Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Chem Commun (Camb) ; 60(11): 1372-1388, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38204416

RESUMO

Biomolecule misfolding and aggregation play a major role in human disease, spanning from neurodegeneration to cancer. Inhibition of these processes is of considerable interest, and due to the multifactorial nature of these diseases, the development of drugs that act on multiple pathways simultaneously is a promising approach. This Feature Article focuses on the development of multifunctional molecules designed to inhibit the misfolding and aggregation of the amyloid-ß (Aß) peptide in Alzheimer's disease (AD), and the mutant p53 protein in cancer. While for the former, the goal is to accelerate the removal of the Aß peptide and associated aggregates, for the latter, the goal is reactivation via stabilization of the active folded form of mutant p53 protein and/or aggregation inhibition. Due to the similar aggregation pathway of the Aß peptide and mutant p53 protein, a common therapeutic approach may be applicable.


Assuntos
Doença de Alzheimer , Neoplasias , Humanos , Peptídeos beta-Amiloides/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/uso terapêutico , Proteína Supressora de Tumor p53/genética , Doença de Alzheimer/metabolismo
2.
J Extracell Vesicles ; 12(2): e12307, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36754903

RESUMO

Extracellular vesicles (EVs) contain more than 100 proteins. Whether there are EVs proteins that act as an 'organiser' of protein networks to generate a new or different biological effect from that identified in EV-producing cells has never been demonstrated. Here, as a proof-of-concept, we demonstrate that EV-G12D-mutant KRAS serves as a leader that forms a protein complex and promotes lung inflammation and tumour growth via the Fn1/IL-17A/FGF21 axis. Mechanistically, in contrast to cytosol derived G12D-mutant KRAS complex from EVs-producing cells, EV-G12D-mutant KRAS interacts with a group of extracellular vesicular factors via fibronectin-1 (Fn1), which drives the activation of the IL-17A/FGF21 inflammation pathway in EV recipient cells. We show that: (i), depletion of EV-Fn1 leads to a reduction of a number of inflammatory cytokines including IL-17A; (ii) induction of IL-17A promotes lung inflammation, which in turn leads to IL-17A mediated induction of FGF21 in the lung; and (iii) EV-G12D-mutant KRAS complex mediated lung inflammation is abrogated in IL-17 receptor KO mice. These findings establish a new concept in EV function with potential implications for novel therapeutic interventions in EV-mediated disease processes.


Assuntos
Vesículas Extracelulares , Neoplasias Pulmonares , Pneumonia , Camundongos , Animais , Interleucina-17/metabolismo , Interleucina-17/uso terapêutico , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Mutantes/metabolismo , Proteínas Mutantes/uso terapêutico , Vesículas Extracelulares/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Pneumonia/genética
3.
Mol Ther ; 27(6): 1195-1205, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31010741

RESUMO

Severe photoreceptor cell death in retinal degenerative diseases leads to partial or complete blindness. Optogenetics is a promising strategy to treat blindness. The feasibility of this strategy has been demonstrated through the ectopic expression of microbial channelrhodopsins (ChRs) and other genetically encoded light sensors in surviving retinal neurons in animal models. A major drawback for ChR-based visual restoration is low light sensitivity. Here, we report the development of highly operational light-sensitive ChRs by optimizing the kinetics of a recently reported ChR variant, Chloromonas oogama (CoChR). In particular, we identified two CoChR mutants, CoChR-L112C and CoChR-H94E/L112C/K264T, with markedly enhanced light sensitivity. The improved light sensitivity of the CoChR mutants was confirmed by ex vivo electrophysiological recordings in the retina. Furthermore, the CoChR mutants restored the vision of a blind mouse model under ambient light conditions with remarkably good contrast sensitivity and visual acuity, as evidenced by the results of behavioral assays. The ability to restore functional vision under normal light conditions with the improved CoChR variants removed a major obstacle for ChR-based optogenetic vision restoration.


Assuntos
Cegueira/terapia , Channelrhodopsins/uso terapêutico , Clorofíceas/química , Sensibilidades de Contraste/efeitos dos fármacos , Terapia Genética/métodos , Optogenética/métodos , Acuidade Visual/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Modelos Animais de Doenças , Vetores Genéticos/uso terapêutico , Células HEK293 , Humanos , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Mutantes/uso terapêutico , Técnicas de Patch-Clamp , Retina/metabolismo
4.
Biomed Pharmacother ; 103: 253-261, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29655167

RESUMO

Inflammatory bowel disease (IBD) remains a major health challenge due in part to unsafe and limited treatment options, hence there is the need for alternatives. CXCL8/interleukin 8 (IL-8) is elevated in inflammation, and binds preferentially to G protein-couple receptors (GPCRs) CXCR1/2 of the CXC chemokine family to initiate cascades of downstream inflammatory signals. A mutant CXCL8 protein, CXCL8(3-72)K11R/G31P (G31P), competitively and selectively binds to CXCR1/2, making CXCL8 redundant. We explore the therapeutic potential of G31P in dextran sulfate sodium (DSS) induced ulcerative colitis (UC), and the corresponding effect if G31P treatment is augmented with Lactobacillus acidophilus (LACT). The treatment options administered significantly reduced TNF-α, IFN-γ, IL-1ß, IL-6, and IL-8, but maintained elevated levels of IL-10. CD68 and F4/80 expressions were down-regulated and showed restricted infiltration to inflamed colon, while IL-17F levels were insignificantly different from the DSS treated mice. Also, we observed up-regulation of IL-17A in G31P + LACT but not G31P treated mice if compared with Control group. The treatments ameliorated colonic fibrosis by reducing VEGF, TGF-ß, MMP-2 and MMP-9. In addition, we observed elevated levels of E-cadherin, and marginal up-regulation of occludin, suggesting the role of the treatments in regulating tight intestinal junction and adherence proteins. Mechanism-wise, G31P interferes with AKT and ERK signaling pathways. Our study suggests that G31P confers protection in IBD, particularly UC, and when G31P treatment is augmented with Lactobacillus acidophilus, the protection is variably enhanced.


Assuntos
Colite Ulcerativa/tratamento farmacológico , Interleucina-8/antagonistas & inibidores , Proteínas Mutantes/uso terapêutico , Probióticos/uso terapêutico , Animais , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Colo/patologia , Citocinas/metabolismo , Sulfato de Dextrana , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Fibrose , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Interleucina-8/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Proteínas Mutantes/farmacologia , Probióticos/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas de Junções Íntimas/metabolismo
6.
J Diabetes Complications ; 31(4): 726-734, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28143735

RESUMO

Fibroblast growth factor 21 (FGF21) is a promising regulator of glucose and lipid metabolism with multiple beneficial effects including hypoglycemic and lipid-lowering. Previous studies have reported that FGF21 is expected to become a new drug for treatment of diabetes. Liraglutide and insulin glargine are the two representative anti-diabetic biological drugs. In the current study, we aim to compare the long-term pharmacological efficacy of mFGF21 (an FGF21 analogue), liraglutide and insulin glargine in type 2 diabetic db/db mice. Db/db mice were initially treated with three kinds of proteins (25nmol/kg/day) by subcutaneous injection once a day for 4weeks, then subsequently be treated with once every two days for next 4weeks. After 8weeks of treatments, the blood glucose levels, body weights, glycosylated hemoglobin levels, fasting insulin levels, serum lipid profiles, hepatic biochemical parameters, oral glucose tolerance tests and hepatic mRNA expression levels of several proteins (GK, G6P, GLUT-1 and GLUT-4) associated with glucose metabolism of the experimental mice were detected. Results demonstrated that three proteins could significantly decrease the fed blood glucose levels of db/db mice. After treatment for 1week, the fed blood glucose levels of db/db mice in liraglutide group were significantly lower than those in mFGF21 and insulin glargine groups. However, after 2weeks of administration, the long-lasting hypoglycemic effect of mFGF21 was superior to liraglutide and insulin glargine up to the end of the experiments. Compared with liraglutide and insulin glargine, mFGF21 significantly reduced the glycosylated hemoglobin levels and improved the ability on glycemic control, insulin resistance, serum lipid and liver function states in db/db mice after 8weeks treatments. In addition, mFGF21 regulated glucose metabolism through increasing the mRNA expression levels of GK and GLUT-1, and decreasing the mRNA expression level of G6P. But liraglutide and insulin glargine could only up-regulate the mRNA expression of GLUT-4. In summary, as a hypoglycemic drug for long-term treatment, mFGF21 has the potential to be an ideal drug candidate for the therapy of type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Drogas em Investigação/uso terapêutico , Fatores de Crescimento de Fibroblastos/uso terapêutico , Hipoglicemiantes/uso terapêutico , Insulina Glargina/uso terapêutico , Liraglutida/uso terapêutico , Proteínas Mutantes/uso terapêutico , Animais , Biomarcadores/sangue , Glicemia/análise , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/metabolismo , Drogas em Investigação/efeitos adversos , Drogas em Investigação/metabolismo , Drogas em Investigação/farmacologia , Endopeptidases/metabolismo , Feminino , Fatores de Crescimento de Fibroblastos/efeitos adversos , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Hiperglicemia/prevenção & controle , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/farmacologia , Insulina Glargina/efeitos adversos , Insulina Glargina/farmacologia , Resistência à Insulina , Liraglutida/efeitos adversos , Liraglutida/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Mutantes/efeitos adversos , Proteínas Mutantes/genética , Proteínas Mutantes/farmacologia , Distribuição Aleatória , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico
7.
FEBS J ; 284(6): 837-850, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27808469

RESUMO

The TP53 tumor suppressor is the most frequently mutated gene in human cancers. In recent years, a blooming of research efforts based on both cell lines and mouse models have highlighted how deeply mutant p53 proteins affect fundamental cellular pathways with cancer-promoting outcomes. Neomorphic mutant p53 activities spread over multiple levels, impinging on chromatin structure, transcriptional regulation and microRNA maturation, shaping the proteome and the cell's metabolic pathways, and also exerting cytoplasmic functions and displaying cell-extrinsic effects. These tumorigenic activities are inextricably linked with the blend of highly corrupted processes that characterize the tumor context. Recent studies indicate that successful strategies to extract core aspects of mutant p53 oncogenic potential and to identify unique tumor dependencies entail the superimposition of large-scale analyses performed in multiple experimental systems, together with a mindful use of animal models. This will hopefully soon lead to the long-awaited inclusion of mutant p53 as an actionable target of clinical antitumor therapies.


Assuntos
Terapia de Alvo Molecular , Proteínas Mutantes/genética , Neoplasias/genética , Proteína Supressora de Tumor p53/genética , Animais , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Camundongos , MicroRNAs/genética , Proteínas Mutantes/uso terapêutico , Neoplasias/terapia , Medicina de Precisão , Transdução de Sinais , Proteína Supressora de Tumor p53/uso terapêutico
8.
J Transl Med ; 14(1): 155, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27246873

RESUMO

BACKGROUND: Cytokine fusion protein that modulates the immune response holds great potential for cancer immunotherapy. IL-2 is an effective treatment against advanced cancers. However, the therapeutic efficacy of IL-2 is limited by severe systemic toxicity. Several mutants recombinant IL-2 can increase antitumor activity and minimize systemic toxicity. Melittin is an attractive anticancer candidate because of its wide-spectrum lytic properties. We previously generated a bifunctional fusion protein melittin-MIL-2, composed of melittin and a mutant IL-2. The melittin-MIL-2 inhibited the growth of human ovarian cancer SKOV3 cells in vitro and in vivo tumor growth. However, whether this antitumor effect could also be used in cancer immunotherapy was unknown. To assess its cancer immunotherapy potential, we further investigated its more effective antitumor immune response and antitumor effect against cancers of different tissue origins in vitro and in vivo. METHODS: The specific IL-2 activity of the melittin-MIL-2 fusion protein was tested on the cytokine growth dependent cell line CTLL-2. The cytolytic activity was detected by standard 4-h (51)Cr-release assays. PBMC stimulation in response to the melittin-MIL-2 was determined by IFN-γ release assay. We observed the cancer cell proliferation of different tissue origins by MTT assay. The ability of melittin-MIL-2 to inhibit tumor growth in vivo was evaluated by using human liver (SMMC-7721 cancer cells), lung (A549 cancer cells) and ovarian (SKOV3 cancer cells) cancer xenograft models. To assess the immunity within the tumor microenvironment, the level of some cytokines including IFN-γ, TNF-α, IL-12 and IL-4 was analyzed by ELISA. We injected the MDA-MB-231 cells and the melittin-MIL-2 into mice, and the anti-metastatic effect was examined by counting nodules in the lung. RESULTS: The melittin-MIL-2 was more effective in inducing T cell and NK-cell cytotoxicity. The fusion protein significantly increased IFN-γ production in PBMCs. In vitro, the melittin-MIL-2 mediated immune cells killing or directly killed the cancer cell lines of different tissue origins. In vivo, the fusion protein exhibited stronger inhibition on the growth of transplanted human tumors compared to rIL-2. The melittin-MIL-2 treatment promoted the IFN-γ secretion in tumor tissues and decreased the immunosuppressive cells in vivo. Furthermore, the fusion protein reduced lung metastasis of breast cancer. CONCLUSIONS: This study provides the evidence that the melittin-MIL-2 can produce stronger immune stimulation and antitumor effects, and the fusion protein is a potent candidate for cancer immunotherapy.


Assuntos
Imunoterapia , Meliteno/uso terapêutico , Proteínas Mutantes/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Imunomodulação/efeitos dos fármacos , Interferon gama/biossíntese , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Ativação Linfocitária/efeitos dos fármacos , Meliteno/farmacologia , Camundongos Endogâmicos BALB C , Proteínas Mutantes/farmacologia , Neoplasias/patologia , Proteínas Recombinantes de Fusão/farmacologia
9.
Proc Natl Acad Sci U S A ; 112(47): E6506-14, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26604307

RESUMO

Signaling through the immune checkpoint programmed cell death protein-1 (PD-1) enables tumor progression by dampening antitumor immune responses. Therapeutic blockade of the signaling axis between PD-1 and its ligand programmed cell death ligand-1 (PD-L1) with monoclonal antibodies has shown remarkable clinical success in the treatment of cancer. However, antibodies have inherent limitations that can curtail their efficacy in this setting, including poor tissue/tumor penetrance and detrimental Fc-effector functions that deplete immune cells. To determine if PD-1:PD-L1-directed immunotherapy could be improved with smaller, nonantibody therapeutics, we used directed evolution by yeast-surface display to engineer the PD-1 ectodomain as a high-affinity (110 pM) competitive antagonist of PD-L1. In contrast to anti-PD-L1 monoclonal antibodies, high-affinity PD-1 demonstrated superior tumor penetration without inducing depletion of peripheral effector T cells. Consistent with these advantages, in syngeneic CT26 tumor models, high-affinity PD-1 was effective in treating both small (50 mm(3)) and large tumors (150 mm(3)), whereas the activity of anti-PD-L1 antibodies was completely abrogated against large tumors. Furthermore, we found that high-affinity PD-1 could be radiolabeled and applied as a PET imaging tracer to efficiently distinguish between PD-L1-positive and PD-L1-negative tumors in living mice, providing an alternative to invasive biopsy and histological analysis. These results thus highlight the favorable pharmacology of small, nonantibody therapeutics for enhanced cancer immunotherapy and immune diagnostics.


Assuntos
Imunoterapia , Proteínas Mutantes/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Tomografia por Emissão de Pósitrons , Receptor de Morte Celular Programada 1/uso terapêutico , Engenharia de Proteínas , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Evolução Molecular Direcionada , Modelos Animais de Doenças , Humanos , Depleção Linfocítica , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/química , Ligação Proteica , Linfócitos T/metabolismo
10.
Blood ; 125(10): 1553-61, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25568350

RESUMO

Emerging successful clinical data on gene therapy using adeno-associated viral (AAV) vector for hemophilia B (HB) showed that the risk of cellular immune response to vector capsid is clearly dose dependent. To decrease the vector dose, we explored AAV-8 (1-3 × 10(12) vg/kg) encoding a hyperfunctional factor IX (FIX-Padua, arginine 338 to leucine) in FIX inhibitor-prone HB dogs. Two naïve HB dogs showed sustained expression of FIX-Padua with an 8- to 12-fold increased specific activity reaching 25% to 40% activity without antibody formation to FIX. A third dog with preexisting FIX inhibitors exhibited a transient anamnestic response (5 Bethesda units) at 2 weeks after vector delivery following by spontaneous eradication of the antibody to FIX by day 70. In this dog, sustained FIX expression reached ∼200% and 30% of activity and antigen levels, respectively. Immune tolerance was confirmed in all dogs after challenges with plasma-derived FIX concentrate. Shortening of the clotting times and lack of bleeding episodes support the phenotypic correction of the severe phenotype, with no clinical or laboratory evidence of risk of thrombosis. Provocative studies in mice showed that FIX-Padua exhibits similar immunogenicity and thrombogenicity compared with FIX wild type. Collectively, these data support the potential translation of gene-based strategies using FIX-Padua for HB.


Assuntos
Fator IX/antagonistas & inibidores , Terapia Genética/métodos , Hemofilia B/genética , Hemofilia B/terapia , Substituição de Aminoácidos , Animais , Capsídeo/imunologia , Citocinas/sangue , Dependovirus/genética , Dependovirus/imunologia , Modelos Animais de Doenças , Cães , Fator IX/genética , Fator IX/imunologia , Fator IX/uso terapêutico , Expressão Gênica , Vetores Genéticos/efeitos adversos , Vetores Genéticos/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Proteínas Mutantes/uso terapêutico , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Trombose/prevenção & controle , Pesquisa Translacional Biomédica
11.
Oncogene ; 34(26): 3413-28, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25174405

RESUMO

Membrane overexpression of ErbB-2/HER2 receptor tyrosine kinase (membrane ErbB-2 (MErbB-2)) has a critical role in breast cancer (BC). We and others have also shown the role of nuclear ErbB-2 (NErbB-2) in BC, whose presence we identified as a poor prognostic factor in MErbB-2-positive tumors. Current anti-ErbB-2 therapies, as with the antibody trastuzumab (Ttzm), target only MErbB-2. Here, we found that blockade of NErbB-2 action abrogates growth of BC cells, sensitive and resistant to Ttzm, in a scenario in which ErbB-2, ErbB-3 and Akt are phosphorylated, and ErbB-2/ErbB-3 dimers are formed. Also, inhibition of NErbB-2 presence suppresses growth of a preclinical BC model resistant to Ttzm. We showed that at the cyclin D1 promoter, ErbB-2 assembles a transcriptional complex with Stat3 (signal transducer and activator of transcription 3) and ErbB-3, another member of the ErbB family, which reveals the first nuclear function of ErbB-2/ErbB-3 dimer. We identified NErbB-2 as the major proliferation driver in Ttzm-resistant BC, and demonstrated that Ttzm inability to disrupt the Stat3/ErbB-2/ErbB-3 complex underlies its failure to inhibit growth. Furthermore, our results in the clinic revealed that nuclear interaction between ErbB-2 and Stat3 correlates with poor overall survival in primary breast tumors. Our findings challenge the paradigm of anti-ErbB-2 drug design and highlight NErbB-2 as a novel target to overcome Ttzm resistance.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Terapia de Alvo Molecular , Proteínas Mutantes/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Feminino , Genes Dominantes/fisiologia , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Terapia de Alvo Molecular/métodos , Proteínas Mutantes/uso terapêutico , Isoformas de Proteínas/farmacologia , Isoformas de Proteínas/uso terapêutico , Transporte Proteico/efeitos dos fármacos , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-2/fisiologia , Trastuzumab , Células Tumorais Cultivadas
12.
Curr Pharm Des ; 20(4): 659-65, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23688008

RESUMO

Random mutagenesis of mouse leptin antagonist (L39A/D40A/F41) followed by selection of high-affinity mutants by yeastsurface display indicated that replacing residue D23 with a non-negatively charged amino acid (most specifically with Leu) leads to dramatically enhanced affinity of leptin toward LEPR leading to development of superactive mouse, human, ovine and rat leptin antagonists (D23L/L39A/D40A/F41A). Superactive leptin antagonist mutants of mouse, human, rat or ovine leptins were developed in our laboratory, expressed in E. coli, refolded and purified to homogeneity as monomeric proteins. Pegylation of leptin antagonists resulted in potent and effective long-acting reagents suitable for in vivo studies or therapies. In the present review we explain the mechanism of leptin inhibition and summarize the possible use of leptin antagonists as possible leptin blockers in various human pathologies such as antiinflammatory and anti-autoimmune diseases, uremic cachexia, and cancer. We also suggest the use of leptin antagonists as research reagents for creation of a novel, fast and reversible model of T2DM in mice.


Assuntos
Desenho de Fármacos , Drogas em Investigação/uso terapêutico , Leptina/antagonistas & inibidores , Receptores para Leptina/antagonistas & inibidores , Substituição de Aminoácidos , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Autoimunidade/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Drogas em Investigação/química , Drogas em Investigação/farmacologia , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Imunossupressores/química , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Leptina/análogos & derivados , Leptina/genética , Ligantes , Proteínas Mutantes/química , Proteínas Mutantes/farmacologia , Proteínas Mutantes/uso terapêutico , Receptores para Leptina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico
13.
Thromb Haemost ; 110(2): 244-56, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23676890

RESUMO

Using gain-of-function factor IX (FIX) for replacement therapy for haemophilia B (HB) is an attractive strategy. We previously reported a high-activity FIX, FIX-Triple (FIX-V86A/E277A/R338A) as a good substitute for FIX-WT (wild-type) in protein replacement therapy, gene therapy, and cell therapy. Here we generated a new recombinant FIX-TripleL (FIX-V86A/E277A/R338L) by replacing the alanine at residue 338 of FIX-Triple with leucine as in FIX-Padua (FIX-R338L). Purified FIX-TripleL exhibited 22-fold higher specific clotting activity and 15-fold increased binding affinity to activated FVIII compared to FIX-WT. FIX-TripleL increased the therapeutic potential of FIX-Triple by nearly 100% as demonstrated with calibrated automated thrombogram and thromboelastography. FIX-TripleL demonstrated a normal clearance rate in HB mice. The clotting activity of FIX-TripleL was consistently 2- to 3-fold higher in these mice than that of FIX-Triple or FIX-R338L. Gene delivery of adeno-associated virus (AAV) in HB mice showed that FIX-TripleL had 15-fold higher specific clotting activity than FIX-WT, and this activity was significantly better than FIX-Triple (10-fold) or FIX-R338L (6-fold). At a lower viral dose, FIX-TripleL improved FIX activity from sub-therapeutic to therapeutic levels. Under physiological conditions, no signs of adverse thrombotic events were observed in long-term AAV-FIX-treated C57Bl/6 mice. Hepatocellular adenomas were observed in the high- but not the medium- or the low-dose AAV-treated mice expressing FIX-WT or FIX-Triple, indicating the advantages of using hyperfunctional FIX variants to reduce viral doses while maintaining therapeutic clotting activity. Thus, incorporation of the FIX Padua mutation significantly improves the clotting function of FIX-Triple so as to optimise protein replacement therapy and gene therapy.


Assuntos
Fator IX/genética , Fator IX/uso terapêutico , Hemofilia B/sangue , Hemofilia B/tratamento farmacológico , Proteínas Mutantes/genética , Proteínas Mutantes/uso terapêutico , Substituição de Aminoácidos , Animais , Coagulação Sanguínea/efeitos dos fármacos , Dependovirus/genética , Modelos Animais de Doenças , Fator IX/metabolismo , Fator VIIIa/metabolismo , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Mutagênese Sítio-Dirigida , Proteínas Mutantes/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Trombina/biossíntese
14.
Sci Transl Med ; 5(174): 174ra28, 2013 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-23447019

RESUMO

Vitiligo is an autoimmune disease characterized by destruction of melanocytes, leaving 0.5% of the population with progressive depigmentation. Current treatments offer limited efficacy. We report that modified inducible heat shock protein 70 (HSP70i) prevents T cell-mediated depigmentation. HSP70i is the molecular link between stress and the resultant immune response. We previously showed that HSP70i induces an inflammatory dendritic cell (DC) phenotype and is necessary for depigmentation in vitiligo mouse models. Here, we observed a similar DC inflammatory phenotype in vitiligo patients. In a mouse model of depigmentation, DNA vaccination with a melanocyte antigen and the carboxyl terminus of HSP70i was sufficient to drive autoimmunity. Mutational analysis of the HSP70i substrate-binding domain established the peptide QPGVLIQVYEG as invaluable for DC activation, and mutant HSP70i could not induce depigmentation. Moreover, mutant HSP70iQ435A bound human DCs and reduced their activation, as well as induced a shift from inflammatory to tolerogenic DCs in mice. HSP70iQ435A-encoding DNA applied months before spontaneous depigmentation prevented vitiligo in mice expressing a transgenic, melanocyte-reactive T cell receptor. Furthermore, use of HSP70iQ435A therapeutically in a different, rapidly depigmenting model after loss of differentiated melanocytes resulted in 76% recovery of pigmentation. Treatment also prevented relevant T cells from populating mouse skin. In addition, ex vivo treatment of human skin averted the disease-related shift from quiescent to effector T cell phenotype. Thus, HSP70iQ435A DNA delivery may offer potent treatment opportunities for vitiligo.


Assuntos
Autoimunidade/imunologia , Terapia Genética , Proteínas de Choque Térmico HSP70/uso terapêutico , Hipopigmentação/imunologia , Proteínas Mutantes/uso terapêutico , Vitiligo/imunologia , Vitiligo/terapia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos/imunologia , Células Dendríticas/imunologia , Progressão da Doença , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Inflamação/patologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mutantes/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Fenótipo , Pele/imunologia , Pele/patologia , Linfócitos T/imunologia , Transcrição Gênica , Transfecção , Vacinação , Vitiligo/patologia
15.
PLoS One ; 8(2): e55892, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23405232

RESUMO

BACKGROUND: Immunotherapy has been used to improve patient immune function, inhibit tumor growth and has become a highly promising method of cancer treatment. Highly agglutinative staphylococcin (HAS), a mixture of Staphylococcus aureus culture filtrates, which include staphylococcal enterotoxin (SE) C as the active ingredient, has been used clinically as an immunomodifier in the treatment of a number of tumors for many years. However, the use of HAS has been associated with some unavoidable side-effects such as fever. Previous studies have shown that SEB stimulates a more potent activation of T lymphocytes than SEC3, and mutations of the histidine residues eliminated the toxicity of SEB. SE mutants with decreased side-effects and/or more potent antitumor activities are required. METHODOLOGY/PRINCIPAL FINDINGS: We built a structural model of the MHC II-SEB-TCR complex and found that a mutation of SEB at Lys173 might decrease the repulsion force between the SEB-TCR, which would facilitate their interaction. From the above results, we designed SEB-H32Q/K173E (mSEB). Analysis of in vitro stimulation of the proliferation of human peripheral blood mononuclear cells (PBMCs), IFN-γ secretion and inhibition of the growth of various tumor cell lines demonstrated that mSEB exhibited higher antitumor activity compared with wild-type SEB (wtSEB). Notably, mSEB inhibited the growth of various tumors at an extremely low concentration with little cytotoxicity against normal cells. Three animal tumor models (C57BL/6 mouse, New Zealand rabbit and a humanized NOD/SCID mouse) were used to evaluate the in vivo immunotherapeutic effects. Compared with wtSEB, mSEB significantly enhanced antitumor effect in more than one animal model with reduced pyrexia toxicity and prolonged the survival of tumor-bearing mice. CONCLUSIONS/SIGNIFICANCE: Our results suggest that SEB-H32Q/K173E retains superantigen (SAg) characteristics and enhances the host immune response to neoplastic diseases while reducing associated pyrogenic toxicity.


Assuntos
Carcinoma Pulmonar de Lewis/terapia , Enterotoxinas/imunologia , Febre/terapia , Genes MHC da Classe II/imunologia , Imunoterapia , Mutação/genética , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/imunologia , Citocinas/metabolismo , Citotoxicidade Imunológica , Enterotoxinas/genética , Enterotoxinas/metabolismo , Feminino , Febre/genética , Febre/imunologia , Genes MHC da Classe II/genética , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Mutantes/uso terapêutico , Coelhos , Receptores de Antígenos de Linfócitos T/genética , Proteínas Recombinantes/uso terapêutico , Staphylococcus aureus/metabolismo , Taxa de Sobrevida
16.
Cytokine ; 61(2): 578-84, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23246116

RESUMO

The cytokine lymphotoxin-α (LTα) is a promising candidate for use in cancer therapy. However, the instability of LTαin vivo and the insufficient levels of tumor necrosis factor receptor 1 (TNFR1)-mediated bioactivity of LTα limit its therapeutic potential. Here, we created LTα mutants with increased TNFR1-mediated bioactivity by using a phage display technique. We constructed a phage library displaying lysine-deficient structural variants of LTα with randomized amino acid residues. After affinity panning, we screened three clones of lysine-deficient LTα mutant, and identified a LTα mutant with TNFR1-mediated bioactivity that was 32 times that of the wild-type LTα (wtLTα). When compared with wtLTα, the selected clone showed augmented affinity to TNFR1 due to slow dissociation rather than rapid association. In contrast, the mutant showed only 4 times the TNFR2-mediated activity of wtLTα. In addition, the LTα mutant strongly and rapidly activated caspases that induce TNFR1-mediated cell death, whereas the mutant and wtLTα activated nuclear factor-kappa B to a similar extent. Our data suggest that the kinetics of LTα binding to TNFR1 play an important role in signal transduction patterns, and a TNFR1-selective LTα mutant with augmented bioactivity would be a superior candidate for cancer therapy.


Assuntos
Linfotoxina-alfa/uso terapêutico , Proteínas Mutantes/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Sequência de Aminoácidos , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Espectroscopia de Ressonância de Spin Eletrônica , Ativação Enzimática/efeitos dos fármacos , Humanos , Ponto Isoelétrico , Cinética , Linfotoxina-alfa/química , Linfotoxina-alfa/farmacologia , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/farmacologia , NF-kappa B/metabolismo , Neoplasias/enzimologia , Ligação Proteica/efeitos dos fármacos , Receptores Tipo I de Fatores de Necrose Tumoral/química
17.
Asian Pac J Cancer Prev ; 13(5): 2121-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22901180

RESUMO

OBJECTIVE: Deoxyribonucleoside kinase of Drosophila melanogaster (Dm-dNK) mutants have been reported to exert suicide gene effects in combined gene/chemotherapy of cancer. Here, we aimed to further evaluate the capacity of the mutanted enzyme and its potential for inhibiting cancer cell growth. METHODS: We altered the sequence of the last 10 amino acids of Dm-dNK to perform site-directed mutagenesis and constructed active site mutanted Dm-dNK (Dm-dNKmut), RT-PCR and western bloting studies were used to reveal the expression of lentivirus mediated Dm-dNKmut in a breast cancer cell line (Bcap37), a gastric cancer cell line (SGC7901) and a colorectal cancer cell line (CCL187). [3H]-labeled substrates were used for enzyme activity assays, cell cytotoxicity was assessed by MTT assays, cell proliferation using a hemocytometer and apoptosis induction by thenannexin-V-FITC labeled FACS method. In vivo, an animal study was set out in which BALB/C nude mice bearing tumors were treated with lentivirus mediated expression of Dm-dNKmut with the pyrimidine nucleoside analog brivudine (BVDU, (E)-5-(2-bromovinyl)-(2-deoxyuridine). RESULTS: The Dm-dNKmut could be stably expressed in the cancer cell lines and retained its enzymatic activity. Moreover, the cells expressing Dm-dNKmut exhibited increased sensitivity in combination with BVDU, with induction of apoptosis in vitro and in vivo. CONCLUSION: These findings underlined the importance of BVDU phosphorylated by Dm-dNKmut in transduced cancer cells and the potential role of Dm-dNKmut as a suicide gene, thus providing the basis for future intensive research for cancer therapy.


Assuntos
Neoplasias da Mama/terapia , Bromodesoxiuridina/análogos & derivados , Neoplasias Colorretais/terapia , Drosophila melanogaster/enzimologia , Lentivirus/genética , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Neoplasias Gástricas/terapia , Sequência de Aminoácidos , Animais , Antivirais/uso terapêutico , Apoptose , Western Blotting , Neoplasias da Mama/genética , Bromodesoxiuridina/uso terapêutico , Proliferação de Células , Neoplasias Colorretais/genética , Terapia Combinada , Feminino , Citometria de Fluxo , Terapia Genética , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/uso terapêutico , Mutação/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proibitinas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Neoplasias Gástricas/genética , Células Tumorais Cultivadas
18.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(3): 313-6, 2011 May.
Artigo em Chinês | MEDLINE | ID: mdl-21826989

RESUMO

OBJECTIVE: To study the antitumor effects and mechanism of cationic liposome-mediated SurvivinT34A mutants in mice with pulmonary metastases of melanoma B16. METHODS: The pulmonary metastases model of B16, a mouse melanoma cell line, was established by the injection of B16 cell suspension via tail vein of C57BL/6 mouse. 3 days later, 18 tumor-bearing mice were randomly divided into 3 groups, which were intravenously administrated with normal saline (100 microL), vector (5 microg DNA, 100 microL) and recombinant plasmid SurvivinT34A mutants (5 microg DNA, 100 microL) every other day for five doses, respectively. After 28 days, the mice were sacrificed, the lung tissue was weighted, and the number of metastasis foci on lung surface was counted and measured. Then, pathological change of lung tissue was studied with HE stainning. The number of apoptotic cells in tumor tissues was assessed by TUNEL assay. RESULTS: Compared with the control, mice treated with mSurvivnT34A had an intact structure of lung, with significant reduction in the number of metastasis foci on lung surface (P < 0.05), and high level of apoptosis in tumor tissue (P < 0. 05). CONCLUSION: Recombinant plasmid SurvivinT34A mutants (pORF9-mSurvivinT34A) may inhibit the formation of pulmonary metastases of melanoma.


Assuntos
Proteínas Inibidoras de Apoptose/uso terapêutico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Melanoma Experimental/patologia , Proteínas Mutantes/uso terapêutico , Proteínas Repressoras/uso terapêutico , Animais , Feminino , Proteínas Inibidoras de Apoptose/genética , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Proteínas Repressoras/genética , Neoplasias Cutâneas/patologia , Survivina
19.
Mol Cancer Ther ; 10(9): 1656-66, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21750221

RESUMO

The fibroblast growth factor (FGF) signaling pathway is a recognized target of cancer therapy. We have developed a strong inhibitor (S252W mutant soluble ectodomain of FGF recptor-2 IIIc, msFGFR2) that binds FGFs and blocks the activation of FGFRs. Thermodynamic binding studies indicated that msFGFR2 bound FGF-2 16.9 times as strongly as wild-type soluble FGFR2IIIc ectodomain (wsFGFR2). It successfully suppressed the growth, angiogenesis, and metastasis of two tumor cell lines in vitro and in vivo, and it potently inhibited cancer cell proliferation but not normal cell proliferation. Therefore, msFGFR2 is a useful probe for FGF-dependent signaling pathways and a potential broad-spectrum antitumor agent.


Assuntos
Antineoplásicos/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/farmacologia , Proteínas Recombinantes/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Estimativa de Kaplan-Meier , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Mutantes/genética , Proteínas Mutantes/farmacologia , Proteínas Mutantes/uso terapêutico , Células NIH 3T3 , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/patologia , Neoplasias/tratamento farmacológico , Neoplasias/mortalidade , Neoplasias/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/uso terapêutico , Ligação Proteica , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/uso terapêutico , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Control Release ; 149(1): 8-14, 2011 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-20036293

RESUMO

Multiple sclerosis (MS) is an inflammatory demyelinating disease, the pathogenesis of which is related to elevated serum levels of tumor necrosis factor-α (TNF). Although anti-TNF therapy has been tested as a potential treatment for MS, no remission of symptoms was observed. Recent reports indicated that the TNFR1 signal was responsible for the pathogenesis of murine experimental autoimmune encephalomyelitis (EAE), while the TNFR2 signal was responsible for recovery of the pathogenesis of EAE. Therefore, selective blocking of TNFR1 appears to be a promising strategy for the treatment of MS. In this regard, we previously succeeded in developing a novel TNFR1-selective antagonistic TNF mutant (R1antTNF) by using phage display technology. Here, we have examined the therapeutic potential of R1antTNF using EAE mice. Treatment with PEGylated R1antTNF (PEG-R1antTNF) significantly improved the clinical score and cerebral demyelination at the onset of EAE. Considerable suppression of Th1 and Th17-type response was also observed in spleen and lymph node cells of mice given PEG-R1antTNF. Moreover, the administration of PEG-R1antTNF suppressed the infiltration of inflammatory cells containing Th1 and Th17 cells into the spinal cord. These results suggest that selective blocking of TNFR1 by PEG-R1antTNF could be an effective therapeutic strategy against MS.


Assuntos
Portadores de Fármacos/química , Encefalomielite Autoimune Experimental/tratamento farmacológico , Proteínas Mutantes/uso terapêutico , Polietilenoglicóis/química , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Fator de Necrose Tumoral alfa/uso terapêutico , Animais , Proliferação de Células/efeitos dos fármacos , Citocinas/sangue , Citocinas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Mutantes/administração & dosagem , Proteínas Mutantes/genética , Mutação , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Medula Espinal/patologia , Baço/efeitos dos fármacos , Baço/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA