Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell Rep ; 32(2): 107886, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32668256

RESUMO

Excessive iron accumulation in the heart causes iron overload cardiomyopathy (IOC), which initially presents as diastolic dysfunction and arrhythmia but progresses to systolic dysfunction and end-stage heart failure when left untreated. However, the mechanisms of iron-related cardiac injury and how iron accumulates in human cardiomyocytes are not well understood. Herein, using human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), we model IOC and screen for drugs to rescue the iron overload phenotypes. Human iPSC-CMs under excess iron exposure recapitulate early-stage IOC, including oxidative stress, arrhythmia, and contractile dysfunction. We find that iron-induced changes in calcium kinetics play a critical role in dysregulation of CM functions. We identify that ebselen, a selective divalent metal transporter 1 (DMT1) inhibitor and antioxidant, could prevent the observed iron overload phenotypes, supporting the role of DMT1 in iron uptake into the human myocardium. These results suggest that ebselen may be a potential preventive and therapeutic agent for treating patients with secondary iron overload.


Assuntos
Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/patologia , Modelos Biológicos , Miócitos Cardíacos/patologia , Arritmias Cardíacas/complicações , Arritmias Cardíacas/fisiopatologia , Azóis/farmacologia , Cálcio/metabolismo , Cardiomiopatias/fisiopatologia , Linhagem Celular , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Ferro/metabolismo , Isoindóis , Cinética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Contração Miocárdica/efeitos dos fármacos , Compostos Organosselênicos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Fatores de Tempo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
2.
Yonsei Med J ; 60(1): 88-97, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30554495

RESUMO

PURPOSE: Sodium chloride (NaCl) has been proposed as a driving factor in autoimmune diseases through the induction of pathogenic CD4+ T helper cells that produce interleukin-17 (Th17 cells). This study investigated the effects of NaCl on inflammatory arthritis in mice and humans. MATERIALS AND METHODS: Collagen-induced arthritis (CIA) mice were fed a normal or high-salt diet ad libitum, and clinical and histologic features of arthritis were evaluated. The proportion of Th17 cells in the spleens of CIA mice fed a normal or high-salt diet was evaluated by flow cytometry, and the expression of IL-17 in joints and intestines was determined by immunohistochemical staining. We also analyzed the effect of NaCl on Th17 differentiation from peripheral blood monocytes of patients with rheumatoid arthritis (RA) and osteoarthritis (OA) and evaluated the contents of sodium and IL-17 in the synovial fluid of RA and OA patients. RESULTS: NaCl increased murine and human Th17 cell differentiation in a dose-dependent manner. Clinical and histological arthritis was more severe in the high-salt-fed CIA mice, compared to control CIA mice. The proportion of Th17 cells among splenocytes was higher in CIA mice fed a high-salt diet. Expression of synovial and intestinal IL-17 was also higher in high-salt-fed CIA mice. Comparison of synovial fluid between RA patients and OA patients revealed that Na+ and IL-17 were more abundant in RA synovial fluid. CONCLUSION: This study suggests that NaCl can aggravate arthritis by affecting Th17 differentiation. Accordingly, limiting salt intake may be helpful for treating inflammatory arthritis, such as RA.


Assuntos
Artrite Experimental/imunologia , Artrite Experimental/patologia , Polaridade Celular , Cloreto de Sódio/efeitos adversos , Células Th17/imunologia , Animais , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Diferenciação Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Humanos , Inflamação/patologia , Interleucina-17/biossíntese , Intestinos/efeitos dos fármacos , Intestinos/patologia , Masculino , Camundongos , Cloreto de Sódio na Dieta/efeitos adversos , Líquido Sinovial/metabolismo , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/patologia , Células Th17/efeitos dos fármacos
3.
Cell Stem Cell ; 22(4): 501-513.e7, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29456158

RESUMO

Cancer cells and embryonic tissues share a number of cellular and molecular properties, suggesting that induced pluripotent stem cells (iPSCs) may be harnessed to elicit anti-tumor responses in cancer vaccines. RNA sequencing revealed that human and murine iPSCs express tumor-associated antigens, and we show here a proof of principle for using irradiated iPSCs in autologous anti-tumor vaccines. In a prophylactic setting, iPSC vaccines prevent tumor growth in syngeneic murine breast cancer, mesothelioma, and melanoma models. As an adjuvant, the iPSC vaccine inhibited melanoma recurrence at the resection site and reduced metastatic tumor load, which was associated with fewer Th17 cells and increased CD11b+GR1hi myeloid cells. Adoptive transfer of T cells isolated from vaccine-treated tumor-bearing mice inhibited tumor growth in unvaccinated recipients, indicating that the iPSC vaccine promotes an antigen-specific anti-tumor T cell response. Our data suggest an easy, generalizable strategy for multiple types of cancer that could prove highly valuable in clinical immunotherapy.


Assuntos
Neoplasias da Mama/imunologia , Vacinas Anticâncer/imunologia , Células-Tronco Pluripotentes Induzidas/imunologia , Melanoma/imunologia , Mesotelioma/imunologia , Animais , Neoplasias da Mama/terapia , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Melanoma/terapia , Mesotelioma/terapia , Camundongos
4.
Sci Rep ; 7: 39593, 2017 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-28084468

RESUMO

Mesenchymal stem cells (MSCs) have multiple properties including anti-inflammatory and immunomodulatory effects in various disease models and clinical treatments. These beneficial effects, however, are sometimes inconsistent and unpredictable. For wider and proper application, scientists sought to improve MSC functions by engineering. We aimed to invent a novel method to produce synthetic biological drugs from engineered MSCs. We investigated the anti-arthritic effect of engineered MSCs in a collagen-induced arthritis (CIA) model. Biologics such as etanercept are the most successful drugs used in anti-cytokine therapy. Biologics are made of protein components, and thus can be theoretically produced from cells including MSCs. MSCs were transfected with recombinant minicircles encoding etanercept (trade name, Enbrel), which is a tumour necrosis factor α blocker currently used to treat rheumatoid arthritis. We confirmed minicircle expression in MSCs in vitro based on GFP. Etanercept production was verified from the conditioned media. We confirmed that self-reproduced etanercept was biologically active in vitro. Arthritis subsided more efficiently in CIA mice injected with mcTNFR2MSCs than in those injected with conventional MSCs or etanercept only. Although this novel strategy is in a very early conceptual stage, it seems to represent a potential alternative method for the delivery of biologics and engineering MSCs.


Assuntos
Antirreumáticos/metabolismo , Artrite/terapia , Colágeno/administração & dosagem , Etanercepte/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Artrite/induzido quimicamente , Artrite Experimental/induzido quimicamente , Artrite Experimental/terapia , Produtos Biológicos/administração & dosagem , Produtos Biológicos/metabolismo , DNA Circular/metabolismo , Sistemas de Liberação de Medicamentos , Vetores Genéticos , Camundongos Transgênicos
5.
Sci Rep ; 6: 32669, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27609119

RESUMO

Cardiovascular disease is a leading cause of morbidity in rheumatoid arthritis (RA) patients. This study aimed to generate and characterise cardiomyocytes from induced pluripotent stem cells (iPSCs) of RA patients. Fibroblast-like synoviocytes (FLSs) from patients with RA and osteoarthritis (OA) were successfully reprogrammed into RA-iPSCs and OA-iPSCs, respectively. The pluripotency of iPSCs was confirmed by quantitative reverse transcription-polymerase chain reaction and immunofluorescence staining. Established iPSCs were differentiated into cardiomyocytes using a small molecule-based monolayer differentiation protocol. Within 12 days of cardiac differentiation from patient-specific and control-iPSCs, spontaneously beating cardiomyocytes (iPSC-CMs) were observed. All iPSC-CMs exhibited a reliable sarcomeric structure stained with antibodies against cardiac markers and similar expression profiles of cardiac-specific genes. Intracellular calcium signalling was recorded to compare calcium-handling properties among cardiomyocytes differentiated from the three groups of iPSCs. RA-iPSC-CMs had a lower amplitude and a shorter duration of calcium transients than the control groups. Peak tangential stress and the maximum contractile rate were also decreased in RA-iPSC-CMs, suggesting that contractility was reduced. This study demonstrates the successful generation of functional cardiomyocytes from pathogenic synovial cells in RA patients through iPSC reprogramming. Research using RA-iPSC-CMs might provide an opportunity to investigate the pathophysiology of cardiac involvement in RA.


Assuntos
Artrite Reumatoide/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Miócitos Cardíacos/patologia , Sinoviócitos/patologia , Diferenciação Celular , Humanos
6.
Stem Cells Int ; 2016: 1329459, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27579041

RESUMO

Human induced pluripotent stem cells (hiPSCs) have demonstrated great potential for differentiation into diverse tissues. We report a straightforward and highly efficient method for the generation of iPSCs from PBMCs. By plating the cells serially to a newly coated plate by centrifugation, this protocol provides multiple healthy iPSC colonies even from a small number of PBMCs. The generated iPSCs expressed pluripotent markers and differentiated into all three germ layer lineages. The protocol can also be used with umbilical cord blood mononuclear cells (CBMCs). In this study, we present a simple and efficient protocol that improved the yield of iPSCs from floating cells such as PBMCs and CBMCs by serial plating and centrifugation.

7.
Sci Rep ; 6: 20150, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26841833

RESUMO

A considerable proportion of patients with rheumatoid arthritis (RA) do not respond to monospecific agents. The purpose of our study was to generate a hybrid form of biologics, targeting tumor-necrosis factor alpha (TNFα) and interleukin-6 receptor (IL-6R), and determine its anti-arthritic properties in vitro and in vivo. A novel dual target-directed agent (DTA(A7/sTNFR2)) was generated by conjugating soluble TNF receptor 2 (sTNFR2) to the Fc region of A7, a new anti-IL-6R antibody obtained by screening the phage display human antibody library. DTA(A7/sTNFR2) inhibited the proliferation and migration of fibroblast-like synoviocytes from patients with RA (RA-FLS) more efficiently than single target-directed agents. DTA(A7/sTNFR2) also blocked osteoclastogenesis from bone marrow cells. The arthritis severity scores of the experimental arthritis mice with DTA(A7/sTNFR2) tended to be lower than those of mice with IgG, A7, or sTNFR2. Histological data suggested that DTA(A7/sTNFR2) is more efficient than single-target drugs in preventing joint destruction and bone loss. These results were confirmed in vivo using the minicircle system. Taken together, the results show that DTA(A7/sTNFR2) may be a promising therapeutic agent for the treatment of RA.


Assuntos
Anticorpos/metabolismo , Artrite Experimental/patologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Receptores de Interleucina-6/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Animais , Anticorpos/genética , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células da Medula Óssea/citologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Camundongos , Camundongos Endogâmicos DBA , Osteogênese/efeitos dos fármacos , Biblioteca de Peptídeos , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Sinoviócitos/citologia , Sinoviócitos/metabolismo
8.
Clin Transl Immunology ; 4(11): e51, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26682058

RESUMO

This study was undertaken to develop a novel anti-citrullinated peptide antibody (ACPA) and to investigate its arthritogenicity in a collagen-induced arthritis (CIA) model. The novel ACPA, 12G1, was developed by injecting cyclic citrullinated antigen in mice and subsequently hybridizing the B cells producing citrullinated peptide-specific antibodies with a myeloma cell line. The arthritic joints of mice with CIA and collagen antibody-induced arthritis (CAIA) as well as interleukin-1 receptor antagonist (IL-1Ra) knockout (KO) mice were stained immunohistochemically using the 12G1 antibody. Confocal immunostaining was used to identify colocalization of 12G1 with various citrullinated proteins. 12G1 in the presence or absence of chelating beads was administered to CIA mice on days 21 and 28 after type II collagen (CII) immunization to investigate 12G1 arthritogenecity. 12G1 detected citrullinated proteins in the arthritic joints of all the experimental arthritis models used. Confocal immunostaining showed that 12G1 was colocalized with well-known citrullinated proteins, including vimentin, collagen, anti-immunoglobulin binding protein and fibronectin. Staining of citrullinated proteins using 12G1 was more diffuse in CIA mice compared with CAIA and IL-1Ra KO mice. 12G1 injection apparently acted as a booster of immunization in CIA mice in combination with a single CII immunization, with this effect being abolished when 12G1 was injected with chelating beads. The novel ACPA, 12G1, identified various citrullinated proteins in the arthritic joints of three experimental arthritis models. 12G1-treated mice developed arthritis following a single CII immunization, suggesting an arthritogenic potential for ACPA in CIA mice.

9.
BMC Musculoskelet Disord ; 16: 170, 2015 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-26210906

RESUMO

BACKGROUND: Adipose-derived stem cells (ASCs) are mesenchymal stem cells (MSCs) derived from adipose tissue. MSCs have multiple properties including anti-inflammatory and immunomodulatory effects in various disease models and human diseases. However, the mechanisms underlying this wide range of effects need to be explored. METHODS: Collagen antibody-induced arthritis (CAIA) is a unique model in which arthritis is rapidly and strongly induced. ASCs were intraperitoneally infused into CAIA mice before or after arthritis induction. The serum levels of various cytokines, adipokines, and chemokines were measured. The expression of FC gamma receptors (FCGRs) was investigated in peritoneal macrophages ex vivo. RAW264.7 cells and ASCs were co-cultured to elucidate the direct and indirect role of ASCs on FCGR expression. RESULTS: ASCs attenuated arthritis in CAIA mice. Serum levels of tumor necrosis factor α, interleukin (IL)-15, resistin, and leptin were reduced in ASC-treated CAIA mice, whereas serum levels of IL-6 and adiponectin were not affected. In peritoneal macrophages isolated from ASC-treated mice, expression of FCGRIIB, which is immunoinhibitory, was higher than that of FCGRI. Co-culture of ASCs with RAW264.7 cells modulated the expression of FCGRs. The expression patterns and timings of peak expression differed among FCGRs. Expression of FCGRIIB was higher and peaked earlier than that of FCGRI. FCGRIII expression was not affected by this co-culture. CONCLUSIONS: This is a study to show that ASCs have anti-arthritic effects in CAIA mice. Modulation of FCGRs by ASCs might be a therapeutic mechanism in this antibody-associated arthritis model.


Assuntos
Tecido Adiposo/metabolismo , Tecido Adiposo/transplante , Artrite Experimental/metabolismo , Artrite Experimental/prevenção & controle , Transplante de Células-Tronco Mesenquimais/métodos , Receptores de IgG/biossíntese , Tecido Adiposo/citologia , Animais , Linhagem Celular , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Receptores de IgG/genética
10.
J Korean Med Sci ; 30(3): 233-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25729243

RESUMO

Eupatilin is the main active component of DA-9601, an extract from Artemisia. Recently, eupatilin was reported to have anti-inflammatory properties. We investigated the anti-arthritic effect of eupatilin in a murine arthritis model and human rheumatoid synoviocytes. DA-9601 was injected into collagen-induced arthritis (CIA) mice. Arthritis score was regularly evaluated. Mouse monocytes were differentiated into osteoclasts when eupatilin was added simultaneously. Osteoclasts were stained with tartrate-resistant acid phosphatase and then manually counted. Rheumatoid synoviocytes were stimulated with TNF-α and then treated with eupatilin, and the levels of IL-6 and IL-1ß mRNA expression in synoviocytes were measured by RT-PCR. Intraperitoneal injection of DA-9601 reduced arthritis scores in CIA mice. TNF-α treatment of synoviocytes increased the expression of IL-6 and IL-1ß mRNAs, which was inhibited by eupatilin. Eupatilin decreased the number of osteoclasts in a concentration dependent manner. These findings, showing that eupatilin and DA-9601 inhibited the expression of inflammatory cytokines and the differentiation of osteoclasts, suggest that eupatilin and DA-9601 is a candidate anti-inflammatory agent.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite Experimental/tratamento farmacológico , Diferenciação Celular/efeitos dos fármacos , Flavonoides/uso terapêutico , Osteoclastos/citologia , Animais , Anti-Inflamatórios/farmacologia , Artrite Experimental/induzido quimicamente , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Células Cultivadas , Colágeno Tipo II , Citocinas/biossíntese , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/uso terapêutico , Feminino , Flavonoides/farmacologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos DBA , Monócitos/citologia , Extratos Vegetais/farmacologia , RNA Mensageiro/biossíntese , Membrana Sinovial/citologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/farmacologia
11.
Sci Rep ; 4: 6935, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25374010

RESUMO

Cytotoxic T lymphocyte-associated antigen 4 immunoglobulin fusion protein (CTLA4Ig, abatacept) is a B7/CD28 costimulation inhibitor that can ward off the immune response by preventing the activation of naïve T cells. This therapeutic agent is administered to patients with autoimmune diseases such as rheumatoid arthritis. Its antiarthritic efficacy is satisfactory, but the limitations are the necessity for frequent injection and high cost. Minicircles can robustly express the target molecule and excrete it outside the cell as an indirect method to produce the protein of interest in vivo. We inserted the sequence of abatacept into the minicircle vector, and by successful in vivo injection the host was able to produce the synthetic protein drug. Intravenous infusion of the minicircle induced spontaneous production of CTLA4Ig in mice with collagen-induced arthritis. Self-produced CTLA4Ig significantly decreased the symptoms of arthritis. Injection of minicircle CTLA4Ig regulated Foxp3(+) T cells and Th17 cells. Parental and mock vectors did not ameliorate arthritis or modify the T cell population. We have developed a new concept of spontaneous protein drug delivery using a minicircle vector. Self in vivo production of a synthetic protein drug may be useful when biological drugs cannot be injected because of manufacturing or practical problems.


Assuntos
Antirreumáticos/farmacologia , Artrite Experimental/terapia , DNA Circular/biossíntese , Vetores Genéticos/biossíntese , Imunoconjugados/farmacologia , Imunossupressores/farmacologia , Abatacepte , Animais , Antirreumáticos/imunologia , Antirreumáticos/metabolismo , Artrite Experimental/genética , Artrite Experimental/imunologia , Artrite Experimental/patologia , DNA Circular/administração & dosagem , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Expressão Gênica , Vetores Genéticos/administração & dosagem , Imunoconjugados/imunologia , Imunoconjugados/metabolismo , Imunossupressores/imunologia , Imunossupressores/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos DBA , Terapia de Alvo Molecular , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia
12.
Sci Rep ; 4: 5961, 2014 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25091294

RESUMO

Biologics are the most successful drugs used in anticytokine therapy. However, they remain partially unsuccessful because of the elevated cost of their synthesis and purification. Development of novel biologics has also been hampered by the high cost. Biologics are made of protein components; thus, theoretically, they can be produced in vivo. Here we tried to invent a novel strategy to allow the production of synthetic drugs in vivo by the host itself. The recombinant minicircles encoding etanercept or tocilizumab, which are synthesized currently by pharmaceutical companies, were injected intravenously into animal models. Self-reproduced etanercept and tocilizumab were detected in the serum of mice. Moreover, arthritis subsided in mice that were injected with minicircle vectors carrying biologics. Self-reproducible biologics need neither factory facilities for drug production nor clinical processes, such as frequent drug injection. Although this novel strategy is in its very early conceptual stage, it seems to represent a potential alternative method for the delivery of biologics.


Assuntos
Anticorpos Monoclonais Humanizados/biossíntese , Artrite Experimental/terapia , Produtos Biológicos/imunologia , DNA Circular/imunologia , Vetores Genéticos/imunologia , Imunoglobulina G/biossíntese , Receptores do Fator de Necrose Tumoral/biossíntese , Animais , Anticorpos Monoclonais Humanizados/genética , Anticorpos Monoclonais Humanizados/imunologia , Antirreumáticos/química , Antirreumáticos/imunologia , Antirreumáticos/metabolismo , Artrite Experimental/genética , Artrite Experimental/imunologia , Artrite Experimental/patologia , Produtos Biológicos/administração & dosagem , Produtos Biológicos/química , DNA Circular/administração & dosagem , DNA Circular/genética , Etanercepte , Feminino , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/química , Células HEK293 , Membro Posterior , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos DBA , Receptores de Interleucina-6/antagonistas & inibidores , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/imunologia , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Resultado do Tratamento
13.
Mol Med Rep ; 9(6): 2301-8, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24718553

RESUMO

Etanercept is a widespread biological drug for the treatment of rheumatoid arthritis, which inhibits tumor necrosis factor-α (TNF-α). Recently, the presence of antibodies targeting TNF-α inhibitors such as infliximab and adalimumab, was reported. However, few reports have studied etanercept in a mouse model of arthritis. We investigated the induction of anti-etanercept antibody production, along with the antibody's potential interfering effects on the biological function of etanercept, in mice with collagen-induced arthritis (CIA). CIA mice received an intraperitoneal injection of etanercept (25, 100 or 400 µg per mouse). The degree of inflammation and cartilage erosion was evaluated, and the number of osteoclasts in the ankle joints was assessed by TRAP staining. The level of pro-inflammatory cytokines in the serum was measured. To analyze the anti-osteoporotic effect of etanercept, microfocal computed tomography analyses of femurs and tibias were performed. Etanercept treatment decreased both the incidence and severity of arthritis in a dose-dependent manner, except for the highest dose of 400 µg. The mice that were treated with 25 and 100 µg etanercept showed an improvement in inflammation, cartilage damage, and even bone loss. However, mice treated with 400 µg etanercept showed no significant improvement in any of the tested parameters. Using a customized enzyme-linked immunosorbent assay (ELISA), the presence of the anti-etanercept antibody was detected in the serum in this treatment-refractory group. The therapeutic effect of etanercept was reduced in the CIA mice that developed the anti-etanercept antibody. In conclusion, the production of an anti-etanercept antibody can be induced in CIA mice, and this antibody can considerably reduce the anti-arthritic and anti-osteoporotic effects of etanercept.


Assuntos
Anticorpos/imunologia , Antirreumáticos/imunologia , Artrite Experimental/imunologia , Imunoglobulina G/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Animais , Antirreumáticos/administração & dosagem , Antirreumáticos/farmacologia , Artrite Experimental/tratamento farmacológico , Artrite Experimental/patologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Desmineralização Patológica Óssea/tratamento farmacológico , Citocinas/sangue , Modelos Animais de Doenças , Etanercepte , Imunoglobulina G/administração & dosagem , Imunoglobulina G/farmacologia , Mediadores da Inflamação/sangue , Masculino , Camundongos , Receptores do Fator de Necrose Tumoral/administração & dosagem
14.
Arthritis Res Ther ; 16(1): R41, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24490617

RESUMO

INTRODUCTION: Since the concept of reprogramming mature somatic cells to generate induced pluripotent stem cells (iPSCs) was demonstrated in 2006, iPSCs have become a potential substitute for embryonic stem cells (ESCs) given their pluripotency and "stemness" characteristics, which resemble those of ESCs. We investigated to reprogram fibroblast-like synoviocytes (FLSs) from patients with rheumatoid arthritis (RA) and osteoarthritis (OA) to generate iPSCs using a 4-in-1 lentiviral vector system. METHODS: A 4-in-1 lentiviral vector containing Oct4, Sox2, Klf4, and c-Myc was transduced into RA and OA FLSs isolated from the synovia of two RA patients and two OA patients. Immunohistochemical staining and real-time PCR studies were performed to demonstrate the pluripotency of iPSCs. Chromosomal abnormalities were determined based on the karyotype. SCID-beige mice were injected with iPSCs and sacrificed to test for teratoma formation. RESULTS: After 14 days of transduction using the 4-in-1 lentiviral vector, RA FLSs and OA FLSs were transformed into spherical shapes that resembled embryonic stem cell colonies. Colonies were picked and cultivated on matrigel plates to produce iPSC lines. Real-time PCR of RA and OA iPSCs detected positive markers of pluripotency. Immunohistochemical staining tests with Nanog, Oct4, Sox2, Tra-1-80, Tra-1-60, and SSEA-4 were also positive. Teratomas that comprised three compartments of ectoderm, mesoderm, and endoderm were formed at the injection sites of iPSCs. Established iPSCs were shown to be compatible by karyotyping. Finally, we confirmed that the patient-derived iPSCs were able to differentiate into osteoblast, which was shown by an osteoimage mineralization assay. CONCLUSION: FLSs derived from RA and OA could be cell resources for iPSC reprogramming. Disease- and patient-specific iPSCs have the potential to be applied in clinical settings as source materials for molecular diagnosis and regenerative therapy.


Assuntos
Artrite Reumatoide , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Osteoartrite , Animais , Diferenciação Celular/fisiologia , Fibroblastos/citologia , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Camundongos SCID , Reação em Cadeia da Polimerase em Tempo Real , Membrana Sinovial/citologia , Transdução Genética
15.
J Korean Med Sci ; 28(8): 1139-44, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23960439

RESUMO

The effects of several antihypertensive drugs on bone mineral density (BMD) and micro-architectural changes in ovariectomized (OVX) mice were investigated. Eight-week-old female C57/BL6 mice were used for this study. Three days after ovariectomy, mice were treated intraperitoneally with nifedipine (15 mg/kg), telmisartan (5 mg/kg), enalapril (20 mg/kg), propranolol (1 mg/kg) or hydrochlorothiazide (12.5 mg/kg) for 35 consecutive days. Uterine atrophy of all mice was confirmed to evaluate estrogen deficiency state. BMD and micro-architectural analyses were performed on tibial proximal ends by micro-computed tomography (micro-CT). When OVX mice with uterine atrophy were compared with mice without atrophy, BMD decreased (P < 0.001). There were significant differences in BMD loss between different antihypertensive drugs (P = 0.005). Enalapril and propranolol increased BMD loss in mice with atrophied uteri compared with control mice. By contrast, thiazide increased BMD in mice with uterine atrophy compared with vehicle-treated mice (P = 0.048). Thiazide (P = 0.032) and telmisartan (P = 0.051) reduced bone loss and bone fraction in mice with uterine atrophy compared with the control. Thiazide affects BMD in OVX mice positively. The reduction in bone loss by thiazide and telmisartan suggest that these drugs may benefit menopausal women with hypertension and osteoporosis.


Assuntos
Anti-Hipertensivos/farmacologia , Densidade Óssea/efeitos dos fármacos , Animais , Atrofia , Benzimidazóis/farmacologia , Benzoatos/farmacologia , Enalapril/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Propranolol/farmacologia , Telmisartan , Tiazidas/farmacologia , Tíbia/diagnóstico por imagem , Tomografia Computadorizada por Raios X , Útero/anatomia & histologia , Útero/patologia
16.
Int Immunopharmacol ; 16(1): 85-92, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23557965

RESUMO

INTRODUCTION: This study was undertaken to determine whether metformin has anti-inflammatory effects in the collagen antibody-induced arthritis (CAIA) murine model. The effect of metformin on Th17 cell differentiation was also investigated. METHODS: CAIA mice were treated with 100 and 150 mg/kg i.p. metformin (low- and high-dose groups, respectively). Arthritis activity and histological joint destruction were studied. Flow cytometry was used to (i) determine RORγt-expressing CD4+ percentages in draining axillary lymph nodes (ALNs) from metformin-treated and untreated mice with CAIA, (ii) determine Th17 percentages in splenic CD4+ T cells cultured ex vivo for 3 days in Th17-differentiation-inducing conditions, and (iii) determine the percentages of RORγt+CD4+ T cells when normal splenic T cells from DBA/1 mice were cultured in Th17-differentiation-inducing conditions together with various metformin doses. Western blot analysis was used to assess the intracellular signaling of the metformin-treated splenocytes. RESULTS: Metformin attenuated both arthritis scores and bone destruction in CAIA mice, decreased the serum levels of the pro-inflammatory cytokines, TNF-α and IL-1, and reduced the number of RORγt+CD4+ T cells in the ALNs. Splenocytes from metformin-treated CAIA mice differentiated less readily into Th17 cells upon ex vivo stimulation. Metformin treatment of normal cells cultured in Th17-differentiation-inducing conditions decreased the number of RORγt-expressing CD4+ cells in a dose-dependent manner and downregulated STAT3 phosphorylation via the AMPK pathway. CONCLUSIONS: Metformin had an anti-inflammatory effect on murine autoimmune arthritis due to the inhibition of Th17 cell differentiation. Metformin may have a possible therapeutic value for treatment of rheumatoid arthritis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite Experimental/tratamento farmacológico , Metformina/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Artrite Experimental/imunologia , Artrite Experimental/patologia , Diferenciação Celular/efeitos dos fármacos , Citocinas/sangue , Masculino , Metformina/farmacologia , Camundongos , Camundongos Endogâmicos DBA , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Fator de Transcrição STAT3/antagonistas & inibidores , Baço/citologia , Células Th17/citologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
17.
Int J Mol Med ; 31(4): 769-77, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23403591

RESUMO

Bone destruction and inflammation are closely linked. Cytokines play an important role in inflammatory bone destruction by upregulating the receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL). The direct role of cytokines that act in a non-RANKL-dependent manner has yet to be elucidated. The aim of this study was to investigate the direct osteoclastogenic properties of inflammatory cytokines at different time-points of osteoclastogenesis. Mouse bone marrow macrophages were stimulated with the macrophage colony-stimulating factor (M-CSF) and various concentrations of RANKL. Inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, IL-17 and IL-23, were added to the culture system of osteoclastogenesis. Two time-points of cytokine treatment were set. The 'early' effect of each cytokine was investigated at the time of first RANKL treatment, whereas the 'late' effect was investigated 48 h after the first RANKL challenge. Osteoclast differentiation and function were assessed using an osteoclast marker [tartrate-resistant acid phosphatase (TRAP)] and by visualization of pit formation. A permissive level of RANKL was required for cytokine-associated osteoclastogenesis in all experiments. In the M-CSF/RANKL monocellular culture system, IL-1ß enhanced and IL-6 decreased osteoclast formation in a dose-dependent manner, regardless of temporal differences. Other cytokines showed various responses according to the phase of osteoclast maturation and the concentration of each cytokine and RANKL. Furthermore, luciferase assays showed that both IL-1ß and RANKL activated the NF-κB signaling pathway. Collectively, our data revealed that targeting IL-1ß may be a promising strategy to inhibit inflammation-associated bone destruction and osteoporosis.


Assuntos
Citocinas/metabolismo , Osteoclastos/citologia , Osteogênese/fisiologia , Animais , Células da Medula Óssea , Sobrevivência Celular , Luciferases/metabolismo , Macrófagos , Masculino , Camundongos , NF-kappa B/metabolismo , Transdução de Sinais/fisiologia , Estatísticas não Paramétricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA