Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Genet Metab ; 114(2): 217-25, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25092414

RESUMO

Recombinant human acid sphingomyelinase (rhASM) is being developed as an enzyme replacement therapy for patients with acid sphingomyelinase deficiency (Niemann-Pick disease types A and B), which causes sphingomyelin to accumulate in lysosomes. In the acid sphingomyelinase knock-out (ASMKO) mouse, intravenously administered rhASM reduced tissue sphingomyelin levels in a dose-dependent manner. When rhASM was administered to normal rats, mice, and dogs, no toxicity was observed up to a dose of 30mg/kg. However, high doses of rhASM≥10mg/kg administered to ASMKO mice resulted in unexpected toxicity characterized by cardiovascular shock, hepatic inflammation, adrenal hemorrhage, elevations in ceramide and cytokines (especially IL-6, G-CSF, and keratinocyte chemoattractant [KC]), and death. The toxicity could be completely prevented by the administration of several low doses (3mg/kg) of rhASM prior to single or repeated high doses (≥20mg/kg). These results suggest that the observed toxicity involves the rapid breakdown of large amounts of sphingomyelin into ceramide and/or other toxic downstream metabolites, which are known signaling molecules with cardiovascular and pro-inflammatory effects. Our results suggest that the nonclinical safety assessment of novel therapeutics should include the use of specific animal models of disease whenever feasible.


Assuntos
Cães , Terapia de Reposição de Enzimas , Doença de Niemann-Pick Tipo A/tratamento farmacológico , Esfingomielina Fosfodiesterase/administração & dosagem , Esfingomielina Fosfodiesterase/deficiência , Administração Intravenosa , Glândulas Suprarrenais , Animais , Ceramidas/sangue , Ceramidas/metabolismo , Citocinas/sangue , Citocinas/imunologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Fígado/metabolismo , Fígado/patologia , Lisossomos/metabolismo , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doença de Niemann-Pick Tipo A/metabolismo , Ratos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/toxicidade , Esfingomielina Fosfodiesterase/toxicidade , Esfingomielinas/metabolismo
2.
PLoS One ; 9(1): e85398, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24416401

RESUMO

TGFß is reportedly responsible for accumulation of CD4(+)Foxp3(+) regulatory T cells (Tregs) in tumor. Thus, we treated mouse 4T1 mammary carcinoma with 1D11, a neutralizing anti-TGFß (1,2,3) antibody. The treatment delayed tumor growth, but unexpectedly increased the proportion of Tregs in tumor. In vitro, 1D11 enhanced while TGFß potently inhibited the proliferation of Tregs. To enhance the anti-tumor effects, 1D11 was administered with cyclophosphamide which was reported to eliminate intratumoral Tregs. This combination resulted in long term tumor-free survival of up to 80% of mice, and the tumor-free mice were more resistant to re-challenge with tumor. To examine the phenotype of tumor infiltrating immune cells, 4T1-tumor bearing mice were treated with 1D11 and a lower dose of cyclophosphamide. This treatment markedly inhibited tumor growth, and was accompanied by massive infiltration of IFNγ-producing T cells. Furthermore, this combination markedly decreased the number of splenic CD11b(+)Gr1(+) cells, and increased their expression levels of MHC II and CD80. In a spontaneous 4T1 lung metastasis model with resection of primary tumor, this combination therapy markedly increased the survival of mice, indicating it was effective in reducing lethal metastasis burden. Taken together, our data show that anti-TGFß antibody and cyclophosphamide represents an effective chemoimmunotherapeutic combination.


Assuntos
Anticorpos Neutralizantes/farmacologia , Antineoplásicos Alquilantes/farmacologia , Carcinoma/terapia , Ciclofosfamida/farmacologia , Imunoterapia , Neoplasias Mamárias Experimentais/terapia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Antígeno B7-1/genética , Antígeno B7-1/imunologia , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Carcinoma/imunologia , Carcinoma/mortalidade , Carcinoma/patologia , Linhagem Celular Tumoral , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Expressão Gênica/imunologia , Humanos , Interferon gama/genética , Interferon gama/imunologia , Contagem de Linfócitos , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/imunologia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/mortalidade , Neoplasias Mamárias Experimentais/patologia , Camundongos , Análise de Sobrevida , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Fator de Crescimento Transformador beta/imunologia
3.
Oncoimmunology ; 2(8): e26218, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24179709

RESUMO

We evaluated a neutralizing anti-TGFß antibody (GC1008) in cancer patients with malignant pleura mesothelioma (MPM). The goal of this study was to assess immunoregulatory effects in relation to clinical safety and clinical response. Patients with progressive MPM and 1-2 prior systemic therapies received GC1008 at 3mg/kg IV over 90 min every 21 d as part of an open-label, two-center Phase II trial. Following TGFß blockade therapy, clinical safety and patient survival were monitored along with the effects of anti-TGFß antibodies on serum biomarkers and peripheral blood mononuclear cells (PBMC). Although designed as a larger trial, only 13 patients were enrolled when the manufacturer discontinued further development of the antibody for oncology indications. All participants tolerated therapy. Although partial or complete radiographic responses were not observed, three patients showed stable disease at 3 mo. GC1008 had no effect in the expression of NK, CD4+, or CD8+ T cell activating and inhibitory markers, other than a decrease in the expression of 2B4 and DNAM-1 on NK cells. However, serum from 5 patients showed new or enhanced levels of antibodies against MPM tumor lysates as measured by immunoblotting. Patients who produced anti-tumor antibodies had increased median overall survival (OS) (15 vs 7.5 mo, p < 0.03) compared with those who did not. To our knowledge, these data represent the first immune analysis of TGFß- blockade in human cancer patients.

4.
Endocrinology ; 154(3): 1373-83, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23389953

RESUMO

Thyrogen (thyrotropin alfa for injection), recombinant human TSH (rhTSH), has been successfully used to enhance diagnostic radioiodine scanning and thyroglobulin testing in the follow-up of patients with thyroid cancer and as an adjunctive treatment for radioiodine thyroid remnant ablation. However, the short half-life of rhTSH in the circulation requires a multidose regimen. We developed novel sialic acid-mediated and galactose-mediated conjugation chemistries for targeting polyethylene glycol (PEG) to the three N-linked glycosylation sites on the protein, to prolong plasma half-life by eliminating kidney filtration and potential carbohydrate-mediated clearance. Conjugates of different PEG sizes and copy numbers were screened for reaction yield, TSH receptor binding, and murine phamacokinetics/pharmacodynamics studies. The best performing of these products, a 40-kDa mono-PEGylated sialic acid-mediated conjugate, exhibited a 3.5-fold longer duration of action than rhTSH in rats, as a 5-fold lower affinity was more than compensated by a 23-fold extension of circulation half-life. Biochemical characterization confirmed conjugation through the sialic acids. Correlation of PEG distribution on the three N-linked glycosylation sites and the PEG effect on receptor binding supported the previously reported structure-function relationship of rhTSH glycosylation. This long-acting rhTSH has the potential to significantly improve patient convenience and provider flexibility while reducing potential side effects associated with a sudden elevation of serum TSH.


Assuntos
Tireotropina/química , Tireotropina/farmacologia , Animais , Carboidratos/química , Feminino , Glicosilação , Meia-Vida , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Modelos Moleculares , Polietilenoglicóis/química , Ratos , Ratos Sprague-Dawley , Receptores da Tireotropina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Ácidos Siálicos/química , Tireotropina/análogos & derivados , Tireotropina/farmacocinética
5.
Bioconjug Chem ; 24(3): 408-18, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23350694

RESUMO

Recombinant human thyroid stimulating hormone (rhTSH or Thyrogen) has been approved for thyroid cancer diagnostics and treatment under a multidose regimen due to its short circulating half-life. To reduce dosing frequency, PEGylation strategies were explored to increase the duration of action of rhTSH. Lysine and N-terminal PEGylation resulted in heterogeneous product profiles with 40% or lower reaction yields of monoPEGylated products. Eleven cysteine mutants were designed based on a structure model of the TSH-TSH receptor (TSHR) complex to create unique conjugation sites on both α and ß subunits for site-specific conjugation. Sequential screening of mutant expression level, oligomerization tendency, and conjugation efficiency resulted in the identification of the αG22C rhTSH mutant for stable expression and scale-up PEGylation. The introduced cysteine in the αG22C rhTSH mutant was partially blocked when isolated from conditioned media and could only be effectively PEGylated after mild reduction with cysteine. This produced a higher reaction yield, ~85%, for the monoPEGylated product. Although the mutation had no effect on receptor binding, PEGylation of αG22C rhTSH led to a PEG size-dependent decrease in receptor binding. Nevertheless, the 40 kDa PEG αG22C rhTSH showed a prolonged duration of action compared to rhTSH in a rat pharmacodynamics model. Reverse-phase HPLC and N-terminal sequencing experiments confirmed site-specific modification at the engineered Cys 22 position on the α-subunit. This work is another demonstration of successful PEGylation of a cysteine-knot protein by an engineered cysteine mutation.


Assuntos
Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Tireotropina/administração & dosagem , Tireotropina/química , Sequência de Aminoácidos , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos , Ratos Sprague-Dawley , Tireotropina/genética , Fatores de Tempo
6.
Biotechnol Bioeng ; 110(5): 1342-53, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23184768

RESUMO

The prevention of adventitious agent contamination is a top priority throughout the entire biopharmaceutical production process. For example, although viral contamination of cell banks or cell cultures is rare, it can result in serious consequences (e.g., shutdown and decontamination of manufacturing facilities). To ensure virus free production, numerous in vivo and in vitro adventitious agent assays and biophysical characterizations such as electron microscopy are conducted on cell banks, raw materials, process materials, and drug substances throughout the manufacturing process. Molecular assays such as PCR and other nucleotide-based techniques are also routinely used for screening and identification of any viral agents. However, modern techniques in protein identification of complex protein mixtures have not yet been effectively integrated throughout the industry into current viral testing strategies. Here, we report the identification and quantitation of Vesivirus 2117 particles in bioreactor fluid from infected Chinese hamster ovary cell cultures by global protein sequencing using mass spectrometry in combination with multi-dimensional liquid-chromatography. Following mass spectrometric data acquisition and rigorous data analysis, six virus specific peptides were identified. These peptides were fragments of two structural proteins, capsid protein pre-cursor (four unique peptides) and small structural protein (two unique peptides), from the same species: Vesivirus 2117. Using stable heavy isotope-labeled peptides as internal standards, we also determined the absolute concentration of Vesivirus particles in the bioreactor fluid and the ratio of two capsid proteins (VP1:VP2) in the particles as approximately 9:1. The positive identification of Vesivirus 2117 was subsequently confirmed by RT-PCR.


Assuntos
Reatores Biológicos/virologia , Biotecnologia/métodos , Técnicas de Cultura de Células/métodos , Vesivirus/isolamento & purificação , Vírion/isolamento & purificação , Sequência de Aminoácidos , Animais , Células CHO , Sobrevivência Celular/fisiologia , Cromatografia por Troca Iônica , Cricetinae , Cricetulus , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Reação em Cadeia da Polimerase , RNA Viral/genética , RNA Viral/metabolismo , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem , Vesivirus/química , Vesivirus/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Vírion/química
7.
Curr Pharm Biotechnol ; 12(12): 2176-89, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21619535

RESUMO

The role of TGF-ß in tumor development and progression is complex. Genetic mutations that disrupt the antiproliferative signaling effects of TGF-ß play a key role in the process of malignant transformation for many types of tumors. Paradoxically, this loss of sensitivity to TGF-ß's inhibitory actions often leads to TGF-ß overexpression by the tumor cells or by normal cells that are recruited to the tumor microenvironment. Elevated concentrations of TGF-ß in the tumor microenvironment have been shown to facilitate tumor growth and metastasis. Numerous published studies have provided evidence that inhibition of TGF-ß using antibodies, soluble receptors and small molecule inhibitors of TGF-ß signal transduction can have beneficial effects in murine models of cancer. Given the pleiotropic nature of TGF-ß and its homeostatic role in numerous biological processes, serious concerns have been expressed regarding the safety of administering TGF-ß antagonists to human patients. Interestingly, the results of numerous animal toxicology studies of TGF-ß antibodies in normal rodents and primates have shown that administration of neutralizing anti-TGF-ß antibodies is well tolerated and any adverse effects were reversible or self-limiting. Likewise, administration of a human anti-TGF-ß antibody (fresolimumab) in three separate human phase 1 clinical trials has also been shown to be well tolerated.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Neoplasias/tratamento farmacológico , Fator de Crescimento Transformador beta/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Modelos Animais de Doenças , Humanos
8.
Mol Genet Metab ; 100(4): 309-15, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20554235

RESUMO

Pompe disease, also known as glycogen storage disease (GSD) type II, is caused by deficiency of lysosomal acid alpha-glucosidase (GAA). The resulting glycogen accumulation causes a spectrum of disease severity ranging from a rapidly progressive course that is typically fatal by 1-2years of age to a more slowly progressive course that causes significant morbidity and early mortality in children and adults. Recombinant human GAA (rhGAA) improves clinical outcomes with variable results. Adjunct therapy that increases the effectiveness of rhGAA may benefit some Pompe patients. Co-administration of the mTORC1 inhibitor rapamycin with rhGAA in a GAA knockout mouse reduced muscle glycogen content more than rhGAA or rapamycin alone. These results suggest mTORC1 inhibition may benefit GSDs that involve glycogen accumulation in muscle.


Assuntos
Doença de Depósito de Glicogênio Tipo II/terapia , Glicogênio/biossíntese , Fatores de Transcrição/antagonistas & inibidores , Envelhecimento/efeitos dos fármacos , Envelhecimento/patologia , Animais , Relação Dose-Resposta a Droga , Terapia de Reposição de Enzimas , Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Doença de Depósito de Glicogênio Tipo II/enzimologia , Glicogênio Sintase/metabolismo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Fosforilação/efeitos dos fármacos , Proteínas , Proteínas Recombinantes/uso terapêutico , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fatores de Transcrição/metabolismo , alfa-Glucosidases/metabolismo , alfa-Glucosidases/uso terapêutico
9.
Int J Cancer ; 126(7): 1666-74, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19830696

RESUMO

Though TGF-beta inhibition enhances antitumor immunity mediated by CD8(+) T cells in several tumor models, it is not always sufficient for rejection of tumors. In this study, to maximize the antitumor effect of TGF-beta blockade, we tested the effect of anti-TGF-beta combined with an irradiated tumor vaccine in a subcutaneous CT26 colon carcinoma tumor model. The irradiated tumor cell vaccine alone in prophylactic setting significantly delayed tumor growth, whereas anti-TGF-beta antibodies alone did not show any antitumor effect. However, tumor growth was inhibited significantly more in vaccinated mice treated with anti-TGF-beta antibodies compared to vaccinated mice without anti-TGF-beta, suggesting that anti-TGF-beta synergistically enhanced irradiated tumor vaccine efficacy. CD8(+) T-cell depletion completely abrogated the vaccine efficacy, and so protection required CD8(+) T cells. Depletion of CD25(+) T regulatory cells led to the almost complete rejection of tumors without the vaccine, whereas anti-TGF-beta did not change the number of CD25(+) T regulatory cells in unvaccinated and vaccinated mice. Though the abrogation of CD1d-restricted NKT cells, which have been reported to induce TGF-beta production by MDSC through an IL-13-IL-4R-STAT6 pathway, partially enhanced antitumor immunity regardless of vaccination, abrogation of the NKT cell-IL-13-IL-4R-STAT-6 immunoregulatory pathway did not enhance vaccine efficacy. Taken together, these data indicated that anti-TGF-beta enhances efficacy of a prophylactic vaccine in normal individuals despite their not having the elevated TGF-beta levels found in patients with cancer and that the effect is not dependent on TGF-beta solely from CD4(+)CD25(+) T regulatory cells or the NKT cell-IL-13-IL-4R-STAT-6 immunoregulatory pathway.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Neoplasias do Colo/prevenção & controle , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Feminino , Citometria de Fluxo , Interleucina-13/fisiologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Interleucina-4/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células T Matadoras Naturais/metabolismo , Receptores de Interleucina-4/metabolismo , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais , Taxa de Sobrevida , Vacinação
10.
Clin Cancer Res ; 15(21): 6560-9, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19861451

RESUMO

PURPOSE: Transforming growth factor-beta (TGF-beta) is an immunosuppressive cytokine, having direct suppressive activity against conventional CD4(+) and CD8(+)T cells and natural killer cells, thereby inhibiting tumor immunosurveillance. Here, we investigated possible synergy between anti-TGF-beta (1D11) and a peptide vaccine on induction of antitumor immunity, and the mechanisms accounting for synergistic efficacy. EXPERIMENTAL DESIGN: The effect of combination treatment with a peptide vaccine and anti-TGF-beta was examined in a subcutaneous TC1 tumor model, as well as the mechanisms of protection induced by this treatment. RESULTS: Anti-TGF-beta significantly and synergistically improved vaccine efficacy as measured by reduction in primary tumor growth, although anti-TGF-beta alone had no impact. The number of tumor antigen-specific CTL with high functional avidity as measured by IFN-gamma production and lytic activity was significantly increased in vaccinated mice by TGF-beta neutralization. Although TGF-beta is known to play a critical role in CD4(+)Foxp3(+) Treg cells, Treg depletion/suppression by an anti-CD25 monoclonal antibody (PC61) before tumor challenge did not enhance vaccine efficacy, and adding anti-TGF-beta did not affect Treg numbers in lymph nodes or tumors or their function. Also, TGF-beta neutralization had no effect on interleukin-17-producing T cells, which are induced by TGF-beta and interleukin-6. Absence of type II NKT cells, which induce myeloid cells to produce TGF-beta, was not sufficient to eliminate all sources of suppressive TGF-beta. Finally, the synergistic protection induced by anti-TGF-beta vaccine augmentation was mediated by CD8(+) T cells since anti-CD8 treatment completely abrogated the effect. CONCLUSIONS: These results suggest that TGF-beta blockade may be useful for enhancing cancer vaccine efficacy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/uso terapêutico , Linfócitos T Citotóxicos/imunologia , Fator de Crescimento Transformador beta/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico , Animais , Linhagem Celular Tumoral , Terapia de Imunossupressão , Camundongos , Camundongos Endogâmicos C57BL
11.
Mol Genet Metab ; 97(1): 35-42, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19231265

RESUMO

Systemic administration of recombinant acid sphingomyelinase (rhASM) into ASM deficient mice (ASMKO) results in hydrolysis of the abnormal storage of sphingomyelin in lysosomes of the liver, spleen and lung. However, the efficiency with which the substrate is cleared from the lung, particularly the alveolar macrophages, appears to be lower than from the other visceral tissues. To determine if delivery of rhASM into the air spaces of the lung could enhance clearance of pulmonary sphingomyelin, enzyme was administered to ASMKO mice by intranasal instillation. Treatment resulted in a significant and dose-dependent reduction in sphingomyelin levels in the lung. Concomitant with this reduction in substrate levels was a decrease in the amounts of the pro-inflammatory cytokine, MIP-1alpha, in the bronchoalveolar lavage fluids and an improvement in lung pathology. Maximal reduction of lung sphingomyelin levels was observed at 7 days post-treatment. However, reaccumulation of the substrate was noted starting at day 14 suggesting that repeated treatments will be necessary to effect a sustained reduction in sphingomyelin levels. In addition to reducing the storage abnormality in the lung, intranasal delivery of rhASM also resulted in clearance of the substrate from the liver and spleen. Hence, pulmonary administration of rhASM may represent an alternative route of delivery to address the visceral pathology associated with ASM deficiency.


Assuntos
Pulmão/metabolismo , Lisossomos/metabolismo , Doenças de Niemann-Pick/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Esfingomielina Fosfodiesterase/administração & dosagem , Esfingomielina Fosfodiesterase/uso terapêutico , Esfingomielinas/metabolismo , Administração Intranasal , Animais , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Feminino , Humanos , Cinética , Fígado/metabolismo , Fígado/patologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doenças de Niemann-Pick/enzimologia , Doenças de Niemann-Pick/metabolismo , Doenças de Niemann-Pick/patologia , Proteínas Recombinantes/administração & dosagem , Esfingomielina Fosfodiesterase/genética , Baço/metabolismo , Baço/patologia
12.
Int Orthop ; 31(6): 753-7, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17611756

RESUMO

Thyroid-stimulating hormone (TSH) and follicle-stimulating hormone (FSH) have both been recently implicated in bone remodelling. Clinical evidence, as well as data from TSH receptor and thyroid hormone receptor knockout mice, suggest that TSH has a direct effect on skeletal homeostasis, although some data are conflicting. Recently, the exogenous administration of TSH has been shown to positively impact bone in oophrectomised rats. These data, along with their potential implications for the treatment of severe osteoporosis, are discussed.


Assuntos
Remodelação Óssea/fisiologia , Hormônio Foliculoestimulante/fisiologia , Tireotropina/fisiologia , Animais , Densidade Óssea/fisiologia , Modelos Animais de Doenças , Camundongos , Osteoporose/fisiopatologia , Ratos
13.
J Bone Miner Res ; 22(6): 849-59, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17352644

RESUMO

UNLABELLED: We show the systemic administration of low levels of TSH increases bone volume and improves bone microarchitecture and strength in aged OVX rats. TSH's actions are mediated by its inhibitory effects on RANKL-induced osteoclast formation and bone resorption coupled with stimulatory effects on osteoblast differentiation and bone formation, suggesting TSH directly affects bone remodeling in vivo. INTRODUCTION: Thyroid-stimulating hormone (TSH) receptor haploinsufficient mice with normal circulating thyroid hormone levels have reduced bone mass, suggesting that TSH directly affects bone remodeling. We examined whether systemic TSH administration restored bone volume in aged ovariectomized (OVX) rats and influenced osteoclast formation and osteoblast differentiation in vitro. MATERIALS AND METHODS: Sprague-Dawley rats were OVX at 6 months, and TSH therapy was started immediately after surgery (prevention mode; n = 80) or 7 mo later (restoration mode; n = 152). Hind limbs and lumbar spine BMD was measured at 2- or 4-wk intervals in vivo and ex vivo on termination at 8-16 wk. Long bones were subjected to microCT, histomorphometric, and biomechanical analyses. The direct effect of TSH was examined in osteoclast and osteoblast progenitor cultures and established rat osteosarcoma-derived osteoblastic cells. Data were analyzed by ANOVA Dunnett test. RESULTS: In the prevention mode, low doses (0.1 and 0.3 microg) of native rat TSH prevented the progressive bone loss, and importantly, did not increase serum triiodothyroxine (T3) and thyroxine (T4) levels in aged OVX rats. In restoration mode, animals receiving 0.1 and 0.3 microg TSH had increased BMD (10-11%), trabecular bone volume (100-130%), trabecular number (25-40%), trabecular thickness (45-60%), cortical thickness (5-16%), mineral apposition and bone formation rate (200-300%), and enhanced mechanical strength of the femur (51-60%) compared with control OVX rats. In vitro studies suggest that TSH's action is mediated by its inhibitory effects on RANKL-induced osteoclast formation, as shown in hematopoietic stem cells cultivated from TSH-treated OVX rats. TSH also stimulates osteoblast differentiation, as shown by effects on alkaline phosphatase activity, osteocalcin expression, and mineralization rate. CONCLUSIONS: These results show for the first time that systemically administered TSH prevents bone loss and restores bone mass in aged OVX rats through both antiresorptive and anabolic effects on bone remodeling.


Assuntos
Osso e Ossos/efeitos dos fármacos , Ovariectomia , Tireotropina/farmacologia , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Fenômenos Biomecânicos , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/anatomia & histologia , Osso e Ossos/química , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Fêmur/anatomia & histologia , Fêmur/química , Fêmur/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Membro Posterior/química , Membro Posterior/efeitos dos fármacos , Vértebras Lombares/química , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/fisiologia , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteocalcina/genética , Osteoclastos/química , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoporose/fisiopatologia , Osteoporose/prevenção & controle , Ratos , Ratos Sprague-Dawley , Tireotropina/uso terapêutico , Tiroxina/sangue , Tíbia/química , Tíbia/efeitos dos fármacos , Tíbia/fisiologia , Tri-Iodotironina/sangue
15.
J Cell Physiol ; 196(1): 70-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12767042

RESUMO

Adult human skeletal muscle-derived cells (HuSkMC) propagated in vitro are under investigation as a cell-based therapy for the treatment of myocardial infarction. We have characterized HuSkMC with respect to cell identity and state of differentiation as a prerequisite to their clinical use. Flow cytometric analysis of propagated HuSkMC revealed a population of cells that expressed the myoblast markers CD56 and desmin. The presence of myoblasts in these cultures was further confirmed by their capacity to form myotubes and increase creatine kinase activity when cultured in low serum conditions. The non-myoblast fraction of these propagated cells expressed TE7, a marker associated with the fibroblast phenotype. Spontaneous differentiation of myoblasts occurred during serial propagation of HuSkMC, as judged by myotube formation, thereby reducing the myoblast representative fraction with continued cell expansion. We examined transforming growth factor beta2 (TGF-beta2) for its utility in controlling this spontaneous differentiation of adult human myoblasts in vitro. Propagation of HuSkMC in the presence of 1 ng/ml TGF-beta2 for 5 days decreased desmin expression within the myoblast population and caused a parallel reduction of creatine kinase activity. CD56 expression was unaffected, indicating a differential regulation of these myoblast markers. The reduction in desmin expression and creatine kinase activity was, however, reversible upon the removal of TGF-beta. These data collectively indicate that TGF-beta2 restrained differentiation of adult human skeletal myoblasts during propagation without causing irreversible loss of the myoblast phenotype, demonstrating the potential utility of using TGF-beta2 during cultivation and expansion of HuSkMC intended for therapeutic implantation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Adulto , Idoso , Biomarcadores/análise , Antígeno CD56/metabolismo , Divisão Celular , Células Cultivadas , Creatina Quinase/metabolismo , Desmina/metabolismo , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Mioblastos Esqueléticos/enzimologia , Mioblastos Esqueléticos/metabolismo , Fator de Crescimento Transformador beta2
16.
Glycobiology ; 13(4): 305-13, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12626384

RESUMO

Fabry disease is a lysosomal storage disease arising from deficiency of the enzyme alpha-galactosidase A. Two recombinant protein therapeutics, Fabrazyme (agalsidase beta) and Replagal (agalsidase alfa), have been approved in Europe as enzyme replacement therapies for Fabry disease. Both contain the same human enzyme, alpha-galactosidase A, but they are produced using different protein expression systems and have been approved for administration at different doses. To determine if there is recognizable biochemical basis for the different doses, we performed a comparison of the two drugs, focusing on factors that are likely to influence biological activity and availability. The two drugs have similar glycosylation, both in the type and location of the oligosaccharide structures present. Differences in glycosylation were mainly limited to the levels of sialic acid and mannose-6-phosphate present, with Fabrazyme having a higher percentage of fully sialylated oligosaccharides and a higher level of phosphorylation. The higher levels of phosphorylated oligomannose residues correlated with increased binding to mannose-6-phosphate receptors and uptake into Fabry fibroblasts in vitro. Biodistribution studies in a mouse model of Fabry disease showed similar organ uptake. Likewise, antigenicity studies using antisera from Fabry patients demonstrated that both drugs were indistinguishable in terms of antibody cross-reactivity. Based on these studies and present knowledge regarding the influence of glycosylation on protein biodistribution and cellular uptake, the two protein preparations appear to be functionally indistinguishable. Therefore, the data from these studies provide no rationale for the use of these proteins at different therapeutic doses.


Assuntos
Isoenzimas/farmacocinética , alfa-Galactosidase/farmacocinética , Animais , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Relação Dose-Resposta a Droga , Doença de Fabry/tratamento farmacológico , Doença de Fabry/enzimologia , Humanos , Imuno-Histoquímica , Isoenzimas/genética , Isoenzimas/farmacologia , Cinética , Camundongos , Camundongos Knockout , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Espectrometria de Massas por Ionização por Electrospray , Fatores de Tempo , Distribuição Tecidual , alfa-Galactosidase/genética , alfa-Galactosidase/farmacologia
17.
Wound Repair Regen ; 10(4): 252-8, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12191008

RESUMO

Tensile strength of 2-cm, full-thickness, surgically incised porcine skin wounds sealed with fibrin sealant was enhanced compared to conventionally sutured wounds at 6 hours postwounding, but was significantly reduced after 3 days. Supplementation of fibrin sealant with transforming growth factor-beta2 (TGF-beta2) reversed the inhibitory effects of fibrin sealant on tensile strength at 3 days, and enhanced tensile strength at 7 days compared to suture or fibrin sealant alone. By 14 days, the tensile strengths of all wounds were similar, although wounds treated with fibrin sealant supplemented with TGF-beta2 showed a small, but statistically significant, improvement in wound strength compared to wounds treated with fibrin sealant alone. Histological assessment at day 7 revealed significant remnants of fibrin sealant at the wound site following fibrin sealant treatment alone, while wounds treated with fibrin sealant supplemented with TGF-beta2 or suture exhibited fibroblast infiltration and extracellular matrix deposition. At day 7, TGF-beta was immunolocalized in the base and margins of only wounds treated with fibrin sealant supplemented with TGF-beta2. A significant increase in matrix metalloproteinase-9 activity was found in fibrin sealant-treated wounds at day 7 as compared to sutured wounds. Addition of TGF-beta to the fibrin sealant suppressed the up-regulation of matrix metalloproteinase-9 in these wounds. These results suggest that fibrin sealant supplemented with TGF-beta may provide superior wound healing as compared to fibrin sealant alone.


Assuntos
Adesivo Tecidual de Fibrina/uso terapêutico , Imunossupressores/uso terapêutico , Pele/lesões , Pele/fisiopatologia , Adesivos Teciduais/uso terapêutico , Fator de Crescimento Transformador beta/uso terapêutico , Cicatrização/fisiologia , Ferimentos Penetrantes/fisiopatologia , Ferimentos Penetrantes/terapia , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Adesivo Tecidual de Fibrina/administração & dosagem , Imunossupressores/administração & dosagem , Imunossupressores/farmacologia , Pele/efeitos dos fármacos , Suínos , Resistência à Tração/efeitos dos fármacos , Resistência à Tração/fisiologia , Adesivos Teciduais/administração & dosagem , Fator de Crescimento Transformador beta/administração & dosagem , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta2 , Falha de Tratamento , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...