Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Gastrointest Liver Physiol ; 323(2): G114-G125, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35727919

RESUMO

Patients with acute-on-chronic liver failure (ACLF) are at risk of developing acute hepatic decompensation and organ failures with an unraveled complex mechanism. An altered immune response toward insults in cirrhotic compared with healthy livers may contribute to the ACLF development. Therefore, we aim to investigate the differences in inflammatory responses between cirrhotic and healthy livers using human precision-cut liver slices (PCLSs) upon the lipopolysaccharide (LPS) challenge. PCLSs prepared from livers of patients with cirrhosis or healthy donors of liver transplantation were incubated ex vivo with or without LPS for up to 48 h. Viability test, qRT-PCR, and multiplex cytokine assay were performed. Regulation of the LPS receptors during incubation or with LPS challenge differed between healthy versus cirrhotic PCLSs. LPS upregulated TLR-2 in healthy PCLSs solely (P < 0.01). Culturing for 48 h induced a stronger inflammatory response in the cirrhotic than healthy PCLS. Upon LPS stimulation, cirrhotic PCLSs secreted more proinflammatory cytokines (IL-8, IL-6, TNF-α, eotaxin, and VEGF) significantly and less anti-inflammatory cytokine (IL-1ra) than those of healthy. In summary, cirrhotic PCLSs released more proinflammatory and less anti-inflammatory cytokines after LPS stimuli than healthy, leading to dysregulated inflammatory response. These events could possibly resemble the liver immune response in ACLF.NEW & NOTEWORTHY Precision-cut liver slices (PCLSs) model provides a unique platform to investigate the different immune responses of healthy versus cirrhotic livers in humans. Our data show that cirrhotic PCLSs exhibit excessive inflammatory response accompanied by a lower anti-inflammatory cytokine release in response to LPS; a better understanding of this alteration may guide the novel therapeutic approaches to mitigate the excessive inflammation during the onset of acute-on-chronic liver failure.


Assuntos
Insuficiência Hepática Crônica Agudizada , Citocinas , Humanos , Lipopolissacarídeos/farmacologia , Fígado , Cirrose Hepática
2.
Cell Physiol Biochem ; 56(1): 28-38, 2022 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-35060690

RESUMO

BACKGROUND/AIMS: Osteoprotegerin (OPG) is a profibrotic mediator produced by myofibro-blasts under influence of transforming growth factor ß (TGFß). Its expression in experimental models of liver fibrosis correlates well with disease severity and treatment responses. The regulation of OPG in liver tissue is largely unknown and we therefore set out to elucidate which growth factors/interleukins associated with fibrosis induce OPG and through which pathways. METHODS: Precision-cut liver slices of wild type and STAT6-deficient mice and 3T3 fibroblasts were used to investigate the effects of TGFß, interleukin (IL) 13 (IL13), IL1ß, and platelet-derived growth factor BB (PDGF-BB) on expression of OPG. OPG protein was measure by ELISA, whereas OPG mRNA and expression of other relevant genes was measured by qPCR. RESULTS: In addition to TGFß, only IL13 and not PDGF-BB or IL1ß could induce OPG expression in 3T3 fibroblasts and liver slices. This IL13-dependent induction was not shown in liver slices of STAT6-deficient mice and when wild type slices were cotreated with TGFß receptor 1 kinase inhibitor galunisertib, STAT6 inhibitor AS1517499, or AP1 inhibitor T5224. This suggests that the OPG-inducing effect of IL13 is mediated through IL13 receptor α1-activation and subsequent STAT6-dependent upregulation of IL13 receptor α2, which in turn activates AP1 and induces production of TGFß and subsequent production of OPG. CONCLUSION: We have shown that IL13 induces OPG release by liver tissue through a TGFß-dependent pathway involving both the α1 and the α2 receptor of IL13 and transcription factors STAT6 and AP1. OPG may therefore be a novel target for the treatment liver fibrosis as it is mechanistically linked to two important regulators of fibrosis in liver, namely IL13 and TGFß1.


Assuntos
Regulação da Expressão Gênica , Interleucina-13/metabolismo , Cirrose Hepática/metabolismo , Fígado/metabolismo , Osteoprotegerina/biossíntese , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Masculino , Camundongos
3.
Am J Physiol Gastrointest Liver Physiol ; 312(3): G219-G227, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28057611

RESUMO

WNT-5A is a secreted growth factor that belongs to the noncanonical members of the Wingless-related MMTV-integration family. Previous studies pointed to a connection between WNT-5A and the fibrogenic factor TGF-ß warranting further studies into the functional role of WNT-5A in liver fibrosis. Therefore, we studied WNT-5A expressions in mouse and human fibrotic livers and examined the relation between WNT-5A and various fibrosis-associated growth factors, cytokines, and extracellular matrix proteins. WNT-5A gene and protein expressions were significantly increased in fibrotic mouse and human livers compared with healthy livers. Regression or therapeutic intervention in mice resulted in decreased hepatic WNT-5A levels paralleled by lower collagen levels. Immunohistochemical analysis showed WNT-5A staining in fibrotic septa colocalizing with desmin staining indicating WNT-5A expression in myofibroblasts. In vitro studies confirmed WNT-5A expression in this cell type and showed that TGF-ß significantly enhanced WNT-5A expression in contrast to PDGF-BB and proinflammatory cytokines IL-1ß and TNF-α. Additionally, TGF-ß induces the expression of the WNT receptors FZD2 and FZD8. After silencing of WNT-5A, reduced levels of collagen type I, vimentin, and fibronectin in TGF-ß-stimulated myofibroblasts were measured compared with nonsilencing siRNA-treated controls. Interestingly, the antifibrotic cytokine IFNγ suppressed WNT-5A in vitro and in vivo. IFNγ-treated fibrotic mice showed significantly less WNT-5A expression compared with untreated fibrotic mice. In conclusion, WNT-5A paralleled collagen I levels in fibrotic mouse and human livers. WNT-5A expression in myofibroblasts is induced by the profibrotic factor TGF-ß and plays an important role in TGF-ß-induced regulation of fibrotic matrix proteins, whereas its expression can be reversed upon treatment, both in vitro and in vivo.NEW & NOTEWORTHY This study describes the localization and functional role of WNT-5A in human and mouse fibrotic livers. Hepatic WNT-5A expression parallels collagen type I expression. In vivo and in vitro, the myofibroblasts were identified as the key hepatic cells producing WNT-5A. WNT-5A is under control of TGF-ß and its activities are primarily profibrotic.


Assuntos
Cirrose Hepática/metabolismo , Fígado/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína Wnt-5a/metabolismo , Animais , Linhagem Celular , Colágeno/metabolismo , Desmina/metabolismo , Inativação Gênica , Humanos , Interferon gama/farmacologia , Interleucina-1beta/metabolismo , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática/patologia , Camundongos , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Wnt-5a/genética
4.
J Pharmacol Exp Ther ; 363(2): 126-135, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28864467

RESUMO

Exchange protein activated by cAMP (Epac-1) is an important signaling mechanism for cAMP-mediated effects, yet factors that change Epac-1 levels are unknown. Such factors are relevant because it has been postulated that Epac-1 directly affects fibrogenesis. Prostaglandin E2 (PGE2) is a well-known cAMP activator, and we therefore studied the effects of this cyclo-oxygenase product on Epac-1 expression and on fibrogenesis within the liver. Liver fibrosis was induced by 8 weeks carbon tetrachloride (CCL4) administration to mice. In the last 2 weeks, mice received vehicle, PGE2, the cyclo-oxygenase-2 inhibitor niflumic acid (NFA), or PGE2 coupled to cell-specific carriers to hepatocytes, Kupffer cells, or hepatic stellate cells (HSC). Results showed antifibrotic effects of PGE2 and profibrotic effects of NFA in CCL4 mice. Western blot analysis revealed reduced Epac-1 protein expression in fibrotic livers of mice and humans compared with healthy livers. PGE2 administration to fibrotic mice completely restored intrahepatic Epac-1 levels and also led to reduced Rho kinase activity, a downstream target of Epac-1. Cell-specific delivery of PGE2 to either hepatocytes, Kupffer cells, or HSC identified the latter cell as the key player in the observed effects on Epac-1 and Rho kinase. No significant alterations in protein kinase A expressions were found. In primary isolated HSC, PGE2 elicited Rap1 translocation reflecting Epac-1 activation, and Epac-1 agonists attenuated platelet-derived growth factor-induced proliferation and migration of these cells. These studies demonstrate that PGE2 enhances Epac-1 activity in HSC, which is associated with significant changes in (myo)fibroblast activities in vitro and in vivo. Therefore, Epac-1 is a potential target for antifibrotic drugs.


Assuntos
Dinoprostona/farmacologia , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/prevenção & controle , Regulação para Cima/fisiologia , Adolescente , Adulto , Idoso , Animais , Células Cultivadas , Criança , Dinoprostona/uso terapêutico , Feminino , Células Hep G2 , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/patologia , Humanos , Cirrose Hepática/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Células NIH 3T3 , Ratos , Ratos Wistar , Regulação para Cima/efeitos dos fármacos , Adulto Jovem
5.
J Hepatol ; 57(6): 1220-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22878469

RESUMO

BACKGROUND & AIMS: Rho-kinase activation mediates cell contraction and increases intrahepatic resistance and consequently portal pressure in liver cirrhosis. Systemic Rho-kinase inhibition decreases portal pressure in cirrhosis, but also arterial pressure. Thus, liver-specific Rho-kinase inhibition is needed. The delivery of Rho-kinase inhibitor to activated hepatic stellate cells reduces fibrosis. It might also relax these contractile cells and therewith decrease intrahepatic resistance. We tested this hypothesis by performing acute experiments in cirrhotic rats. METHODS: Cirrhosis models were CCl(4)-intoxication and bile duct ligation. Three hours after injection of the Rho-kinase inhibitor (Y26732) coupled with a carrier (mannose-6-phosphate modified human serum albumin), which targets activated hepatic stellate cells, hemodynamics were analyzed by the colored microsphere technique and direct pressure measurements. The delivery site and effect of Rho-kinase inhibitor were investigated by immunohistochemical stainings, as well as Western blot. Experiments with Rho-kinase inhibitor coupled with unmodified human serum albumin served as untargeted control. RESULTS: In both models of cirrhosis, the carrier coupled Rho-kinase inhibitor lowered the portal pressure and decreased the hepatic-portal resistance. Immunohistochemical desmin-staining showed the carrier in hepatic stellate cells. The targeted therapy decreased the expression of the phosphorylated substrate of Rho-kinase (moesin) and abolished myosin light chains phosphorylation in fibrotic septae (collagen-staining). The targeted Rho-kinase inhibitor showed no major extrahepatic effects. By contrast, the untargeted Rho-kinase inhibitor elicited severe systemic hypotension. CONCLUSIONS: Activated hepatic stellate cells are crucially involved in portal hypertension in cirrhosis. Targeting of Rho-kinase in hepatic stellate cells not only decreased fibrosis, as previously shown, but also lowers portal pressure acutely without major systemic effects as demonstrated in this study.


Assuntos
Amidas/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Cirrose Hepática Experimental/tratamento farmacológico , Pressão na Veia Porta/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Animais , Células Estreladas do Fígado/enzimologia , Cirrose Hepática Experimental/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley , Quinases Associadas a rho/metabolismo
6.
Hepatology ; 54(2): 586-96, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21538439

RESUMO

UNLABELLED: Liver fibrogenesis is a process tightly controlled by endogenous anti- and pro-fibrogenic factors. Interferon gamma (IFNγ) is a potent antifibrogenic cytokine in vitro and might therefore represent a powerful therapeutic entity. However, its poor pharmacokinetics and adverse effects, due to the presence of IFNγ receptors on nearly all cells, prevented its clinical application so far. We hypothesized that delivery of IFNγ specifically to the disease-inducing cells and concurrently avoiding its binding to nontarget cells might increase therapeutic efficacy and avoid side effects. We conjugated IFNγ to a cyclic peptide recognizing the platelet-derived growth factor beta receptor (PDGFßR) which is strongly up-regulated on activated hepatic stellate cells (HSC), the key effector cells responsible for hepatic fibrogenesis. The IFNγ conjugates were analyzed in vitro for PDGFßR-specific binding and biological effects and in vivo in acute (early) and chronic (progressive and established) carbon-tetrachloride-induced liver fibrosis in mice. The targeted-IFNγ construct showed PDGFßR-specific binding to fibroblasts and HSC and inhibited their activation in vitro. In vivo, the targeted-IFNγ construct attenuated local HSC activation in an acute liver injury model. In the established liver fibrosis model, it not only strongly inhibited fibrogenesis but also induced fibrolysis. In contrast, nontargeted IFNγ was ineffective in both models. Moreover, in contrast to unmodified IFNγ, our engineered targeted-IFNγ did not induce IFNγ-related side effects such as systemic inflammation, hyperthermia, elevated plasma triglyceride levels, and neurotropic effects. CONCLUSION: This study presents a novel HSC-targeted engineered-IFNγ, which in contrast to systemic IFNγ, blocked liver fibrogenesis and is devoid of side effects, by specifically acting on the key pathogenic cells within the liver.


Assuntos
Interferon gama/uso terapêutico , Cirrose Hepática/prevenção & controle , Terapia de Alvo Molecular , Animais , Células Cultivadas , Células Estreladas do Fígado , Humanos , Camundongos
7.
Hepatology ; 51(3): 942-52, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20044807

RESUMO

UNLABELLED: There is no effective therapy for advanced liver fibrosis. Angiotensin type 1 (AT1) receptor blockers attenuate liver fibrogenesis, yet their efficacy in reversing advanced fibrosis is unknown. We investigated whether the specific delivery of an AT1 receptor blocker to activated hepatic stellate cells (HSCs) reduces established liver fibrosis. We used a platinum-based linker to develop a conjugate of the AT1 receptor blocker losartan and the HSC-selective drug carrier mannose-6-phosphate modified human serum albumin (losartan-M6PHSA). An average of seven losartan molecules were successfully coupled to M6PHSA. Rats with advanced liver fibrosis due to prolonged bile duct ligation or carbon tetrachloride administration were treated with daily doses of saline, losartan-M6PHSA, M6PHSA or oral losartan during 3 days. Computer-based morphometric quantification of inflammatory cells (CD43), myofibroblasts (smooth muscle alpha-actin [alpha-SMA]) and collagen deposition (Sirius red and hydroxyproline content) were measured. Hepatic expression of procollagen alpha2(I) and genes involved in fibrogenesis was assessed by quantitative polymerase chain reaction. Losartan-M6PHSA accumulated in the fibrotic livers and colocalized with HSCs, as assessed by immunostaining of anti-HSA and anti-alpha-SMA. Losartan-M6PHSA, but not oral losartan, reduced collagen deposition, accumulation of myofibroblasts, inflammation and procollagen alpha2(I) gene expression. Losartan-M6PHSA did not affect metalloproteinase type 2 and 9 activity and did not cause apoptosis of activated HSCs. CONCLUSION: Short-term treatment with HSC-targeted losartan markedly reduces advanced liver fibrosis. This approach may provide a novel means to treat chronic liver diseases.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/patologia , Losartan/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/administração & dosagem , Animais , Células Cultivadas , Progressão da Doença , Sistemas de Liberação de Medicamentos , Losartan/administração & dosagem , Masculino , Ratos , Ratos Wistar , Fatores de Tempo
8.
Mol Pharm ; 8(5): 1899-909, 2011 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-21800888

RESUMO

Excessive accumulation of the extracellular matrix proteins primarily produced by activated hepatic stellate cells (HSC) leads to liver fibrosis. To date, no successful therapeutic intervention is available for the treatment of this disease. Platelet derived growth factor beta receptor (PDGFßR) is highly upregulated on disease-inducing activated HSC and thus can be used for delivery of antifibrotic drugs to increase therapeutic efficacy with reduced adverse effects. Interferon gamma (IFNγ) has been recognized as a potent antifibrotic cytokine; however, poor pharmacokinetics and side effects due to frequent administration have limited its clinical use. For HSC-specific delivery, a PDGFßR-specific drug delivery carrier (PPB-HSA) was developed by modifying albumin with PDGFßR-recognizing cyclic peptides. Subsequently, IFNγ was conjugated to PPB-HSA via bifunctional PEG linkers to synthesize PPB-HSA-PEG-IFNγ. In vitro, PPB-HSA-PEG-IFNγ retained complete biological activity similar to unmodified IFNγ and showed PDGFßR-specific binding to human HSC and primary culture-activated rat HSC. In TGFß-stimulated mouse fibroblasts and human HSC, PPB-HSA-PEG-IFNγ induced significant reduction in crucial fibrotic parameters. In vivo, the conjugate rapidly accumulated into PDGFßR-expressing HSC in fibrotic livers and activated IFNγ-mediated pstat1α signaling pathway. Furthermore, in a CCl(4)-induced acute liver injury model in mice, treatment with HSC-targeted IFNγ strongly ameliorated hepatic fibrogenesis by inducing significant reduction (about 60%; p < 0.01) in collagen I and α-SMA expression as well as enhanced fibrolysis (increased MMP/TIMP ratio; p < 0.05) while free unmodified IFNγ was ineffective. Furthermore, in contrast to free native IFNγ, the conjugate did not induce macrophage infiltration and IL-1ß expression in the liver. In conclusion, these data demonstrate the enhanced antifibrotic efficacy and reduced off-target effects of PPB-HSA-PEG-IFNγ conjugate showing the potential of cell-specific targeting of IFNγ for the treatment of liver fibrosis.


Assuntos
Portadores de Fármacos/uso terapêutico , Células Estreladas do Fígado/metabolismo , Interferon gama/administração & dosagem , Cirrose Hepática/tratamento farmacológico , Peptídeos Cíclicos/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Albumina Sérica/química , Animais , Transporte Biológico , Linhagem Celular , Células Cultivadas , Reagentes de Ligações Cruzadas/química , Portadores de Fármacos/efeitos adversos , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Feminino , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/patologia , Humanos , Interferon gama/química , Interferon gama/farmacologia , Interferon gama/uso terapêutico , Ligantes , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos Cíclicos/química , Fator de Transcrição STAT1/metabolismo , Albumina Sérica Humana , Transdução de Sinais/efeitos dos fármacos , Distribuição Tecidual
9.
Pharm Res ; 28(8): 2045-54, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21442374

RESUMO

PURPOSE: Rho-kinase regulates activation of hepatic stellate cells (HSC) during liver fibrosis, but the ubiquitous presence of this kinase may hinder examination of its exact role and the therapeutic use of inhibitors. We therefore coupled the Rho-kinase inhibitor Y27632 to a drug carrier that binds the mannose-6-phosphate insulin-like growth factor II (M6P/IGFII)-receptor which is upregulated on activated HSC. METHODS: Y27632 was coupled to mannose-6-phosphate human serum albumin (M6PHSA), and in vitro experiments were performed on primary rat HSC. Biodistribution and effect studies were performed in an acute CCl(4) model in mice. RESULTS: Y27-conjugate remained stable in serum, while drug was efficiently released in liver homogenates. Receptor-blocking studies revealed that it was specifically taken up through the M6P/IGFII-receptor on fibroblasts, and it inhibited expression of fibrotic markers in activated HSC. In vivo, liver drug levels were significantly higher after injection of Y27-conjugate as compared to Y27632, and the conjugate accumulated specifically in HSC. After acute CCl(4)-induced liver injury, Y27-conjugate reduced the local activation of HSC, whereas an equimolar dose of free drug did not. CONCLUSIONS: We conclude that specific targeting of a Rho-kinase inhibitor to HSC leads to enhanced accumulation of the drug in HSC, reducing early fibrogenesis in the liver.


Assuntos
Amidas/farmacologia , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Fígado/metabolismo , Piridinas/farmacologia , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/metabolismo , Amidas/química , Animais , Intoxicação por Tetracloreto de Carbono/metabolismo , Células Cultivadas , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/farmacocinética , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/enzimologia , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Cirrose Hepática/induzido quimicamente , Masculino , Manosefosfatos/administração & dosagem , Manosefosfatos/química , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Piridinas/química , Ratos , Ratos Wistar , Receptor IGF Tipo 2/química , Receptor IGF Tipo 2/metabolismo , Albumina Sérica/química , Albumina Sérica/metabolismo , Distribuição Tecidual
10.
Int J Cancer ; 126(8): 1966-1981, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19795464

RESUMO

Tumor-targeting of anticancer drugs is an interesting approach for the treatment of cancer since chemotherapies possess several adverse effects. In the present study, we propose a novel strategy to deliver anticancer drugs to the tumor cells through the mannose-6-phosphate/insulin-like growth factor receptor (M6P/IGF-IIR) which are abundantly expressed in several human tumors. We developed a drug carrier against M6P/IGF-II receptor by modifying human serum albumin (HSA) with M6P moieties. M6P-HSA specifically bound and internalized into M6P/IGF-IIR-expressing B16 melanoma cells as demonstrated with radioactive studies and anti-HSA immunostaining. In vivo, M6P-HSA rapidly accumulated in subcutaneous tumors in tumor and stromal components after an intravenous injection. To demonstrate the application of M6P-HSA as a drug carrier, we coupled doxorubicin to it. Dox-HSA-M6P conjugate could release doxorubicin at lysosomal pH and showed M6P-specific binding and uptake in tumor cells. In vitro, a short exposure with Dox-HSA-M6P induced killing of tumor cells, which could be blocked by excess M6P-HSA. In vivo, Dox-HSA-M6P distributed to tumors and some other organs while free doxorubicin distributed to all organs but slightly to tumors. In B16 tumor-bearing mice, Dox-HSA-M6P significantly inhibited the tumor growth whereas an equimolar dose of free doxorubicin did not show any anti-tumor effect. In addition, targeted doxorubicin did not show any side-effects on liver and kidney function tests, body weight and blood cell counts. In conclusion, M6P-HSA is a suitable carrier for delivery of anticancer drugs to tumors through M6P/IGF-IIR. Improved antitumor effects of the targeted doxorubicin by M6P-HSA suggest that this novel approach may be applied to improve the therapeutic efficacy of anticancer drugs.


Assuntos
Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Manosefosfatos/administração & dosagem , Neoplasias Experimentais/tratamento farmacológico , Receptor IGF Tipo 2/metabolismo , Albumina Sérica/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Western Blotting , Linhagem Celular Tumoral , Doxorrubicina/farmacocinética , Portadores de Fármacos/farmacologia , Sistemas de Liberação de Medicamentos , Imunofluorescência , Humanos , Imuno-Histoquímica , Manosefosfatos/farmacocinética , Camundongos , Albumina Sérica/farmacocinética
11.
Front Med (Lausanne) ; 7: 617261, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33409288

RESUMO

Biliary atresia (BA) is a rare cholangiopathy of infancy in which the bile ducts obliterate, leading to profound cholestasis and liver fibrosis. BA is hypothesized to be caused by a viral insult that leads to over-activation of the immune system. Patients with BA are surgically treated with a Kasai portoenterostomy (KPE), which aims to restore bile flow from the liver to the intestines. After KPE, progressive liver fibrosis is often observed in BA patients, even despite surgical success and clearance of their jaundice. The innate immune response is involved during the initial damage to the cholangiocytes and further differentiation of the adaptive immune response into a T-helper 1 cell (Th1) response. Multiple studies have shown that there is continuing elevation of involved cytokines that can lead to the progressive liver fibrosis. However, the mechanism by which the progressive injury occurs is not fully elucidated. Recently, matrix metalloproteinase-7 (MMP-7) has been investigated to be used as a biomarker to diagnose BA. MMPs are involved in extracellular matrix (ECM) turnover, but also have non-ECM related functions. The role of MMP-7 and other MMPs in liver fibrosis is just starting to be elucidated. Multiple studies have shown that serum MMP-7 measurements are able to accurately diagnose BA in a cohort of cholestatic patients while hepatic MMP-7 expression correlated with BA-related liver fibrosis. While the mechanism by which MMP-7 can be involved in the pathophysiology of BA is unclear, MMP-7 has been investigated in other fibrotic pathologies such as renal and idiopathic pulmonary fibrosis. MMP-7 is involved in Wnt/ß-catenin signaling, reducing cell-to-cell contact by shedding of E-cadherin, amplifying inflammation and fibrosis via osteopontin (OPN) and TNF-α while it also appears to play a role in induction of angiogenesis This review aims to describe the current understandings of the pathophysiology of BA. Subsequently, we describe how MMP-7 is involved in other pathologies, such as renal and pulmonary fibrosis. Then, we propose how MMP-7 can potentially be involved in BA. By doing this, we aim to describe the putative role of MMP-7 as a prognostic biomarker in BA and to provide possible new therapeutic and research targets that can be investigated in the future.

12.
Pharmaceutics ; 12(3)2020 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-32244897

RESUMO

The pivotal cell involved in the pathogenesis of liver fibrosis, i.e., the activated hepatic stellate cell (HSC), has a wide range of activities during the initiation, progression and even regression of the disease. These HSC-related activities encompass cellular activation, matrix synthesis and degradation, proliferation, contraction, chemotaxis and inflammatory signaling. When determining the in vitro and in vivo effectivity of novel antifibrotic therapies, the readout is currently mainly based on gene and protein levels of α-smooth muscle actin (α-SMA) and the fibrillar collagens (type I and III). We advocate for a more comprehensive approach in addition to these markers when screening potential antifibrotic drugs that interfere with HSCs. Therefore, we aimed to develop a gene panel for human in vitro and ex vivo drug screening models, addressing each of the HSC-activities with at least one gene, comprising, in total, 16 genes. We determined the gene expression in various human stellate cells, ranging from primary cells to cell lines with an HSC-origin, and human liver slices and stimulated them with two key profibrotic factors, i.e., transforming growth factor ß (TGFß) or platelet-derived growth factor BB (PDGF-BB). We demonstrated that freshly isolated HSCs showed the strongest and highest variety of responses to these profibrotic stimuli, in particular following PDGF-BB stimulation, while cell lines were limited in their responses. Moreover, we verified these gene expression profiles in human precision-cut liver slices and showed similarities with the TGFß- and PDGF-BB-related fibrotic responses, as observed in the primary HSCs. With this study, we encourage researchers to get off the beaten track when testing antifibrotic compounds by including more HSC-related markers in their future work. This way, potential compounds will be screened more extensively, which might increase the likelihood of developing effective antifibrotic drugs.

13.
Cells ; 9(12)2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33348845

RESUMO

Alkaline phosphatase (AP) activity is highly upregulated in plasma during liver diseases. Previously, we demonstrated that AP is able to detoxify lipopolysaccharide (LPS) by dephosphorylating its lipid A moiety. Because a role of gut-derived LPS in liver fibrogenesis has become evident, we now examined the relevance of phosphate groups in the lipid A moiety in this process. The effects of mono-phosphoryl and di-phosphoryl lipid A (MPLA and DPLA, respectively) were studied in vitro and LPS-dephosphorylating activity was studied in normal and fibrotic mouse and human livers. The effects of intestinal AP were studied in mice with CCL4-induced liver fibrosis. DPLA strongly stimulated fibrogenic and inflammatory activities in primary rat hepatic stellate cells (rHSCs) and RAW264.7 macrophages with similar potency as full length LPS. However, MPLA did not affect any of the parameters. LPS-dephosphorylating activity was found in mouse and human livers and was strongly increased during fibrogenesis. Treatment of fibrotic mice with intravenous intestinal-AP significantly attenuated intrahepatic desmin+- and αSMA+ -HSC and CD68+- macrophage accumulation. In conclusion, the lack of biological activity of MPLA, contrasting with the profound activities of DPLA, shows the relevance of LPS-dephosphorylating activity. The upregulation of LPS-dephosphorylating activity in fibrotic livers and the protective effects of exogenous AP during fibrogenesis indicate an important physiological role of intestinal-derived AP during liver fibrosis.


Assuntos
Células Estreladas do Fígado/efeitos dos fármacos , Lipídeo A/metabolismo , Lipopolissacarídeos/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Células Estreladas do Fígado/citologia , Células Estreladas do Fígado/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Óxido Nítrico/metabolismo , Fosforilação/efeitos dos fármacos , Células RAW 264.7 , Ratos , Regulação para Cima/efeitos dos fármacos
14.
Chem Commun (Camb) ; 56(44): 5941-5944, 2020 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-32347235

RESUMO

Targeted epitope-based mass spectrometry imaging (MSI) utilizes laser cleavable mass-tags bound to targeting moieties for detecting proteins in tissue sections. Our work constitutes the first proof-of-concept of a novel laser desorption ionization (LDI)-MSI strategy using photocleavable Ru(ii) polypyridine complexes as mass-tags for imaging of integrins αvß3 in human cancer tissues.


Assuntos
Neoplasias de Cabeça e Pescoço/metabolismo , Integrina alfaVbeta3/metabolismo , Peptídeos Cíclicos/farmacologia , Piridinas/farmacologia , Rutênio/farmacologia , Humanos , Espectrometria de Massas/métodos , Peptídeos Cíclicos/química , Piridinas/química , Rutênio/química
15.
Pharmaceutics ; 12(5)2020 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-32455750

RESUMO

Osteoprotegerin (OPG) serum levels are associated with liver fibrogenesis and have been proposed as a biomarker for diagnosis. However, the source and role of OPG in liver fibrosis are unknown, as is the question of whether OPG expression responds to treatment. Therefore, we aimed to elucidate the fibrotic regulation of OPG production and its possible function in human and mouse livers. OPG levels were significantly higher in lysates of human and mouse fibrotic livers compared to healthy livers. Hepatic OPG expression localized in cirrhotic collagenous bands in and around myofibroblasts. Single cell sequencing of murine liver cells showed hepatic stellate cells (HSC) to be the main producers of OPG in healthy livers. Using mouse precision-cut liver slices, we found OPG production induced by transforming growth factor ß1 (TGFß1) stimulation. Moreover, OPG itself stimulated expression of genes associated with fibrogenesis in liver slices through TGFß1, suggesting profibrotic activity of OPG. Resolution of fibrosis in mice was associated with decreased production of OPG compared to ongoing fibrosis. OPG may stimulate fibrogenesis through TGFß1 and is associated with the degree of fibrogenesis. It should therefore be investigated further as a possible drug target for liver fibrosis or biomarker for treatment success of novel antifibrotics.

17.
Sci Rep ; 9(1): 2256, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30783172

RESUMO

Rho-kinase (ROCK) activation in hepatic stellate cells (HSC) is a key mechanism promoting liver fibrosis and portal hypertension (PTH). Specific delivery of ROCK-inhibitor Y-27632 (Y27) to HSC targeting mannose-6-phosphate-receptors reduces portal pressure and fibrogenesis. In decompensated cirrhosis, presence of ascites is associated with reduced renal perfusion. Since in cirrhosis, platelet-derived growth factor receptor beta (PDGFRß) is upregulated in the liver as well as the kidney, this study coupled Y27 to human serum albumin (HSA) substituted with PDGFRß-recognizing peptides (pPB), and investigated its effect on PTH in cirrhotic rats. In vitro collagen contraction assays tested biological activity on LX2 cells. Hemodynamics were analyzed in BDL and CCl4 cirrhotic rats 3 h, 6 h and 24 h after i.v. administration of Y27pPBHSA (0.5/1 mg/kg b.w). Phosphorylation of moesin and myosin light chain (MLC) assessed ROCK activity in liver, femoral muscle, mesenteric artery, kidney and heart. Three Y27 molecules were coupled to pPBHSA as confirmed by HPLC/MS, which was sufficient to relax LX2 cells. In vivo, Y27pPBHSA-treated rats exhibited lower portal pressure, hepatic vascular resistance without effect on systemic vascular resistance, but a tendency towards lower cardiac output compared to non-treated cirrhotic rats. Y27pPBHSA reduced intrahepatic resistance by reduction of phosphorylation of moesin and MLC in Y27pPBHSA-treated cirrhotic rats. Y27pPBHSA was found in the liver of rats up to 6 hours after its injection, in the HSC demonstrated by double-immunostainings. Interestingly, Y27pPBHSA increased renal arterial flow over time combined with an antifibrotic effect as shown by decreased renal acta2 and col1a1 mRNA expression. Therefore, targeting the ROCK inhibitor Y27 to PDGFRß decreases portal pressure with potential beneficial effects in the kidney. This unique approach should be tested in human cirrhosis.


Assuntos
Portadores de Fármacos , Inibidores Enzimáticos , Rim/irrigação sanguínea , Cirrose Hepática , Pressão na Veia Porta/efeitos dos fármacos , Albumina Sérica Humana , Quinases Associadas a rho/antagonistas & inibidores , Animais , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Humanos , Rim/metabolismo , Rim/patologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Perfusão , Ratos , Ratos Sprague-Dawley , Albumina Sérica Humana/química , Albumina Sérica Humana/farmacologia , Quinases Associadas a rho/metabolismo
18.
Antivir Ther ; 12(2): 273-8, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17503670

RESUMO

BACKGROUND: Succinylated human serum albumin (Suc-HAS) is a negatively charged neo-glycoprotein that binds to the positively charged V3-loop of HIV-1 gp120, acting as HIV-1-fusion inhibitor in vitro (IC50: 0.5-5.0 microg/ml). Suc-HSA was safe in rats and monkeys, and showed antiretroviral effect in a human-to-mouse model. We evaluated safety and pharmacokinetics of single and multiple doses of Suc-HSA in HIV-1-infected individuals. METHODS: First, six untreated, chronically HIV-1-infected patients were randomized to a single dose of 1 or 10 mg/kg Suc-HSA intravenously. Second, five consecutive daily doses (10 mg/kg, based on the results of the single dose study) were given to four patients. Safety laboratory assessments, Suc-HSA plasma levels, plasma HIV-1 RNA (pVL), and CD4+ T-cell counts were determined. RESULTS: Increase of liver transaminases (grade 1/2) occurred in one of six patients in the single-dose phase and in three of four patients in the multiple-dosing phase. Suc-HSA plasma levels were undetectable 4 h after a single dose of 1 mg/kg. After a dose of 10 mg/kg, plasma levels were more sustained, but declined under the target plasma concentration (10 microg/ml) 12-24 h post-dosing. After multiple dosing, plasma levels reached peak values 2h post-dosing as predicted by our kinetic model. However, trough levels were below the target concentrations. There was no change in pVL or CD4+ T-cell count in either the single- or multiple-dosing phase. CONCLUSIONS: At the chosen dosing regimens, adequate antiviral plasma levels were not maintained, probably because the hepatic clearance was more rapid than expected. This may partially explain the lack of effect on pVL and CD4+ T-cell count. The observed liver transaminase increases prohibit further dose escalation.


Assuntos
Inibidores da Fusão de HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , HIV-1/genética , RNA Viral/sangue , Albumina Sérica/uso terapêutico , Succinatos/uso terapêutico , Adulto , Idoso , Alanina Transaminase/sangue , Aspartato Aminotransferases/sangue , Contagem de Linfócito CD4 , Doença Hepática Induzida por Substâncias e Drogas , Esquema de Medicação , Feminino , Inibidores da Fusão de HIV/administração & dosagem , Inibidores da Fusão de HIV/efeitos adversos , Inibidores da Fusão de HIV/farmacocinética , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Injeções Intravenosas , Hepatopatias/enzimologia , Masculino , Pessoa de Meia-Idade , Albumina Sérica/administração & dosagem , Albumina Sérica/efeitos adversos , Albumina Sérica/farmacocinética , Albumina Sérica Humana , Succinatos/administração & dosagem , Succinatos/efeitos adversos , Succinatos/farmacocinética , Falha de Tratamento , Carga Viral
19.
Biochem Pharmacol ; 73(9): 1455-62, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17239825

RESUMO

BACKGROUND/AIMS: Inhibition of hepatic stellate cell (HSC) proliferation is a relevant strategy to inhibit liver fibrosis. Coupling of antiproliferative drugs to the HSC-selective drug carrier mannose-6-phosphate-modified human serum albumin (M6PHSA) may lead to cell-selective inhibition of HSC proliferation. We coupled the antiproliferative drug doxorubicin (DOX) to this drug carrier and investigated the pharmacokinetics of this construct in a rat model of liver fibrosis, as well as in cultured HSC. METHODS/RESULTS: M6PHSA-DOX was cleared from the plasma in a biphasic manner. Upon i.v. injection of 4 microg kg(-1) (tracer), 2 and 20 mg kg(-1), the clearance in the distribution phase of drug disposition (CL(d)) significantly decreased from 9.7+/-0.7 to 4.7+/-2.3 and 1.0+/-0.1 ml kg(-1)min(-1), respectively. This indicates that saturation of clearance mechanisms occurs in this phase of drug disposition, likely reflecting saturable receptor-mediated uptake in the target cells. Gamma-camera studies revealed that the majority of the conjugate accumulated in the liver within 5 min, and immunohistochemical double-staining of liver sections demonstrated co-localization of the construct with HSC-markers. Simulation of the release of DOX from the carrier, after cellular uptake by HSC, showed that a gradual release of the drug takes place over a 9h period. Studies in cultured HSC illustrated that after 24h incubation with the conjugate, DOX was associated with the cell nucleus. CONCLUSIONS: The rapid distribution of M6PHSA-DOX from the blood to HSC, in combination with the expected gradual release of DOX within these cells, make this construct a promising tool for achieving sustained and selective inhibition of HSC proliferation.


Assuntos
Ductos Biliares/metabolismo , Doxorrubicina/farmacocinética , Hepatócitos/metabolismo , Ligadura , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Ductos Biliares/fisiopatologia , Modelos Animais de Doenças , Doxorrubicina/metabolismo , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos , Hepatócitos/citologia , Fígado/citologia , Masculino , Ratos , Ratos Wistar , Distribuição Tecidual
20.
J Control Release ; 111(1-2): 193-203, 2006 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-16466667

RESUMO

Targeting of antifibrotic drugs to hepatic stellate cells (HSC) is a promising strategy to block fibrotic processes leading to liver cirrhosis. For this purpose, we utilized the neo-glycoprotein mannose-6-phosphate-albumin (M6PHSA) that accumulates efficiently in HSC during liver fibrosis. Pentoxifylline (PTX), an antifibrotic compound that inhibits HSC proliferation and activation in vitro, was conjugated to M6PHSA. We employed a new type of platinum-based linker, which conjugates PTX via coordination chemistry rather than via covalent linkage. When incubated in plasma or in the presence of thiol compounds, free PTX was released from PTX-M6PHSA at a sustained slow rate. PTX-M6PHSA displayed pharmacological activity in cultured HSC as evidenced by changes in cell morphology and reduction of collagen I production. PTX-M6PHSA and platinum coupled PTX did not induce platinum-related toxicity (Alamar Blue viability assay) or apoptosis (caspase activation and TUNEL staining). In vivo distribution studies in fibrotic rats demonstrated specific accumulation of the conjugate in nonparenchymal cells in the fibrotic liver. In conclusion, we have developed PTX-M6PHSA employing a novel type of platinum linker, which allows sustained delivery of the drug to HSC in the fibrotic liver.


Assuntos
Química Farmacêutica/métodos , Fígado/metabolismo , Compostos Organoplatínicos/química , Pentoxifilina/farmacocinética , Actinas/análise , Animais , Disponibilidade Biológica , Caspase 3 , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Colágeno Tipo I/análise , Sistemas de Liberação de Medicamentos/métodos , Ativação Enzimática/efeitos dos fármacos , Fígado/química , Fígado/citologia , Cirrose Hepática/metabolismo , Cirrose Hepática/prevenção & controle , Masculino , Camundongos , Músculo Liso/química , Células NIH 3T3 , Pentoxifilina/química , Pentoxifilina/farmacologia , Monoéster Fosfórico Hidrolases/química , Ratos , Ratos Wistar , Tecnologia Farmacêutica/métodos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA