Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Lab Anim Res ; 40(1): 10, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38532510

RESUMO

The ferret (Mustela putorius furo) is a small domesticated species of the family Mustelidae within the order Carnivora. The present article reviews and discusses the current state of knowledge about housing, care, breeding, and biomedical uses of ferrets. The management and breeding procedures of ferrets resemble those used for other carnivores. Understanding its behavior helps in the use of environmental enrichment and social housing, which promote behaviors typical of the species. Ferrets have been used in research since the beginning of the twentieth century. It is a suitable non-rodent model in biomedical research because of its hardy nature, social behavior, diet and other habits, small size, and thus the requirement of a relatively low amount of test compounds and early sexual maturity compared with dogs and non-human primates. Ferrets and humans have numerous similar anatomical, metabolic, and physiological characteristics, including the endocrine, respiratory, auditory, gastrointestinal, and immunological systems. It is one of the emerging animal models used in studies such as influenza and other infectious respiratory diseases, cystic fibrosis, lung cancer, cardiac research, gastrointestinal disorders, neuroscience, and toxicological studies. Ferrets are vulnerable to many human pathogenic organisms, like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), because air transmission of this virus between them has been observed in the laboratory. Ferrets draw the attention of the medical community compared to rodents because they occupy a distinct niche in biomedical studies, although they possess a small representation in laboratory research.

2.
Sci Adv ; 8(25): eabm6504, 2022 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-35749495

RESUMO

Aging has been reported to deteriorate the quantity and quality of mesenchymal stem cells (MSCs), which affect their therapeutic use in regenerative medicine. A dearth of age-related stem cell research further restricts their clinical applications. The present study explores the possibility of using MSCs derived from human gingival tissues (GMSCs) for studying their ex vivo growth characteristics and differentiation potential with respect to donor age. GMSCs displayed decreased in vitro adipogenesis and in vitro and in vivo osteogenesis with age, but in vitro neurogenesis remained unaffected. An increased expression of p53 and SIRT1 with donor age was correlated to their ability of eliminating tumorigenic events through apoptosis or autophagy, respectively. Irrespective of donor age, GMSCs displayed effective immunoregulation and regenerative potential in a mouse model of LPS-induced acute lung injury. Thus, we suggest the potential of GMSCs for designing cell-based immunomodulatory therapeutic approaches and their further extrapolation for acute inflammatory conditions such as acute respiratory distress syndrome and COVID-19.


Assuntos
COVID-19 , Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Gengiva , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Osteogênese
3.
J Immunol ; 204(4): 819-831, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31900339

RESUMO

IL-3, a cytokine secreted by activated T lymphocytes, is known to regulate the proliferation, survival, and differentiation of hematopoietic cells. However, the role of IL-3 in regulation of T cell functions is not fully delineated. Previously, we have reported that IL-3 plays an important role in development of regulatory T cells in mice. In this study, we investigated the regulation of IL-3R expression on human Th cells and also examined the role of IL-3 in effector functions of these cells. We found that human peripheral blood Th cells in resting state do not show surface expression of IL-3R; however, its expression was observed at transcript and intracellular protein levels. The functional IL-3R expression on the surface was seen only after antigenic stimulation. When naive Th cells were activated in the presence of various cytokines, we found that IL-4 significantly increases the surface expression of IL-3R and also increases the number of IL-3R+ Th cells. Interestingly, IL-3R+ cells exhibit a Th2 cell-like phenotype and show high GATA-3 expression. Moreover, Th2 cells in presence of IL-3 show increased expression of type 2 effector cytokines, such as IL-4, IL-5, and IL-13. Furthermore, IL-3R expressing and IL-3-secreting Th cells were high in house dust mite-allergic patients. Thus, to our knowledge, we provide the first evidence that the expression of IL-3R on activated human Th cells is modulated by IL-4, and IL-3 regulates the effector functions of Th2 cells. Our results suggest that IL-3 may play an important role in regulating allergic immune responses.


Assuntos
Diferenciação Celular/imunologia , Interleucina-3/imunologia , Interleucina-4/imunologia , Receptores de Interleucina-3/imunologia , Células Th2/imunologia , Humanos , Hipersensibilidade/imunologia , Interleucina-3/metabolismo , Interleucina-4/metabolismo , Ativação Linfocitária/imunologia , Receptores de Interleucina-3/metabolismo
4.
J Immunol ; 200(2): 595-606, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29203513

RESUMO

Bone remodeling comprises balanced activities between osteoclasts and osteoblasts, which is regulated by various factors, including hormones and cytokines. We previously reported that IL-3 inhibits osteoclast differentiation and pathological bone loss. IL-3 also enhances osteoblast differentiation and bone formation from mesenchymal stem cells. However, the role of IL-3 in regulation of osteoblast-osteoclast interactions and underlying mechanisms is not yet delineated. In this study, we investigated the role of IL-3 on the regulation of osteoblast-specific molecules, receptor activator of NF-κB ligand (RANKL), and osteoprotegerin (OPG) that modulate bone homeostasis. We found that IL-3 increases RANKL expression at both the transcriptional and translational levels, and it showed no effect on OPG expression in calvarial osteoblasts. The increased RANKL expression by IL-3 induces mononuclear osteoclasts; however, it does not induce multinuclear osteoclasts. Interestingly, IL-3 decreases soluble RANKL by reducing ectodomain shedding of membrane RANKL through downregulation of metalloproteases mainly a disintegrin and metalloproteinase (ADAM)10, ADAM17, ADAM19, and MMP3. Moreover, IL-3 increases membrane RANKL by activating the JAK2/STAT5 pathway. Furthermore, IL-3 enhances RANKL expression in mesenchymal stem cells of wild-type mice but not in STAT5a knockout mice. Interestingly, IL-3 restores RANKL expression in adult mice by enhancing bone-specific RANKL and decreasing serum RANKL. Furthermore, IL-3 increases the serum OPG level in adult mice. Thus, our results reveal, to our knowledge for the first time, that IL-3 differentially regulates two functional forms of RANKL through metalloproteases and the JAK2/STAT5 pathway, and it helps in restoring the decreased RANKL/OPG ratio in adult mice. Notably, our studies indicate the novel role of IL-3 in regulating bone homeostasis in important skeletal disorders.


Assuntos
Interleucina-3/metabolismo , Janus Quinase 2/metabolismo , Metaloproteinases da Matriz/metabolismo , Osteoblastos/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Células Cultivadas , Técnicas de Cocultura , Expressão Gênica , Interleucina-3/farmacologia , Camundongos , Camundongos Transgênicos , Osteoblastos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Ligante RANK/sangue , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Transdução de Sinais/efeitos dos fármacos
5.
Stem Cell Res Ther ; 8(1): 168, 2017 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-28705238

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) represent an important source for cell therapy in regenerative medicine. MSCs have shown promising results for repair of damaged tissues in various degenerative diseases in animal models and also in human clinical trials. However, little is known about the factors that could enhance the migration and tissue-specific engraftment of exogenously infused MSCs for successful regenerative cell therapy. Previously, we have reported that interleukin-3 (IL-3) prevents bone and cartilage damage in animal models of rheumatoid arthritis and osteoarthritis. Also, IL-3 promotes the differentiation of human MSCs into functional osteoblasts and increases their in-vivo bone regenerative potential in immunocompromised mice. However, the role of IL-3 in migration of MSCs is not yet known. In the present study, we investigated the role of IL-3 in migration of human MSCs under both in-vitro and in-vivo conditions. METHODS: MSCs isolated from human bone marrow, adipose and gingival tissues were used for in-vitro cell migration, motility and wound healing assays in the presence or absence of IL-3. The effect of IL-3 preconditioning on expression of chemokine receptors and integrins was examined by flow cytometry and real-time PCR. The in-vivo migration of IL-3-preconditioned MSCs was investigated using a subcutaneous matrigel-releasing stromal cell-derived factor-1 alpha (SDF-1α) model in immunocompromised mice. RESULTS: We observed that human MSCs isolated from all three sources express IL-3 receptor-α (IL-3Rα) both at gene and protein levels. IL-3 significantly enhances in-vitro migration, motility and wound healing abilities of MSCs. Moreover, IL-3 preconditioning upregulates expression of chemokine (C-X-C motif) receptor 4 (CXCR4) on MSCs, which leads to increased migration of cells towards SDF-1α. Furthermore, CXCR4 antagonist AMD3100 decreases the migration of IL-3-treated MSCs towards SDF-1α. Importantly, IL-3 also induces in-vivo migration of MSCs towards subcutaneously implanted matrigel-releasing-SDF-1α in immunocompromised mice. CONCLUSIONS: The present study demonstrates for the first time that IL-3 has an important role in enhancing the migration of human MSCs through regulation of the CXCR4/SDF-1α axis. These findings suggest a potential role of IL-3 in improving the efficacy of MSCs in regenerative cell therapy.


Assuntos
Movimento Celular , Regulação da Expressão Gênica , Interleucina-3/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptores CXCR4/biossíntese , Animais , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
6.
J Immunol ; 196(12): 5024-35, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27183574

RESUMO

Osteoarthritis (OA) is a chronic disease of articular joints that leads to degeneration of both cartilage and subchondral bone. These degenerative changes are further aggravated by proinflammatory cytokines including IL-1ß and TNF-α. Previously, we have reported that IL-3, a cytokine secreted by activated T cells, protects cartilage and bone damage in murine models of inflammatory and rheumatoid arthritis. However, how IL-3 protects cartilage degeneration is not yet known. In this study, we investigated the role of IL-3 on cartilage degeneration under both in vitro and in vivo conditions. We found that both mouse and human chondrocytes show strong expression of IL-3R at gene and protein levels. IL-3 increases the expression of mouse chondrocyte-specific genes, Sox9 and collagen type IIa, which were downregulated by IL-1ß. Moreover, IL-3 downregulated IL-1ß- and TNF-α-induced expression of matrix metalloproteinases in both mouse and human chondrocytes. Interestingly, IL-3 reduces the degeneration of articular cartilage and subchondral bone microarchitecture in a mouse model of human OA. Moreover, IL-3 showed the preventive and therapeutic effects on cartilage degeneration induced by IL-1ß in micromass pellet cultures of human mesenchymal stem cells. Thus, to our knowledge, we provide the first evidence that IL-3 has therapeutic potential in amelioration of degeneration of articular cartilage and subchondral bone microarchitecture associated with OA.


Assuntos
Cartilagem Articular/patologia , Regulação para Baixo , Interleucina-3/uso terapêutico , Metaloproteinases da Matriz/genética , Osteoartrite/tratamento farmacológico , Animais , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/imunologia , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/imunologia , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Modelos Animais de Doenças , Humanos , Interleucina-1beta/farmacologia , Interleucina-3/administração & dosagem , Interleucina-3/farmacologia , Subunidade alfa de Receptor de Interleucina-3/genética , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Metaloproteinases da Matriz/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , Osteoartrite/imunologia , Osteoartrite/fisiopatologia , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA