Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Small ; 19(23): e2207201, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36899444

RESUMO

Insufficienct T lymphocyte infiltration and unresponsiveness to immune checkpoint blockade therapy are still major difficulties for the clinical treatment of pancreatic ductal adenocarcinoma (PDAC). Although econazole has shown promise in inhibiting PDAC growth, its poor bioavailability and water solubility limit its potential as a clinical therapy for PDAC. Furthermore, the synergistic role of econazole and biliverdin in immune checkpoint blockade therapy in PDAC remains elusive and challenging. Herein, a chemo-phototherapy nanoplatform is designed by which econazole and biliverdin can be co-assembled (defined as FBE NPs), which significantly improve the poor water solubility of econazole and enhance the efficacy of PD-L1 checkpoint blockade therapy against PDAC. Mechanistically, econazole and biliverdin are directly released into the acidic cancer microenvironment, to activate immunogenic cell death via biliverdin-induced PTT/PDT and boost the immunotherapeutic response of PD-L1 blockade. In addition, econazole simultaneously enhances PD-L1 expression to sensitize anti-PD-L1 therapy, leading to suppression of distant tumors, long-term immune memory effects, improved dendritic cell maturation, and tumor infiltration of CD8+ T lymphocytes. The combined FBE NPs and α-PDL1 show synergistic antitumor efficacy. Collectively, FBE NPs show excellent biosafety and antitumor efficacy by combining chemo-phototherapy with PD-L1 blockade, which has promising potential in a precision medicine approach as a PDAC treatment strategy.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Econazol/uso terapêutico , Biliverdina/uso terapêutico , Inibidores de Checkpoint Imunológico/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Imunoterapia , Água , Microambiente Tumoral , Linhagem Celular Tumoral , Neoplasias Pancreáticas
2.
Int J Mol Sci ; 23(19)2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36233341

RESUMO

A diabetic foot ulcer (DFU) is one of the major complications of diabetes. Wound healing under diabetic conditions is often impaired. This is in part due to the excessive oxidative stress, prolonged inflammation, immune cell dysfunction, delayed re-epithelialization, and decreased angiogenesis present at the wound site. As a result of these multifactorial impaired healing pathways, it has been difficult to develop effective therapeutic strategies for DFU. Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme degradation generating carbon monoxide (CO), biliverdin (BV) which is converted into bilirubin (BR), and iron. HO-1 is a potent antioxidant. It can act as an anti-inflammatory, proliferative, angiogenic and cytoprotective enzyme. Due to its biological functions, HO-1 plays a very important role in wound healing, in part mediated through the biologically active end products generated by its enzymatic activity, particularly CO, BV, and BR. Therapeutic strategies involving the activation of HO-1, or the topical application of its biologically active end products are important in diabetic wound healing. Therefore, HO-1 is an attractive therapeutic target for DFU treatment. This review will provide an overview and discussion of the importance of HO-1 as a therapeutic target for diabetic wound healing.


Assuntos
Diabetes Mellitus , Pé Diabético , Anti-Inflamatórios , Antioxidantes , Biliverdina/metabolismo , Biliverdina/uso terapêutico , Monóxido de Carbono/metabolismo , Pé Diabético/tratamento farmacológico , Heme/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1/metabolismo , Humanos , Ferro/metabolismo
3.
Med Hypotheses ; 149: 110534, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33640714

RESUMO

Bilirubin has been proven to possess significant anti-inflammatory, antioxidant and antiviral activities. Recently, it has been postulated as a metabolic hormone. Further, moderately higher levels of bilirubin are positively associated with reduced risk of cardiovascular diseases, diabetes, metabolic syndrome and obesity. However, due to poor solubility the therapeutic delivery of bilirubin remains a challenge. Nanotechnology offers unique advantages which may be exploited for improved delivery of bilirubin to the target organ with reduced risk of systemic toxicity. Herein, we postulate the use of intravenous administration or inhalational delivery of bilirubin nanomedicine (BNM) to combat systemic dysfunctions associated with COVID-19, owing to the remarkable preclinical efficacy and optimistic results of various clinical studies of bilirubin in non-communicable disorders. BNM may be used to harness the proven preclinical pharmacological efficacy of bilirubin against COVID-19 related systemic complications.


Assuntos
Bilirrubina/uso terapêutico , COVID-19/terapia , Nanomedicina/métodos , Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Antivirais/uso terapêutico , Biliverdina/uso terapêutico , Síndrome da Liberação de Citocina , Humanos , Inflamação , Sistema de Sinalização das MAP Quinases , Modelos Teóricos , Subunidade p50 de NF-kappa B/metabolismo , Risco , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Tratamento Farmacológico da COVID-19
4.
Chin Med J (Engl) ; 130(7): 859-865, 2017 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-28345551

RESUMO

BACKGROUND: Biliverdin (BV) has a protective role against ischemia-reperfusion injury (IRI). However, the protective role and potential mechanisms of BV on lung IRI (LIRI) remain to be elucidated. Thus, we aimed to investigate the protective role and potential mechanisms of BV on LIRI. METHODS: Lungs were isolated from Sprague-Dawley rats to establish an ex vivo LIRI model. After an initial 15 min stabilization period, the isolated lungs were subjected to ischemia for 60 min, followed by 90 min of reperfusion with or without BV treatment. RESULTS: Lungs in the I/R group exhibited significant decrease in tidal volume (1.44 ± 0.23 ml/min in I/R group vs. 2.41 ± 0.31 ml/min in sham group; P< 0.001), lung compliance (0.27 ± 0.06 ml/cmH2O in I/R group vs. 0.44 ± 0.09 ml/cmH2O in sham group; P< 0.001; 1 cmH2O=0.098 kPa), and oxygen partial pressure (PaO2) levels (64.12 ± 12 mmHg in I/R group vs. 114 ± 8.0 mmHg in sham group; P< 0.001; 1 mmHg = 0.133 kPa). In contrast, these parameters in the BV group (2.27 ± 0.37 ml/min of tidal volume, 0.41 ± 0.10 ml/cmH2O of compliance, and 98.7 ± 9.7 mmHg of PaO2) were significantly higher compared with the I/R group (P = 0.004, P< 0.001, and P< 0.001, respectively). Compared to the I/R group, the contents of superoxide dismutase were significantly higher (47.07 ± 7.91 U/mg protein vs. 33.84 ± 10.15 U/mg protein; P = 0.005) while the wet/dry weight ratio (P < 0.01), methane dicarboxylic aldehyde (1.92 ± 0.25 nmol/mg protein vs. 2.67 ± 0.46 nmol/mg protein; P< 0.001), and adenosine triphosphate contents (297.05 ± 47.45 nmol/mg protein vs. 208.09 ± 29.11 nmol/mg protein; P = 0.005) were markedly lower in BV-treated lungs. Histological analysis revealed that BV alleviated LIRI. Furthermore, the expression of inflammatory cytokines (interleukin-1ß, interleukin-6, and tumor necrosis factor-ß) was downregulated and the expression of cyclooxygenase-2, inducible nitric oxide synthase, and Jun N-terminal kinase was significantly reduced in BV group (all P< 0.01 compared to I/R group). Finally, the apoptosis index in the BV group was significantly decreased (P < 0.01 compared to I/R group). CONCLUSION: BV protects lung IRI through its antioxidative, anti-inflammatory, and anti-apoptotic effects.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Biliverdina/uso terapêutico , Pulmão/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo-Oxigenase 2/metabolismo , Marcação In Situ das Extremidades Cortadas , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Pulmão/patologia , Linfotoxina-alfa/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Superóxido Dismutase/metabolismo
5.
Liver Int ; 37(8): 1128-1137, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27992676

RESUMO

BACKGROUND & AIMS: Enhancement of host anti-oxidant enzymes, such as haemoxygenase-1, may attenuate virus-mediated hepatocyte injury, while the induction of HO-1 by cobalt-protoporphyrin-IX (CoPP) administration, as the application of its haem degradation product biliverdin (BV), was shown to hinder HCV replication in vitro. In addition, (GT)n -repeats length in the polymorphic region of the HO-1 promoter may affect HO-1 expression and responsiveness to infection and disease severity. Aim of this study was to investigate the antiviral and hepatoprotective effects of CoPP-mediated HO-1 induction, alone or in combination with interferon alpha (peg-IFNα), in HCV-infected mice harbouring hepatocytes from donors with different HO-1-promoter polymorphisms. METHODS: Upon establishment of HCV infection, CoPP, BV and peg-IFNα were given alone or in combination. Viraemia changes and intrahepatic human gene expression were determined by qRT-PCR and immunohistochemistry. RESULTS: CoPP administration increased human HO-1 expression and significantly reduced viraemia, although changes correlated with promoter length (Δ0.5log and Δ2log reduction with medium- and short-polymorphism respectively). Polymorphisms did not influence BV-mediated antiviral effects (Δ1log). Notably, HO-1 induction attenuated basal HCV-driven enhancement of interferon genes and pro-inflammatory cytokines, both in cells with short- or medium-polymorphisms. Moreover, simultaneous administration of CoPP and peg-IFNα reduced viraemia even stronger (median 3log), whereas 1log viraemia reduction was determined in mice receiving peg-IFNα monotherapy. CONCLUSIONS: Although the protective function of HO-1 could be elicited in vivo with both host polymorphisms, the strength of HO-1 induction and suppression of HCV occurred in a polymorphism-dependent manner, indicating that host-genetic determinants may affect disease progression and infection outcome.


Assuntos
Heme Oxigenase-1/metabolismo , Hepacivirus/imunologia , Hepatite C/terapia , Animais , Antivirais/farmacologia , Antivirais/uso terapêutico , Biliverdina/farmacologia , Biliverdina/uso terapêutico , Heme Oxigenase-1/genética , Hepacivirus/efeitos dos fármacos , Hepatite C/genética , Hepatite C/virologia , Humanos , Interferon-alfa/farmacologia , Interferon-alfa/uso terapêutico , Camundongos , Polimorfismo Genético , Ativação Transcricional , Replicação Viral/efeitos dos fármacos
6.
Transl Res ; 178: 81-94.e2, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27497182

RESUMO

The main limitation to successful transplantation is the antigraft response developed by the recipient immune system, and the adverse side effects of immunosuppressive agents which are associated with significant toxicity and counter indications such as infection and cancer. Furthermore, immunosuppressants do little to prevent ischemia-reperfusion injury during the transplantation procedure itself hence there is a growing need to develop novel immunosuppressive drugs specifically aimed at prolonging graft survival. Linear tetrapyrroles derived from the breakdown of mammalian heme have been shown in numerous studies to play a protective role in allograft transplantation and ischemia-reperfusion injury; however, commercial sources of these products have not been approved for use in humans. Plants and algae produce equivalent linear tetrapyrroles called bilins that serve as chromophores in light-sensing. One such marine-derived tetrapyrrole, phycocyanobilin (PCB), shows significant structural similarity to mammalian biliverdin (BV) and may prove to be a safer alternative for use in the clinic if it can exert direct effects on human immune cells. Using a mixed lymphocyte reaction, we quantified the allogeneic responses of recipient cells to donor cells and found that PCB, like BV, effectively suppressed proliferation and proinflammatory cytokine production. In addition, we found that BV and PCB can directly downregulate the proinflammatory responses of both innate dendritic cells and adaptive T cells. We therefore propose that PCB may be an effective therapeutic drug in the clinical setting of transplantation and may also have wider applications in regulating inappropriate inflammation.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Tetrapirróis/farmacologia , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Biliverdina/farmacologia , Biliverdina/uso terapêutico , Complexo CD3/metabolismo , Proliferação de Células/efeitos dos fármacos , Citocinas/biossíntese , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Feminino , Humanos , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Inflamação/patologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Ficobilinas/farmacologia , Ficobilinas/uso terapêutico , Ficocianina/farmacologia , Ficocianina/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Transplante Homólogo
8.
PLoS One ; 8(7): e69972, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23922878

RESUMO

Ischemia reperfusion injury (IRI) in organ transplantation remains a serious and unsolved problem. Organs that undergo significant damage during IRI, function less well immediately after reperfusion and tend to have more problems at later times when rejection can occur. Biliverdin has emerged as an agent that potently suppress IRI in rodent models. Since the use of biliverdin is being developed as a potential therapeutic modality for humans, we tested the efficacy for its effects on IRI of the liver in swine, an accepted and relevant pre-clinical animal model. Administration of biliverdin resulted in rapid appearance of bilirubin in the serum and significantly suppressed IRI-induced liver dysfunction as measured by multiple parameters including urea and ammonia clearance, neutrophil infiltration and tissue histopathology including hepatocyte cell death. Taken together, our findings, in a large animal model, provide strong support for the continued evaluation of biliverdin as a potential therapeutic in the clinical setting of transplantation of the liver and perhaps other organs.


Assuntos
Biliverdina/uso terapêutico , Fígado/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Fígado/efeitos dos fármacos , Suínos
9.
PLoS One ; 8(5): e63606, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667646

RESUMO

Hemorrhagic shock and resuscitation induces pulmonary inflammation that leads to acute lung injury. Biliverdin, a metabolite of heme catabolism, has been shown to have potent cytoprotective, anti-inflammatory, and anti-oxidant effects. This study aimed to examine the effects of intravenous biliverdin administration on lung injury induced by hemorrhagic shock and resuscitation in rats. Biliverdin or vehicle was administered to the rats 1 h before sham or hemorrhagic shock-inducing surgery. The sham-operated rats underwent all surgical procedures except bleeding. To induce hemorrhagic shock, rats were bled to achieve a mean arterial pressure of 30 mmHg that was maintained for 60 min, followed by resuscitation with shed blood. Histopathological changes in the lungs were evaluated by histopathological scoring analysis. Inflammatory gene expression was determined by Northern blot analysis, and oxidative DNA damage was assessed by measuring 8-hydroxy-2' deoxyguanosine levels in the lungs. Hemorrhagic shock and resuscitation resulted in prominent histopathological damage, including congestion, edema, cellular infiltration, and hemorrhage. Biliverdin administration prior to hemorrhagic shock and resuscitation significantly ameliorated these lung injuries as judged by histopathological improvement. After hemorrhagic shock and resuscitation, inflammatory gene expression of tumor necrosis factor-α and inducible nitric oxide synthase were increased by 18- and 8-fold, respectively. Inflammatory gene expression significantly decreased when biliverdin was administered prior to hemorrhagic shock and resuscitation. Moreover, after hemorrhagic shock and resuscitation, lung 8-hydroxy-2' deoxyguanosine levels in mitochondrial DNA expressed in the pulmonary interstitium increased by 1.5-fold. Biliverdin administration prior to hemorrhagic shock and resuscitation decreased mitochondrial 8-hydroxy-2' deoxyguanosine levels to almost the same level as that in the control animals. We also confirmed that biliverdin administration after hemorrhagic shock and resuscitation had protective effects on lung injury. Our findings suggest that biliverdin has a protective role, at least in part, against hemorrhagic shock and resuscitation-induced lung injury through anti-inflammatory and anti-oxidant mechanisms.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/etiologia , Biliverdina/administração & dosagem , Biliverdina/uso terapêutico , Ressuscitação , Choque Hemorrágico/complicações , 8-Hidroxi-2'-Desoxiguanosina , Lesão Pulmonar Aguda/sangue , Animais , Aquaporina 5/metabolismo , Bilirrubina/sangue , Biliverdina/farmacologia , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Edema Pulmonar/tratamento farmacológico , Edema Pulmonar/etiologia , Edema Pulmonar/patologia , Ratos , Ratos Sprague-Dawley , Choque Hemorrágico/sangue , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
10.
Clin Exp Rheumatol ; 30(1): 73-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22325451

RESUMO

OBJECTIVES: Heme oxygenase-1 (HO-1) which degrades Heme to free iron, biliverdin and carbon monoxide (CO) plays an important role in inflammation. There are, however, conflicting data concerning the role of HO-1 in rheumatoid arthritis (RA) and the therapeutic potential of individual heme degradation products remains to be determined. We therefore investigated the effect of CO and biliverdin upon therapeutic administration in the murine collagen induced arthritis (CIA) model of RA. METHODS: CIA was induced in DBA/1 mice. Anti-CII antibody levels were determined by ELISA. Mice were scored for paw swelling and grip strength. After the first clinical signs of arthritis one group of animals was treated with biliverdin, the second group was treated with CO. After 60 days all animals were sacrificed and analysed for histomorphological signs of arthritis. RESULTS: All animals immunised with CII developed serum anti-CII antibodies. Antibody levels were decreased in the CO-treated group. Both, Biliverdin and the CO-treated animals, showed an improvement in clinical disease activity. Histological analysis revealed significantly less inflammation, erosion and reduced numbers of osteoclasts in CO-treated animals only, whereas cartilage degradation was prevented in both biliverdin and CO-treated animals. CONCLUSIONS: Our data demonstrate a beneficial effect of CO, in particular, and biliverdin, on inflammation and bone destruction in the CIA mouse model.


Assuntos
Artrite Experimental/tratamento farmacológico , Biliverdina/uso terapêutico , Monóxido de Carbono/uso terapêutico , Heme Oxigenase-1/metabolismo , Articulações/efeitos dos fármacos , Administração por Inalação , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Biliverdina/administração & dosagem , Biliverdina/metabolismo , Monóxido de Carbono/administração & dosagem , Monóxido de Carbono/metabolismo , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Articulações/metabolismo , Articulações/patologia , Camundongos
11.
Diabetologia ; 54(8): 2183-91, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21614569

RESUMO

AIMS/HYPOTHESIS: We have previously shown a negative correlation between serum bilirubin levels and prevalence of type 2 diabetes, suggesting that bilirubin inhibits development of this disease. To confirm this hypothesis, we investigated whether administration of biliverdin, the precursor of bilirubin, protects against the deterioration of glucose tolerance in db/db mice, a rodent model of type 2 diabetes. METHODS: Biliverdin (20 mg/kg daily) was orally administered to 5-week-old db/db mice for 4 weeks. After 4 weeks of treatment, i.p. glucose tolerance and insulin tolerance tests were performed. Insulin content was evaluated by immunostaining and ELISA. Oxidative stress markers (8-hydroxy-2'-deoxyguansosine and dihydroethidium staining) and expression of NADPH oxidase components Pdx1 and Bax were also evaluated in isolated islets. RESULTS: Treatment with biliverdin partially prevented worsening of hyperglycaemia and glucose intolerance in db/db mice. This effect was accompanied by a significant increase in insulin content and Pdx1 expression, and a significant decrease of apoptosis and Bax expression in pancreatic islets from db/db mice. At the same time, levels of oxidative stress markers and NADPH oxidase component production in islets were normalised. Biliverdin had little effect on HOMA of insulin resistance or insulin resistance evaluated by insulin tolerance tests. CONCLUSIONS/INTERPRETATION: Biliverdin may protect against progressive worsening of glucose tolerance in db/db mice, mainly via inhibition of oxidative stress-induced beta cell damage.


Assuntos
Biliverdina/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Intolerância à Glucose/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Transplant Proc ; 42(5): 1602-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20620483

RESUMO

BACKGROUND: Biliverdin, a product of heme oxygenase-1 (HO-1), ameliorates the posttransplant functions of heart, kidney, and liver. In this study, we investigated the effects of biliverdin on lung grafts from brain dead (BD) rat donors. METHODS: Male Wistar rats were randomly divided into 3 groups. The sham group (n = 7), did not undergo BD. Both donor and recipient rats in the BD biliverdin group (n = 8) were injected with biliverdin (35 mg/kg in 1 mL) intraperitoneally after confirmed BD and transplantation. In the BD group (n = 8), both donor and recipient rats received the same volume of saline (35 mg/kg in 1 mL) as the BD biliverdin group. All donor rats were observed for 1.5 hours before undergoing lung transplantation. Two hours after transplantation, we obtained blood and lung graft samples. RESULTS: Biliverdin reversed the aggravation of Pa(O(2)) in recipients, reduced the grafts wet/dry ratio, decreased the severity of lung injury measured by histologic examination, reduced serum tumor necrosis factor-alpha and interleukin-8 levels and inhibited myeloperoxidase activity (MPO) in the grafts. Furthermore, it significantly decreased malonaldehyde levels and increased superoxide dismutase levels. Biliverdin reduced cell apoptosis, activated protein expression of biliverdin reductase, and inhibited expression of HO-1 and nuclear factor (NF)-kappaB in lung grafts. CONCLUSION: Biliverdin exerts protective effects on lung grafts from BD donors through anti-inflammatory, antioxidant, and anti-apoptotic mechanisms.


Assuntos
Biliverdina/uso terapêutico , Transplante de Pulmão/fisiologia , Animais , Apoptose , Bilirrubina/sangue , Pressão Sanguínea , Morte Encefálica , Ensaio de Imunoadsorção Enzimática , Marcação In Situ das Extremidades Cortadas , Inflamação/patologia , Interleucina-8/sangue , Transplante de Pulmão/métodos , Transplante de Pulmão/patologia , Masculino , NF-kappa B/metabolismo , Peroxidase/metabolismo , Ratos , Ratos Wistar , Organismos Livres de Patógenos Específicos , Traqueostomia , Fator de Necrose Tumoral alfa/sangue
13.
Cell Transplant ; 18(9): 999-1002, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19523331

RESUMO

Possible strategies for treating ischemic stroke include neuroprotection (preventing injured neurons from undergoing apoptosis in the acute phase of cerebral ischemia) and stem cell therapy (the repair of disrupted neuronal networks with newly born neurons in the chronic phase of cerebral ischemia). First, we estimated the neuroprotective effect of glial cell line-derived neurotrophic factor (GDNF) by administration of GFNF protein. GDNF protein showed a direct protective effect against ischemic brain damage. Pretreatment of animals with adenoviral vector containing GDNF gene (Ad-GDNF) 24 h before the subsequent transient middle cerebral artery occlusion (MCAO) effectively reduced infarcted volume. Secondly, we studied the neuroprotective effect of a calcium channel blocker, azelnidipine, or a by-product of heme degradation, biliverdin. Both azelnidipine and biliverdin had a neuroprotective effect in the ischemic brain through their antioxidative property. Lastly, we developed a restorative stroke therapy with a bioaffinitive scaffold, which is able to provide an appropriate platform for newly born neurons. In the future, we will combine these strategies to develop more effective therapies for treatment of strokes.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/terapia , Terapia Genética , Transplante de Células-Tronco/métodos , Animais , Antioxidantes/uso terapêutico , Ácido Azetidinocarboxílico/análogos & derivados , Ácido Azetidinocarboxílico/uso terapêutico , Biliverdina/uso terapêutico , Di-Hidropiridinas/uso terapêutico , Modelos Animais de Doenças , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Humanos , Modelos Biológicos , Fármacos Neuroprotetores/uso terapêutico
14.
Neurochem Res ; 34(4): 707-10, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18770029

RESUMO

Possible strategies for treating ischemic stroke include: (1) Neuroprotection: preventing damaged neurons from undergoing apoptosis in the acute phase of cerebral ischemia; (2) Stem cell therapy: the repair of broken neuronal networks with newly born neurons in the chronic phase of cerebral ischemia. Firstly, we studied the neuroprotective effect of a calcium channel blocker, azelnidipine, or a by-product of heme degradation, biliverdin, in the ischemic brain. These results revealed both azelnidipine and biliverdin had a neuroprotective effect in the ischemic brain through their anti-oxidative property. Secondly, we investigated the role of granulocyte colony-stimulating factor (G-CSF) by administering G-CSF to rats after cerebral ischemia and found G-CSF plays a critical role in neuroprotection. Lastly, we developed a restorative stroke therapy with a bio-affinitive scaffold, which is able to provide an appropriate environment for newly born neurons. In the future, we will combine these strategies to develop more effective therapies for treatment of strokes.


Assuntos
Isquemia Encefálica/terapia , Acidente Vascular Cerebral/terapia , Animais , Ácido Azetidinocarboxílico/análogos & derivados , Ácido Azetidinocarboxílico/uso terapêutico , Biliverdina/uso terapêutico , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Di-Hidropiridinas/uso terapêutico , Sequestradores de Radicais Livres/uso terapêutico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Ratos , Células-Tronco/patologia , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/patologia , Tecidos Suporte
15.
Exp Eye Res ; 87(3): 268-78, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18602389

RESUMO

Heme oxygenase (HO-1 and HO-2) represents an intrinsic cytoprotective and anti-inflammatory system based on its ability to modulate leukocyte migration and to inhibit expression of inflammatory cytokines and proteins. HO-2 deletion leads to unresolved corneal inflammation and chronic inflammatory complications including ulceration, perforation and neovascularization. We examined the consequences of HO-2 deletion on hemangiogenesis and lymphangiogenesis in the model of suture-induced inflammatory neovascularization. An 8.0 silk suture was placed at the corneal apex of wild type and HO-2 null mice. Neovascularization was assessed by vital microscopy and quantified by image analysis. Hemangiogenesis and lymphangiogenesis were determined by immunofluorescence staining using anti-CD31 and anti-LYVE-1 antibodies, respectively. Inflammation was quantified by histology and myeloperoxidase activity. The levels of HO-1 expression and inflammatory cytokines were determined by real time PCR and ELISA, respectively. Corneal sutures produced a consistent inflammatory response and a time-dependent neovascularization. The response in HO-2 null mice was associated with a greater increase compared to the wild type in the number of leukocytes (827,600+/-129,000 vs. 294,500+/-57,510; p<0.05), neovessels measured by vital microscopy (21.91+/-1.05 vs. 12.77+/-1.55 mm; p<0.001) 4 days after suture placement. Hemangiogenesis but not lymphangiogenesis was more pronounced in HO-2 null mice compared to wild type mice. Induction of HO-1 in sutured corneas was greatly attenuated in HO-2 null corneas and treatment with biliverdin diminished the exaggerated inflammatory and neovascular response in HO-2 null mice. The demonstration that the inflammatory responses, including expression of proinflammatory proteins, inflammatory cell influx and hemangiogenesis are exaggerated in HO-2 knockout mice strongly supports the notion that the HO system is critical for controlling the inflammatory and neovascular response in the cornea. Hence, pharmacological amplification of this system may constitute a novel therapeutic strategy for the treatment of corneal disorders associated with excessive inflammation and neovascularization.


Assuntos
Biliverdina/uso terapêutico , Neovascularização da Córnea/prevenção & controle , Ceratite/prevenção & controle , Animais , Córnea/enzimologia , Neovascularização da Córnea/enzimologia , Neovascularização da Córnea/patologia , Progressão da Doença , Avaliação Pré-Clínica de Medicamentos , Feminino , Heme Oxigenase (Desciclizante)/deficiência , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase (Desciclizante)/fisiologia , Heme Oxigenase-1/metabolismo , Ceratite/enzimologia , Ceratite/patologia , Masculino , Camundongos , Camundongos Knockout , Peroxidase/metabolismo
16.
Antioxid Redox Signal ; 9(12): 2175-85, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17919067

RESUMO

Bilirubin is the end product of heme catabolism by heme oxygenases. The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Biliverdin is very rapidly converted to bilirubin by the enzyme biliverdin reductase, and free iron upregulates the expression of ferritin. HO-1 is a ubiquitous stress protein and is induced in many cell types by various stimuli. Induced HO-1 exerts antiinflammatory effects and modulates apoptosis. Expression of HO-1 in vivo suppresses the inflammatory responses in endotoxic shock, hyperoxia, acute pleurisy, and organ transplantation, as well as ischemia-reperfusion injury, and thereby provides salutary effects in these conditions. Accumulating evidence indicates that biliverdin/bilirubin can mediate the protective effects of HO-1 in many disease models, such as IRI and organ transplantation, via its antiinflammatory, antiapoptotic, antiproliferative, and antioxidant properties, as well as its effects on the immune response. This review attempts to summarize these protective roles as well as the molecular mechanisms by which biliverdin/bilirubin benefit IRI and solid-organ transplantation, including chronic rejection, and islet transplantation.


Assuntos
Bilirrubina/uso terapêutico , Biliverdina/uso terapêutico , Transplante de Órgãos/fisiologia , Animais , Bilirrubina/metabolismo , Biliverdina/metabolismo , Citoproteção , Rejeição de Enxerto/enzimologia , Rejeição de Enxerto/genética , Heme/química , Heme/metabolismo , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/fisiologia , Heme Oxigenase-1/uso terapêutico , Humanos , Transplante das Ilhotas Pancreáticas , Modelos Biológicos , Estrutura Molecular , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Transplante Homólogo
17.
Transplant Proc ; 39(5): 1338-44, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17580135

RESUMO

OBJECTIVE: This study sought to investigate the protective potential of exogenous biliverdin (BV) for small-for-size rat liver transplants. METHODS AND RESULTS: We employed a rat orthotopic liver transplantation model using small-for-size grafts. BV (50 mumol/kg, intravenously) given to the recipient immediately before reperfusion increased 7-day survival rates (90% vs 40% in controls) and significantly diminished hepatocyte injury, as compared with a control group. These effects correlated with improved liver function and preserved hepatic architecture. BV adjuvant increased antioxidant ability, suppressed proinflammatory tumor necrosis factor-alpha expression, down-regulated proapoptotic molecules (cytochrome C and caspase-3), and inhibited most apoptotic cells. After reperfusion, there was a significant increase of c-Jun NH(2)-terminal kinase (JNK) activation and AP-1 binding ability. BV treatment effectively repressed JNK/AP-1 activation, indicating that a beneficial effect of BV treatment may be related to suppression of the JNK/AP-1 pathway. CONCLUSIONS: BV treatment alleviated ischemia-reperfusion injury at least in part via inhibition of the proinflammatory and proapoptotic JNK/AP-1 pathway. Our findings provide a rationale for a novel therapeutic approach using BV to maximize the availability of small-for-size liver grafts.


Assuntos
Biliverdina/uso terapêutico , Transplante de Fígado/efeitos adversos , Fígado/anatomia & histologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose , Ensaio de Imunoadsorção Enzimática/métodos , Sobrevivência de Enxerto/fisiologia , Transplante de Fígado/patologia , Masculino , Malondialdeído/análise , Ratos , Ratos Sprague-Dawley , Transplante Isogênico , Fator de Necrose Tumoral alfa/análise
18.
Trends Pharmacol Sci ; 28(5): 200-5, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17416426

RESUMO

Inflammation and immunity result in a wide range of disease processes, including atherosclerosis, vascular thrombosis and sepsis. Heme oxygenase-1 (HO-1) is a key enzyme that is integral to the temporal and spatial regulation of the host response and, together with its products carbon monoxide (CO) and bilirubin, is crucial for maintaining homeostasis and the preservation of function and life. An increasing number of reports demonstrates that HO-1, CO and bilirubin regulate the immune response. As CO and bilirubin enter clinical trials, there are obstacles to be addressed before their full therapeutic potential can be achieved. In this article, we delineate the challenges that lie ahead regarding toxicity, pharmacokinetics and mechanisms of action to be able to take full advantage of the powerful cytoprotective properties of these agents for clinical benefit.


Assuntos
Bilirrubina/farmacologia , Biliverdina/farmacologia , Monóxido de Carbono/farmacologia , Heme Oxigenase-1/metabolismo , Substâncias Protetoras/farmacologia , Aterosclerose/fisiopatologia , Bilirrubina/efeitos adversos , Bilirrubina/farmacocinética , Bilirrubina/uso terapêutico , Biliverdina/uso terapêutico , Monóxido de Carbono/efeitos adversos , Monóxido de Carbono/farmacocinética , Monóxido de Carbono/uso terapêutico , Citoproteção/efeitos dos fármacos , Heme Oxigenase-1/fisiologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Humanos , Substâncias Protetoras/efeitos adversos , Substâncias Protetoras/farmacocinética , Substâncias Protetoras/uso terapêutico , Sepse/fisiopatologia , Trombose/fisiopatologia
19.
Cell Cycle ; 6(1): 39-43, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17245120

RESUMO

We have recently shown that the natural bile pigment bilirubin has antiproliferative effects on vascular smooth muscle cells (VSMCs). Bilirubin is the end product of heme catabolism mediated by heme oxygenases and has for decades been considered a toxic waste product of our bodies. However, 14 separate studies and a meta-analysis have documented an inverse correlation between atherosclerosis and the levels of bilirubin in normal individuals. Having high normal or supranormal levels of bilirubin is associated with less atherosclerotic-type disease as compared with that in individuals with low normal levels of bilirubin. This combined with experimental data showing anti-atherosclerotic properties of the enzyme heme oxygenase-1 encouraged us to hypothesize that bilirubin and its precursor biliverdin, would act to ameliorate components of atherosclerosis, in a manner similar to what has been shown with HO-1. Both did so in an animal model of restenosis in which vascular smooth muscle cell proliferation leads to intimal proliferation and causes narrowing of the vessels. We also analyzed the antiproliferative effects of the bile pigments in an in vitro system where bilirubin/biliverdin caused p53 dependent cell cycle arrest by hypophosphorylation of the retinoblastoma tumor suppressor protein in growth factor stimulated VSMCs.


Assuntos
Aterosclerose/tratamento farmacológico , Bilirrubina/uso terapêutico , Biliverdina/uso terapêutico , Animais , Aterosclerose/patologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Masculino , Camundongos , Ratos , Ratos Endogâmicos Lew
20.
Am J Physiol Renal Physiol ; 290(3): F563-71, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16461755

RESUMO

Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. BV is then converted to bilirubin (BR) by the enzyme biliverdin reductase. Experimental evidence suggests that induction of the HO system is an important endogenous mechanism for cytoprotection and that the downstream products of heme degradation, CO, BR, and BV, may mediate these powerful beneficial effects. These molecules, which were once considered to be toxic metabolic waste products, have recently been shown to have dose-dependent vasodilatory, antioxidant, and anti-inflammatory properties that are particularly desirable for tissue protection during organ transplantation. In fact, recent work has demonstrated that administration of exogenous CO, BR, or BV may offer a simple, inexpensive method to substitute for the cytoprotective effects of HO-1 in a variety of clinically applicable models. This review will attempt to summarize the relevant biochemical and cytoprotective properties of CO, BR, and BV, and will discuss emerging studies involving the therapeutic applications of these molecules in the kidney and other organ systems.


Assuntos
Heme Oxigenase-1/metabolismo , Heme/metabolismo , Adulto , Biliverdina/metabolismo , Biliverdina/uso terapêutico , Monóxido de Carbono/metabolismo , Heme/uso terapêutico , Humanos , Ferro/metabolismo , Ferro/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...