Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Biomater Sci Eng ; 7(12): 5598-5610, 2021 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-34788004

RESUMO

Biodegradable and implantable materials having elastomeric properties are highly desirable for many biomedical applications. Here, we report that poly(lactide)-co-poly(ß-methyl-δ-valerolactone)-co-poly(lactide) (PLA-PßMδVL-PLA), a thermoplastic triblock poly(α-ester), has combined favorable properties of elasticity, biodegradability, and biocompatibility. This material exhibits excellent elastomeric properties in both dry and aqueous environments. The elongation at break is approximately 1000%, and stretched specimens completely recover to their original shape after force is removed. The material is degradable both in vitro and in vivo; it degrades more slowly than poly(glycerol sebacate) and more rapidly than poly(caprolactone) in vivo. Both the polymer and its degradation product show high cytocompatibility in vitro. The histopathological analysis of PLA-PßMδVL-PLA specimens implanted in the gluteal muscle of rats for 1, 4, and 8 weeks revealed similar tissue responses as compared with poly(glycerol sebacate) and poly(caprolactone) controls, two widely accepted implantable polymers, suggesting that PLA-PßMδVL-PLA can potentially be used as an implantable material with favorable in vivo biocompatibility. The thermoplastic nature allows this elastomer to be readily processed, as demonstrated by the facile fabrication of the substrates with topographical cues to enhance muscle cell alignment. These properties collectively make this polymer potentially highly valuable for applications such as medical devices and tissue engineering scaffolds.


Assuntos
Elastômeros , Alicerces Teciduais , Animais , Elasticidade , Ratos , Engenharia Tecidual
2.
J Pharmacol Exp Ther ; 376(1): 74-83, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33127750

RESUMO

Acute respiratory distress syndrome (ARDS) is a severe, life-threatening form of respiratory failure characterized by pulmonary edema, inflammation, and hypoxemia due to reduced alveolar fluid clearance (AFC). Alveolar fluid clearance is required for recovery and effective gas exchange, and higher rates of AFC are associated with reduced mortality. Thyroid hormones play multiple roles in lung function, and L-3,5,3'-triiodothyronine (T3) has multiple effects on lung alveolar type II cells. T3 enhances AFC in normal adult rat lungs when administered intramuscularly and in normal or hypoxia-injured lungs when given intratracheally. The safety of a commercially available formulation of liothyronine sodium (synthetic T3) administered intratracheally was assessed in an Investigational New Drug Application-enabling toxicology study in healthy rats. Instillation of the commercial formulation of T3 without modification rapidly caused tracheal injury and often mortality. Intratracheal instillation of T3 that was reformulated and brought to a neutral pH at the maximum feasible dose of 2.73 µg T3 in 300 µl for 5 consecutive days had no clinically relevant T3-related adverse clinical, histopathologic, or clinical pathology findings. There were no unscheduled deaths that could be attributed to the reformulated T3 or control articles, no differences in the lung weights, and no macroscopic or microscopic findings considered to be related to treatment with T3. This preclinical safety study has paved the way for a phase I/II study to determine the safety and tolerability of a T3 formulation delivered into the lungs of patients with ARDS, including coronavirus disease 2019-associated ARDS, and to measure the effect on extravascular lung water in these patients. SIGNIFICANCE STATEMENT: There is growing interest in treating lung disease with thyroid hormone [triiodothyronine (T3)] in pulmonary edema and acute respiratory distress syndrome (ARDS). However, there is not any published experience on the impact of direct administration of T3 into the lung. An essential step is to determine the safety of multiple doses of T3 administered in a relevant animal species. This study enabled Food and Drug Administration approval of a phase I/II clinical trial of T3 instillation in patients with ARDS, including coronavirus disease 2019-associated ARDS (T3-ARDS ClinicalTrials.gov Identifier NCT04115514).


Assuntos
Instilação de Medicamentos , Pulmão/efeitos dos fármacos , Síndrome do Desconforto Respiratório/tratamento farmacológico , Tri-Iodotironina/efeitos adversos , Animais , Avaliação Pré-Clínica de Medicamentos , Feminino , Intubação Intratraqueal/efeitos adversos , Intubação Intratraqueal/métodos , Masculino , Ratos , Ratos Sprague-Dawley , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/uso terapêutico
3.
PLoS One ; 14(5): e0216669, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31071184

RESUMO

Antimicrobial peptides have been evaluated as possible alternatives to traditional antibiotics. The translational potential of the antimicrobial peptide DGL13K was tested with focus on peptide toxicity and in vivo activity in two animal models. DGL13K was effective against Pseudomonas aeruginosa, Staphylococcus aureus and methicillin-resistant S. aureus with minimal bactericidal concentrations similar to the minimal inhibitory concentration. The peptide showed low toxicity to human red blood cells and HEK cells with median lethal dose around 1 mg/ml. The median lethal dose in greater wax moth larvae (Galleria mellonella) was about 125mg/kg while the peptide caused no skin toxicity in a mouse model. A novel high-throughput luminescence assay was used to test peptide activity in infected G. mellonella, thus reducing vertebrate animal use. DGL13K killed P. aeruginosa in both the G. mellonella model and a mouse burn wound infection model, with bacterial viability 3-10-fold lower than in untreated controls. Future experiments will focus on optimizing peptide delivery, dose and frequency to further improve the antibacterial effect.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Sequência de Aminoácidos , Animais , Antibacterianos/química , Antibacterianos/toxicidade , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/toxicidade , Eritrócitos/efeitos dos fármacos , Feminino , Células HEK293 , Humanos , Dose Letal Mediana , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Mariposas , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Pele/efeitos dos fármacos , Pele/patologia , Staphylococcus aureus/efeitos dos fármacos , Infecção dos Ferimentos/tratamento farmacológico , Infecção dos Ferimentos/microbiologia
4.
Invest New Drugs ; 37(2): 378-383, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30198058

RESUMO

Fluoroquinolone-class agents selectively target the bacterial type IIA topoisomerases DNA gyrase and topoisomerase IV, with a few exceptions that target eukaryotic type IIA topoisomerases. Fluoroquinolones bind and stabilize type IIA topoisomerase-DNA covalent complexes that contain a double-strand break. This unique mode of action is referred to as 'topoisomerase poisoning'. We discovered that two novel fluoroquinolones having aryl functionality at the N-1 position, UITT-3-217 (217) and UITT-3-227 (227), could inhibit the catalytic activity of human topoisomerase II without stabilizing topoisomerase-DNA complexes, i.e., without poisoning it. Surprisingly, these compounds are more effective in inhibiting the catalytic activities of human and bacterial topoisomerase I. The National Cancer Institute's 60 human tumor cell lines screen revealed significant anti-proliferative activities with 217 and 227 against the majority of 60 cancer cell lines. A proof of concept in vivo efficacy study using an HT-29 xenograft model of human colorectal cancer showed that 217 could inhibit the proliferation of human colorectal cancer cells to a degree comparable to fluorouracil in mice. Although 227 also exhibited anti-proliferative activity, it was not as effective as 217 in this xenograft model. These novel fluoroquinolones may serve as promising lead compounds for the development of new anticancer drugs.


Assuntos
Antineoplásicos/farmacologia , Neoplasias do Colo/tratamento farmacológico , DNA Topoisomerases Tipo I/química , Fluoroquinolonas/farmacologia , Inibidores da Topoisomerase I/farmacologia , Animais , Antineoplásicos/química , Apoptose , Proliferação de Células , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Feminino , Fluoroquinolonas/química , Humanos , Camundongos , Camundongos Nus , Inibidores da Topoisomerase I/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cell Chem Biol ; 24(10): 1259-1275.e6, 2017 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-28919040

RESUMO

The mechanisms by which cancer cell-intrinsic CYP monooxygenases promote tumor progression are largely unknown. CYP3A4 was unexpectedly associated with breast cancer mitochondria and synthesized arachidonic acid (AA)-derived epoxyeicosatrienoic acids (EETs), which promoted the electron transport chain/respiration and inhibited AMPKα. CYP3A4 knockdown activated AMPKα, promoted autophagy, and prevented mammary tumor formation. The diabetes drug metformin inhibited CYP3A4-mediated EET biosynthesis and depleted cancer cell-intrinsic EETs. Metformin bound to the active-site heme of CYP3A4 in a co-crystal structure, establishing CYP3A4 as a biguanide target. Structure-based design led to discovery of N1-hexyl-N5-benzyl-biguanide (HBB), which bound to the CYP3A4 heme with higher affinity than metformin. HBB potently and specifically inhibited CYP3A4 AA epoxygenase activity. HBB also inhibited growth of established ER+ mammary tumors and suppressed intratumoral mTOR. CYP3A4 AA epoxygenase inhibition by biguanides thus demonstrates convergence between eicosanoid activity in mitochondria and biguanide action in cancer, opening a new avenue for cancer drug discovery.


Assuntos
Biguanidas/metabolismo , Biguanidas/farmacologia , Citocromo P-450 CYP3A/metabolismo , Heme/metabolismo , Mitocôndrias/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Biguanidas/química , Neoplasias da Mama/patologia , Domínio Catalítico , Respiração Celular/efeitos dos fármacos , Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/deficiência , Citocromo P-450 CYP3A/genética , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Humanos , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Modelos Moleculares , Transporte Proteico/efeitos dos fármacos
7.
Ann Rheum Dis ; 69(5): 898-902, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19497915

RESUMO

OBJECTIVE: To evaluate the decrease of cartilage destruction by a novel orally active and specific matrix metalloproteinase 13 (MMP-13) inhibitor in three different animal models of rheumatoid arthritis (RA). MATERIALS AND METHODS: The SCID mouse co-implantation model of RA, the collagen-induced arthritis (CIA) model in mice and the antigen-induced arthritis model (AIA) in rabbits were used. RESULTS: In the SCID mouse co-implantation model, the MMP-13 inhibitor reduced cartilage destruction by 75%. In the CIA model of RA, the MMP-13 inhibitor resulted in a significant and dose-dependent decrease in clinical symptoms as well as of cartilage erosion by 38% (30 mg/kg), 28% (10 mg/kg) and 21% (3 mg/kg). No significant effects were seen in the AIA model. No toxic effects were seen in all three animal models. CONCLUSION: Although several MMPs in concert with other proteinases have a role in the process of cartilage destruction, there is a need for highly selective MMP inhibitors to reduce severe side effects that occur with non-specific inhibitors. Significant inhibition of MMP-13 reduced cartilage erosions in two of three tested animal models of RA. These results strongly support the development of this class of drugs to reduce or halt joint destruction in patients with RA.


Assuntos
Antirreumáticos/uso terapêutico , Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Inibidores Enzimáticos/uso terapêutico , Inibidores de Metaloproteinases de Matriz , Administração Oral , Animais , Antirreumáticos/administração & dosagem , Artrite Experimental/enzimologia , Artrite Experimental/patologia , Artrite Reumatoide/enzimologia , Artrite Reumatoide/patologia , Cartilagem Articular/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/administração & dosagem , Feminino , Humanos , Masculino , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos SCID , Coelhos , Membrana Sinovial/enzimologia
8.
Arthritis Rheum ; 56(10): 3347-57, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17907188

RESUMO

OBJECTIVE: To evaluate the role of the MEK/ERK MAP kinase pathway in murine collagen-induced arthritis (CIA) using the selective MEK inhibitor PD184352. We examined the effects of the inhibitor in cytokine-stimulated synovial fibroblasts and in cytokine-induced arthritis in rabbits to investigate its antiinflammatory mechanisms. METHODS: Murine CIA was used to assess the effects of the selective MEK inhibitor on paw edema, clinical scores, weight loss, histopathologic features, and joint levels of p-ERK. Western blotting and immunohistochemistry techniques were used to assess p-ERK in human and rabbit synovial fibroblasts and synovial tissue from rheumatoid arthritis (RA) patients. Interleukin-1alpha (IL-1alpha)-stimulated stromelysin production in rabbit synovial fibroblasts was assessed by enzyme-linked immunosorbent assay. A rabbit IL-1alpha-induced arthritis model was used to assess the effects of the inhibitor on IL-1alpha-induced MEK activity, stromelysin production, and cartilage degradation. RESULTS: In the CIA model, PD184352 inhibited paw edema and clinical arthritis scores in a dose-dependent manner. Disease-induced weight loss and histopathologic changes were also significantly improved by treatment. Inhibition of disease-induced p-ERK levels in the joints was seen with the inhibitor. Levels of p-ERK in the synovium were higher in RA patients than in normal individuals. PD184352 reduced IL-1alpha-induced p-ERK levels in human RA synovial fibroblasts. The production of p-ERK and stromelysin was also inhibited in IL-1alpha-stimulated rabbit synovial fibroblasts. We observed IL-1alpha-induced p-ERK in the synovial lining, subsynovial vasculature, and articular chondrocytes. IL-1alpha-induced stromelysin production and proteoglycan loss from the articular cartilage were reduced by PD184352. CONCLUSION: These data demonstrate the inhibition of murine CIA by PD184352, support the hypothesis that antiinflammatory activity contributes to the mechanism of action of the inhibitor, and suggest that a selective inhibitor may effectively treat RA and other inflammatory disorders.


Assuntos
Artrite Reumatoide/enzimologia , Benzamidas/farmacologia , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinase Quinase Quinases/efeitos dos fármacos , Animais , Artrite Reumatoide/tratamento farmacológico , Western Blotting , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/enzimologia , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Imuno-Histoquímica , Técnicas In Vitro , MAP Quinase Quinase Quinases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos DBA , Coelhos
9.
Arthritis Rheum ; 52(2): 488-500, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15693013

RESUMO

OBJECTIVE: To examine the in vivo effects of PD-0200347, an alpha(2)delta ligand of voltage-activated Ca(2+) channels and a compound chemically related to pregabalin and gabapentin, on the development of cartilage structural changes in an experimental dog model of osteoarthritis (OA). The effects of PD-0200347 on the major pathways involved in OA cartilage degradation, including matrix metalloproteinases (MMPs) and the inducible form of nitric oxide synthase (iNOS), were also studied. METHODS: OA was surgically induced in dogs by sectioning the anterior cruciate ligament. OA dogs were randomly distributed into 3 groups and treated orally with either 1) placebo, 2) 15 mg/kg/day of PD-0200347, or 3) 90 mg/kg/day of PD-0200347. Dogs were killed 12 weeks after surgery. The severity of the lesions was scored macroscopically and histologically. Cartilage specimens from the femoral condyles and tibial plateaus were processed for RNA extraction and quantitative reverse transcription-polymerase chain reaction (RT-PCR) or immunohistochemistry. Specific probes and antibodies were used to study the messenger RNA and protein levels of iNOS, MMP-1, MMP-3, and MMP-13. RESULTS: No clinical signs of drug toxicity were noted in the treated animals. Treatment with PD-0200347 at both dosages tested (15 and 90 mg/kg/day) reduced the development of cartilage lesions. There was a reduction in the score of lesions, with a statistically significant (P = 0.01) difference when the highest dosage of the drug was administered. The reduction in the score was mainly related to a decrease in the surface size of the lesions. Quantitative RT-PCR showed that PD-0200347 significantly reduced the expression of MMP-13, a key mediator in OA. Immunohistochemical analyses showed that treatment with PD-0200347 significantly reduced the synthesis of all key OA mediators studied. CONCLUSION: This study demonstrated the efficacy of PD-0200347 in reducing the progression of cartilage structural changes in a dog model of OA. It also showed that this effect is linked to the inhibition of the major pathophysiologic mediators responsible for cartilage degradation.


Assuntos
Aminas/administração & dosagem , Cartilagem Articular/enzimologia , Condrócitos/enzimologia , Ácidos Cicloexanocarboxílicos/administração & dosagem , Metaloproteases/biossíntese , Metaloproteases/genética , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase/genética , Osteoartrite/prevenção & controle , Ácido gama-Aminobutírico/análogos & derivados , Ácido gama-Aminobutírico/administração & dosagem , Administração Oral , Animais , Cartilagem Articular/patologia , Modelos Animais de Doenças , Progressão da Doença , Cães , Gabapentina , Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Metaloproteases/antagonistas & inibidores , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II , Osteoartrite/patologia , Pregabalina , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ácido gama-Aminobutírico/farmacologia
10.
Arthritis Rheum ; 48(6): 1582-93, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12794826

RESUMO

OBJECTIVE: The primary aim of this study was to investigate, using an experimental rabbit model of osteoarthritis (OA), the effect of a selective mitogen-activated protein kinase kinase 1/2 (MEK-1/2) inhibitor, PD 198306, on the development of structural changes. Additional aims were to assess the effects of the inhibitor on levels of phosphorylated extracellular signal-regulated kinase 1/2 (phospho-ERK-1/2) and matrix metalloproteinase 1 (MMP-1; collagenase 1) in OA chondrocytes. METHODS: After surgical sectioning of the anterior cruciate ligament of the right knee joint, rabbits with OA were separated into 3 experimental groups: oral treatment with placebo or with PD 198306 at a therapeutic concentration of 10 mg/kg/day or 30 mg/kg/day. Each treatment started immediately after surgery. The animals were killed 8 weeks after surgery. Macroscopic and histologic studies were performed on the cartilage and synovial membrane. The levels of phospho-ERK-1/2 and MMP-1 in OA cartilage chondrocytes were evaluated by immunohistochemistry. Normal, untreated rabbits were used as controls. RESULTS: OA rabbits treated with the highest dosage of MEK-1/2 inhibitor showed decreases in the surface area (size) of cartilage macroscopic lesions (P < 0.002) and in osteophyte width on the lateral condyles (P = 0.05). Histologically, the severity of synovial inflammation (villous hyperplasia) was also reduced (P < 0.02). In cartilage from placebo-treated OA rabbits, a significantly higher percentage of chondrocytes in the superficial layer stained positive for phospho-ERK-1/2 and MMP-1 compared with normal controls. Rabbits treated with the highest dosage of PD 198306 demonstrated a significant and dose-dependent reduction in the level of phospho-ERK-1/2 and a lower level of MMP-1. CONCLUSION: This study demonstrates that, in vivo, PD 198306, a selective inhibitor of MEK-1/2, can partially decrease the development of some of the structural changes in experimental OA. This effect was associated with a reduction in the level of phospho-ERK-1/2 in OA chondrocytes, which probably explains the action of the drug.


Assuntos
Inibidores Enzimáticos/farmacologia , Fluorbenzenos/farmacologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Osteoartrite do Joelho/enzimologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Animais , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Colagenases/metabolismo , Modelos Animais de Doenças , Membro Posterior/cirurgia , Imuno-Histoquímica , Ligamentos Longitudinais/cirurgia , MAP Quinase Quinase 1 , MAP Quinase Quinase 2 , Masculino , Metaloproteinase 1 da Matriz/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteoartrite do Joelho/tratamento farmacológico , Osteoartrite do Joelho/patologia , Fosforilação , Coelhos , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/patologia , Sinovite/tratamento farmacológico , Sinovite/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA