Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 82(23): 4303-4312, 2022 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-36191083

RESUMO

Obesity induces multifactorial effects such as dyslipidemia, insulin resistance, and arterial hypertension that influence the progression of many diseases. Obesity is associated with an increased incidence of cancers, and multiple mechanisms link obesity with cancer initiation and progression. Macrophages participate in the homeostasis of adipose tissue and play an important role in cancer. Adipose tissue expansion in obesity alters the balance between pro- and anti-inflammatory macrophages, which is a primary cause of inflammation. Chronic low-grade inflammation driven by macrophages is also an important characteristic of cancer. Adipocytes secrete various adipokines, including adiponectin, leptin, IL6, and TNFα, that influence macrophage behavior and tumor progression. Furthermore, other metabolic effects of obesity, such as hyperlipidemia, hyperglycemia, and hypercholesterolemia, can also regulate macrophage functionality in cancer. This review summarizes how obesity influences macrophage-tumor cell interactions and the role of macrophages in the response to anticancer therapies under obese conditions.


Assuntos
Resistência à Insulina , Neoplasias , Humanos , Obesidade/patologia , Macrófagos/metabolismo , Tecido Adiposo/metabolismo , Inflamação/patologia , Neoplasias/patologia
2.
Eur J Immunol ; 52(11): 1842-1858, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36074916

RESUMO

IL-17-producing Th17 cells play an important role in pathogenesis of rheumatoid arthritis (RA). Aberrant immune activation due to an imbalance between Th17 and regulatory T (Treg) cells is associated with the development of RA and other autoimmune diseases. Targeting pathogenic Th17 cells and their associated molecules is emerging as a promising strategy to treat and reverse RA. Here, we demonstrate that IL-3 inhibits the differentiation of Th17 cells and promotes the development of Treg cells in IL-2-dependent manner. In IL-2 KO mice, we observed that IL-3 has no effect on differentiation of both Th17 and Treg cells. In addition, IL-3 decreases pathogenic IL-17A+ TNF-α+ , IL-17A+ IFN-γ+ and IL-23R+ Th17 cells, secretion of GM-CSF and IFN-γ, and osteoclastogenesis when presented in the culture together with Th17 polarizing cytokines. Mechanistically, IL-3 regulates the development of Th17 cells through the inhibition of STAT3 phosphorylation. IL-3 treatment significantly decreases the pathogenic Th17 cell responses and arthritic scores in the mouse model of RA. Importantly, IL-3 inhibits the differentiation of human Th17 cells. Thus, our results suggest a novel therapeutic role of IL-3 in the regulation of Th17 cell-mediated pathophysiology of RA.


Assuntos
Artrite Reumatoide , Diferenciação Celular , Interleucina-3 , Células Th17 , Animais , Humanos , Camundongos , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Interleucina-3/metabolismo , Linfócitos T Reguladores/citologia , Células Th17/citologia
3.
J Biosci ; 462021.
Artigo em Inglês | MEDLINE | ID: mdl-34183475

RESUMO

IL-3, a haematopoiesis regulatory factor, has previously been shown to inhibit both mouse and human osteoclast differentiation and bone resorption. Here, the role of rat IL-3 on rat osteoclast differentiation was evaluated to address whether the inhibitory action of IL-3 on osteoclastogenesis is conserved in various species. It was observed that IL-3 inhibited rat osteoclast differentiation induced by both TNF-α and receptor activator of NF-ĸB ligand (RANKL). TNF-α is known to induce bone loss in postmenopausal osteoporotic women and it also synergise with many pro-osteoclastogenic cytokines to cause huge pathological bone loss. Importantly, it was found that rat IL-3 inhibits the synergistic action of TNF-α with RANKL and IL-1ß, TGFß1 and TGF-ß3. IL-3 downregulates the TNF-α-induced nuclear translocation of NF-ĸB-p65 and c-fos without affecting c-jun. Interestingly, we observed that IL-3 also inhibits osteoclast differentiation in vivo in rats induced by TNF-α. All these results suggest that inhibitory action of IL-3 on osteoclastogenesis is conserved in various species including mice, rats and humans. Thus, our results clearly indicate that IL-3 has therapeutic potential to treat pathological bone loss in important skeletal diseases.


Assuntos
Diferenciação Celular , Interleucina-3/fisiologia , Osteoclastos , Osteogênese , Animais , NF-kappa B/metabolismo , Ligante RANK/metabolismo , Ratos Wistar , Fator de Transcrição AP-1/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
Ultrasonics ; 104: 106110, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32146383

RESUMO

Surface mechanical attrition treatment (SMAT) of metallic biomaterials has gained significant importance due to its ability to develop nano structure in the surface region. In the present study, the microstructural changes and corrosion behavior of the commercially pure titanium (cp-Ti), following different durations of ultrasonic shot peening (USSP) has been investigated. cp-Ti was shot peened for different durations from 0 to 120 s and the treated samples were examined for microstructural changes in the surface region, cell viability and corrosion behavior. Cell viability was considerably increased after USSP for 60-120 s, exhibiting maximum for the 90 s of USSP. The passivation tendency was also improved with peening duration up to 90 s, however, it declined for longer duration of USSP. The beneficial effects of USSP may be attributed to nano structuring in the surface region and development of higher positive potentials at the USSP treated surface. Transmission Electron Microscope (TEM) examination of the USSPed surface revealed dislocation entanglement and substructure. Also, higher surface volta potential was observed over the USSPed sample exhibiting better cell proliferation. The present work is corollary to previous work of the group and mainly discusses the role of USSP duration, as a process parameter, on the cell viability and corrosion resistance of cp-Ti.


Assuntos
Células-Tronco Mesenquimais/fisiologia , Nanoestruturas/química , Titânio/química , Ultrassom/métodos , Proliferação de Células , Sobrevivência Celular , Corrosão , Técnicas Eletroquímicas , Humanos , Teste de Materiais , Microscopia de Força Atômica , Microscopia Eletrônica , Propriedades de Superfície , Difração de Raios X
5.
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
6.
J Biol Chem ; 294(41): 14879-14895, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31413110

RESUMO

F-box protein 31 (FBXO31) is a reported putative tumor suppressor, and its inactivation due to loss of heterozygosity is associated with cancers of different origins. An emerging body of literature has documented FBXO31's role in preserving genome integrity following DNA damage and in the cell cycle. However, knowledge regarding the role of FBXO31 during normal cell-cycle progression is restricted to its functions during the G2/M phase. Interestingly, FBXO31 levels remain high even during the early G1 phase, a crucial stage for preparing the cells for DNA replication. Therefore, we sought to investigate the functions of FBXO31 during the G1 phase of the cell cycle. Here, using flow cytometric, biochemical, and immunofluorescence techniques, we show that FBXO31 is essential for maintaining optimum expression of the cell-cycle protein cyclin A for efficient cell-cycle progression. Stable FBXO31 knockdown led to atypical accumulation of cyclin A during the G1 phase, driving premature DNA replication and compromised loading of the minichromosome maintenance complex, resulting in replication from fewer origins and DNA double-strand breaks. Because of these inherent defects in replication, FBXO31-knockdown cells were hypersensitive to replication stress-inducing agents and displayed pronounced genomic instability. Upon entering mitosis, the cells defective in DNA replication exhibited a delay in the prometaphase-to-metaphase transition and anaphase defects such as lagging and bridging chromosomes. In conclusion, our findings establish that FBXO31 plays a pivotal role in preserving genomic integrity by maintaining low cyclin A levels during the G1 phase for faithful genome duplication and segregation.


Assuntos
Ciclina A/metabolismo , Replicação do DNA/genética , Proteínas F-Box/metabolismo , Genoma Humano/genética , Proteínas Supressoras de Tumor/metabolismo , Ciclo Celular/genética , Cromatina/genética , Proteínas F-Box/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Cinética , Células MCF-7 , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Ubiquitinação/genética
7.
Biochim Biophys Acta Mol Cell Res ; 1866(9): 1498-1507, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31255720

RESUMO

Estrogen increases bone formation by promoting mineralization and prolonging the lifespan of osteoblasts. To understand the underlying molecular mechanism/s, we identified estrogen-regulated proteins at different stages of human osteoblast differentiation using differential proteomics approach. Among the identified proteins, we observed that estrogen upregulated RAB3GAP1 on day 1 and 5 of differentiation. RAB3GAP1 is critically involved in the process of autophagy, a eukaryotic degradative pathway essential for cell survival. We, therefore, investigated the effect of estrogen on autophagy in differentiating human osteoblasts and their precursors, the mesenchymal stem cells (MSCs). MSCs exhibited high autophagic flux which declined during osteoblast differentiation, resulting in high basal apoptosis in osteoblasts. Estrogen reduced apoptosis in differentiating osteoblasts by promoting autophagy, thus contributing towards their longer lifespan. Further, MSCs were resistant against starvation-induced apoptosis, whereas, differentiating osteoblasts showed significant susceptibility towards it. Estrogen, in addition to promoting mineralization, protected differentiating osteoblasts from starvation-induced apoptosis by increasing autophagic flux. Autophagic flux in RAB3GAP1 knockdown osteoblasts appeared diminished, and showed increased apoptosis even in nutrient-rich conditions, and exhibited significantly impaired mineralization. However, irrespective of the presence of estrogen, starvation further enhanced apoptosis in these cells. Furthermore, estrogen failed to promote mineralization in these osteoblasts. Our study illustrates that autophagy is essential for human osteoblast survival and mineralization, and osteoblasts are susceptible to apoptosis due to reduced autophagy during differentiation. Estrogen, via upregulation of RAB3GAP1, promotes autophagy in osteoblasts during differentiation thereby increasing their survival and mineralization capacity. Our study demonstrates the positive role of autophagy in bone homeostasis.


Assuntos
Autofagia/efeitos dos fármacos , Estrogênios/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Apoptose/efeitos dos fármacos , Calcificação Fisiológica , Diferenciação Celular/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Homeostase , Humanos , Células-Tronco Mesenquimais , Osteogênese/efeitos dos fármacos , Osteoporose , Regulação para Cima , Proteínas rab3 de Ligação ao GTP/genética , Proteínas rab3 de Ligação ao GTP/metabolismo
8.
Biochim Biophys Acta Mol Basis Dis ; 1865(3): 547-557, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30579930

RESUMO

Estrogen insufficiency at menopause cause accelerated bone loss due to unwarranted differentiation and function of osteoclasts. Unraveling the underlying mechanism/s may identify mediators of estrogen action which can be targeted for improved management of osteoporosis. Towards this, we analyzed the effect of 17ß-estradiol on the proteomes of differentiating human osteoclasts. The major proteomic changes observed included upregulation of LYN by estrogen. We, therefore, investigated the effect of estrogen on osteoclast differentiation, survival, and function in control and LYN knockdown conditions. In control condition, estrogen treatment increased the apoptosis rate and suppressed the calcium signaling by reducing the intracellular Ca2+ levels as well as expression and activation of NFATc1 and c-Src during differentiation, resulting in reduced osteoclastogenesis. These osteoclasts were smaller in size with reduced extent of multinuclearity and produced significantly low levels of bone resorbing enzymes. They also exhibited disrupted sealing zone formation with low podosome density, impaired cell polarization and reduced resorption of dentine slices. Interestingly, in LYN knockdown condition, estrogen failed to induce apoptosis and inhibit activation of NFATc1 and c-Src. Compared to effect of estrogen on osteoclast in control condition, LYN knockdown osteoclasts did not show reduction in production of bone resorbing enzymes and had defined sealing zone formation with high podosome density with no impairment in cell polarization. They resorbed significant area on dentine slices. Thus, the inhibitory action of estrogen on osteoclast was severely restrained in LYN knockdown condition, demonstrating the importance of LYN as a key mediator of the effect of estrogen on osteoclastogenesis.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Estradiol/farmacologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Quinases da Família src/fisiologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Desenvolvimento Ósseo/efeitos dos fármacos , Desenvolvimento Ósseo/genética , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Diferenciação Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Osteoclastos/metabolismo , Quinases da Família src/genética
9.
J Cell Commun Signal ; 12(3): 615-624, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29705949

RESUMO

Interleukin-33 (IL-33) is a member of the IL-1 family of cytokines that play a central role in the regulation of immune responses. Its release from epithelial and endothelial cells is mediated by pro-inflammatory cytokines, cell damage and by recognition of pathogen-associated molecular patterns (PAMPs). The activity of IL-33 is mediated by binding to the IL-33 receptor complex (IL-33R) and activation of NF-κB signaling via the classical MyD88/IRAK/TRAF6 module. IL-33 also induces the phosphorylation and activation of ERK1/2, JNK, p38 and PI3K/AKT signaling modules resulting in the production and release of pro-inflammatory cytokines. Aberrant signaling by IL-33 has been implicated in the pathogenesis of several acute and chronic inflammatory diseases, including asthma, atopic dermatitis, rheumatoid arthritis and ulcerative colitis among others. Considering the biomedical importance of IL-33, we developed a pathway resource of signaling events mediated by IL-33/IL-33R in this study. Using data mined from the published literature, we describe an integrated pathway reaction map of IL-33/IL-33R consisting of 681 proteins and 765 reactions. These include information pertaining to 19 physical interaction events, 740 enzyme catalysis events, 6 protein translocation events, 4 activation/inhibition events, 9 transcriptional regulators and 2492 gene regulation events. The pathway map is publicly available through NetPath ( http://www.netpath.org /), a resource of human signaling pathways developed previously by our group. This resource will provide a platform to the scientific community in facilitating identification of novel therapeutic targets for diseases associated with dysregulated IL-33 signaling. Database URL: http://www.netpath.org/pathways?path_id=NetPath_120 .

10.
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
11.
Int J Nanomedicine ; 12: 6437-6459, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28919746

RESUMO

Bacterial cellulose (BC) is a naturally occurring nanofibrous biomaterial which exhibits unique physical properties and is amenable to chemical modifications. To explore whether this versatile material can be used in the treatment of osteochondral defects (OCD), we developed and characterized novel BC-based nanocomposite scaffolds, for example, BC-hydroxyapatite (BC-HA) and BC-glycosaminoglycans (BC-GAG) that mimic bone and cartilage, respectively. In vitro biocompatibility of BC-HA and BC-GAG scaffolds was established using osteosarcoma cells, human articular chondrocytes, and human adipose-derived mesenchymal stem cells. On subcutaneous implantation, the scaffolds allowed tissue ingrowth and induced no adverse immunological reactions suggesting excellent in vivo biocompatibility. Implantation of acellular bilayered scaffolds in OCD created in rat knees induced progressive regeneration of cartilage tissue, deposition of extracellular matrix, and regeneration of subchondral bone by the host cells. The results of micro-CT revealed that bone mineral density and ratio of bone volume to tissue volume were significantly higher in animals receiving bilayered scaffold as compared to the control animals. To the best of our knowledge, this study proves for the first time, the functional performance of acellular BC-based bilayered scaffolds. Thus, this strategy has great potential for clinical translation and can be used in repair of OCD.


Assuntos
Materiais Biocompatíveis/química , Condrócitos/citologia , Nanocompostos/química , Alicerces Teciduais , Acetobacteraceae/química , Animais , Regeneração Óssea/fisiologia , Cartilagem/citologia , Celulose/química , Condrócitos/química , Modelos Animais de Doenças , Durapatita/química , Glicosaminoglicanos/química , Humanos , Teste de Materiais , Células-Tronco Mesenquimais/citologia , Ratos , Ratos Wistar , Engenharia Tecidual/métodos , Alicerces Teciduais/química
12.
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
13.
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
14.
Pigment Cell Melanoma Res ; 29(4): 465-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27124831

RESUMO

Vitiligo is a multifactorial acquired depigmenting disorder. Recent insights into the molecular mechanisms driving the gradual destruction of melanocytes in vitiligo will likely lead to the discovery of novel therapies, which need to be evaluated in animal models that closely recapitulate the pathogenesis of human vitiligo. In humans, vitiligo is characterized by a spontaneous loss of functional melanocytes from the epidermis, but most animal models of vitiligo are either inducible or genetically programmed. Here, we report that acquired depigmentation in water buffalo recapitulates molecular, histological, immunohistochemical, and ultrastructural changes observed in human vitiligo and hence could be used as a model to study vitiligo pathogenesis and facilitate the discovery and evaluation of therapeutic interventions for vitiligo.


Assuntos
Modelos Animais de Doenças , Epiderme/patologia , Melanócitos/patologia , Vitiligo/veterinária , Animais , Búfalos , Células Cultivadas , Feminino , Humanos , Vitiligo/patologia
15.
J Immunol ; 195(11): 5136-48, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26538398

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammatory synovitis leading to joint destruction and systemic bone loss. The inflammation-induced bone loss is mediated by increased osteoclast formation and function. Current antirheumatic therapies primarily target suppression of inflammatory cascade with limited or no success in controlling progression of bone destruction. Mesenchymal stem cells (MSCs) by virtue of their tissue repair and immunomodulatory properties have shown promising results in various autoimmune and degenerative diseases. However, the role of MSCs in prevention of bone destruction in RA is not yet understood. In this study, we investigated the effect of adipose-derived MSCs (ASCs) on in vitro formation of bone-resorbing osteoclasts and pathological bone loss in the mouse collagen-induced arthritis (CIA) model of RA. We observed that ASCs significantly inhibited receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis in both a contact-dependent and -independent manner. Additionally, ASCs inhibited RANKL-induced osteoclastogenesis in the presence of proinflammatory cytokines such as TNF-α, IL-17, and IL-1ß. Furthermore, treatment with ASCs at the onset of CIA significantly reduced clinical symptoms and joint pathology. Interestingly, ASCs protected periarticular and systemic bone loss in CIA mice by maintaining trabecular bone structure. We further observed that treatment with ASCs reduced osteoclast precursors in bone marrow, resulting in decreased osteoclastogenesis. Moreover, ASCs suppressed autoimmune T cell responses and increased the percentages of peripheral regulatory T and B cells. Thus, we provide strong evidence that ASCs ameliorate inflammation-induced systemic bone loss in CIA mice by reducing osteoclast precursors and promoting immune tolerance.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Reabsorção Óssea/imunologia , Células-Tronco Mesenquimais/imunologia , Osteoclastos/imunologia , Ligante RANK/antagonistas & inibidores , Tecido Adiposo/citologia , Animais , Artrite Experimental/patologia , Artrite Reumatoide/patologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Osso e Ossos/imunologia , Osso e Ossos/patologia , Diferenciação Celular/imunologia , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Feminino , Tolerância Imunológica/imunologia , Interleucina-17/metabolismo , Interleucina-1beta/metabolismo , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/metabolismo
16.
Biochem Biophys Res Commun ; 455(1-2): 133-8, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25450704

RESUMO

The relationship between obesity and bone is complex. Epidemiological studies demonstrate positive as well as negative correlation between obesity and bone health. In the present study, we investigated the impact of high fat diet-induced obesity on peak bone mass. After 9 months of feeding young rats with high fat diet, we observed obesity phenotype in rats with increased body weight, fat mass, serum triglycerides and cholesterol. There were significant increases in serum total alkaline phosphatase, bone mineral density and bone mineral content. By micro-computed tomography (µ-CT), we observed a trend of better trabecular bones with respect to their microarchitecture and geometry. This indicated that high fat diet helps in achieving peak bone mass and microstructure at younger age. We subsequently shifted rats from high fat diet to normal diet for 6 months and evaluated bone/obesity parameters. It was observed that after shifting rats from high fat diet to normal diet, fat mass, serum triglycerides and cholesterol were significantly decreased. Interestingly, the gain in bone mineral density, bone mineral content and trabecular bone parameters by HFD was retained even after body weight and obesity were normalized. These results suggest that fat rich diet during growth could accelerate achievement of peak bone mass that is sustainable even after withdrawal of high fat diet.


Assuntos
Densidade Óssea , Dieta Hiperlipídica , Obesidade/etiologia , Animais , Osso e Ossos/diagnóstico por imagem , Dieta Hiperlipídica/efeitos adversos , Masculino , Radiografia , Ratos , Ratos Wistar
17.
J Oral Implantol ; 40 Spec No: 347-55, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25020216

RESUMO

This investigation was carried out to study the effect of a novel process of surface modification, surface nanostructuring by ultrasonic shot peening, on osteoblast proliferation and corrosion behavior of commercially pure titanium (c p-Ti) in simulated body fluid. A mechanically polished disc of c p-Ti was subjected to ultrasonic shot peening with stainless steel balls to create nanostructure at the surface. A nanostructure (<20 nm) with inhomogeneous distribution was revealed by atomic force and scanning electron microscopy. There was an increase of approximately 10% in cell proliferation, but there was drastic fall in corrosion resistance. Corrosion rate was increased by 327% in the shot peened condition. In order to examine the role of residual stresses associated with the shot peened surface on these aspects, a part of the shot peened specimen was annealed at 400°C for 1 hour. A marked influence of annealing treatment was observed on surface structure, cell proliferation, and corrosion resistance. Surface nanostructure was much more prominent, with increased number density and sharper grain boundaries; cell proliferation was enhanced to approximately 50% and corrosion rate was reduced by 86.2% and 41% as compared with that of the shot peened and the as received conditions, respectively. The highly significant improvement in cell proliferation, resulting from annealing of the shot peened specimen, was attributed to increased volume fraction of stabilized nanostructure, stress recovery, and crystallization of the oxide film. Increase in corrosion resistance from annealing of shot peened material was related to more effective passivation. Thus, the surface of c p-Ti, modified by this novel process, possessed a unique quality of enhancing cell proliferation as well as the corrosion resistance and could be highly effective in reducing treatment time of patients adopting dental and orthopedic implants of titanium and its alloys.


Assuntos
Materiais Dentários/química , Nanoestruturas/química , Osteoblastos/fisiologia , Titânio/química , Ultrassom/métodos , Adesão Celular/fisiologia , Técnicas de Cultura de Células , Linhagem Celular , Proliferação de Células , Corantes , Corrosão , Cristalização , Polimento Dentário/métodos , Temperatura Alta , Humanos , Microscopia de Força Atômica , Microscopia Eletrônica de Varredura , Nitritos/análise , Óxidos/química , Potenciometria , Aço Inoxidável/química , Estresse Mecânico , Propriedades de Superfície , Sais de Tetrazólio , Tiazóis , Fatores de Tempo
18.
EMBO J ; 33(9): 994-1010, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24431222

RESUMO

There is emerging evidence that stem cells can rejuvenate damaged cells by mitochondrial transfer. Earlier studies show that epithelial mitochondrial dysfunction is critical in asthma pathogenesis. Here we show for the first time that Miro1, a mitochondrial Rho-GTPase, regulates intercellular mitochondrial movement from mesenchymal stem cells (MSC) to epithelial cells (EC). We demonstrate that overexpression of Miro1 in MSC (MSCmiro(Hi)) leads to enhanced mitochondrial transfer and rescue of epithelial injury, while Miro1 knockdown (MSCmiro(Lo)) leads to loss of efficacy. Treatment with MSCmiro(Hi) was associated with greater therapeutic efficacy, when compared to control MSC, in mouse models of rotenone (Rot) induced airway injury and allergic airway inflammation (AAI). Notably, airway hyperresponsiveness and remodeling were reversed by MSCmiro(Hi) in three separate allergen-induced asthma models. In a human in vitro system, MSCmiro(Hi) reversed mitochondrial dysfunction in bronchial epithelial cells treated with pro-inflammatory supernatant of IL-13-induced macrophages. Anti-inflammatory MSC products like NO, TGF-ß, IL-10 and PGE2, were unchanged by Miro1 overexpression, excluding non-specific paracrine effects. In summary, Miro1 overexpression leads to increased stem cell repair.


Assuntos
Lesão Pulmonar/terapia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Mitocôndrias/metabolismo , Proteínas rho de Ligação ao GTP/fisiologia , Animais , Transporte Biológico/genética , Células Cultivadas , Terapia Genética/métodos , Humanos , Pulmão/patologia , Lesão Pulmonar/patologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Mitocôndrias/transplante , Células NIH 3T3 , Nanotubos , Resultado do Tratamento , Proteínas rho de Ligação ao GTP/genética
19.
Biochem Biophys Res Commun ; 418(4): 669-75, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22293197

RESUMO

IL-3 is an important cytokine that regulates hematopoiesis. We have previously demonstrated that IL-3 is a potent inhibitor of osteoclastogenesis and bone resorption. In the present study, we have investigated the role of IL-3 on human osteoblast differentiation and bone formation. We found that IL-3 in a dose-dependent manner increases osteoblast differentiation and matrix mineralization in human mesenchymal stem cells (MSCs). IL-3 significantly enhances the expression of osteoblast specific genes such as alkaline phosphatase, collagen type-I, osteocalcin and osteopontin; and Runx-2 and osterix transcription factors. Moreover, IL-3 induces the expression of bone morphogenetic protein-2 (BMP-2), and activates smad1/5/8. IL-3 enhances osteoblast differentiation and BMP-2 secretion through JAK/STAT pathway. Interestingly, IL-3 promotes in vivo bone regeneration ability of MSCs. Thus, we reveal for the first time that IL-3 enhances human osteoblast differentiation and bone formation in both in vitro and in vivo conditions, and suggest its therapeutic potential for bone formation in important bone diseases.


Assuntos
Diferenciação Celular , Interleucina-3/fisiologia , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osteogênese , Fosfatase Alcalina/biossíntese , Animais , Proteína Morfogenética Óssea 2/biossíntese , Regeneração Óssea , Colágeno Tipo I/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Humanos , Interleucina-3/farmacologia , Subunidade alfa de Receptor de Interleucina-3/biossíntese , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Osteocalcina/biossíntese , Osteopontina/biossíntese , Fator de Transcrição Sp7 , Fatores de Transcrição/biossíntese
20.
Database (Oxford) ; 2011: bar021, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21742767

RESUMO

Receptor activator of nuclear factor-kappa B ligand (RANKL) is a member of tumor necrosis factor (TNF) superfamily that plays a key role in the regulation of differentiation, activation and survival of osteoclasts and also in tumor cell migration and bone metastasis. Osteoclast activation induced by RANKL regulates hematopoietic stem cell mobilization as part of homeostasis and host defense mechanisms thereby linking regulation of hematopoiesis with bone remodeling. Binding of RANKL to its receptor, Receptor activator of nuclear factor-kappa B (RANK) activates molecules such as NF-kappa B, mitogen activated protein kinase (MAPK), nuclear factor of activated T cells (NFAT) and phosphatidyl 3-kinase (PI3K). Although the molecular and cellular roles of these molecules have been reported previously, a systematic cataloging of the molecular events induced by RANKL/RANK interaction has not been attempted. Here, we present a comprehensive reaction map of the RANKL/RANK-signaling pathway based on an extensive manual curation of the published literature. We hope that the curated RANKL/RANK-signaling pathway model would enable new biomedical discoveries, which can provide novel insights into disease processes and development of novel therapeutic interventions.


Assuntos
Bases de Dados Factuais , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Sistemas de Gerenciamento de Base de Dados , Humanos , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...