Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biomaterials ; 102: 209-19, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27343468

RESUMO

Mesenchymal stem cells (MSCs) are undergoing intensive testing in clinical trials as a promising new therapy for many inflammatory diseases and for regenerative medicine, but further optimization of current MSC-based therapies is required. In this study, we found that in addition to direct complement-mediated attack through the assembly of membrane attack complexes (MACs) that we and others have recently reported, of the released complement activation products, C5a, but not C3a, activates neutrophils in the blood to further damage MSCs through oxidative burst. In addition, we have developed a simple method for painting factor H, a native complement inhibitor, onto MSCs to locally inhibit complement activation on MSCs. MSCs painted with factor H are protected from both MAC- and neutrophil-mediated attack and are significantly more effective in inhibiting antigen-specific T cell responses than the mock-painted MSCs both in vitro and in vivo.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Fator H do Complemento/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/imunologia , Neutrófilos/efeitos dos fármacos , Animais , Células Cultivadas , Fator H do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Receptor da Anafilatoxina C5a/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
2.
PLoS One ; 10(3): e0120225, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25781901

RESUMO

Cutaneous wound continues to cause significant morbidity and mortality in the setting of diseases such as diabetes and cardiovascular diseases. Despite advances in wound care management, there is still an unmet medical need exists for efficient therapy for cutaneous wound. Combined treatment of adrenomedullin (AM) and its binding protein-1 (AMBP-1) is protective in various disease conditions. To examine the effect of the combination treatment of AM and AMBP-1 on cutaneous wound healing, full-thickness 2.0-cm diameter circular excision wounds were surgically created on the dorsum of rats, saline (vehicle) or AM/AMBP-1 (96/320 µg kg BW) was topically applied to the wound daily and wound size measured. At days 3, 7, and 14, skin samples were collected from the wound sites. AM/AMBP-1 treated group had significantly smaller wound surface area than the vehicle group over the 14-day time course. At day 3, AM/AMBP-1 promoted neutrophil infiltration (MPO), increased cytokine levels (IL-6 and TNF-α), angiogenesis (CD31, VEGF and TGFß-1) and cell proliferation (Ki67). By day 7 and 14, AM/AMBP-1 treatment decreased MPO, followed by a rapid resolution of inflammation characterized by a decrease in cytokines. At the matured stage, AM/AMBP-1 treatment increased the alpha smooth muscle actin expression (mature blood vessels) and Masson-Trichrome staining (collagen deposition) along the granulation area, and increased MMP-9 and decreased MMP-2 mRNA expressions. TGFß-1 mRNA levels in AM/AMBP-1 group were 5.3 times lower than those in the vehicle group. AM/AMBP-1 accelerated wound healing by promoting angiogenesis, collagen deposition and remodeling. Treatment also shortened the days to reach plateau for wound closure. Thus, AM/AMBP-1 may be further developed as a therapeutic for cutaneous wound healing.


Assuntos
Adrenomedulina/farmacologia , Fator H do Complemento/farmacologia , Pele/metabolismo , Cicatrização/efeitos dos fármacos , Animais , Masculino , Neovascularização Fisiológica/efeitos dos fármacos , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/patologia , Ratos , Ratos Sprague-Dawley , Pele/patologia
3.
Haematologica ; 98(12): 1939-47, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23850806

RESUMO

The antitumor activity of monoclonal antibodies in the treatment of chronic lymphocytic leukemia is mediated mainly by antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. Unfortunately, the efficacy of complement-dependent cytotoxicity is strongly restricted due to the expression and acquisition of regulators of complement activation by lymphocytic leukemia cells. Whereas the role of membrane regulators of complement activation, such as CD55 and CD59, has been investigated in detail in chronic lymphocytic leukemia, the involvement of soluble regulators of complement activation, such as complement factor H, has not yet been reported. Propidium iodide staining was performed to investigate the efficacy of ofatumumab and factor H-derived short-consensus-repeat 18-20 in the induction of complement-dependent cytotoxicity on primary chronic lymphocytic leukemia cells from 20 patients. Deposition of complement C3 fragments was monitored by western blot analysis. Expression of CD20, CD55 or CD59 was determined by FACS analysis. Replacement of factor H with short consensus repeat 18-20 significantly increased the susceptibility of primary chronic lymphocytic leukemia cells to ofatumumab-induced complement-dependent cytotoxicity. More importantly, addition of short-consensus-repeat 18-20 was able to overcome complement- resistance occurring during treatment with ofatumumab alone. Use of short consensus repeat 18-20 is likely to prolong the turnover time of active C3b fragments generated on the target cells following ofatumumab-induced complement activation, thereby improving specific killing of chronic lymphocytic leukemia cells by complement-dependent cytotoxicity. The relative contribution of factor H to the protection of chronic lymphocytic leukemia cells against complement-dependent cytotoxicity was comparable to that of CD55. Our data suggest that, by abrogating factor H function, short consensus repeat 18-20 may provide a novel approach that improves the complement-dependent efficacy of therapeutic monoclonal antibodies.


Assuntos
Anticorpos Monoclonais/farmacologia , Fator H do Complemento/farmacologia , Sequência Consenso/efeitos dos fármacos , Citotoxinas/farmacologia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Células Cultivadas , Fator H do Complemento/genética , Sequência Consenso/genética , Citotoxinas/uso terapêutico , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Leucemia Linfocítica Crônica de Células B/patologia
4.
Blood ; 119(26): 6307-16, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22577173

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by complement-mediated intravascular hemolysis because of the lack from erythrocyte surface of the complement regulators CD55 and CD59, with subsequent uncontrolled continuous spontaneous activation of the complement alternative pathway (CAP), and at times of the complement classic pathway. Here we investigate in an in vitro model the effect on PNH erythrocytes of a novel therapeutic strategy for membrane-targeted delivery of a CAP inhibitor. TT30 is a 65 kDa recombinant human fusion protein consisting of the iC3b/C3d-binding region of complement receptor 2 (CR2) and the inhibitory domain of the CAP regulator factor H (fH). TT30 completely inhibits in a dose-dependent manner hemolysis of PNH erythrocytes in a modified extended acidified serum assay, and also prevents C3 fragment deposition on surviving PNH erythrocytes. The efficacy of TT30 derives from its direct binding to PNH erythrocytes; if binding to the erythrocytes is disrupted, only partial inhibition of hemolysis is mediated by TT30 in solution, which is similar to that produced by the fH moiety of TT30 alone, or by intact human fH. TT30 is a membrane-targeted selective CAP inhibitor that may prevent both intravascular and C3-mediated extravascular hemolysis of PNH erythrocytes and warrants consideration for the treatment of PNH patients.


Assuntos
Fator H do Complemento/química , Eritrócitos/efeitos dos fármacos , Hemoglobinúria Paroxística/sangue , Hemólise/efeitos dos fármacos , Proteínas de Fusão Oncogênica/farmacologia , Receptores de Complemento 3d/química , Proteínas Recombinantes de Fusão/farmacologia , Estudos de Casos e Controles , Células Cultivadas , Complemento C3/efeitos adversos , Complemento C3/antagonistas & inibidores , Complemento C3/farmacologia , Fator H do Complemento/metabolismo , Fator H do Complemento/farmacologia , Proteínas do Sistema Complemento/efeitos adversos , Proteínas do Sistema Complemento/fisiologia , Citoproteção/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Eritrócitos/fisiologia , Hemoglobinúria Paroxística/patologia , Humanos , Proteínas de Fusão Oncogênica/metabolismo , Ligação Proteica , Receptores de Complemento 3d/metabolismo , Receptores de Complemento 3d/fisiologia , Proteínas Recombinantes de Fusão/metabolismo
5.
J Trauma ; 69(6): 1415-21; discussion 1421-2, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21057332

RESUMO

BACKGROUND: The liver is a major organ that is susceptible to injury after blunt or penetrating trauma to the abdomen. No specific nonoperative treatment exists for traumatic hepatic injury (THI). Adrenomedullin (AM), a vasoactive peptide, combined with its binding protein, AM protein (AMBP-1), is beneficial in various disease conditions. In this study, we propose to analyze whether human AM combined with human AMBP-1 provides benefit in a model of THI in the rat. METHODS: Male adult rats were subjected to trauma hemorrhage by resection of ∼50% of total liver tissues and allowed bleeding for 15 minutes. Immediately thereafter, human AM (48 µg/kg birth weight) plus human AMBP-1 (160 µg/kg birth weight) were given intravenously over 30 minutes in 1-mL normal saline. After 4 hours, the rats were killed, blood was collected, and tissue injury indicators were assessed. A 10-day survival study was also conducted. RESULTS: At 4 hours after THI, plasma AMBP-1 levels were markedly decreased. Plasma levels of liver injury indicators (i.e., aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase) were significantly increased after THI. Similarly, lactate, creatinine, and tumor necrosis factor-α levels were significantly increased after THI. Administration of human AM/AMBP-1 after THI produced significant decreases of 64%, 23%, and 19% of plasma aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase levels, respectively. Similarly, plasma levels of lactate, creatinine, and tumor necrosis factor-α were also decreased by 42%, 28%, and 46% after human AM/AMBP-1 treatment, respectively. In a 10-day survival study, although vehicle treatment produced 41% survival, human AM/AMBP-1 treatment improved the survival rate to 81%. CONCLUSIONS: Administration of human AM/AMBP-1 significantly attenuated tissue injury and inflammation and improved survival after THI. Thus, human AM/AMBP-1 can be developed as a novel treatment for victims with uncontrolled traumatic hemorrhage.


Assuntos
Adrenomedulina/farmacologia , Fator H do Complemento/farmacologia , Hemorragia/tratamento farmacológico , Fígado/lesões , Ressuscitação/métodos , Adrenomedulina/sangue , Análise de Variância , Animais , Fator H do Complemento/metabolismo , Creatinina/sangue , Modelos Animais de Doenças , Hemorragia/etiologia , Técnicas Imunoenzimáticas , Lactatos/sangue , Masculino , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Fator de Necrose Tumoral alfa/sangue
6.
Regul Pept ; 152(1-3): 82-7, 2009 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18948146

RESUMO

Previous studies have demonstrated that co-administration of rat adrenomedullin (AM) and human AM binding protein-1 (AMBP-1) has various beneficial effects following adverse circulatory conditions. In order to reduce rat proteins to elicit possible immune responses in humans, we determined the effect of human AM combined with human AMBP-1 after intestinal ischemia and reperfusion (I/R). Intestinal ischemia was induced in the rat by occluding the superior mesenteric artery for 90 min. At 60 min after the beginning of reperfusion, human AM/AMBP-1 at 3 different dosages was administered intravenously over 30 min. At 240 min after the treatment, blood and tissue samples were harvested and measured for pro-inflammatory cytokines (i.e., TNF-alpha and IL-6), myeloperoxidase activities in the gut and lungs, and cleaved caspase-3 expression in the lungs, as well as serum levels of hepatic enzymes and lactate. In additional groups of animals, a 10-day survival study was conducted. Results showed that administration of human AM/AMBP-1 reduced pro-inflammatory cytokines, attenuated organ injury, and improved the survival rate in a seemingly dose-response fashion. Co-administration of the highest dose of human AM/AMBP-1 in this study had the optimal therapeutic effect in the rat model of intestinal I/R.


Assuntos
Adrenomedulina/farmacologia , Fator H do Complemento/farmacologia , Intestinos/irrigação sanguínea , Isquemia/metabolismo , Traumatismo por Reperfusão/metabolismo , Adrenomedulina/administração & dosagem , Adrenomedulina/metabolismo , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Fator H do Complemento/administração & dosagem , Fator H do Complemento/metabolismo , Citocinas/metabolismo , Humanos , Ácido Láctico/sangue , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Fator de Necrose Tumoral alfa/metabolismo
7.
Mol Med ; 14(7-8): 443-50, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18496585

RESUMO

We recently discovered that the vascular responsiveness to adrenomedullin (AM), a potent vasoactive peptide, decreased during sepsis and hemorrhage in the rat and was markedly improved by its novel binding protein (AMBP-1). Moreover, AM/AMBP-1 appears to be one of the leading candidates for further development to treat sepsis and hemorrhage. However, the extremely high cost of commercial AMBP-1 limits the development of human AM and AMBP-1 as therapeutic agents. The purpose of this study was to isolate and purify AMBP-1 from normal human serum and test its stability and biological activity under in vitro and in vivo conditions. AMBP-1 was isolated and purified from normal human serum with a yield of about 3.0 mg per 100 mL and purity of >99%. The purified AMBP-1 has a AM-binding capacity similar to that of the commercial AMBP-1. Human AM and human AMBP-1 in combination significantly inhibited lipopolysaccharide-induced tumor necrosis factor (TNF)-alpha and interleukin (IL)-6 production from macrophages. The biological activity of the purified human AMBP-1 was well preserved when stored at 45 degrees C for 5 d in solution or at 100 degrees C for 1 h in powder. Moreover, administration of AM and purified AMBP-1 to hemorrhaged rats attenuated tissue injury and neutrophil accumulation. Purified AMBP-1 in combination with AM also suppressed the hemorrhage-induced rise in serum cytokines TNF-alpha and IL-6. Thus, we have successfully purified biologically active AMBP-1 from human normal serum and demonstrated the stability of purified human AMBP-1. This technique will enable us to further develop human AM/AMBP-1 as a novel treatment for safe and effective therapy of patients with hemorrhagic shock, sepsis, and ischemic injury.


Assuntos
Fator H do Complemento/química , Fator H do Complemento/isolamento & purificação , Animais , Células Cultivadas , Quimiocina CCL2/metabolismo , Fator H do Complemento/metabolismo , Fator H do Complemento/farmacologia , Avaliação Pré-Clínica de Medicamentos , Hemorragia/complicações , Hemorragia/patologia , Hemorragia/prevenção & controle , Humanos , Interleucina-6/metabolismo , Isquemia/patologia , Lipopolissacarídeos/farmacologia , Hepatopatias/etiologia , Hepatopatias/patologia , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Soro/química , Fator de Necrose Tumoral alfa/metabolismo
8.
J Immunol ; 179(9): 6263-72, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17947702

RESUMO

Sepsis is a critical inflammatory condition from which numerous patients die due to multiple organ failure and septic shock. The vasoactive hormone adrenomedullin (AM) and its binding protein (AMBP-1) are beneficial in sepsis by abrogating the progression to irreversible shock and decreasing proinflammatory cytokine release. To investigate the anti-inflammatory mechanism, we studied to determine the effect of the AM/AMBP-1 complex on peroxisome proliferator-activated receptor-gamma (PPAR-gamma) expression and activation by using RAW264.7 cells and a rat endotoxemia model. LPS treatment significantly decreased PPAR-gamma expression in vivo and in vitro and was associated with increased TNF-alpha production. Treatment with AM/AMBP-1 for 4 h completely restored PPAR-gamma levels in both models, resulting in TNF-alpha suppression. In a knockdown model using small interfering RNA in RAW264.7 macrophages, AM/AMBP-1 failed to suppress TNF-alpha production in the absence of PPAR-gamma. LPS caused the suppression of intracellular cyclic AMP (cAMP), which was prevented by simultaneous AM/AMBP-1 treatment. Although incubation with dibutyryl cAMP significantly decreased LPS-induced TauNuF-alpha release, it did not alter PPAR-gamma expression. Through inhibition studies using genistein and PD98059 we found that the Pyk-2 tyrosine kinase-ERK1/2 pathway is in part responsible for the AM/AMBP-1-mediated induction of PPAR-gamma and the anti-inflammatory effect. We conclude that AM/AMBP-1 is protective in sepsis due to its vasoactive properties and direct anti-inflammatory effects mediated through both the cAMP-dependent pathway and Pyk-2-ERK1/2-dependent induction of PPAR-gamma.


Assuntos
Adrenomedulina/farmacologia , Fator H do Complemento/farmacologia , PPAR gama/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Endotoxemia/induzido quimicamente , Endotoxemia/genética , Endotoxemia/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Inflamação/genética , Inflamação/metabolismo , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , PPAR gama/genética , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
9.
Ann Surg ; 242(1): 115-23, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15973109

RESUMO

OBJECTIVE: Irreversible hypovolemia remains a major clinical problem. Preliminary studies indicate that administration of adrenomedullin and adrenomedullin binding protein-1 in combination (AM/AMBP-1) after hemorrhage, improves cardiovascular function despite the increased levels of AM. Our aim was to determine whether vascular responsiveness to AM is reduced after hemorrhage and, if so, to elucidate the possible mechanism responsible for such hyporesponsiveness. METHODS: Male rats were bled to and maintained at a mean arterial pressure of 40 mm Hg for 90 minutes. The animals were then resuscitated with 4 times the volume of shed blood with lactated Ringer's solution over 60 minutes. At 1.5 hours postresuscitation, vascular responses to AM and AMBP-1, plasma levels of AM and AMBP-1, AMBP-1 and AM receptor gene expression were measured. In additional animals, AM and AMBP-1 were administered intravenously at 15 minutes after resuscitation over 45 minutes. Serum levels of liver enzymes, lactate, creatinine, TNF-alpha, IL-6, and IL-10 were measured at 1.5 hours postresuscitation. RESULTS: AM-induced vascular relaxation decreased significantly after hemorrhage and resuscitation, which was markedly improved by AMBP-1. However, AM receptor gene expression did not change under such conditions. Hemorrhage-induced AM hyporesponsiveness was accompanied by the decreased expression and release of AMBP-1. Moreover, AM/AMBP-1 treatment down-regulated TNF-alpha and IL-6, up-regulated IL-10, and attenuated organ injury. CONCLUSIONS: The decreased AMBP-1 levels rather than alterations in AM receptors are responsible for producing AM hyporesponsiveness after hemorrhage. Thus, administration of AMBP-1 in combination with AM can be useful to reduce organ injury after severe hypovolemia.


Assuntos
Fator H do Complemento/farmacologia , Hemorragia/sangue , Intestino Delgado/irrigação sanguínea , Peptídeos/farmacologia , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Adrenomedulina , Alanina Transaminase/metabolismo , Animais , Sequência de Bases , Biomarcadores/análise , Western Blotting , Creatinina/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Hemorragia/fisiopatologia , Hemorragia/terapia , Lactatos/metabolismo , Masculino , Dados de Sequência Molecular , Peptídeos/metabolismo , Reação em Cadeia da Polimerase/métodos , Probabilidade , RNA Mensageiro/análise , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Valores de Referência , Ressuscitação/métodos , Sensibilidade e Especificidade , Choque Hemorrágico , Vasodilatadores/metabolismo
10.
Crit Care Med ; 33(2): 391-8, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15699844

RESUMO

OBJECTIVE: The neuroendocrine response to hemorrhage is to maintain perfusion to the heart and brain, often at the expense of other organ systems. Systemic inflammation and tissue injury are important components of pathophysiologic consequences of hemorrhage. We have recently shown that administration of adrenomedullin (AM, a potent vasodilator peptide) and adrenomedullin binding protein-1 (AMBP-1) prevented the transition from the hyperdynamic to the hypodynamic stage in the progression of sepsis. However, the effect of AM/AMBP-1 on the inflammatory response after hemorrhage remains unknown. We therefore hypothesized that administration of AM/AMBP-1 during fluid resuscitation in hemorrhaged animals (i.e., posttreatment) attenuates tissue injury and the proinflammatory response. DESIGN: Prospective, controlled, and randomized animal study. SETTING: A research institute laboratory. SUBJECTS: Male adult rats. INTERVENTIONS: Rats were bled, and then a mean arterial pressure was maintained at 40 mm Hg for 90 mins. They were then resuscitated by infusion of four times the volume of shed blood using Ringer's lactate solution for 60 mins. MEASUREMENTS AND MAIN RESULTS: Fifteen minutes after the beginning of resuscitation, AM (12 microg/kg of body weight) in combination with AMBP-1 (40 microg/kg of body weight) was administered via a femoral venous catheter for 45 mins. Blood samples were collected 4 hrs postresuscitation and assayed for levels of liver enzymes (i.e., alanine aminotransferase and aspartate aminotransferase), lactate, creatinine, proinflammatory cytokines tumor necrosis factor and high mobility group box 1, and anti-inflammatory cytokine interleukin-10. The results indicate that levels of alanine aminotransferase, aspartate aminotransferase, creatinine, lactate, tumor necrosis factor, and high mobility group box 1 markedly elevated after hemorrhage and resuscitation, and AM/AMBP-1 treatment significantly attenuated these increases. In contrast, the serum concentration of anti-inflammatory cytokine interleukin-10 was increased by the treatment of AM/AMBP-1. Moreover, AM/AMBP-1 treatment significantly improved the survival rate from 35% in vehicle-treated animals to 73% in AM/AMBP-1-treated animals in a low-volume resuscitation model of hemorrhage. CONCLUSION: The combined administration of AM and AMBP-1 effectively suppresses hemorrhage-elicited organ injury and reduces hemorrhage-induced mortality, partly through down-regulation of proinflammatory cytokines (tumor necrosis factor and high mobility group box 1) and up-regulation of the anti-inflammatory cytokine interleukin-10.


Assuntos
Fator H do Complemento/farmacologia , Mediadores da Inflamação/metabolismo , Peptídeos/farmacologia , Choque Hemorrágico/metabolismo , Vasodilatadores/farmacologia , Adrenomedulina , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Creatinina/metabolismo , Proteínas HMGB/metabolismo , Interleucina-10/metabolismo , Ácido Láctico/sangue , Masculino , Ratos , Ratos Sprague-Dawley , Ressuscitação , Choque Hemorrágico/terapia , Fator de Necrose Tumoral alfa/metabolismo
11.
FASEB J ; 18(6): 734-6, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14766790

RESUMO

Matrix metalloproteinases (MMPs) are critical for development, wound healing, and for the progression of cancer. It is generally accepted that MMPs are secreted in a latent form (proMMP) and are activated only upon removal of their inhibitory propeptides. This report shows that three members of the SIBLING (Small, Integrin-Binding LIgand, N-linked Glycoprotein) family can specifically bind (Kd approximately equal nM) and activate three different MMPs. Binding of SIBLING to their corresponding proMMPs is associated with structural changes as indicated by quenching of intrinsic tryptophan fluorescence, increased susceptibility to plasmin cleavage, and decreased inhibition by specific natural and synthetic inhibitors. Activation includes both making the proMMPs enzymatically active and the reactivation of the TIMP (tissue inhibitors of MMP) inhibited MMPs. Bone sialoprotein specifically binds proMMP-2 and active MMP-2, while osteopontin binds proMMP-3 and active MMP-3, and dentin matrix protein-1 binds proMMP-9 and active MMP-9. Both pro and active MMP-SIBLING complexes are disrupted by the abundant serum protein, complement Factor H, thereby probably limiting SIBLING-mediated activation to regions immediately adjacent to sites of secretion in vivo. These data suggest that the SIBLING family offers an alternative method of controlling the activity of at least three MMPs.


Assuntos
Glicoproteínas/metabolismo , Metaloproteinases da Matriz/metabolismo , Fator H do Complemento/farmacologia , Ativação Enzimática , Proteínas da Matriz Extracelular , Glicoproteínas/química , Humanos , Sialoproteína de Ligação à Integrina , Integrinas/metabolismo , Ligantes , Substâncias Macromoleculares , Metaloproteinases da Matriz/isolamento & purificação , Modelos Biológicos , Osteopontina , Fosfoproteínas/isolamento & purificação , Fosfoproteínas/metabolismo , Sialoglicoproteínas/isolamento & purificação , Sialoglicoproteínas/metabolismo
12.
Regul Pept ; 112(1-3): 19-26, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12667621

RESUMO

Recent studies have demonstrated that administration of adrenomedullin (AM) and AM binding protein-1 (AMBP-1) maintains cardiovascular stability and reduces mortality in sepsis. However, the mechanism responsible for the beneficial effect of AM/AMBP-1 remains unknown. The aim of this study therefore was to determine whether AM/AMBP-1 directly reduces lipopolysaccharide (LPS)-induced secretion of TNF-alpha from murine macrophage-like cell line RAW 264.7 cells and Kupffer cells isolated from normal rats. TNF-alpha release and gene expression were determined by ELISA and RT-PCR, respectively. The results indicated that LPS increased TNF-alpha production from RAW cells by 38-63-fold in a dose- and time-dependent manner. Although incubation with AM or AMBP-1 alone inhibited LPS-induced TNF-alpha release by 14-22% and 13-22%, respectively, AM and AMBP-1 in combination significantly suppressed TNF-alpha production (by 24-35%). Moreover, the upregulated TNF-alpha mRNA by LPS stimulation was significantly reduced by AM/AMBP-1, but not by AM or AMBP-1 alone. In the Kupffer cells primary culture, AM or AMBP-1 alone inhibited LPS-induced TNF-alpha production by 52% and 44%, respectively. Co-culture with AM/AMBP-1 markedly reduced TNF-alpha production (by 90%). Moreover, AM or AMBP-1 alone decreased TNF-alpha mRNA expression by 41% and 36%, respectively, whereas the combination of AM/AMBP-1 decreased its expression by 63%. These results indicate that AM and AMBP-1 in combination effectively suppress LPS-induced TNF-alpha expression and release especially from primary cultured Kupffer cells, suggesting that the downregulatory effect of AM/AMBP-1 on proinflammatory cytokine TNF-alpha may represent a mechanism responsible for their beneficial effects in preventing inflammatory responses and tissue damage in sepsis.


Assuntos
Fator H do Complemento/farmacologia , Células de Kupffer/imunologia , Macrófagos/imunologia , Peptídeos/farmacologia , Fator de Necrose Tumoral alfa/biossíntese , Adrenomedulina , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Regulação para Baixo , Células de Kupffer/efeitos dos fármacos , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Fator de Necrose Tumoral alfa/genética
13.
Crit Care Med ; 30(12): 2729-35, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12483065

RESUMO

OBJECTIVE: Our recent study indicates that administration of adrenomedullin (AM) in combination with AM-binding protein-1 (AMBP-1) before sepsis (i.e., pretreatment) maintains cardiovascular stability and reduces the mortality rate. The aim of the present study was to determine whether administration of AM/AMBP-1 after the onset of sepsis (posttreatment) has any salutary effects on the septic host, and if so, whether AM/AMBP-1 down-regulates proinflammatory cytokines, such as tumor necrosis factor-alpha, interleukin-1beta, and interleukin-6. DESIGN: Prospective, controlled, randomized animal study. SETTING: A university research laboratory. SUBJECTS: Male adult Sprague-Dawley rats. INTERVENTIONS: Rats were subjected either to polymicrobial sepsis by cecal ligation and puncture or to sham operation followed by the administration of normal saline solution (i.e., fluid resuscitation). MEASUREMENTS AND MAIN RESULTS: At 5 hrs after cecal ligation and puncture, AM (12 microg/kg body weight) and AMBP-1 (40 microg/kg body weight) were administered intravenously over 1 hr. At 20 hrs after cecal ligation and puncture (i.e., the late, hypodynamic stage of sepsis), cardiac output, stroke volume, total peripheral resistance, systemic oxygen delivery, and organ blood flow were determined by radioactive microspheres, and circulating concentrations of proinflammatory cytokines were measured using enzyme-linked immunosorbent assay kits. Moreover, plasma concentrations of transaminases and lactate were measured. The results indicated that administration of AM/AMBP-1 at 5 hrs after cecal ligation and puncture prevented the decrease in measured systemic and regional hemodynamic variables and reduced plasma concentrations of tumor necrosis factor-alpha, interleukin-1beta, and interleukin-6 at 20 hrs after the onset of sepsis. Moreover, administration of AM/AMBP-1 attenuated hepatic damage and the increase in plasma lactate and prevented hemoconcentration. CONCLUSION: Administration of AM/AMBP-1 may provide a novel approach to the treatment of sepsis. Moreover, because AM/AMBP-1 significantly reduced circulating concentrations of tumor necrosis factor-alpha, interleukin-1beta, and interleukin-6, down-regulation of those proinflammatory cytokines by AM/AMBP-1 appears to play an important role for the beneficial effects of these agents in polymicrobial sepsis.


Assuntos
Fator H do Complemento/farmacologia , Citocinas/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Peptídeos/farmacologia , Sepse/tratamento farmacológico , Vasodilatadores/farmacologia , Adrenomedulina , Análise de Variância , Animais , Débito Cardíaco/efeitos dos fármacos , Citocinas/sangue , Interleucina-1/sangue , Interleucina-6/sangue , Ácido Láctico/sangue , Masculino , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Sepse/imunologia , Transaminases/sangue , Fator de Necrose Tumoral alfa/metabolismo
14.
Am J Physiol Regul Integr Comp Physiol ; 283(3): R553-60, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12184987

RESUMO

Adrenomedullin (AM), a potent vasodilatory peptide, plays an important role in initiating the hyperdynamic response during the early stage of sepsis. Moreover, the reduced vascular responsiveness to AM appears to be responsible for the transition from the early, hyperdynamic to the late, hypodynamic phase of sepsis. Although the novel specific AM binding protein-1 (AMBP-1) enhances AM-mediated action in a cultured cell line, it remains to be determined whether AMBP-1 plays any role in modulating vascular responsiveness to AM during sepsis. To study this, adult male rats were subjected to sepsis by cecal ligation and puncture (CLP). The thoracic aorta was harvested for determination of AM-induced vascular relaxation. Aortic levels of AMBP-1 were determined by Western blot analysis, and AM receptor gene expression in the aortic tissue was assessed by RT-PCR. The results indicate that AMBP-1 significantly enhanced AM-induced vascular relaxation in aortic rings from sham-operated animals. Although vascular responsiveness to AM decreased at 20 h after CLP (i.e., the late, hypodynamic stage of sepsis), addition of AMBP-1 in vitro restored the vascular relaxation induced by AM. Moreover, the aortic level of AMBP-1 decreased significantly at 20 h after CLP. In contrast, AM receptor gene expression was not altered under such conditions. These results, taken together, suggest that AMBP-1 plays an important role in modulating vascular responsiveness to AM, and the reduced AMBP-1 appears to be responsible for the vascular AM hyporesponsiveness observed during the hypodynamic phase of sepsis.


Assuntos
Fator H do Complemento/farmacologia , Peptídeos/metabolismo , Sepse/metabolismo , Adrenomedulina , Animais , Aorta Torácica/efeitos dos fármacos , Ceco , Fator H do Complemento/metabolismo , Expressão Gênica/fisiologia , Ligadura , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Adrenomedulina , Receptores de Peptídeos/genética , Vasodilatação/efeitos dos fármacos
15.
Biochem J ; 341 ( Pt 1): 61-9, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10377245

RESUMO

The selectin family of adhesion molecules (E-, P- and L-selectins) is involved in leukocyte recruitment to sites of inflammation and tissue damage. Recently it has been shown that L-selectin is involved not only in leukocyte tethering and rolling, but also plays an important role in leukocyte activation. For example, glycosylation-dependent cell-adhesion molecule 1 (GlyCAM-1), a known ligand for L-selectin, has been shown to enhance beta2-integrin function. GlyCAM-1 is a secreted protein and is present in mouse serum at a concentration of approx. 1.5 microg/ml. There is no obvious GlyCAM-1 homologue in man and, to date, L-selectin ligand(s) from human serum have not been characterized. Therefore we have used L-selectin affinity chromatography, followed by ion-exchange chromatography, to isolate specific ligand(s) for L-selectin. Using this procedure, we have isolated three major glycoproteins of apparent molecular masses 170 kDa, 70kDa and 50 kDa. The 170 kDa protein band was digested with trypsin and peptides were analysed by delayed extraction matrix-assisted laser desorption ionization MS and protein database searching. The 170 kDa protein was identified as the human complement protein Factor H. Human Factor H, isolated by a different method, was shown to bind specifically to L-selectin in the presence of CaCl2, and binding was inhibited by anti-L-selectin antibodies, fucoidan and lipopolysaccharide. Only a part of the purified Factor H preparation bound to immobilized L-selectin. The interaction of Factor H with leukocyte L-selectin was shown to induce the secretion of tumour necrosis factor-alpha (TNF-alpha). Pretreatment of Factor H with sialidase reduced both the binding of L-selectin to Factor H and the Factor H-induced L-selectin-mediated TNF-alpha secretion by leukocytes. Taken together, these results demonstrate that a post-translationally modified form of human plasma Factor H is a potential physiological ligand for L-selectin.


Assuntos
Fator H do Complemento/metabolismo , Glicoproteínas/metabolismo , Selectina L/metabolismo , Proteínas Sanguíneas/isolamento & purificação , Cromatografia de Afinidade , Fator H do Complemento/isolamento & purificação , Fator H do Complemento/farmacologia , Glicoproteínas/isolamento & purificação , Humanos , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Ligantes , Dados de Sequência Molecular , Ácido N-Acetilneuramínico , Polissacarídeos/metabolismo , Ligação Proteica , Análise de Sequência , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Fator de Necrose Tumoral alfa/metabolismo
16.
J Immunol ; 160(8): 4057-66, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9558116

RESUMO

The work presented here demonstrates that human complement factor H is an adhesion ligand for human neutrophils but not for eosinophils. The adherence of polymorphonuclear leukocytes (PMNs) to plastic wells coated with factor H depended on divalent metal ions and was augmented by C5a and TNF-alpha. PMN adhesion to factor H in the presence or absence of C5a was blocked specifically by mAbs against CD11b or CD18. Affinity purification using factor H Sepharose followed by immunoprecipitation using mAbs to various integrin chains identified Mac-1 (CD11b/CD18) as a factor H binding receptor. The presence of surface bound factor H enhanced neutrophil activation resulting in a two- to fivefold increase in the generation of hydrogen peroxide by PMNs stimulated by C5a or TNF-alpha. When factor H was mixed with PMNs, 1.4 to 3.8-fold more cells adhered to immobilized heparin or chondroitin A. In addition, augmented adhesion of PMNs was measured when factor H, but not HSA or C9, was absorbed to wells that were first coated with heparin or chondroitin A. The adhesion of PMNs to glycosaminoglycan-factor H was blocked by mAbs to CD11b and CD18. These studies demonstrate that factor H is an adhesion molecule for human neutrophils and suggest that the interaction of factor H with glycosaminoglycans may facilitate the tethering of this protein in tissues allowing factor H to serve as a neutrophil adhesion ligand in vivo.


Assuntos
Antígenos CD18/metabolismo , Fator H do Complemento/metabolismo , Antígeno de Macrófago 1/metabolismo , Neutrófilos/imunologia , Anticorpos Monoclonais/farmacologia , Proteínas Sanguíneas/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Complemento C5a/farmacologia , Fator H do Complemento/farmacologia , Glicosaminoglicanos/imunologia , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/farmacologia , Humanos , Técnicas In Vitro , Interleucina-8/farmacologia , Lactoferrina/metabolismo , Ligantes , Antígeno de Macrófago 1/isolamento & purificação , Neutrófilos/citologia , Neutrófilos/metabolismo , Oxidantes/metabolismo , Ligação Proteica , Fator de Necrose Tumoral alfa/farmacologia
17.
Infect Immun ; 66(2): 399-405, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9453587

RESUMO

The alternative complement pathway (ACP) functions as a surveillance mechanism by which microorganisms are opsonized with C3b in the absence of specific antibodies. The effectiveness of the ACP relies on its ability to distinguish self from non-self. This recognition function is mediated by C3 regulatory proteins including serum factor H, membrane cofactor protein (MCP), and membrane decay-accelerating factor (DAF). H activity against bound C3b can be increased by host components such as sialic acid and decreased by microbial polysaccharides. DAF and MCP may also recognize cell surface changes such as the presence of viral glycoproteins, since some virus-infected and tumor cells activate the ACP. In the present study, liposomes containing wild-type and mutant Salmonella minnesota lipopolysaccharide (LPS) were tested for ACP activation in serum. LPS-containing liposomes with bound C3b were then tested for their susceptibility to C3 convertase regulation by H and membrane DAF and for the sensitivity of their bound C3b to the cofactor activity of H. The results indicate that while the shortest mutant, Re595 LPS, did not induce ACP activation, R7 LPS containing an additional disaccharide did. This activation was poorly regulated by DAF but was inhibited by H. The regulatory activity of H for liposome-bound C3b, however, decreased when LPS of greater polysaccharide size was present in the membrane. In contrast the ACP activation induced by the phospholipid phosphatidylethanolamine was effectively inhibited by DAF but only poorly inhibited by H.


Assuntos
Antígenos CD55/farmacologia , Fator H do Complemento/farmacologia , Via Alternativa do Complemento/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , Lipossomos/farmacologia , Fosfolipídeos/farmacologia
18.
Biochem J ; 326 ( Pt 2): 321-7, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9291099

RESUMO

Complement factor H (FH) and factor-H-like protein 1 (FHL-1) are human plasma proteins with regulatory functions in the alternative pathway of complement activation. FH and FHL-1 are organized in repetitive elements termed short consensus repeats (SCRs) and the seven SCRs of FHL-1 are identical with the N-terminal domain of the 20 SCRs of FH. The fourth SCR of both proteins (SCR 4) includes the sequence Arg-Gly-Asp (RGD), a motif that is responsible for the major adhesive activity of matrix proteins like fibronectin. A synthetic hexapeptide with the sequence ERGDAV derived from the RGD domain of FH/FHL-1 interferes with cell attachment to a fibronectin matrix. Although the identical motif is present in both FH and FHL-1, only FHL-1 acts as a matrix for cell spreading and attachment, thus the two proteins differ in function. The adhesive activity of FHL-1 is localized to the RGD-containing SCR 4 by the use of recombinant fragments. All three analysed anchorage-dependent cell lines (CCl64, C32 and MRC-5) adhere to an FHL-1 matrix. The use of synthetic peptides in competition assays, on either FHL-1-derived or fibronectin matrices, shows that the cellular receptors binding to the FH/FHL-1-derived RGD motif are related to or identical with integrin receptors which interact with fibronectin. The identification of a functional adhesive domain in the FH/FHL-1 sequence demonstrates, at least for FHL-1, a role in cell attachment and adhesion.


Assuntos
Proteínas Sanguíneas/metabolismo , Adesão Celular , Animais , Proteínas Sanguíneas/química , Proteínas Sanguíneas/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Fator H do Complemento/farmacologia , Ácido Edético/farmacologia , Ácido Egtázico/farmacologia , Epitélio , Fibroblastos , Humanos , Melanoma , Vison , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Estrutura Terciária de Proteína , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA