Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
JCI Insight ; 3(8)2018 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-29669932

RESUMO

Colon cancer is a complex disease affected by a combination of genetic and epigenetic factors. Here we demonstrate that nardilysin (N-arginine dibasic convertase; NRDC), a metalloendopeptidase of the M16 family, regulates intestinal tumorigenesis via its nuclear functions. NRDC is highly expressed in human colorectal cancers. Deletion of the Nrdc gene in ApcMin mice crucially suppressed intestinal tumor development. In ApcMin mice, epithelial cell-specific deletion of Nrdc recapitulated the tumor suppression observed in Nrdc-null mice. Moreover, epithelial cell-specific overexpression of Nrdc significantly enhanced tumor formation in ApcMin mice. Notably, epithelial NRDC controlled cell apoptosis in a gene dosage-dependent manner. In human colon cancer cells, nuclear NRDC directly associated with HDAC1, and controlled both acetylation and stabilization of p53, with alterations of p53 target apoptotic factors. These findings demonstrate that NRDC is critically involved in intestinal tumorigenesis through its epigenetic regulatory function, and targeting NRDC may lead to a novel prevention or therapeutic strategy against colon cancer.


Assuntos
Carcinogênese/genética , Neoplasias Colorretais/metabolismo , Metaloendopeptidases/metabolismo , Metaloendopeptidases/uso terapêutico , Adulto , Idoso , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Modelos Animais de Doenças , Epigenômica , Feminino , Deleção de Genes , Histona Desacetilase 1 , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteína Supressora de Tumor p53/metabolismo
2.
Curr Top Med Chem ; 17(32): 3425-3443, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29357802

RESUMO

Overexpressed cell-surface receptors are hallmarks of many disease states and are often used as markers for targeting diseased cells over healthy counterparts. Cell adhesion peptides, which are often derived from interacting regions of these receptor-ligand proteins, mimic surfaces of intact proteins and, thus, have been studied as targeting agents for various payloads to certain cell targets for cancers and autoimmune diseases. Because many cytotoxic agents in the free form are often harmful to healthy cells, the use of cell adhesion peptides in targeting their delivery to diseased cells has been studied to potentially reduce required effective doses and associated harmful side-effects. In this review, multiple cell adhesion peptides from extracellular matrix and ICAM proteins were used to selectively direct drug payloads, signal-inhibitor peptides, and diagnostic molecules, to diseased cells over normal counterparts. RGD constructs have been used to improve the selectivity and efficacy of diagnostic and drug-peptide conjugates against cancer cells. From this precedent, novel conjugates of antigenic and cell adhesion peptides, called Bifunctional Peptide Inhibitors (BPIs), have been designed to selectively regulate immune cells and suppress harmful inflammatory responses in autoimmune diseases. Similar peptide conjugations with imaging agents have delivered promising diagnostic methods in animal models of rheumatoid arthritis. BPIs have also been shown to generate immune tolerance and suppress autoimmune diseases in animal models of type-1 diabetes, rheumatoid arthritis, and multiple sclerosis. Collectively, these studies show the potential of cell adhesion peptides in improving the delivery of drugs and diagnostic agents to diseased cells in clinical settings.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/química , ATPases Associadas a Diversas Atividades Celulares/farmacocinética , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Metaloendopeptidases/química , Metaloendopeptidases/farmacocinética , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , ATPases Associadas a Diversas Atividades Celulares/administração & dosagem , ATPases Associadas a Diversas Atividades Celulares/uso terapêutico , Doenças Autoimunes/metabolismo , Humanos , Metaloendopeptidases/administração & dosagem , Metaloendopeptidases/uso terapêutico , Neoplasias/metabolismo
3.
BMC Complement Altern Med ; 14: 446, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25407317

RESUMO

BACKGROUND: Malignant melanoma is a less common but highly dangerous form of skin cancer; it starts in the melanocytes cells found in the outer layer of the skin. Jararhagin toxin, a metalloproteinase isolated from Bothrops jararaca snake venom acts upon several biological processes, as inflammation, pain, platelet aggregation, proliferation and apoptosis, though not yet approved for use, may one day be employed to treat tumors. METHODS: B16F10 murine melanoma cells were treated with jararhagin (jara), a disintegrin-like metalloproteinase isolated from Bothrops jararaca snake venom, and jari (catalytic domain inactivated with 1,10-phenanthroline). Viability and adhesion cells were evaluated by MTT assay. The expression of caspase-3 active, phases of the cell cycle and apoptosis were assessed by flow cytometry. We analyze in vivo the effects of jararhagin on melanoma growth, apoptosis and metastasis. RESULTS: The tumor cells acquired round shapes, lost cytoplasmic expansions, formed clusters in suspension and decreased viability. Jari was almost 20 times more potent toxin than jara based on IC50 values and on morphological changes of the cells, also observed by scanning electron microscopy. Flow cytometry analysis showed 48.3% decrease in the proliferation rate of cells and 47.2% increase in apoptosis (jara) and necrosis (jari), following 1.2 µM jara and 0.1 µM jari treatments. Caspase-3 activity was increased whereas G0/G1 cell cycle phase was on the decline. Proliferative rate was assessed by staining with 5,6-carboxyfluoresceindiacetate succinimidyl ester, showing a significant decrease in proliferation at all concentrations of both toxins. CONCLUSIONS: In vivo treatment of the toxins was observed reduction in the incidence of nodules, and metastasis and antiproliferative inhibition capacity. This data strengthens the potential use jararhagin as an anti-neoplastic drug.


Assuntos
Antineoplásicos/uso terapêutico , Bothrops , Venenos de Crotalídeos/uso terapêutico , Melanoma/tratamento farmacológico , Metaloendopeptidases/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Adesão Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Venenos de Crotalídeos/isolamento & purificação , Venenos de Crotalídeos/farmacologia , Melanoma/metabolismo , Metaloendopeptidases/isolamento & purificação , Metaloendopeptidases/farmacologia , Metaloproteases/farmacologia , Metaloproteases/uso terapêutico , Camundongos , Agregação Plaquetária , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Neoplasias Cutâneas/metabolismo , Veneno de Bothrops jararaca , Melanoma Maligno Cutâneo
4.
Science ; 346(6213): 1118-23, 2014 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-25430769

RESUMO

Tetanus neurotoxin (TeNT) is among the most poisonous substances on Earth and a major cause of neonatal death in nonvaccinated areas. TeNT targets the neuromuscular junction (NMJ) with high affinity, yet the nature of the TeNT receptor complex remains unknown. Here, we show that the presence of nidogens (also known as entactins) at the NMJ is the main determinant for TeNT binding. Inhibition of the TeNT-nidogen interaction by using small nidogen-derived peptides or genetic ablation of nidogens prevented the binding of TeNT to neurons and protected mice from TeNT-induced spastic paralysis. Our findings demonstrate the direct involvement of an extracellular matrix protein as a receptor for TeNT at the NMJ, paving the way for the development of therapeutics for the prevention of tetanus by targeting this protein-protein interaction.


Assuntos
Glicoproteínas de Membrana/metabolismo , Metaloendopeptidases/uso terapêutico , Neurônios Motores/efeitos dos fármacos , Junção Neuromuscular/efeitos dos fármacos , Toxina Tetânica/uso terapêutico , Tétano/prevenção & controle , Animais , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/química , Camundongos , Camundongos Endogâmicos , Neurônios Motores/metabolismo , Junção Neuromuscular/metabolismo , Peptídeos/farmacologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Toxina Tetânica/antagonistas & inibidores , Toxina Tetânica/química
5.
Drugs R D ; 9(3): 185-90, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18457471

RESUMO

Alfimeprase, a fibrolase derivative with thrombolytic activity produced by recombinant DNA technology, was discovered by Amgen and was in development with Nuvelo for the treatment of stroke and catheter occlusion. However, development has been discontinued. Fibrolase is a zinc-containing metalloendopeptidase that was first isolated from the venom of the Southern copperhead snake, Agkistrodon contortrix contortrix. Alfimeprase directly degrades fibrin to break down clots. Alfimeprase is infused directly into the thrombus (side-hole catheter pushed through the entire clot) via multiple manual pulsed infusions. Alfimeprase degrades fibrin directly and entrapped blood cells are freed. Excess alfimeprase is rapidly inactivated by alpha-2 macroglobulin through an irreversible, covalent interaction. Phase III development of alfimeprase for peripheral arterial occlusion was discontinued based on poor results from the NAPA-2 and SONOMA-2 trials; however, Nuvelo resumed development of alfimeprase in 2007 for stroke and catheter occlusion.Alfimeprase's thrombolytic activity appears to be localized to the site of delivery because it is rapidly inactivated by alpha-2 macroglobulin, a naturally occurring protein in the blood, as it moves away from the site of delivery and into general blood circulation. In January 2002, Amgen and Hyseq Pharmaceuticals (now Nuvelo) entered into a collaboration to develop and commercialize alfimeprase. Nuvelo is to develop the product through clinical trials and Amgen was to be responsible for its manufacture. Both companies were to participate in commercial activities; Amgen was to have the option to lead these. Full financial terms were not disclosed. Further to this agreement, in November 2004 Amgen granted Nuvelo worldwide rights to develop and commercialize alfimeprase in exchange for milestone and royalty payments. In August 2007, Nuvelo decided to focus on core development programmes that it believed would produce the nearest-term proof-of-concept data. As a result of this realignment of organizational expenses, Nuvelo decided to continue to pursue the development of alfimeprase. In February 2003, Hyseq Pharmaceuticals merged with Variagenics Inc. to form Nuvelo Inc. In January 2006, Nuvelo and Bayer HealthCare entered a collaboration to develop and commercialize alfimeprase. Bayer was to commercialize the drug in all territories outside the US, whilst Nuvelo retains full US rights. Nuvelo was to receive other territory royalties and milestone payments to a total of $US385 million. A $US50 million upfront payment will be made to Nuvelo, while Bayer was to be responsible for 40% of the commercialization cost for global development. This partnership was to also develop stroke and deep vein thrombosis therapies. Data from the SONOMA-3 trial fell short of the company's expectations and so Nuvelo has decided to discontinue further development of alfimeprase. Nuvelo previously had decided to resume development of alfimeprase for the treatment of multiple coagulation-related disorders, including acute ischaemic stroke, catheter occlusion (CO) and acute peripheral arterial occlusion.Previously, results from the NAPA-2 (Novel Arterial Perfusion with Alfimeprase-2) trial and SONOMA-2 (Speedy Opening of Non-functional and Occluded catheters with Mini-dose Alfimeprase) phase III trial of alfimeprase in patients with acute peripheral arterial occlusion and catheter occlusion did not meet their primary endpoints. The primary endpoint of the NAPA-2 trial was the avoidance of surgery within 30 days of treatment with alfimeprase, whilst the SONOMA-2 trial's endpoint was the restoration of function at 15 minutes after dosing. In addition, these trials did not meet their secondary endpoints. As a result, Nuvelo and Bayer suspended enrolment in the NAPA-3 and SONOMA-3 phase III trials until additional analysis was complete; the SONOMA-3 trial was re-initiated. Nuvelo concluded that the delivery method for alfimeprase in the treatment of peripheral arterial occlusive disorders was suboptimal. The company closed the suspended NAPA-3 trial and planned to initiate preclinical studies focused on identifying optimized delivery methods in acute PAO in the second half of 2007. Data from the NAPA-2 trial suggested that efficacy could potentially be enhanced by maintaining alfimeprase longer at the site of the thrombus. Nuvelo's multinational phase III programme for peripheral arterial occlusion consisted of the NAPA-2 and NAPA-3 trials. Both trials were randomized, double-blind studies comparing 0.3 mg/kg of alfimeprase with placebo in a total of 600 patients in 100 centres. The primary endpoint of the NAPA-2 trial was the avoidance of surgery within 30 days of treatment; secondary endpoints included safety and pharmacoeconomics such as length of hospital and intensive care unit stay. Results from both trials have been presented. The phase II trial (NAPA-1) was completed in June 2004. This open-label, dose-escalation trial assessed the safety and efficacy of alfimeprase in 115 patients with acute peripheral arterial occlusion and was conducted in centres across the US, Europe and South Africa. Full data from the trial have been presented, which indicated that the 0.3 mg/kg dose appeared to be the optimal dose for investigation in phase III trials. In March 2003, Nuvelo announced the positive results of a phase I trial initiated in the US in July 2002; an IND was transferred from Amgen to Nuvelo in January 2002. In the SONOMA-2 trial, alfimeprase restored catheter function in patients with occluded catheters within 15 minutes with a p-value of 0.022. However, it did not meet the company's target product profile for commercial success with a p-value < 0.00125. Data from a phase II trial were presented at the 46th Annual Meeting of the American Society of Hematology held in December 2004. The study was closed in July 2004 with 55 patients enrolled. Initially the phase II study was to compare three doses of alfimeprase with the approved dose of alteplase in over 90 patients in the US. Nuvelo initiated the phase II CARNEROS-1 (Catheter Directed Alfimeprase for Restoration of Neurologic Function and Rapid Opening of Arteries in Stroke) study of alfimeprase in the treatment of acute ischaemic stroke in June 2007. CARNEROS-1 was a multicentre, open-label, dose-escalation study beginning with doses of 1, 5 and 10 mg of alfimeprase in 100 patients within 3-9 hours of stroke onset. The primary endpoints were recanalization (unblocking) of the occlusive lesion within 120 minutes of treatment, and symptomatic intracerebral haemorrhage. The first patient was dosed in December 2007; enrolment was taking place in the US and Canada. In the US, three patents have been issued relating to alfimeprase; US Patent Nos 6 261 820 (alfimeprase protein sequence), 6 440 414 (formulation of alfimeprase with a zinc stabilizer) and 6 455 269 (methods for localized administration of alfimeprase).


Assuntos
Desenho de Fármacos , Fibrinolíticos/farmacologia , Metaloendopeptidases/farmacologia , Arteriopatias Oclusivas/tratamento farmacológico , Ensaios Clínicos como Assunto , Fibrinolíticos/efeitos adversos , Fibrinolíticos/uso terapêutico , Humanos , Metaloendopeptidases/efeitos adversos , Metaloendopeptidases/uso terapêutico
6.
Expert Opin Biol Ther ; 8(5): 683-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18407770

RESUMO

BACKGROUND: Catheter-directed thrombolysis for the management of acute peripheral arterial occlusion emerged as a viable treatment option in the 1990s. It offers a less invasive approach than traditional open surgery for correcting acute limb ischemia. Nonetheless, thrombolysis is plagued by a relatively high rate of bleeding complications as well as long infusion times. OBJECTIVE: To review the clinical experience with alfimeprase, a new thrombolytic agent. METHODS: All published data on alfimeprase were reviewed. Review articles, press releases and web-based data were also included. RESULTS/CONCLUSIONS: Alfimeprase is a novel agent with a unique mechanism of action compared with currently available thrombolytic agents. It is a direct-acting fibrinolytic agent that does not require activation of plasminogen. This mechanism may potentially reduce the number of bleeding complications. Current clinical data are limited, but ongoing clinical trials may demonstrate that this compelling agent represents a clinical advance.


Assuntos
Arteriopatias Oclusivas/tratamento farmacológico , Metaloendopeptidases/uso terapêutico , Doenças Vasculares Periféricas/tratamento farmacológico , Doença Aguda , Ensaios Clínicos como Assunto , Fibrinólise/efeitos dos fármacos , Humanos , Metaloendopeptidases/farmacologia
7.
J Clin Oncol ; 24(19): 3056-60, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16809729

RESUMO

PURPOSE: Alfimeprase is a recombinantly produced, genetically modified variant of the metalloproteinase, fibrolase. Alfimeprase proteolytically cleaves fibrin, independent of plasminogen activation to plasmin, and directly dissolves thrombi. Based on the direct fibrin degradation effect of alfimeprase, rapid activity in patients with occluded central venous access devices (CVADs) was hypothesized. PATIENTS AND METHODS: We performed a phase II, randomized, double-blind, active-control, multicenter, dose-ranging study to compare the safety and efficacy of one or two instillations of three intraluminal doses of alfimeprase (0.3, 1.0, and 3.0 mg) and alteplase 2.0 mg in re-establishing patency to occluded CVADs in 55 adult patients. RESULTS: All three alfimeprase doses were more successful than alteplase during the first 15 and 30 minutes of treatment. The alfimeprase 3.0-mg dose resulted in 40%, 50%, and 60% patency restoration rates at 5, 15, and 30 minutes, respectively, compared with 0%, 0%, and 23% for alteplase. The difference at 15 minutes was highly significant (P = .0075). Alfimeprase 3.0 mg produced the highest patency rate at 120 minutes after the first (60%) and second (80%) doses. No major hemorrhagic or embolic events were reported. CONCLUSION: A single 1- or 3-mg dose of alfimeprase has the potential to restore function to occluded CVADs rapidly and safely, and to facilitate on-time infusion of vital therapies.


Assuntos
Cateterismo Venoso Central/efeitos adversos , Metaloendopeptidases/uso terapêutico , Trombose/tratamento farmacológico , Adulto , Idoso , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Fibrinolíticos/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Trombose/etiologia , Ativador de Plasminogênio Tecidual/uso terapêutico
8.
J Clin Invest ; 116(1): 202-8, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16357941

RESUMO

Degeneration of peripheral motor axons is a common feature of several debilitating diseases including complicated forms of hereditary spastic paraplegia. One such form is caused by loss of the mitochondrial energy-dependent protease paraplegin. Paraplegin-deficient mice display a progressive degeneration in several axonal tracts, characterized by the accumulation of morphological abnormal mitochondria. We show that adenoassociated virus-mediated (AAV-mediated) intramuscular delivery of paraplegin halted the progression of neuropathological changes and rescued mitochondrial morphology in the peripheral nerves of paraplegin-deficient mice. One single injection before onset of symptoms improved the motor performance of paraplegin-deficient mice for up to 10 months, indicating that the peripheral neuropathy contributes to the clinical phenotype. This study provides a proof of principle that gene transfer may be an effective therapeutic option for patients with paraplegin deficiency and demonstrates that AAV vectors can be successfully employed for retrograde delivery of an intracellular protein to spinal motor neurons, opening new perspectives for several hereditary axonal neuropathies of the peripheral nerves.


Assuntos
Terapia Genética , Metaloendopeptidases/genética , Paraplegia/genética , ATPases Associadas a Diversas Atividades Celulares , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Dependovirus , Terapia Genética/métodos , Vetores Genéticos , Metaloendopeptidases/administração & dosagem , Metaloendopeptidases/deficiência , Metaloendopeptidases/uso terapêutico , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Paraplegia/tratamento farmacológico , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/patologia , Medula Espinal/metabolismo
9.
Otolaryngol Head Neck Surg ; 133(6): 836-8, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16360499

RESUMO

OBJECTIVE: The purpose of this study was to find the conversion factor, safety, and efficacy of type A to type B toxin for laryngeal muscles. METHODS: Thirty-two patients with adductor spasmodic dysphonia with stable doses of A toxin to manage their symptom were given type B toxin starting at a conversion of 1 U of BTX-A to 50 U of BTX-B. The patients were followed for 1 year, and doses adjusted according to response. RESULTS: The conversion factor was found to be 52.3 U:1 U. The onset of action of type B was more rapid (2.09 days vs 3.2 days [P = 0.028]), with a shorter duration of benefit (10.8 weeks vs 17 weeks [P = 0.002). The safety profile for A and B toxin appeared the same, with 3 patients receiving Myobloc reporting dry mouth. CONCLUSION: This study shows that a conversion factor of 52.3:1 Myobloc (BTX-B) to Botox (BTX-A) and that Myobloc is an effective alternative to Botox (BTX-A) for patients with spasmodic dysphonia.


Assuntos
Toxinas Botulínicas Tipo A/administração & dosagem , Toxinas Botulínicas/administração & dosagem , Metaloendopeptidases/administração & dosagem , Fármacos Neuromusculares/administração & dosagem , Distúrbios da Voz/tratamento farmacológico , Toxinas Botulínicas/uso terapêutico , Toxinas Botulínicas Tipo A/uso terapêutico , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Humanos , Músculos Laríngeos/efeitos dos fármacos , Músculos Laríngeos/fisiopatologia , Masculino , Metaloendopeptidases/uso terapêutico , Contração Muscular/efeitos dos fármacos , Fármacos Neuromusculares/uso terapêutico , Resultado do Tratamento , Distúrbios da Voz/fisiopatologia
10.
Dermatol Surg ; 31(3): 263-70, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15841624

RESUMO

BACKGROUND: Palmar hyperhidrosis is a problem of unknown etiology that affects patients both socially and professionally. Botulinum toxin type B (Myobloc), approved by the Food and Drug Administration for use in the treatment of cervical dystonia in the United States in December 2000, has subsequently been used effectively in an off-label indication to treat hyperhidrosis. There are sparse data, however, in the literature evaluating the safety and efficacy of BTX-B for the treatment of palmar hyperhidrosis. OBJECTIVE: We evaluated the safety and efficacy of Myobloc in the treatment of bilateral palmar hyperhidrosis. This was a double-blind, randomized, placebo-controlled study to report on the safety and efficacy of Myobloc. METHODS: Twenty participants (10 men, 10 women) diagnosed with palmar hyperhidrosis were injected with either Myobloc (5,000 U per palm) or a 1.0 mL vehicle (100 mM NaCl, 10 mM succinate, and 0.5 mg/mL human albumin) into bilateral palms (15 Myobloc, 5 placebo). The participants were followed until sweating returned to baseline levels. The main outcome measures were safety, efficacy versus placebo, and duration of effect. RESULTS: A significant difference was found in treatment response at day 30, as determined by participant assessments, between 15 participants injected with Myobloc and 3 participants injected with placebo. The duration of action, calculated in the 17 participants who received Myobloc injections and completed the study, ranged from 2.3 to 4.9 months, with a mean duration of 3.8 months. The single most reported adverse event was dry mouth or throat, which was reported by 18 of 20 participants. The adverse event profile also included indigestion or heartburn (60%), excessively dry hands (60%), muscle weakness (60%), and decreased grip strength (50%). CONCLUSION: Myobloc proved to be efficacious for the treatment of palmar hyperhidrosis. Myobloc had a rapid onset, with most participants responding within 1 week. The duration of action ranged from 2.3 to 4.9 months, with a mean of 3.8 months. The adverse event profile included dry mouth, indigestion or heartburn, excessively dry hands, muscle weakness, and decreased grip strength.


Assuntos
Toxinas Botulínicas , Toxinas Botulínicas/uso terapêutico , Hiperidrose/tratamento farmacológico , Metaloendopeptidases/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Toxinas Botulínicas/efeitos adversos , Toxinas Botulínicas Tipo A , Método Duplo-Cego , Feminino , Humanos , Masculino , Metaloendopeptidases/efeitos adversos , Pessoa de Meia-Idade , Projetos Piloto , Qualidade de Vida
11.
Int J Immunopathol Pharmacol ; 17(3): 343-52, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15461868

RESUMO

The need for a vaccine against Leishmania spp., a major cause of worldwide morbidity and mortality, is urgent. We tested the efficacy of an experimental vaccination in murine models of cutaneous leishmaniasis, using dendritic cells (DCs) pulsed with synthetic or native parasite antigens. DCs pulsed with peptide 154-169aa of gp63 or soluble promastigote lysate (SPL) triggered antigen-specific immune responses and efficiently reduced lesion formation and parasite load of genetically susceptible BALB/c mice infected with Leishmania major. This effect was accompanied by a modulation of the cellular immune response towards a Th1 profile. Vaccination of genetically resistant CBA mice with DCs pulsed with peptide 154-169aa or SPL did not affect the course of the disease, whereas pulsing with the epitope 467-482aa of gp63 resulted in disease exacerbation, accompanied by a switch to a Th2 profile. In view of our continuously growing knowledge about the immunobiology of DCs, these findings suggest that vaccination with DCs pulsed with defined peptides could be a strategy against infectious diseases. Peptide selection is a prerequisite as they can differentially regulate the type of immune response in susceptible or resistant hosts.


Assuntos
Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Leishmaniose Cutânea/prevenção & controle , Metaloendopeptidases/imunologia , Metaloendopeptidases/uso terapêutico , Peptídeos/uso terapêutico , Vacinas Protozoárias/uso terapêutico , Animais , Antígenos de Protozoários/imunologia , Carga Corporal (Radioterapia) , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Citocinas/biossíntese , Imunidade Celular , Imunoglobulina G/biossíntese , Imunoglobulina G/genética , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Células Th1/imunologia , Vacinação
12.
Cancer Res ; 64(6): 2062-9, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15026344

RESUMO

Metargidin, a transmembrane protein of the adamalysin family, and integrins, e.g., alpha5beta1 and alphav, are preferentially expressed on endothelial cells on angiogenesis. Furthermore, metargidin interacts with these integrins via its disintegrin domain. In this study, recombinant human disintegrin domain (RDD) was produced in Escherichia coli by subcloning its cDNA into the pGEX-2T vector, and the effect of purified RDD on different steps of angiogenesis was evaluated. At concentrations of 2-10 micro g/ml, RDD exhibited inhibitory activities in a variety of in vitro functional assays, including endothelial cell proliferation and adhesion on the integrin substrates fibronectin, vitronectin, and fibrinogen. RDD (10 micro g/ml) totally abrogated endothelial cell migration and blocked most capillary formation in a three-dimensional fibrin gel. To test RDD efficacy in vivo, the RDD gene inserted into pBi vector containing a tetracycline-inducible promoter was electrotransferred into nude mouse muscle. RDD was successfully synthesized by muscle cells in vivo as shown by immunolabeling and Western blotting. In addition, 78% less MDA-MB-231 tumor growth, associated with strong inhibition of tumor angiogenesis, was observed in athymic mice bearing electrotransferred RDD. Moreover, in the presence of RDD, 74% fewer B16F10 melanoma lung metastases were found in C57BL/6 mice. Taken together, these results identified this RDD as a potent intrinsic inhibitor of angiogenesis, tumor growth, and metastasis, making it a promising tool for use in anticancer treatment.


Assuntos
Antineoplásicos/uso terapêutico , Desintegrinas/uso terapêutico , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/prevenção & controle , Proteínas de Membrana/uso terapêutico , Metaloendopeptidases/uso terapêutico , Neovascularização Patológica/prevenção & controle , Proteínas ADAM , Animais , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Escherichia coli/genética , Feminino , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/secundário , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/secundário , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Músculo Esquelético/patologia , Proteínas Recombinantes/uso terapêutico , Células Tumorais Cultivadas
13.
Ann N Y Acad Sci ; 1035: 1-20, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15681797

RESUMO

The levels of amyloid beta-peptides (Abeta) in the brain represent a dynamic equilibrium state as a result of their biosynthesis from the amyloid precursor protein (APP) by beta- and gamma-secretases, their degradation by a team of amyloid-degrading enzymes, their subsequent oligomerization, and deposition into senile plaques. While most therapeutic attention has focused on developing inhibitors of secretases to prevent Abeta formation, enhancing the rate of Abeta degradation represents an alternative and viable strategy. Current evidence both in vivo and in vitro suggests that there are three major players in amyloid turnover: neprilysin, endothelin converting enzyme(s), and insulin-degrading enzyme, all of which are zinc metallopeptidases. Other proteases have also been implicated in amyloid metabolism, including angiotensin-converting enzyme, and plasmin but for these the evidence is less compelling. Neprilysin and endothelin converting enzyme(s) are homologous membrane proteins of the M13 peptidase family, which normally play roles in the biosynthesis and/or metabolism of regulatory peptides. Insulin-degrading enzyme is structurally and mechanistically distinct. The regional, cellular, and subcellular localizations of these enzymes differ, providing an efficient and diverse mechanism for protecting the brain against the normal accumulation of toxic Abeta peptides. Reduction in expression levels of some of these proteases following insults (e.g., hypoxia and ischemia) or aging might predispose to the development of Alzheimer's disease. Conversely, enhancement of their levels by gene delivery or pharmacological means could be neuroprotective. Even a relatively small enhancement of Abeta metabolism could slow the inexorable progression of the disease. The relative merits of targeting these enzymes for the treatment of Alzheimer's disease will be reviewed and possible side-effects of enhancing their activity evaluated.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Amiloide/metabolismo , Ácidos Docosa-Hexaenoicos/uso terapêutico , Metaloendopeptidases/uso terapêutico , Doenças Neurodegenerativas/tratamento farmacológico , Animais , Ácido Aspártico Endopeptidases/uso terapêutico , Enzimas Conversoras de Endotelina , Fibrinolisina , Humanos , Insulisina/uso terapêutico , Modelos Biológicos , Neprilisina/uso terapêutico , Doenças Neurodegenerativas/enzimologia
14.
Surg Technol Int ; 13: 249-52, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15744697

RESUMO

Between 5% to 10% of tibial fractures progress to nonunion, causing substantial disability. Bone autografts, along with internal fixation, are the usual treatment for these failures, but the morbidity associated with autogenous tissues remains problematic. Bone morphogenetic proteins are currently available for clinical use and preclinical models, as well as an increasing number of patients treated with these molecules demonstrate their safety and efficacy. Osteogenic Protein-1, OP-1, has been evaluated in a randomized, prospective, multi-institution study of tibial nonunions. Sixty-one patients with 63 nonunions received OP-1 and intramedullary rod fixation, and were compared with 61 patients with 61 nonunions treated with fresh autogenous bone graft and the same fixation. Clinical outcomes (success in 81% of OP-1 and 85% of autograft-treated patients) and radiographic evaluation (healing in 75% of OP-1 and 84% of autograft-treated patients) were statistically indistinguishable at 9 months following treatment. No OP-1 or graft-related adverse events occurred. More than 20% of the autograft group had significant donor-site pain 6 months following surgery. OP-1 is a safe and effective alternative to autogenous bone in treatment of tibial nonunions.


Assuntos
Proteínas Morfogenéticas Ósseas/uso terapêutico , Fraturas não Consolidadas/terapia , Metaloendopeptidases/uso terapêutico , Fraturas da Tíbia/terapia , Adolescente , Adulto , Proteína Morfogenética Óssea 1 , Regeneração Óssea/efeitos dos fármacos , Regeneração Óssea/fisiologia , Feminino , Fixação Intramedular de Fraturas/efeitos adversos , Fixação Intramedular de Fraturas/métodos , Consolidação da Fratura/efeitos dos fármacos , Consolidação da Fratura/fisiologia , Fraturas não Consolidadas/diagnóstico por imagem , Humanos , Injeções Intralesionais , Escala de Gravidade do Ferimento , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Radiografia , Recuperação de Função Fisiológica , Fraturas da Tíbia/diagnóstico por imagem , Resultado do Tratamento
15.
Arch Mal Coeur Vaiss ; 95(9): 821-6, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12407798

RESUMO

The angiotensin converting enzyme (ACE), endothelin (ET) converting enzyme (ECE) and neutral endopeptidase (NEP) are all zinc-metallopeptidases expressed in almost all the organs, such as heart, vessels and kidneys. While ACE and ECE are respectively involved in the transformation of angiotensin I and Big-ET into angiotensin II and ET-1 respectively, which possess vasoconstrictor and mitogenic properties, NEP is involved in the degradation of atrial natriuric factor (ANF), which possesses vasorelaxant, diuretic/natriuretic and antihypertrophic properties. These three systems are activated in heart failure and modulate the progression of heart failure. This article will discuss preliminary date concerning simultaneous inhibition of ACE, ECE and/or NEP and their therapeutic potential interest in the treatment of heart failure.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Ácido Aspártico Endopeptidases/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/fisiopatologia , Neprilisina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Ácido Aspártico Endopeptidases/uso terapêutico , Progressão da Doença , Enzimas Conversoras de Endotelina , Humanos , Metaloendopeptidases/farmacologia , Metaloendopeptidases/uso terapêutico , Neprilisina/uso terapêutico
16.
Nursing ; 32(4): 56-62; quiz 62-3, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11969021

Assuntos
Tratamento Farmacológico , Hirudinas/análogos & derivados , Interferon-alfa , Peptídeos Cíclicos , Peptídeos , Polietilenoglicóis , Agonistas Adrenérgicos beta/farmacologia , Agonistas Adrenérgicos beta/uso terapêutico , Alemtuzumab , Androstenos/farmacologia , Androstenos/uso terapêutico , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Anticorpos Antineoplásicos/farmacologia , Anticorpos Antineoplásicos/uso terapêutico , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico , Antifúngicos/farmacologia , Antifúngicos/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Benzamidas , Toxinas Botulínicas , Toxinas Botulínicas Tipo A , Caspofungina , Aprovação de Drogas , Equinocandinas , Etanolaminas/farmacologia , Etanolaminas/uso terapêutico , Fumarato de Formoterol , Terapia com Hirudina , Hirudinas/farmacologia , Humanos , Mesilato de Imatinib , Interferon alfa-2 , Interferon-alfa/farmacologia , Interferon-alfa/uso terapêutico , Lipopeptídeos , Metaloendopeptidases/farmacologia , Metaloendopeptidases/uso terapêutico , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Piperazinas/farmacologia , Piperazinas/uso terapêutico , Congêneres da Progesterona/farmacologia , Congêneres da Progesterona/uso terapêutico , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Estados Unidos
17.
Int J Cancer ; 93(5): 731-5, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11477587

RESUMO

We previously demonstrated that gene replacement of mouse macrophage metalloelastase (MME) into murine melanoma cells that grow rapidly and are MME deficient suppresses the primary tumor growth in vivo by halting angiogenesis. The aim of the present study was to evaluate the effectiveness of gene therapy against cancer using a cDNA-encoding MME gene. In a subcutaneous tumor model of CT-26 mouse colon cancer cells that are MME deficient, syngeneic mice repetitively treated with direct injections into the tumors of MME- hemagglutinating virus of Japan (HVJ), a type of HVJ-cationic liposome encapsulating a plasmid expressing MME, developed smaller tumors (210 +/- 47.2 mm(3) versus 925 +/- 156 mm(3) mean +/- SEM; p = 0.0004) with fewer microvessels (10.25 +/- 1.03 vs. 17.25 +/- 2.14; p = 0.03) than control mice. TUNEL staining revealed a significant increase of apoptotic cells in the MME-HVJ liposomes-treated tumors compared with control tumors. MME was effectively expressed in the s.c. tumors treated with MME-HVJ liposomes, inducing angiostatin generation in those tumors, as demonstrated by Western blot analysis. In conclusion, our study demonstrated that repeated in vivo transduction of the MME gene directly into the tumors using HVJ-cationic liposomes suppressed the tumor growth by an antiangiogenic mechanism, providing, then, a feasible strategy for gene therapy of cancer.


Assuntos
Neoplasias do Colo/terapia , Terapia Genética , Metaloendopeptidases/uso terapêutico , Angiostatinas , Animais , Antineoplásicos/farmacologia , Apoptose , Neoplasias do Colo/genética , Modelos Animais de Doenças , Portadores de Fármacos , Ativação Enzimática , Técnicas de Transferência de Genes , Lipossomos/química , Masculino , Metaloproteinase 12 da Matriz , Metaloendopeptidases/administração & dosagem , Metaloendopeptidases/genética , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Neoplasias Experimentais/genética , Neoplasias Experimentais/terapia , Fragmentos de Peptídeos/farmacologia , Plasminogênio/farmacologia , Respirovirus/química , Transfecção , Células Tumorais Cultivadas
18.
Semin Cutan Med Surg ; 20(2): 127-36, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11474745

RESUMO

Produced by Clostridium botulinum, botulinum toxins are high molecular weight protein complexes consisting of the neurotoxin and additional nontoxic proteins that function to protect the toxin molecule. The neurotoxin acts to inhibit the release of acetylcholine at the neuromuscular junction, causing muscle paralysis. Purified toxin complexes have found a niche in the treatment of clinical disorders involving muscle hyperactivity. The different serotypes are structurally and functionally similar; however, specific differences in neuronal acceptor binding sites, intracellular enzymatic sites, and species sensitivities suggest that each serotype is its own unique pharmacologic entity. Recently, botulinum toxin type B has been developed as a liquid formulation to avoid the lyophilization (vacuum-drying) and reconstitution processes associated with decreasing the potency and stability of current type A toxin preparations. Biochemical tests were conducted to evaluate the quality of toxin in this formulation. In 3 consecutive manufacturing lots, the botulinum toxin type B complex was found to be highly purified, intact, uniform, and consistent from lot to lot. Also, it showed long-term stability at refrigerator and room temperatures (2 to 25 degrees C). Electrophysiologic studies in cynomolgus monkeys showed that botulinum toxin type B is effective in paralyzing injected muscle groups, with minimal spread to relatively distant noninjected muscles.


Assuntos
Acetilcolina/antagonistas & inibidores , Toxinas Botulínicas/farmacologia , Animais , Toxinas Botulínicas/química , Toxinas Botulínicas/uso terapêutico , Toxinas Botulínicas Tipo A , Química Farmacêutica , Estabilidade de Medicamentos , Eletrofisiologia , Humanos , Metaloendopeptidases/química , Metaloendopeptidases/farmacologia , Metaloendopeptidases/uso terapêutico
19.
Circulation ; 102(15): 1766-72, 2000 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-11023930

RESUMO

BACKGROUND: Thrombolytic therapy of acute myocardial infarction (AMI) is evolving toward bolus administration. Derivatization of proteins with polyethylene glycol (PEG) may reduce their clearance. METHODS AND RESULTS: A staphylokinase (SakSTAR) variant with 12 amino acid substitutions to reduce its antigenicity, SakSTAR (K35A, E65Q, K74R, E80A, D82A, T90A, E99D, T101S, E108A, K109A, K130T, K135R), and with Ser in position 3 mutated into Cys (code SY161), was derivatized with maleimide-PEG with M:(r) of 5,000 (P5), 10,000 (P10), or 20,000 (P20). The PEGylated variants recognized only one third of the antibodies elicited with wild-type SakSTAR in AMI patients. In experimental animals, plasma clearances were reduced 2. 5- to 5-fold with P5, 5- to 20-fold with P10, and 20-fold with P20, and bolus injection induced pulmonary plasma clot lysis at doses inversely related to their clearance. Intravenous bolus injection of 5 mg of the P5, P10, or P20 variants in AMI patients was associated with plasma half-lives (t(1/2alpha)) of 13, 30, and 120 minutes and clearances of 75, 43, and 8 mL/min, respectively, compared with 3 minutes and 360 mL/min for SakSTAR. Injection of 5 mg P5 variant restored TIMI-3 flow within 60 minutes in 14 of 18 AMI patients (78%, 95% CI 55% to 91%) and of 2.5 mg in 7 of 11 patients (63%, 95% CI 35% to 85%), both in the absence of fibrinogen degradation. The immunogenicity of the variants was significantly (P:<0.002) reduced. CONCLUSIONS: The staphylokinase variant SY161-P5, derivatized with one linear polyethylene glycol molecule of M:(r) 5000, is a promising fibrin-selective agent for single-bolus coronary thrombolysis.


Assuntos
Fibrinolíticos/uso terapêutico , Metaloendopeptidases/uso terapêutico , Infarto do Miocárdio/tratamento farmacológico , Doença Aguda , Idoso , Cisteína/química , Estabilidade Enzimática , Fibrinolíticos/imunologia , Fibrinolíticos/farmacocinética , Meia-Vida , Humanos , Metaloendopeptidases/química , Metaloendopeptidases/imunologia , Metaloendopeptidases/farmacocinética , Infarto do Miocárdio/metabolismo , Polietilenoglicóis/química , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapêutico
20.
Jpn J Antibiot ; 53(3): 135-56, 2000 Mar.
Artigo em Japonês | MEDLINE | ID: mdl-10834146

RESUMO

Chronic hepatitis B and C virus infections have been characterized by the pathophysiological features with a high incidence of progression to cirrhosis and development of hepatocellular carcinoma. The viral persistence produced by escape mutations from virus-specific cytotoxic T lymphocytes (CTL) response may lead to upregulation of delayed-type hypersensitivity immune response, which causes hepatic tissue damage through non specific macrophage activation and CTL response and promotes pathogenesis of hepatic fibrosis. In a preliminary clinical study, a novel metalloendopeptidase-F (MEP-F) has been shown to be effective in the treatment of patients with either chronic hepatitis B or C infection. Oral administration of MEP-F resulted in a significant reduction of the serum levels of HBs antigen and HCV RNA and improvement in the liver function abnormalities. However, the mechanism of action of MEP-F is not yet well understood. There are accumulating evidences showing an important role of alpha 2-macroglobulin-proteinase complexes in regulatory mechanisms of immune response and repairing within impaired and inflammatory tissues. In this article, reviewing the pharmacological and biological properties of alpha 2-macroglobulin-proteinase complexes, the mechanism of anti-viral effect of MEP-F is examined based on the clinical findings. It is indicated that alpha 2-macroglobulin-MEP-F complexes may induce macrophage/Kuppfer cell activation and proliferation through binding their receptors on the cells and activating signaling cascades, which enhance both anti-viral specific and nonspecific immune responses. alpha 2-Macroglobulin-MEP-F complexes may also augment cellular immunity and hepatic regeneration by neutralizing the immunosuppressive and fibrogenic activities of transforming growth factor-beta.


Assuntos
Hepatite B Crônica/terapia , Hepatite C Crônica/terapia , Metaloendopeptidases/uso terapêutico , Animais , Citocinas/metabolismo , Citocinas/fisiologia , Hepatite B Crônica/imunologia , Hepatite C Crônica/imunologia , Humanos , Fígado/metabolismo , Fígado/fisiologia , Regeneração Hepática , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Macrófagos/imunologia , Metaloendopeptidases/metabolismo , Metaloendopeptidases/farmacologia , Receptores Imunológicos/metabolismo , Receptores Imunológicos/fisiologia , Linfócitos T Citotóxicos/imunologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , alfa-Macroglobulinas/metabolismo , alfa-Macroglobulinas/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA