Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Hepatology ; 72(5): 1771-1785, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32060938

RESUMO

BACKGROUND AND AIMS: This study examined whether enhanced susceptibility of steatotic liver to ischemia-reperfusion (I/R) injury is due to impaired recruitment of bone marrow (BM) progenitors of liver sinusoidal endothelial cells (LSECs, also called sinusoidal endothelial cell progenitor cells [sprocs]) with diminished repair of injured LSECs and whether restoring signaling to recruit BM sprocs reduces I/R injury. APPROACH AND RESULTS: Hepatic vessels were clamped for 1 hour in rats fed a high-fat, high-fructose (HFHF) diet for 5, 10, or 15 weeks. Matrix metalloproteinase 9 (MMP-9) antisense oligonucleotides (ASO) or an MMP inhibitor were used to induce liver-selective MMP-9 inhibition. HFHF rats had mild, moderate, and severe steatosis, respectively, at 5, 10, and 15 weeks. I/R injury was enhanced in HFHF rats; this was accompanied by complete absence of hepatic vascular endothelial growth factor (VEGF)-stromal cell-derived factor 1 (sdf1) signaling, leading to lack of BM sproc recruitment. Liver-selective MMP-9 inhibition to protect against proteolytic cleavage of hepatic VEGF using either MMP-9 ASO or intraportal MMP inhibitor in 5-week and 10-week HFHF rats enhanced hepatic VEGF-sdf1 signaling, increased BM sproc recruitment, and reduced alanine aminotransferase (ALT) by 92% and 77% at 5 weeks and by 80% and 64% at 10 weeks of the HFHF diet, respectively. After I/R injury in 15-week HFHF rats, the MMP inhibitor reduced active MMP-9 expression by 97%, ameliorated histologic evidence of injury, and reduced ALT by 58%, which is comparable to control rats sustaining I/R injury. Rescue therapy with intraportal MMP inhibitor, given after ischemia, in the 5-week HFHF rat reduced ALT by 71% and reduced necrosis. CONCLUSIONS: Lack of signaling to recruit BM sprocs that repair injured LSECs renders steatotic liver more susceptible to I/R injury. Liver-selective MMP-9 inhibition enhances VEGF-sdf1 signaling and recruitment of BM sprocs, which markedly protects against I/R injury, even in severely steatotic rats.


Assuntos
Células Progenitoras Endoteliais/efeitos dos fármacos , Fígado Gorduroso/etiologia , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Transplante de Medula Óssea , Dieta Hiperlipídica , Açúcares da Dieta/efeitos adversos , Modelos Animais de Doenças , Suscetibilidade a Doenças/terapia , Células Progenitoras Endoteliais/patologia , Fígado Gorduroso/diagnóstico , Fígado Gorduroso/tratamento farmacológico , Frutose/efeitos adversos , Humanos , Fígado/irrigação sanguínea , Fígado/diagnóstico por imagem , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Inibidores de Metaloproteinases de Matriz/uso terapêutico , Microvasos/citologia , Microvasos/efeitos dos fármacos , Microvasos/patologia , Ratos , Traumatismo por Reperfusão/etiologia
3.
Hepatology ; 69(2): 831-844, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30120894

RESUMO

Treatment of hematological malignancy with antibody-drug conjugates (ADCs) may cause liver injury. ADCs deliver a toxic moiety into antigen-expressing tumor cells, but may also injure hepatic sinusoids (sinusoidal obstruction syndrome; SOS). We studied patients who received an anti-CD22/calicheamicin conjugate (inotuzumab ozogamicin; InO) to gain insight into mechanisms of sinusoidal injury, given that there are no CD22+ cells in the normal liver, but nonspecific uptake of ADCs by liver sinusoidal endothelial cells (LSECs). Six hundred thirty-eight patients (307 with acute lymphocytic leukemia [ALL], 311 with non-Hodgkin's lymphoma [NHL]) were randomized to either InO or standard chemotherapy (controls). While blinded to treatment assignment, we reviewed all cases with hepatobiliary complications to adjudicate the causes. Frequency of SOS among patients who received InO was 5 of 328 (1.5%), compared to no cases among 310 control patients. Drug-induced liver injury (DILI) developed in 26 (7.9%) InO recipients and 3 (1%) controls. Intrahepatic cholestasis (IHC) was observed in 4.9% of InO recipients and in 5.5% of controls. Subsequent to the randomization study, 113 patients with ALL underwent allogeneic hematopoietic cell transplantation (HCT); frequency of SOS in those previously exposed to InO was 21 of 79 (27%) versus 3 of 34 (9%) in controls. An exploratory multivariate model identified a past history of liver disease and thrombocytopenia before conditioning therapy as dominant risk factors for SOS after transplant. Conclusion: Frequencies of SOS and DILI after inotuzumab ozogamicin treatment were 1.5% and 7.9%, respectively, compared to none and 1% among controls who received standard chemotherapy. These data suggest that ADCs that do not target antigens present in the normal liver have a relatively low frequency of SOS, but a relatively high frequency of DILI.


Assuntos
Antineoplásicos Imunológicos/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Inotuzumab Ozogamicina/efeitos adversos , Linfoma não Hodgkin/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Gastroenterologistas , Transplante de Células-Tronco Hematopoéticas , Hepatopatia Veno-Oclusiva/etiologia , Humanos
4.
Hepatology ; 69(3): 1259-1272, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30141211

RESUMO

Normal liver sinusoidal endothelial cells (LSECs) promote quiescence of hepatic stellate cells (HSCs). Prior to fibrosis, LSECs undergo capillarization, which is permissive for HSC activation, the proximate event in hepatic fibrosis. The aims of this study were to elucidate the nature of and mechanisms leading to capillarization and to determine how LSECs promote HSC quiescence and why "capillarized LSECs" lose control of HSC activation. The contribution of bone marrow (BM) endothelial progenitor cells to capillarization was identified using rats transplanted with transgenic enhanced green fluorescent protein-positive BM. Shotgun proteomics and informatics were used to identify the LSEC mediator that maintains HSC quiescence. The study shows that capillarization is due to repair of injured LSECs by BM endothelial progenitors that engraft but fail to fully mature. Lack of maturation of BM-derived LSECs is due to cell autonomous pathways that inhibit the nitric oxide pathway. We identify heparin binding epidermal growth factor-like growth factor (HB-EGF) as the signal that maintains HSC quiescence and show that immature LSECs are unable to shed HB-EGF from the cytosolic membrane. Conclusion: Chronic liver injury can recruit BM progenitors of LSECs that engraft and fail to fully differentiate, which creates an environment that is permissive for hepatic fibrosis; elucidation of these early events in the fibrotic process will provide targets for treatment of hepatic fibrosis.


Assuntos
Diferenciação Celular , Células Progenitoras Endoteliais/citologia , Cirrose Hepática/etiologia , Animais , Células Progenitoras Endoteliais/transplante , Masculino , Ratos , Ratos Endogâmicos Lew
5.
Hepatology ; 69(1): 314-328, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30019419

RESUMO

Recruitment of liver sinusoidal endothelial cell progenitor cells (sprocs) from the bone marrow by vascular endothelial growth factor-stromal cell-derived factor-1 (VEGF-sdf-1) signaling promotes recovery from injury and drives liver regeneration. Matrix metalloproteinases (MMPs) can proteolytically cleave VEGF, which might inhibit progenitor cell recruitment, but systemic matrix metalloproteinase inhibition might prevent efflux of progenitors from the bone marrow. The hypothesis for this study was that liver-selective MMP-9 inhibition would protect the hepatic VEGF-sdf-1 signaling pathway, enhance bone marrow sproc recruitment, and thereby ameliorate liver injury and accelerate liver regeneration, whereas systemic MMP inhibition would impair bone marrow sproc mobilization and therefore have less benefit or be detrimental. We found that liver-selective MMP-9 inhibition accelerated liver regeneration after partial hepatectomy by 40%, whereas systemic MMP inhibition impaired liver regeneration. Liver-selective MMP-9 inhibition largely abolished warm ischemia-reperfusion injury. In the extended hepatectomy model, liver-selective MMP-9 inhibition restored liver sinusoidal endothelial cell integrity, enhanced liver regeneration, and reduced ascites. Liver-selective MMP-9 inhibition markedly increased recruitment and engraftment of bone marrow sprocs, whereas systemic MMP inhibition impaired mobilization of bone marrow sprocs and their hepatic engraftment. Hepatic MMP-9 proteolytically cleaved VEGF after partial hepatectomy. Liver-selective MMP-9 inhibition prevented VEGF cleavage and doubled protein expression of VEGF and its downstream signaling partner sdf-1. In contrast, systemic MMP inhibition enhanced recruitment and engraftment of infused allogeneic progenitors. Conclusion: Liver-selective MMP inhibition prevents proteolytic cleavage of hepatic VEGF, which enhances recruitment and engraftment of bone marrow sprocs after liver injury. This ameliorates injury and accelerates liver regeneration. Liver-selective MMP-9 inhibition may be a therapeutic tool for liver injury that damages the vasculature, whereas systemic MMP inhibition can enhance the benefit of stem cell therapy with endothelial progenitor cells.


Assuntos
Regeneração Hepática/efeitos dos fármacos , Fígado/irrigação sanguínea , Metaloproteinase 9 da Matriz/efeitos dos fármacos , Inibidores de Metaloproteinases de Matriz/farmacocinética , Inibidores de Metaloproteinases de Matriz/uso terapêutico , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos Antissenso/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Animais , Masculino , Metaloproteinase 9 da Matriz/fisiologia , Ratos , Ratos Endogâmicos Lew , Fatores de Tempo
6.
Semin Liver Dis ; 37(4): 377-387, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29272898

RESUMO

This update focuses on two main topics. First, recent developments in our understanding of liver sinusoidal endothelial cell (LSEC) function will be reviewed, specifically elimination of blood-borne waste, immunological function of LSECs, interaction of LSECs with liver metastases, LSECs and liver regeneration, and LSECs and hepatic fibrosis. Second, given the current emphasis on rigor and transparency in biomedical research, the update discusses the need for standardization of methods to demonstrate identity and purity of isolated LSECs, pitfalls in methods that might lead to a selection bias in the types of LSECs isolated, and questions about long-term culture of LSECs. Various surface markers used for immunomagnetic selection are reviewed.


Assuntos
Capilares/metabolismo , Células Endoteliais/metabolismo , Hepatopatias/metabolismo , Fígado/irrigação sanguínea , Animais , Biomarcadores/metabolismo , Capilares/imunologia , Capilares/patologia , Técnicas de Cultura de Células , Separação Celular/métodos , Células Endoteliais/imunologia , Células Endoteliais/patologia , Humanos , Hepatopatias/imunologia , Hepatopatias/patologia , Fenótipo , Transdução de Sinais
7.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G739-46, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26939868

RESUMO

In liver injury, recruitment of bone marrow (BM) progenitors of liver sinusoidal endothelial cells (sprocs) is necessary for normal liver regeneration. Hepatic vascular endothelial growth factor (VEGF) is a central regulator of the recruitment process. We examine whether stromal cell-derived factor 1 [sdf1, or CXC ligand 12 (CXCL12)] acts downstream from VEGF to mediate recruitment of BM sprocs, what the sdf1 receptor type [CXC receptor (CXCR)-4 or CXCR7] is on sprocs, and whether sdf1 signaling is required for normal liver regeneration. Studies were performed in the rat partial hepatectomy model. Tracking studies of BM sprocs were performed in wild-type Lewis rats that had undergone BM transplantation from transgenic enhanced green fluorescent protein-positive Lewis rats. Knockdown studies were performed using antisense oligonucleotides (ASOs). Expression of sdf1 doubles in liver and liver sinusoidal endothelial cells (LSECs) after partial hepatectomy. Upregulation of sdf1 expression increases proliferation of sprocs in the BM, mobilization of CXCR7(+) BM sprocs to the circulation, and engraftment of CXCR7(+) BM sprocs in the liver and promotes liver regeneration. Knockdown of hepatic VEGF with ASOs decreases hepatic sdf1 expression and plasma sdf1 levels. When the effect of VEGF knockdown on sdf1 is offset by infusion of sdf1, VEGF knockdown-induced impairment of BM sproc recruitment after partial hepatectomy is completely attenuated and liver regeneration is normalized. These data demonstrate that the VEGF-sdf1 pathway regulates recruitment of CXCR7(+) BM sprocs to the hepatic sinusoid after partial hepatectomy and is required for normal liver regeneration.


Assuntos
Quimiocina CXCL12/metabolismo , Células Progenitoras Endoteliais/metabolismo , Regeneração Hepática , Fígado/metabolismo , Receptores CXCR/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Transplante de Medula Óssea , Células Cultivadas , Quimiocina CXCL12/genética , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Fígado/irrigação sanguínea , Fígado/citologia , Fígado/fisiologia , Masculino , Ratos , Ratos Endogâmicos Lew , Fator A de Crescimento do Endotélio Vascular/genética
8.
Hepatology ; 61(5): 1740-6, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25131509

RESUMO

Capillarization, lack of liver sinusoidal endothelial cell (LSEC) fenestration, and formation of an organized basement membrane not only precedes fibrosis, but is also permissive for hepatic stellate cell activation and fibrosis. Thus, dysregulation of the LSEC phenotype is a critical step in the fibrotic process. Both a vascular endothelial growth factor (VEGF)-stimulated, nitric oxide (NO)-independent pathway and a VEGF-stimulated NO-dependent pathway are necessary to maintain the differentiated LSEC phenotype. The NO-dependent pathway is impaired in capillarization and activation of this pathway downstream from NO restores LSEC differentiation in vivo. Restoration of LSEC differentiation in vivo promotes HSC quiescence, enhances regression of fibrosis, and prevents progression of cirrhosis.


Assuntos
Células Endoteliais , Cirrose Hepática/patologia , Diferenciação Celular , Humanos
9.
J Control Release ; 191: 15-23, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25016969

RESUMO

L4F, an alpha helical peptide inspired by the lipid-binding domain of the ApoA1 protein, has potential applications in the reduction of inflammation involved with cardiovascular disease as well as an antioxidant effect that inhibits liver fibrosis. In addition to its biological activity, amphipathic peptides such as L4F are likely candidates to direct the molecular assembly of peptide nanostructures. Here we describe the stabilization of the amphipathic L4F peptide through fusion to a high molecular weight protein polymer. Comprised of multiple pentameric repeats, elastin-like polypeptides (ELPs) are biodegradable protein polymers inspired from the human gene for tropoelastin. Dynamic light scattering confirmed that the fusion peptide forms nanoparticles with a hydrodynamic radius of approximately 50nm, which is unexpectedly above that observed for the free ELP (~5.1nm). To further investigate their morphology, conventional and cryogenic transmission electron microscopy were used to reveal that they are unilamellar vesicles. On average, these vesicles are 49nm in radius with lamellae 8nm in thickness. To evaluate their therapeutic potential, the L4F nanoparticles were incubated with hepatic stellate cells. Stellate cell activation leads to hepatic fibrosis; furthermore, their activation is suppressed by anti-oxidant activity of ApoA1 mimetic peptides. Consistent with this observation, L4F nanoparticles were found to suppress hepatic stellate cell activation in vitro. To evaluate the in vivo potential for these nanostructures, their plasma pharmacokinetics were evaluated in rats. Despite the assembly of nanostructures, both free L4F and L4F nanoparticles exhibited similar half-lives of approximately 1h in plasma. This is the first study reporting the stabilization of peptide-based vesicles using ApoA1 mimetic peptides fused to a protein polymer; furthermore, this platform for peptide-vesicle assembly may have utility in the design of biodegradable nanostructures.


Assuntos
Anti-Inflamatórios/química , Antioxidantes/química , Apolipoproteína A-I/química , Portadores de Fármacos , Fragmentos de Peptídeos/química , Tropoelastina/química , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/sangue , Anti-Inflamatórios/farmacocinética , Antioxidantes/administração & dosagem , Antioxidantes/farmacocinética , Apolipoproteína A-I/administração & dosagem , Apolipoproteína A-I/sangue , Apolipoproteína A-I/genética , Apolipoproteína A-I/farmacocinética , Células Cultivadas , Química Farmacêutica , Engenharia Genética , Meia-Vida , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Injeções Intravenosas , Camundongos , Nanopartículas , Nanotecnologia , Tamanho da Partícula , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacocinética , Estabilidade Proteica , Estrutura Secundária de Proteína , Ratos , Proteínas Recombinantes de Fusão/química , Tecnologia Farmacêutica/métodos , Tropoelastina/genética , Tropoelastina/metabolismo
10.
J Clin Invest ; 123(5): 1861-6, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23635783

RESUMO

Liver sinusoidal endothelial cells (LSECs) have long been noted to contribute to liver regeneration after liver injury. In normal liver, the major cellular source of HGF is the hepatic stellate cell, but after liver injury, HGF expression has been thought to increase markedly in proliferating LSECs. However, emerging data suggest that even after injury, LSEC expression of HGF does not increase greatly. In contrast, bone marrow progenitor cells of LSECs (BM SPCs), which are rich in HGF, are recruited to the liver after injury. This Review examines liver regeneration from the perspective that BM SPCs that have been recruited to the liver, rather than mature LSECs, drive liver regeneration.


Assuntos
Células Endoteliais/citologia , Regeneração Hepática , Fígado/citologia , Animais , Células da Medula Óssea/citologia , Proliferação de Células , Fator de Crescimento de Hepatócito/metabolismo , Hepatócitos/citologia , Humanos , Fígado/metabolismo , Hepatopatias/metabolismo , Ratos , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Int J Hematol ; 97(4): 456-64, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23460018

RESUMO

The purpose of this prospective observational study was to determine the incidence of hepatic sinusoidal obstruction syndrome (SOS), following gemtuzumab ozogamicin (GO) therapy in routine clinical practice. Patients receiving GO for acute myeloid leukemia (AML) were eligible. Assessments were requested to be performed weekly for 6 weeks after the start of GO therapy or 4 weeks after the last dose (whichever was later), and after 6 months. The primary outcome variable was the incidence of SOS as judged by a panel of independent experts. A total of 512 patients were enrolled at 54 US centers and 482 were evaluable. The incidence of SOS in this study population was 9.1 % (44/482; 95 % confidence interval 6.9-12.0 %). Of the 44 patients classified as having SOS, 8 were mild, 17 moderate, and 19 severe; 33 died within 6 months (20 of disease progression and 13 of SOS and multiorgan failure). Most (68 %) patients in the study died within 6 months; most of these deaths (73 %) were due to progression of AML. Serious adverse events occurred in 85 % of patients, most (81 %) due to AML, febrile neutropenia, pyrexia, and sepsis. GO administered in routine clinical practice carries an overall 9.1 % risk of SOS and a 2.7 % risk of death from SOS and multiorgan failure. No risk factors were identified for the development of SOS.


Assuntos
Aminoglicosídeos/efeitos adversos , Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos/efeitos adversos , Hepatopatia Veno-Oclusiva/induzido quimicamente , Hepatopatia Veno-Oclusiva/epidemiologia , Adulto , Idoso , Aminoglicosídeos/administração & dosagem , Aminoglicosídeos/uso terapêutico , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Feminino , Gemtuzumab , Hepatopatia Veno-Oclusiva/diagnóstico , Humanos , Incidência , Leucemia Mieloide Aguda/complicações , Leucemia Mieloide Aguda/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Mortalidade , Estudos Prospectivos , Fatores de Risco
12.
Gastroenterology ; 143(6): 1555-1563.e2, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22902870

RESUMO

BACKGROUND & AIMS: After liver injury, bone marrow-derived liver sinusoidal endothelial cell progenitor cells (BM SPCs) repopulate the sinusoid as liver sinusoidal endothelial cells (LSECs). After partial hepatectomy, BM SPCs provide hepatocyte growth factor, promote hepatocyte proliferation, and are necessary for normal liver regeneration. We examined how hepatic vascular endothelial growth factor (VEGF) regulates recruitment of BM SPCs and their effects on liver injury. METHODS: Rats were given injections of dimethylnitrosamine to induce liver injury, which was assessed by histology and transaminase assays. Recruitment of SPCs was analyzed by examining BM SPC proliferation, mobilization to the circulation, engraftment in liver, and development of fenestration (differentiation). RESULTS: Dimethylnitrosamine caused extensive denudation of LSECs at 24 hours, followed by centrilobular hemorrhagic necrosis at 48 hours. Proliferation of BM SPCs, the number of SPCs in the bone marrow, and mobilization of BM SPCs to the circulation increased 2- to 4-fold by 24 hours after injection of dimethylnitrosamine; within 5 days, 40% of all LSECs came from engrafted BM SPCs. Allogeneic resident SPCs, infused 24 hours after injection of dimethylnitrosamine, repopulated the sinusoid as LSECs and reduced liver injury. Expression of hepatic VEGF messenger RNA and protein increased 5-fold by 24 hours after dimethylnitrosamine injection. Knockdown of hepatic VEGF with antisense oligonucleotides completely prevented dimethylnitrosamine-induced proliferation of BM SPCs and their mobilization to the circulation, reduced their engraftment by 46%, completely prevented formation of fenestration after engraftment as LSECs, and exacerbated dimethylnitrosamine injury. CONCLUSIONS: BM SPC recruitment is a repair response to dimethylnitrosamine liver injury in rats. Hepatic VEGF regulates recruitment of BM SPCs to liver and reduces this form of liver injury.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Células Endoteliais/patologia , Fígado/metabolismo , Fígado/patologia , Células-Tronco/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células da Medula Óssea/patologia , Transplante de Medula Óssea , Movimento Celular , Proliferação de Células , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Dimetilnitrosamina/efeitos adversos , Hepatectomia , Modelos Animais , Ratos , Ratos Endogâmicos Lew , Fatores de Tempo
13.
J Clin Invest ; 122(4): 1567-73, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22406533

RESUMO

The ability of the liver to regenerate is crucial to protect liver function after injury and during chronic disease. Increases in hepatocyte growth factor (HGF) in liver sinusoidal endothelial cells (LSECs) are thought to drive liver regeneration. However, in contrast to endothelial progenitor cells, mature LSECs express little HGF. Therefore, we sought to establish in rats whether liver injury causes BM LSEC progenitor cells to engraft in the liver and provide increased levels of HGF and to examine the relative contribution of resident and BM LSEC progenitors. LSEC label-retaining cells and progenitors were identified in liver and LSEC progenitors in BM. BM LSEC progenitors did not contribute to normal LSEC turnover in the liver. However, after partial hepatectomy, BM LSEC progenitor proliferation and mobilization to the circulation doubled. In the liver, one-quarter of the LSECs were BM derived, and BM LSEC progenitors differentiated into fenestrated LSECs. When irradiated rats underwent partial hepatectomy, liver regeneration was compromised, but infusion of LSEC progenitors rescued the defect. Further analysis revealed that BM LSEC progenitors expressed substantially more HGF and were more proliferative than resident LSEC progenitors after partial hepatectomy. Resident LSEC progenitors within their niche may play a smaller role in recovery from partial hepatectomy than BM LSEC progenitors, but, when infused after injury, these progenitors engrafted and expanded markedly over a 2-month period. In conclusion, LSEC progenitor cells are present in liver and BM, and recruitment of BM LSEC progenitors is necessary for normal liver regeneration.


Assuntos
Células Endoteliais/fisiologia , Regeneração Hepática/fisiologia , Fígado/irrigação sanguínea , Animais , Transplante de Medula Óssea , Divisão Celular , Linhagem da Célula , Movimento Celular , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas/classificação , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Hematopoéticas/ultraestrutura , Hepatectomia , Fator de Crescimento de Hepatócito/biossíntese , Fator de Crescimento de Hepatócito/fisiologia , Masculino , Quimera por Radiação , Ratos , Ratos Endogâmicos F344 , Ratos Endogâmicos Lew , Ratos Sprague-Dawley
14.
Gastroenterology ; 142(4): 918-927.e6, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22178212

RESUMO

BACKGROUND & AIMS: Capillarization, characterized by loss of differentiation of liver sinusoidal endothelial cells (LSECs), precedes the onset of hepatic fibrosis. We investigated whether restoration of LSEC differentiation would normalize crosstalk with activated hepatic stellate cells (HSC) and thereby promote quiescence of HSC and regression of fibrosis. METHODS: Rat LSECs were cultured with inhibitors and/or agonists and examined by scanning electron microscopy for fenestrae in sieve plates. Cirrhosis was induced in rats using thioacetamide, followed by administration of BAY 60-2770, an activator of soluble guanylate cyclase (sGC). Fibrosis was assessed by Sirius red staining; expression of α-smooth muscle actin was measured by immunoblot analysis. RESULTS: Maintenance of LSEC differentiation requires vascular endothelial growth factor-A stimulation of nitric oxide-dependent signaling (via sGC and cyclic guanosine monophosphate) and nitric oxide-independent signaling. In rats with thioacetamide-induced cirrhosis, BAY 60-2770 accelerated the complete reversal of capillarization (restored differentiation of LSECs) without directly affecting activation of HSCs or fibrosis. Restoration of differentiation to LSECs led to quiescence of HSCs and regression of fibrosis in the absence of further exposure to BAY 60-2770. Activation of sGC with BAY 60-2770 prevented progression of cirrhosis, despite continued administration of thioacetamide. CONCLUSIONS: The state of LSEC differentiation plays a pivotal role in HSC activation and the fibrotic process.


Assuntos
Capilares/patologia , Diferenciação Celular , Células Endoteliais/patologia , Células Estreladas do Fígado/patologia , Cirrose Hepática Experimental/patologia , Fígado/irrigação sanguínea , Fígado/patologia , Comunicação Parácrina , Actinas/metabolismo , Animais , Benzoatos/farmacologia , Compostos de Bifenilo , Western Blotting , Capilares/efeitos dos fármacos , Capilares/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , GMP Cíclico/metabolismo , Progressão da Doença , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Guanilato Ciclase/metabolismo , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Hidrocarbonetos Fluorados/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/metabolismo , Cirrose Hepática Experimental/prevenção & controle , Masculino , Microscopia Eletrônica de Varredura , Óxido Nítrico , Comunicação Parácrina/efeitos dos fármacos , Fenótipo , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Guanilil Ciclase Solúvel , Tioacetamida , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Curr Gerontol Geriatr Res ; 2011: 439835, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21687587

RESUMO

Aging is associated with marked changes in the hepatic sinusoid, yet the effect of old age on hepatic stellate cells (HSC) has not been well described. Transmission electron microscopy and immunohistochemistry were used to study the effects of aging on HSC in livers from rats (3-4 mths versus 24-27 mths) and mice (2-3 mths versus 20-22 mths). Desmin-positive HSC doubled in old age in both mice and rats. Alpha-smooth muscle actin- (αSMA-) positive cells did not increase significantly and remained only a small percentage of desmin-positive cells. Electron microscopy revealed that old age is associated with HSC that have a substantial increase in the number of lipid droplets which are larger in diameter. There was also a marked increase of HSC that protruded into the sinusoidal lumen in old mice. In conclusion, old age is associated with hyperplasia of HSC that are not activated and are engorged with lipid droplets.

16.
Liver Int ; 31(6): 762-72, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21645207

RESUMO

This is a meeting report of the presentations given at the 15th International Symposium on Cells of the Hepatic Sinusoid, held in 2010. The areas covered include the contributions of the various liver cell populations to liver disease, molecular and cellular targets involved in steatohepatitis, hepatic fibrosis and cancer and regenerative medicine. In addition to a review of the science presented at the meeting, this report provides references to recent literature on the topics covered at the meeting.


Assuntos
Células Endoteliais/patologia , Células Estreladas do Fígado/patologia , Hepatócitos/patologia , Fígado/patologia , Células-Tronco/patologia , Animais , Carcinoma Hepatocelular/patologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Fígado Gorduroso/patologia , Células Estreladas do Fígado/imunologia , Células Estreladas do Fígado/metabolismo , Hepatócitos/imunologia , Hepatócitos/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Fígado/irrigação sanguínea , Fígado/imunologia , Fígado/metabolismo , Cirrose Hepática/patologia , Neoplasias Hepáticas/patologia , Regeneração Hepática , Transdução de Sinais , Células-Tronco/imunologia , Células-Tronco/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 299(5): G1204-10, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20813915

RESUMO

Many liver sinusoidal endothelial cell (LSEC)-dependent processes, including drug-induced liver injury, ischemia-reperfusion injury, acute and chronic rejection, fibrosis, and the HELLP (hemolytic anemia, elevated liver enzymes, low platelet count) syndrome, may have a lobular distribution. Studies of the mechanism of this distribution would benefit from a reliable method to isolate LSEC populations from different regions. We established and verified a simple method to isolate periportal, midlobular, and centrilobular LSEC. Three subpopulations of LSEC were isolated by immunomagnetic separation on the basis of CD45 expression. Flow cytometry showed that 78.2 ± 2.3% of LSEC were CD45 positive and that LSEC could be divided into CD45 bright (28.6 ± 2.7% of total population), dim (49.6 ± 1.0%), and negative populations (21.8 ± 2.3%). Immunohistochemistry confirmed that in vivo expression of CD45 in LSEC had a lobular distribution with enhanced CD45 staining in periportal LSEC. Cell diameter, fenestral diameter, number of fenestrae per sieve plate and per cell, porosity, and lectin uptake were significantly different in the subpopulations, consistent with the literature. Endocytosis of low concentrations of the LSEC-specific substrate, formaldehyde-treated serum albumin, was restricted to CD45 bright and dim LSEC. Acetaminophen was more toxic to the CD45 dim and negative populations than to the CD45 bright population. In conclusion, CD45 is highly expressed in periportal LSEC, low in midlobular LSEC, and negative in centrilobular LSEC, and this provides an easy separation method to isolate LSEC from the three different hepatic regions. The LSEC subpopulations obtained by this method are adequate for functional studies and drug toxicity testing.


Assuntos
Células Endoteliais/citologia , Antígenos Comuns de Leucócito/imunologia , Fígado/citologia , Acetaminofen/farmacologia , Análise de Variância , Animais , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Endocitose/imunologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Citometria de Fluxo , Imuno-Histoquímica , Fígado/efeitos dos fármacos , Fígado/imunologia , Masculino , Microscopia Eletrônica de Varredura , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Struct Biol ; 171(3): 382-8, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20570732

RESUMO

Fenestrations are pores in liver sinusoidal endothelial cells that filter substrates and debris between the blood and hepatocytes. Fenestrations have significant roles in aging and the regulation of lipoproteins. However their small size (<200 nm) has prohibited any functional analysis by light microscopy. We employed structured illumination light microscopy to observe fenestrations in isolated rat liver sinusoidal endothelial cells with great clarity and spatial resolution. With this method, the three-dimensional structure of fenestrations (diameter 123+/-24 nm) and sieve plates was elucidated and it was shown that fenestrations occur in areas of abrupt cytoplasmic thinning (165+/-54 nm vs. 292+/-103 nm in non-fenestrated regions, P<0.0001). Sieve plates were not preferentially co-localized with fluorescently labeled F-actin stress fibers and endothelial nitric oxide synthase but appeared to occur in primarily attenuated non-raft regions of the cell membrane. Labyrinthine structures were not seen and all fenestrations were short cylindrical pores. In conclusion, three-dimensional structured illumination microscopy has enabled the unlimited power of fluorescent immunostaining and co-localization to reveal new structural and functional information about fenestrations and sieve plates.


Assuntos
Células Endoteliais/citologia , Hepatócitos/citologia , Actinas , Animais , Membrana Celular/metabolismo , Células Endoteliais/metabolismo , Imunofluorescência , Hepatócitos/metabolismo , Fígado/citologia , Fígado/metabolismo , Masculino , Microscopia , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos , Ratos Sprague-Dawley
19.
Gastroenterology ; 137(2): 704-12, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19447108

RESUMO

BACKGROUND & AIMS: Damage to hepatic sinusoidal endothelial cells (SECs) initiates sinusoidal obstruction syndrome (SOS), which is most commonly a consequence of myeloablative chemoirradiation or ingestion of pyrrolizidine alkaloids such as monocrotaline (Mct). This study examines whether SECs are of bone marrow origin, whether bone marrow repair can be a determinant of severity of liver injury, and whether treatment with progenitor cells is beneficial. METHODS: Mct-treated female rats received infusion of male whole bone marrow or CD133(+) cells at the peak of sinusoidal injury. The Y chromosome was identified in isolated SECs by fluorescent in situ hybridization. Bone marrow suppression was induced by irradiation of both lower extremities with shielding of the abdomen. RESULTS: SECs in uninjured liver have both hematopoietic (CD45, CD33) and endothelial (CD31) markers. After Mct-induced SOS, infusion of bone marrow-derived CD133(+) progenitor cells replaces more than one quarter of SECs. All CD133(+) cells recovered from the SEC fraction after injury are CD45(+). CD133(+)/CD45(+) progenitors also repaired central vein endothelium. Mct suppresses CD133(+)/CD45(+) progenitors in bone marrow by 50% and in the circulation by 97%. Irradiation-induced bone marrow suppression elicited SOS from a subtoxic dose of Mct, whereas infusion of bone marrow during the necrotic phase of SOS nearly eradicates histologic features of SOS. CONCLUSIONS: SECs have both hematopoietic and endothelial markers. Bone marrow-derived CD133(+)/CD45(+) progenitors replace SECs and central vein endothelial cells after injury. Toxicity to bone marrow progenitors impairs repair and contributes to the pathogenesis of SOS, whereas timely infusion of bone marrow has therapeutic benefit.


Assuntos
Transplante de Medula Óssea/métodos , Células Endoteliais/fisiologia , Hepatopatia Veno-Oclusiva/patologia , Hepatopatia Veno-Oclusiva/terapia , Células-Tronco/fisiologia , Análise de Variância , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Biomarcadores/análise , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/citologia , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Regeneração Hepática/fisiologia , Masculino , Probabilidade , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Sensibilidade e Especificidade , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...