Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
J Thorac Cardiovasc Surg ; 160(3): 777-790.e5, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32222412

RESUMO

OBJECTIVES: Pulmonary vein obstruction (PVO) frequently occurs after repair of total anomalous pulmonary vein connection with progression of intimal hyperplasia from the anastomotic site toward upstream pulmonary veins (PVs). However, the understanding of mechanism in PVO progression is constrained by lack of data derived from a physiological model of the disease, and no prophylaxis has been established. We developed a new PVO animal model, investigated the mechanisms of PVO progression, and examined a new prophylactic strategy. METHODS: We developed a chronic PVO model using infant domestic pigs by cutting and resuturing the left lower PV followed by weekly hemodynamic parameter measurement and angiographic assessment of the anastomosed PV. Subsequently, we tested a novel therapeutic strategy with external application of rapamycin-eluting film to the anastomotic site. RESULTS: We found the pig PVO model mimicked human PVO hemodynamically and histopathologically. This model exhibited increased expression levels of Ki-67 and phospho-mammalian target of rapamycin in smooth muscle-like cells at the anastomotic neointima. In addition, contractile to synthetic phenotypic transition; that is, dedifferentiation of smooth muscle cells and mammalian target of rapamycin pathway activation in the neointima of upstream PVs were observed. Rapamycin-eluting films externally applied around the anastomotic site inhibited the activation of mammalian target of rapamycin in the smooth muscle-like cells of neointima, and delayed PV anastomotic stenosis. CONCLUSIONS: We demonstrate the evidence on dedifferentiation of smooth muscle-like cells and mammalian target of rapamycin pathway activation in the pathogenesis of PVO progression. Delivery of rapamycin to the anastomotic site from the external side delayed PV anastomotic stenosis, implicating a new therapeutic strategy to prevent PVO progression.


Assuntos
Veias Pulmonares , Pneumopatia Veno-Oclusiva/prevenção & controle , Pneumopatia Veno-Oclusiva/fisiopatologia , Sirolimo/farmacologia , Remodelação Vascular , Angiografia , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Músculo Liso/citologia , Neointima , Pneumopatia Veno-Oclusiva/metabolismo , Suínos , Serina-Treonina Quinases TOR/metabolismo
2.
Am J Respir Cell Mol Biol ; 63(1): 118-131, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32209028

RESUMO

Pulmonary veno-occlusive disease (PVOD) occurs in humans either as a heritable form (hPVOD) due to biallelic inactivating mutations of EIF2AK4 (encoding GCN2) or as a sporadic form in older age (sPVOD). The chemotherapeutic agent mitomycin C (MMC) is a potent inducer of PVOD in humans and in rats (MMC-PVOD). Here, we compared human hPVOD and sPVOD, and MMC-PVOD pathophysiology at the histological, cellular, and molecular levels to unravel common altered pathomechanisms. MMC exposure in rats was associated primarily with arterial and microvessel remodeling, and secondarily by venous remodeling, when PVOD became symptomatic. In all forms of PVOD tested, there was convergent GCN2-dependent but eIF2α-independent pulmonary protein overexpression of HO-1 (heme oxygenase 1) and CHOP (CCAAT-enhancer-binding protein [C/EBP] homologous protein), two downstream effectors of GCN2 signaling and endoplasmic reticulum stress. In human PVOD samples, CHOP immunohistochemical staining mainly labeled endothelial cells in remodeled veins and arteries. Strong HO-1 staining was observed only within capillary hemangiomatosis foci, where intense microvascular proliferation occurs. HO-1 and CHOP stainings were not observed in control and pulmonary arterial hypertension lung tissues, supporting the specificity for CHOP and HO-1 involvement in PVOD pathobiology. In vivo loss of GCN2 (EIF2AK4 mutations carriers and Eif2ak4-/- rats) or in vitro GCN2 inhibition in cultured pulmonary artery endothelial cells using pharmacological and siRNA approaches demonstrated that GCN2 loss of function negatively regulates BMP (bone morphogenetic protein)-dependent SMAD1/5/9 signaling. Exogenous BMP9 was still able to reverse GCN2 inhibition-induced proliferation of pulmonary artery endothelial cells. In conclusion, we identified CHOP and HO-1 inhibition, and BMP9, as potential therapeutic options for PVOD.


Assuntos
Pneumopatia Veno-Oclusiva/metabolismo , Pneumopatia Veno-Oclusiva/patologia , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Pulmão/metabolismo , Pulmão/patologia , Mutação/genética , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Ratos , Transdução de Sinais/fisiologia , Fator de Transcrição CHOP/metabolismo
3.
Vet Pathol ; 53(4): 813-22, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26926086

RESUMO

Pulmonary hypertension is a well-known though poorly characterized disease in veterinary medicine. In humans, pulmonary veno-occlusive disease (PVOD) is a rare cause of severe pulmonary hypertension with a mean survival time of 2 years without lung transplantation. Eleven adult dogs (5 males, 6 females; median age 10.5 years, representing various breeds) were examined following the development of severe respiratory signs. Lungs of affected animals were evaluated morphologically and with immunohistochemistry for alpha smooth muscle actin, desmin, CD31, CD3, CD20, and CD204. All dogs had pulmonary lesions consistent with PVOD, consisting of occlusive remodeling of small- to medium-sized pulmonary veins, foci of pulmonary capillary hemangiomatosis (PCH), and accumulation of hemosiderophages; 6 of 11 dogs had substantial pulmonary arterial medial and intimal thickening. Ultrastructural examination and immunohistochemistry showed that smooth muscle cells contributed to the venous occlusion. Increased expression of CD31 was evident in regions of PCH indicating increased numbers of endothelial cells in these foci. Spindle cells strongly expressing alpha smooth muscle actin and desmin co-localized with foci of PCH; similar cells were present but less intensely labeled elsewhere in non-PCH alveoli. B cells and macrophages, detected by immunohistochemistry, were not co-localized with the venous lesions of canine PVOD; small numbers of CD3-positive T cells were occasionally in and around the wall of remodeled veins. These findings indicate a condition in dogs with clinically severe respiratory disease and pathologic features resembling human PVOD, including foci of pulmonary venous remodeling and PCH.


Assuntos
Hemangioma Capilar/veterinária , Hipertensão Pulmonar/veterinária , Neoplasias Pulmonares/veterinária , Pneumopatia Veno-Oclusiva/veterinária , Animais , Cães , Feminino , Hemangioma Capilar/metabolismo , Hemangioma Capilar/patologia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Pneumopatia Veno-Oclusiva/complicações , Pneumopatia Veno-Oclusiva/metabolismo , Pneumopatia Veno-Oclusiva/patologia
4.
PLoS One ; 9(12): e115219, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25501010

RESUMO

AIMS: Pulmonary veno-occlusive disease (PVOD) is a rare condition of pulmonary arterial hypertension (PAH), in which post-capillary veins are affected. Since the therapeutic approach in PVOD differs from other forms of PAH, it is crucial to establish the diagnosis. Due to the fact that affected patients are often hemodynamically unstable, minimal invasive procedures are necessary for the diagnostic work-up. Chronic alveolar haemorrhage has been observed during bronchoalveolar lavage in PVOD cases. This study therefore investigates whether signs of alveolar haemorrhage can also be found in the sputum of these patients. METHODS AND RESULTS: Six patients suffering from PVOD were included in this analysis. As controls, patients with idiopathic PAH (n = 11), chronic thromboembolic PH (n = 9) and with sclerodermia-associated PH (n = 10) were assessed. Sputum from every patient was obtained by a non-invasive manner. The amount of haemosiderin-laden macrophages was determined using the Golde score. There were statistically significant more haemosiderin-laden macrophages in the sputum of patients suffering from PVOD as compared to the other groups (P<0.05). Assuming a cut-off of 200 on the Golde score, all of the 6 PVOD patients surpassed this value compared with only 1 out of the 30 cases with precapillary PH. Thus, sensitivity and specificity with respect to the diagnosis of PVOD was 100% and 97%, respectively. CONCLUSION: The content of haemosiderin-laden macrophages in the sputum of patients suffering from PVOD is significantly higher as compared to other forms of PH and may be useful in the non-invasive diagnostic work-up of these patients.


Assuntos
Hemossiderina/metabolismo , Macrófagos/metabolismo , Pneumopatia Veno-Oclusiva/diagnóstico , Escarro/metabolismo , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Humanos , Hipertensão Pulmonar/diagnóstico , Hipertensão Pulmonar/metabolismo , Masculino , Pessoa de Meia-Idade , Pneumopatia Veno-Oclusiva/metabolismo , Escarro/citologia
5.
J Thorac Cardiovasc Surg ; 148(6): 2550-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25172325

RESUMO

OBJECTIVES: Pulmonary vein stenosis (PVS) is a relentless disease with a poor prognosis. Although surgical repair can effectively treat "downstream" (near left atrial junction) PVS, residual "upstream" (deep in lung parenchyma) PVS commonly dictates long-term survival. Our initial studies revealed an association between PVS and transforming growth factor-ß signaling, which led us to investigate the effect of losartan on upstream pulmonary vein vasculopathy in a piglet model of PVS. METHODS: Neonatal Yorkshire piglets underwent sham surgical banding (sham, n = 6), staged bilateral pulmonary vein banding of all pulmonary veins except the right middle pulmonary vein (banded, n = 6), and staged pulmonary vein banding with losartan treatment (losartan, 1 mg/kg/d, n = 7). After 7 weeks, the hemodynamic data were obtained and the piglets killed. RESULTS: Pulmonary vein banding (compared with sham) was associated with continuous turbulent flow in banded pulmonary veins, pulmonary hypertension (pulmonary artery/systemic blood pressure ratio 0.51 ± 0.06 vs 0.23 ± 0.02, P < .001), and diffuse pulmonary vein intimal hyperplasia in the upstream pulmonary veins (P < .001). Losartan administration decreased the pulmonary artery/systemic blood pressure ratios compared with those in the banded piglets (0.36 ± 0.08 vs 0.51 ± 0.06, P = .007) but it remained greater than those in the sham group (P = .001). Losartan was also associated with diminished pulmonary vein intimal hyperplasia compared with that in the banded piglets (P < .001) but still remained more than that in the sham group (P = .035). Pulmonary vein banding reduced vascular endothelial-cadherin expression, indicative of diminished endothelial integrity, which was restored with losartan. CONCLUSIONS: Losartan treatment improved PVS-associated pulmonary hypertension and intimal hyperplasia and might be a beneficial prophylactic therapy for patients at high risk of developing PVS after pulmonary vein surgery.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Hipertensão Pulmonar/tratamento farmacológico , Losartan/farmacologia , Veias Pulmonares/efeitos dos fármacos , Pneumopatia Veno-Oclusiva/tratamento farmacológico , Túnica Íntima/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Antígenos CD/metabolismo , Caderinas/metabolismo , Constrição Patológica , Modelos Animais de Doenças , Hemodinâmica/efeitos dos fármacos , Hiperplasia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/tratamento farmacológico , Hipertrofia Ventricular Direita/patologia , Hipertrofia Ventricular Direita/fisiopatologia , Neointima , Veias Pulmonares/metabolismo , Veias Pulmonares/patologia , Veias Pulmonares/fisiopatologia , Pneumopatia Veno-Oclusiva/metabolismo , Pneumopatia Veno-Oclusiva/patologia , Pneumopatia Veno-Oclusiva/fisiopatologia , Suínos , Fatores de Tempo , Túnica Íntima/metabolismo , Túnica Íntima/patologia
6.
J Thorac Cardiovasc Surg ; 148(1): 245-53, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24084286

RESUMO

BACKGROUND: Surgical and catheter-based interventions on pulmonary veins are associated with pulmonary vein stenosis (PVS), which can progress diffusely through the "upstream" pulmonary veins. The mechanism has been rarely studied. We used a porcine model of PVS to assess disease progression with emphasis on the potential role of endothelial-mesenchymal transition (EndMT). METHODS: Neonatal piglets underwent bilateral pulmonary vein banding (banded, n = 6) or sham operations (sham, n = 6). Additional piglets underwent identical banding and stent implantation in a single-banded pulmonary vein 3 weeks postbanding (stented, n = 6). At 7 weeks postbanding, hemodynamics and upstream PV pathology were assessed. RESULTS: Banded piglets developed pulmonary hypertension. The upstream pulmonary veins exhibited intimal thickening associated with features of EndMT, including increased transforming growth factor (TGF)-ß1 and Smad expression, loss of endothelial and gain of mesenchymal marker expression, and coexpression of endothelial and mesenchymal markers in banded pulmonary vein intimal cells. These immunopathologic changes and a prominent myofibroblast phenotype in the remodeled pulmonary veins were consistently identified in specimens from patients with PVS, in vitro TGF-ß1-stimulated cells isolated from piglet and human pulmonary veins, and human umbilical vein endothelial cells. After stent implantation, decompression of a pulmonary vein was associated with reappearance of endothelial marker expression, suggesting the potential for plasticity in the observed pathologic changes, followed by rapid in-stent restenosis. CONCLUSIONS: Neonatal pulmonary vein banding in piglets recapitulates critical aspects of clinical PVS and highlights a pathologic profile consistent with EndMT, supporting the rationale for evaluating therapeutic strategies designed to exploit reversibility of upstream pulmonary vein pathology.


Assuntos
Veias Pulmonares/fisiopatologia , Pneumopatia Veno-Oclusiva/fisiopatologia , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Células Cultivadas , Constrição Patológica , Modelos Animais de Doenças , Progressão da Doença , Transição Epitelial-Mesenquimal , Hemodinâmica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Hiperplasia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Neointima , Fenótipo , Veias Pulmonares/metabolismo , Veias Pulmonares/patologia , Pneumopatia Veno-Oclusiva/complicações , Pneumopatia Veno-Oclusiva/metabolismo , Pneumopatia Veno-Oclusiva/patologia , Recidiva , Proteínas Smad/metabolismo , Suínos , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo
7.
Arthritis Res Ther ; 13(2): R61, 2011 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-21492463

RESUMO

INTRODUCTION: Systemic sclerosis (SSc) complicated by pulmonary arterial hypertension (PAH) carries a poor prognosis, despite pulmonary vascular dilating therapy. Platelet-derived growth factor receptor-ß (PDGFR-ß) and epidermal growth factor receptor (EGFR) are potential therapeutic targets for PAH because of their proliferative effects on vessel remodelling. To explore their role in SScPAH, we compared PDGFR- and EGFR-mmunoreactivity in lung tissue specimens from SScPAH. We compared staining patterns with idiopathic PAH (IPAH) and pulmonary veno-occlusive disease (PVOD), as SScPAH vasculopathy differs from IPAH and sometimes displays features of PVOD. Immunoreactivity patterns of phosphorylated PDGFR-ß (pPDGFR-ß) and the ligand PDGF-B were evaluated to provide more insight into the patterns of PDGFR-b activation. METHODS: Lung tissue specimens from five SScPAH, nine IPAH, six PVOD patients and five controls were examined. Immunoreactivity was scored for presence, distribution and intensity. RESULTS: All SScPAH and three of nine IPAH cases (P = 0.03) showed PDGFR-ß-immunoreactivity in small vessels (arterioles/venules); of five SScPAH vs. two of nine IPAH cases (P = 0.02) showed venous immunoreactivity. In small vessels, intensity was stronger in SScPAH vs. IPAH. No differences were found between SScPAH and PVOD. One of five normal controls demonstrated focally mild immunoreactivity. There were no differences in PDGF-ligand and pPDGFR-b-immunoreactivity between patient groups; however, pPDGFR-b-immunoreactivity tended to be more prevalent in SScPAH small vasculature compared to IPAH. Vascular EGFR-immunoreactivity was limited to arterial and arteriolar walls, without differences between groups. No immunoreactivity was observed in vasculature of normals. CONCLUSIONS: PDGFR-ß-immunoreactivity in SScPAH is more common and intense in small- and post-capillary vessels than in IPAH and does not differ from PVOD, fitting in with histomorphological distribution of vasculopathy. PDGFR-ß immunoreactivity pattern is not paralleled by pPDGFR-ß or PDGF-B patterns. PDGFR-ß- and EGFR-immunoreactivity of pulmonary vessels distinguishes PAH patients from controls.


Assuntos
Biomarcadores/análise , Receptores ErbB/biossíntese , Hipertensão Pulmonar/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/biossíntese , Escleroderma Sistêmico/metabolismo , Adulto , Idoso , Biomarcadores/metabolismo , Estudos de Casos e Controles , Receptores ErbB/análise , Hipertensão Pulmonar Primária Familiar , Feminino , Humanos , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Imuno-Histoquímica , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Masculino , Pessoa de Meia-Idade , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Veias Pulmonares/metabolismo , Veias Pulmonares/fisiopatologia , Pneumopatia Veno-Oclusiva/metabolismo , Pneumopatia Veno-Oclusiva/fisiopatologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/análise , Escleroderma Sistêmico/complicações , Escleroderma Sistêmico/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA