism remains unknown. Here, we investigated the role of extraparathyroid CaSR using thyroparathyroidectomized, PTH-supplemented rats. Chronic inhibition of CaSR selectively increased renal tubular calcium absorption and blood calcium concentration independent of PTH secretion change and without altering intestinal calcium absorption. CaSR inhibition increased blood calcium concentration in animals pretreated with a bisphosphonate, indicating that the increase did not result from release of bone calcium. Kidney CaSR was expressed primarily in the thick ascending limb of the loop of Henle (TAL). As measured by in vitro microperfusion of cortical TAL, CaSR inhibitors increased calcium reabsorption and paracellular pathway permeability but did not change NaCl reabsorption. We conclude that CaSR is a direct determinant of blood calcium concentration, independent of PTH, and modulates renal tubular calcium transport in the TAL via the permeability of the paracellular pathway. These findings suggest that CaSR inhibitors may provide a new specific treatment for disorders related to impaired PTH secretion, such as primary hypoparathyroidism.


Assuntos
Cálcio/sangue , Hormônio Paratireóideo/metabolismo , Receptores de Detecção de Cálcio/fisiologia , Aminoácidos/urina , Animais , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/uso terapêutico , Cálcio/metabolismo , Cálcio/urina , Creatinina/urina , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Hipoparatireoidismo/sangue , Hipoparatireoidismo/tratamento farmacológico , Alça do Néfron/metabolismo , Masculino , Naftalenos/farmacologia , Naftalenos/uso terapêutico , Osteocalcina/sangue , Pamidronato , Paratireoidectomia , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de Detecção de Cálcio/antagonistas & inibidores , Receptores de Detecção de Cálcio/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto
18.
Nat Genet ; 44(4): 456-60, S1-3, 2012 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-22406640

RESUMO

Familial hyperkalemic hypertension (FHHt) is a Mendelian form of arterial hypertension that is partially explained by mutations in WNK1 and WNK4 that lead to increased activity of the Na(+)-Cl(-) cotransporter (NCC) in the distal nephron. Using combined linkage analysis and whole-exome sequencing in two families, we identified KLHL3 as a third gene responsible for FHHt. Direct sequencing of 43 other affected individuals revealed 11 additional missense mutations that were associated with heterogeneous phenotypes and diverse modes of inheritance. Polymorphisms at KLHL3 were not associated with blood pressure. The KLHL3 protein belongs to the BTB-BACK-kelch family of actin-binding proteins that recruit substrates for Cullin3-based ubiquitin ligase complexes. KLHL3 is coexpressed with NCC and downregulates NCC expression at the cell surface. Our study establishes a role for KLHL3 as a new member of the complex signaling pathway regulating ion homeostasis in the distal nephron and indirectly blood pressure.


Assuntos
Proteínas de Transporte/genética , Transporte de Íons/genética , Néfrons/metabolismo , Pseudo-Hipoaldosteronismo/genética , Simportadores de Cloreto de Sódio/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Sequência de Bases , Pressão Sanguínea/genética , Criança , Feminino , Humanos , Rim/metabolismo , Masculino , Proteínas dos Microfilamentos , Pessoa de Meia-Idade , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Pseudo-Hipoaldosteronismo/metabolismo , Pseudo-Hipoaldosteronismo/fisiopatologia , Análise de Sequência de DNA , Transdução de Sinais , Simportadores de Cloreto de Sódio/genética , Adulto Jovem
19.
Virchows Arch ; 456(3): 325-37, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19806361

RESUMO

The present work identifies a new mouse model of inductible acute glomerular injury leading to focal segmental glomerulonephritis. We take advantage of the suicide gene/prodrug nitroreductase/CB1954 combination, in which nitroreductase converts CB1954, a monofunctional alkylating agent, into its toxic form. We generate two lines of transgenic mice in which the nitroreductase gene was placed under the control of the podocyte-specific gene podocin. The functional analysis of transgenic mice lines showed that CB1954 treatment induced a severe but transitory proteinuria. Sequential histopathological analysis was performed on serial kidney biopsies. Injured glomeruli showed acute lesions with early podocyte vacuolization and detachment, podocyte apoptosis, and cellular proliferation leading to a marked hypercellularity of the urinary space that was associated with collapsing of the glomerular tuft. After 1 month, progressive scarring lead to focal segmental glomerulosclerosis with fibrous capsular adhesion, hyalinosis, and podocytosis associated with interstitial fibrosis. The phenotype of podocytes was changed exhibiting dedifferentiation characterized by the loss of podocyte specific proteins/transcription factor and the expression of injury markers. Bowman's capsule cells were also involved in the cellular changes in a manner suggesting epithelial to mesenchymal transition. This model of podocyte injury in transgenic mice provides new insights into the cellular mechanisms of podocytopathies and their progression to scarring.


Assuntos
Glomerulosclerose Segmentar e Focal/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Glomérulos Renais/metabolismo , Proteínas de Membrana/genética , Nitrorredutases/genética , Animais , Aziridinas/metabolismo , Modelos Animais de Doenças , Feminino , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/patologia , Camundongos , Camundongos Transgênicos , Podócitos/patologia , Regiões Promotoras Genéticas/fisiologia
20.
Cardiovasc Res ; 83(2): 345-53, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19196828

RESUMO

AIMS: We investigated whether rapid cooling instituted by total liquid ventilation (TLV) improves cardiac and mitochondrial function in rabbits submitted to ischaemia-reperfusion. METHODS AND RESULTS: Rabbits were chronically instrumented with a coronary artery occluder and myocardial ultrasonic crystals for assessment of segment length-shortening. Two weeks later they were re-anaesthetized and underwent either a normothermic 30-min coronary artery occlusion (CAO) (Control group, n = 7) or a comparable CAO with cooling initiated by a 10-min hypothermic TLV and maintained by a cold blanket placed on the skin. Cooling was initiated after 5 or 15 min of CAO (Hypo-TLV and Hypo-TLV(15') groups, n = 6 and 5, respectively). A last group underwent normothermic TLV during CAO (Normo-TLV group, n = 6). Wall motion was measured in the conscious state over three days of reperfusion before infarct size evaluation and histology. Additional experiments were done for myocardial sampling in anaesthetized rabbits for mitochondrial studies. The Hypo-TLV procedure induced a rapid decrease in myocardial temperature to 32-34 degrees C. Throughout reperfusion, segment length-shortening was significantly increased in Hypo-TLV and Hypo-TLV(15') vs. Control and Normo-TLV (15.1 +/- 3.3%, 16.4 +/- 2.3%, 1.8 +/- 0.6%, and 1.1 +/- 0.8% at 72 h, respectively). Infarct sizes were also considerably attenuated in Hypo-TLV and Hypo-TLV(15') vs. Control and Normo-TLV (4 +/- 1%, 11 +/- 5%, 39 +/- 2%, and 42 +/- 5% infarction of risk zones, respectively). Mitochondrial function in myocardial samples obtained at the end of ischaemia or after 10 min of reperfusion was improved by Hypo-TLV with respect to ADP-stimulated respiration and calcium-induced opening of mitochondrial permeability transition pores (mPTP). Calcium concentration opening mPTP was, e.g., increased at the end of ischaemia in the risk zone in Hypo-TLV vs. Control (157 +/- 12 vs. 86 +/- 12 microM). Histology and electron microscopy also revealed better preservation of lungs and of cardiomyocyte ultrastructure in Hypo-TLV when compared with Control. CONCLUSION: Institution of rapid cooling by TLV during ischaemia reduces infarct size as well as other sequelae of ischaemia, such as post-ischaemic contractile and mitochondrial dysfunction.


Assuntos
Hipotermia Induzida , Mitocôndrias Cardíacas/patologia , Isquemia Miocárdica/terapia , Miocárdio/patologia , Traumatismo por Reperfusão/prevenção & controle , Disfunção Ventricular Esquerda/prevenção & controle , Difosfato de Adenosina/metabolismo , Animais , Roupas de Cama, Mesa e Banho , Cálcio/metabolismo , Modelos Animais de Doenças , Hemodinâmica , Hipotermia Induzida/instrumentação , Hipotermia Induzida/métodos , Ventilação Líquida , Masculino , Mitocôndrias Cardíacas/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Poro de Transição de Permeabilidade Mitocondrial , Contração Miocárdica , Isquemia Miocárdica/complicações , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/metabolismo , Coelhos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Fatores de Tempo , Disfunção Ventricular Esquerda/etiologia , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA