Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.279
Filtrar
1.
Sci Rep ; 13(1): 14556, 2023 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-37666868

RESUMO

Deriving stem cells to regenerate full-thickness human skin is important for treating skin disorders without invasive surgical procedures. Our previous protocol to differentiate human induced pluripotent stem cells (iPSCs) into skin-derived precursor cells (SKPs) as a source of dermal stem cells employs mouse fibroblasts as feeder cells and is therefore unsuitable for clinical use. Herein, we report a feeder-free method for differentiating iPSCs into SKPs by customising culture substrates. We immunohistochemically screened for laminins expressed in dermal papillae (DP) and explored the conditions for inducing the differentiation of iPSCs into SKPs on recombinant laminin E8 (LM-E8) fragments with or without conjugation to domain I of perlecan (PDI), which binds to growth factors through heparan sulphate chains. Several LM-E8 fragments, including those of LM111, 121, 332, 421, 511, and 521, supported iPSC differentiation into SKPs without PDI conjugation. However, the SKP yield was significantly enhanced on PDI-conjugated LM-E8 fragments. SKPs induced on PDI-conjugated LM111-E8 fragments retained the gene expression patterns characteristic of SKPs, as well as the ability to differentiate into adipocytes, osteocytes, and Schwann cells. Thus, PDI-conjugated LM-E8 fragments are promising agents for inducing iPSC differentiation into SKPs in clinical settings.


Assuntos
Diferenciação Celular , Proteoglicanas de Heparan Sulfato , Células-Tronco Pluripotentes Induzidas , Peptídeos e Proteínas de Sinalização Intercelular , Laminina , Fragmentos de Peptídeos , Domínios Proteicos , Pele , Humanos , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proteoglicanas de Heparan Sulfato/química , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Laminina/química , Laminina/farmacologia , Osteócitos/citologia , Osteócitos/efeitos dos fármacos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Células de Schwann/citologia , Células de Schwann/efeitos dos fármacos , Pele/citologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacologia
2.
Cell Tissue Bank ; 24(2): 417-434, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36434165

RESUMO

Bone grafts are widely used to successfully restore structure and function to patients with a broad range of musculoskeletal ailments and bone defects. Autogenous bone grafts are historically preferred because they theoretically contain the three essential components of bone healing (ie, osteoconductivity, osteoinductivity, and osteogenicity), but they have inherent limitations. Allograft bone derived from deceased human donors is one alternative that is also capable of providing both an osteoconductive scaffold and osteoinductive potential but, until recently, lacked the osteogenic component of bone healing. Relatively new, cellular bone allografts (CBAs) were designed to address this need by preserving viable cells. Although most commercially-available CBAs feature mesenchymal stem cells (MSCs), osteogenic differentiation is time-consuming and complex. A more advanced graft, a viable bone allograft (VBA), was thus developed to preserve lineage-committed bone-forming cells, which may be more suitable than MSCs to promote bone fusion. The purpose of this paper was to present the results of preclinical research characterizing VBA. Through a comprehensive series of in vitro and in vivo assays, the present results demonstrate that VBA in its final form is capable of providing all three essential bone remodeling properties and contains viable lineage-committed bone-forming cells, which do not elicit an immune response. The results are discussed in the context of clinical evidence published to date that further supports VBA as a potential alternative to autograft without the associated drawbacks.


Assuntos
Aloenxertos , Transplante Ósseo , Transplante Ósseo/economia , Transplante Ósseo/métodos , Humanos , Transplante Autólogo , Matriz Óssea/química , Osteócitos/citologia , Proliferação de Células , Cálcio/metabolismo , Células da Medula Óssea/metabolismo , Aloenxertos/citologia , Aloenxertos/imunologia , Histocompatibilidade
3.
Sci Rep ; 12(1): 19, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34997043

RESUMO

Invasive dental treatment such as tooth extraction following treatment with strong anti-bone resorptive agents, including bisphosphonates and denosumab, reportedly promotes osteonecrosis of the jaw (ONJ) at the extraction site, but strategies to prevent ONJ remain unclear. Here we show that in mice, administration of either active vitamin D analogues, antibiotics or anti-inflammatory agents can prevent ONJ development induced by tooth extraction during treatment with the bisphosphonate zoledronate. Specifically, tooth extraction during treatment with zoledronate induced osteonecrosis in mice, but administration of either 1,25(OH)2D3 or ED71, both active vitamin D analogues, significantly antagonized osteonecrosis development, even under continuous zoledronate treatment. 1,25(OH)2D3 or ED71 administration also significantly inhibited osteocyte apoptosis induced by tooth extraction and bisphosphonate treatment. Administration of either active vitamin D analogue significantly inhibited elevation of serum inflammatory cytokine levels in mice in response to injection of lipopolysaccharide, an infection mimetic. Furthermore, administration of either anti-inflammatory or antibiotic reagents significantly blocked ONJ development following tooth extraction and zoledronate treatment. These findings suggest that administration of active vitamin D, anti-inflammatory agents or antibiotics could prevent ONJ development induced by tooth extraction in patients treated with zoledronate.


Assuntos
Antibacterianos/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/prevenção & controle , Extração Dentária/efeitos adversos , Vitamina D/administração & dosagem , Ácido Zoledrônico/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/sangue , Osteonecrose da Arcada Osseodentária Associada a Difosfonatos/etiologia , Citocinas/sangue , Difosfonatos/efeitos adversos , Feminino , Humanos , Camundongos Endogâmicos C57BL , Osteócitos/citologia , Osteócitos/efeitos dos fármacos , Vitamina D/análogos & derivados
4.
Biochem Biophys Res Commun ; 590: 132-138, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-34974301

RESUMO

Osteocytes are master regulators of skeletal homeostasis. However, little is known about the molecular mechanism of their differentiation. Epigenetic regulations, especially H3K27me3 modification, play critical roles in cell differentiation. Here, we found that H3K27me3 in the loci of osteocyte-expressing genes decreased during osteocyte differentiation and that H3K27me3 demethylase, Utx, was bound to the loci of those genes. To investigate the physiological functions of Utx in vivo, we generated late osteoblast-to-osteocyte specific Utx knockout mice using Dmp1-cre mice (UtxΔOcy/ΔOcy). Micro CT analyses showed that UtxΔOcy/ΔOcy displayed osteopenic phenotypes with lower bone volume and trabecular number, and greater trabecular separation. Bone histomorphometric analysis showed that bone mineralization and formation were significantly lower in UtxΔOcy/ΔOcy. Furthermore, Dmp1 expression and the number of osteocytes were significantly decreased in UtxΔOcy/ΔOcy. These results suggest that Utx in Dmp1-expressing osteoblast/osteocyte positively regulates osteoblast-to-osteocyte differentiation through H3K27me3 modifications in osteocyte genes. Our results provide new insight into the molecular mechanism of osteocyte differentiation.


Assuntos
Diferenciação Celular , Histona Desmetilases/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Osteoblastos/citologia , Osteócitos/citologia , Animais , Sequência de Bases , Doenças Ósseas Metabólicas/genética , Osso Esponjoso/diagnóstico por imagem , Osso Esponjoso/patologia , Contagem de Células , Diferenciação Celular/genética , Regulação para Baixo/genética , Epigenoma , Loci Gênicos , Histona Desmetilases/deficiência , Metilação , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/metabolismo , Osteócitos/metabolismo , Fenótipo , Processamento de Proteína Pós-Traducional , Transcriptoma/genética
5.
Endocrinology ; 163(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34669927

RESUMO

Thyroid hormones (TH) are essential for skeletal development and adult bone homeostasis. Their bioavailability is determined by specific transporter proteins at the cell surface. The TH-specific transporter monocarboxylate transporter 8 (MCT8) was recently reported as a regulator of bone mass in mice. Given that high systemic triiodothyronine (T3) levels in Mct8 knockout (KO) mice are still able to cause trabecular bone loss, alternative TH transporters must substitute for MCT8 function in bone. In this study, we analyzed the skeletal phenotypes of male Oatp1c1 KO and Mct10 KO mice, which are euthyroid, and male Mct8/Oatp1c1 and Mct8/Mct10 double KO mice, which have elevated circulating T3 levels, to unravel the role of TH transport in bone. MicroCT analysis showed no significant trabecular bone changes in Oatp1c1 KO mice at 4 weeks and 16 weeks of age compared with wild-type littermate controls, whereas 16-week-old Mct8/Oatp1c1 double KO animals displayed trabecular bone loss. At 12 weeks, Mct10 KO mice, but not Mct8/Mct10 double KO mice, had decreased trabecular femoral bone volume with reduced osteoblast numbers. By contrast, lack of Mct10 in 24-week-old mice led to trabecular bone gain at the femur with increased osteoblast numbers and decreased osteoclast numbers whereas Mct8/Mct10 double KO did not alter bone mass. Neither Mct10 nor Mct8/Mct10 deletion affected vertebral bone structures at both ages. In vitro, osteoblast differentiation and activity were impaired by Mct10 and Mct8/Mct10-deficiency. These data demonstrate that MCT10, but not OATP1C1, is a site- and age-dependent regulator of bone mass and turnover in male mice.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Osso e Ossos/metabolismo , Animais , Transporte Biológico , Fenômenos Biomecânicos , Osso Esponjoso/metabolismo , Diferenciação Celular , Fêmur/fisiologia , Homeostase , Masculino , Camundongos , Camundongos Knockout , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteócitos/citologia , Fenótipo , Simportadores/metabolismo , Glândula Tireoide/metabolismo , Hormônios Tireóideos/metabolismo , Tri-Iodotironina/metabolismo , Microtomografia por Raio-X
6.
Biomed Mater Eng ; 33(3): 209-219, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34744060

RESUMO

BACKGROUND: Magnesium (Mg) alloy have biodegradation and mechanical properties that are similar to those of human bone, making it a promising candidate material for inclusion in implantable medical devices. OBJECTIVE: The osteointegration effect of Mg alloy scaffolds with different corrosion rates were studied and evaluated in large bone defect models. METHOD: Mg-Sr and Mg-Ca alloy scaffolds with a 20-µm Micro-arc oxidation (MAO) coating were used to repair critical bone defects for subsequent assessment of each alloy's degradation and osteointegration by X-ray, Micro-CT, fluorescence and histological examination. RESULTS: At 12 weeks post-implantation, each defect was found to be effectively reconstructed by either of the Mg alloys based on X-ray and Micro-CT images. The corrosion rate (CR) of each Mg alloy - as calculated based on micro-computed tomography information - demonstrated that the MAO coating could provide effective protection for only 4 weeks post-surgery. From weeks 8 to 12, the CR of the Mg-Ca alloy scaffold increased from 1.34 ± 0.23 mm/y to 1.57 ± 0.16 mm/y. In contrast, the CR of the Mg-Sr alloy scaffold decreased from 0.58 ± 0.14 mm/y to 0.54 ± 0.16 mm/y. However, fluorescence and histological examination revealed more mature, closely and regularly arranged newborn osteocytes at the Mg-Ca scaffold-fracture interface e from weeks 8 to 12 after surgery. CONCLUSION: The Mg-Sr scaffold was more corrosion resistant and the Mg-Ca scaffold yielded a better overall repair, which indicates that the CR of magnesium alloys matches the rate of new bone formation and is the key to repair bone defects as a bone substitute.


Assuntos
Ligas , Materiais Revestidos Biocompatíveis , Magnésio , Osteócitos , Corrosão , Humanos , Osteócitos/citologia , Osteogênese , Microtomografia por Raio-X
7.
Physiol Rev ; 102(1): 379-410, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34337974

RESUMO

Osteocytes, former osteoblasts encapsulated by mineralized bone matrix, are far from being passive and metabolically inactive bone cells. Instead, osteocytes are multifunctional and dynamic cells capable of integrating hormonal and mechanical signals and transmitting them to effector cells in bone and in distant tissues. Osteocytes are a major source of molecules that regulate bone homeostasis by integrating both mechanical cues and hormonal signals that coordinate the differentiation and function of osteoclasts and osteoblasts. Osteocyte function is altered in both rare and common bone diseases, suggesting that osteocyte dysfunction is directly involved in the pathophysiology of several disorders affecting the skeleton. Advances in osteocyte biology initiated the development of novel therapeutics interfering with osteocyte-secreted molecules. Moreover, osteocytes are targets and key distributors of biological signals mediating the beneficial effects of several bone therapeutics used in the clinic. Here we review the most recent discoveries in osteocyte biology demonstrating that osteocytes regulate bone homeostasis and bone marrow fat via paracrine signaling, influence body composition and energy metabolism via endocrine signaling, and contribute to the damaging effects of diabetes mellitus and hematologic and metastatic cancers in the skeleton.


Assuntos
Remodelação Óssea/fisiologia , Osteoclastos/citologia , Osteócitos/citologia , Osteogênese/fisiologia , Animais , Reabsorção Óssea/metabolismo , Diferenciação Celular/fisiologia , Humanos
8.
PLoS One ; 16(12): e0261660, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34941939

RESUMO

Earlier, we proposed the "mechanosome" concept as a testable model for understanding how mechanical stimuli detected by cell surface adhesion molecules are transmitted to modulate gene expression inside cells. Here, for the first time we document a putative mechanosome involving Src, Pyk2 and MBD2 in MLO-Y4 osteocytes with high spatial resolution using FRET-FLIM. Src-Pyk2 complexes were concentrated at the periphery of focal adhesions and the peri-nuclear region. Pyk2-MBD2 complexes were located primarily in the nucleus and peri-nuclear region. Lifetime measurements indicated that Src and MBD2 did not interact directly. Finally, mechanical stimulation by fluid flow induced apparent accumulation of Src-Pyk2 protein complexes in the peri-nuclear/nuclear region, consistent with the proposed behavior of a mechanosome in response to a mechanical stimulus.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Osteócitos/metabolismo , Quinases da Família src/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/análise , Transferência Ressonante de Energia de Fluorescência , Quinase 2 de Adesão Focal/análise , Adesões Focais/metabolismo , Mecanotransdução Celular , Camundongos , Osteócitos/citologia , Quinases da Família src/análise
9.
Sci Rep ; 11(1): 22593, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34799645

RESUMO

Mesenchymal progenitors differentiate into several tissues including bone, cartilage, and adipose. Targeting these cells in vivo is challenging, making mesenchymal progenitor cell lines valuable tools to study tissue development. Mesenchymal stem cells (MSCs) can be isolated from humans and animals; however, obtaining homogenous, responsive cells in a reproducible fashion is challenging. As such, we developed two mesenchymal progenitor cell (MPC) lines, MPC1 and MPC2, generated from bone marrow of male C57BL/6 mice. These cells were immortalized using the temperature sensitive large T-antigen, allowing for thermal control of proliferation and differentiation. Both MPC1 and MPC2 cells are capable of osteogenic, adipogenic, and chondrogenic differentiation. Under osteogenic conditions, both lines formed mineralized nodules, and stained for alizarin red and alkaline phosphatase, while expressing osteogenic genes including Sost, Fgf23, and Dmp1. Sost and Dmp1 mRNA levels were drastically reduced with addition of parathyroid hormone, thus recapitulating in vivo responses. MPC cells secreted intact (iFGF23) and C-terminal (cFGF23) forms of the endocrine hormone FGF23, which was upregulated by 1,25 dihydroxy vitamin D (1,25D). Both lines also rapidly entered the adipogenic lineage, expressing adipose markers after 4 days in adipogenic media. MPC cells were also capable of chondrogenic differentiation, displaying increased expression of cartilaginous genes including aggrecan, Sox9, and Comp. With the ability to differentiate into multiple mesenchymal lineages and mimic in vivo responses of key regulatory genes/proteins, MPC cells are a valuable model to study factors that regulate mesenchymal lineage allocation as well as the mechanisms that dictate transcription, protein modification, and secretion of these factors.


Assuntos
Adipócitos/citologia , Técnicas de Cultura de Células , Condrócitos/citologia , Células-Tronco Mesenquimais/citologia , Osteócitos/citologia , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Proliferação de Células , Fator de Crescimento de Fibroblastos 23/metabolismo , Imunofenotipagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo
10.
Curr Osteoporos Rep ; 19(6): 604-615, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34757588

RESUMO

PURPOSE OF THE REVIEW: Osteocytes are cells embedded within the bone matrix, but their function and specific patterns of gene expression remain only partially defined; this is beginning to change with recent studies using transcriptomics. This unbiased approach can generate large amounts of data and is now being used to identify novel genes and signalling pathways within osteocytes both at baseline conditions and in response to stimuli. This review outlines the methods used to isolate cell populations containing osteocytes, and key recent transcriptomic studies that used osteocyte-containing preparations from bone tissue. RECENT FINDINGS: Three common methods are used to prepare samples to examine osteocyte gene expression: digestion followed by sorting, laser capture microscopy, and the isolation of cortical bone shafts. All these methods present challenges in interpreting the data generated. Genes previously not known to be expressed by osteocytes have been identified and variations in osteocyte gene expression have been reported with age, sex, anatomical location, mechanical loading, and defects in bone strength. A substantial proportion of newly identified transcripts in osteocytes remain functionally undefined but several have been cross-referenced with functional data. Future work and improved methods (e.g. scRNAseq) likely provide useful resources for the study of osteocytes and important new information on the identity and functions of this unique cell type within the skeleton.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/metabolismo , Técnicas de Cultura de Células/métodos , Expressão Gênica , Osteócitos/citologia , Osteócitos/metabolismo , Humanos , Transcriptoma
11.
Biomolecules ; 11(10)2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34680033

RESUMO

Adipose-derived mesenchymal stromal cells (ASCs) are multipotent stem cells which can differentiate into various cell types, including osteocytes and adipocytes. Due to their ease of harvesting, multipotency, and low tumorigenicity, they are a prime candidate for the development of novel interventional approaches in regenerative medicine. ASCs exhibit slow, spontaneous Ca2+ oscillations and the manipulation of Ca2+ signalling via electrical stimulation was proposed as a potential route for promoting their differentiation in vivo. However, the effects of differentiation-inducing treatments on spontaneous Ca2+ oscillations in ASCs are not yet fully characterised. In this study, we used 2-photon live Ca2+ imaging to assess the fraction of cells showing spontaneous oscillations and the frequency of the oscillation (measured as interpeak interval-IPI) in ASCs undergoing osteogenic or adipogenic differentiation, using undifferentiated ASCs as controls. The measurements were carried out at 7, 14, and 21 days in vitro (DIV) to assess the effect of time in culture on Ca2+ dynamics. We observed that both time and differentiation treatment are important factors associated with a reduced fraction of cells showing Ca2+ oscillations, paralleled by increased IPI times, in comparison with untreated ASCs. Both adipogenic and osteogenic differentiation resulted in a reduction in Ca2+ dynamics, such as the fraction of cells showing intracellular Ca2+ oscillations and their frequency. Adipogenic differentiation was associated with a more pronounced reduction of Ca2+ dynamics compared to cells differentiating towards the osteogenic fate. Changes in Ca2+ associated oscillations with a specific treatment had already occurred at 7 DIV. Finally, we observed a reduction in Ca2+ dynamics over time in untreated ASCs. These data suggest that adipogenic and osteogenic differentiation cell fates are associated with specific changes in spontaneous Ca2+ dynamics over time. While this observation is interesting and provides useful information to understand the functional correlates of stem cell differentiation, further studies are required to clarify the molecular and mechanistic correlates of these changes. This will allow us to better understand the causal relationship between Ca2+ dynamics and differentiation, potentially leading to the development of novel, more effective interventions for both bone regeneration and control of adipose growth.


Assuntos
Adipogenia/genética , Diferenciação Celular/genética , Células-Tronco Mesenquimais/citologia , Osteogênese/genética , Adipócitos/citologia , Cálcio/metabolismo , Sinalização do Cálcio/genética , Linhagem Celular , Linhagem da Célula/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteócitos/citologia , Medicina Regenerativa
12.
Cells ; 10(9)2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34572070

RESUMO

Mesenchymal stem cells (MSCs) are accessible, abundantly available, and capable of regenerating; they have the potential to be developed as therapeutic agents for diseases. However, concerns remain in their further application. In this study, we developed a SMall cell+Ultra Potent+Scale UP cell (SMUP-Cell) platform to improve whole-cell processing, including manufacturing bioreactors and xeno-free solutions for commercialization. To confirm the superiority of SMUP-Cell improvements, we demonstrated that a molecule secreted by SMUP-Cells is capable of polarizing inflammatory macrophages (M1) into their anti-inflammatory phenotype (M2) at the site of injury in a pain-associated osteoarthritis (OA) model. Lipopolysaccharide-stimulated macrophages co-cultured with SMUP-Cells expressed low levels of M1-phenotype markers (CD11b, tumor necrosis factor-α, interleukin-1α, and interleukin-6), but high levels of M2 markers (CD163 and arginase-1). To identify the paracrine action underlying the anti-inflammatory effect of SMUP-Cells, we employed a cytokine array and detected increased levels of pentraxin-related protein-3 (PTX-3). Additionally, PTX-3 mRNA silencing was applied to confirm PTX-3 function. PTX-3 silencing in SMUP-Cells significantly decreased their therapeutic effects against monosodium iodoacetate (MIA)-induced OA. Thus, PTX-3 expression in injected SMUP-Cells, applied as a therapeutic strategy, reduced pain in an OA model.


Assuntos
Proteína C-Reativa/metabolismo , Macrófagos/imunologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Osteoartrite/terapia , Osteócitos/citologia , Dor/prevenção & controle , Componente Amiloide P Sérico/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Inflamação/terapia , Injeções Intra-Articulares , Ácido Iodoacético/toxicidade , Ativação de Macrófagos/imunologia , Masculino , Osteoartrite/induzido quimicamente , Osteoartrite/metabolismo , Osteoartrite/patologia , Dor/etiologia , Dor/metabolismo , Dor/patologia , Ratos , Ratos Sprague-Dawley
13.
Mol Med ; 27(1): 122, 2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34583640

RESUMO

BACKGROUND: Osteoporosis is a widespread and serious metabolic bone disease. At present, revealing the molecular mechanisms of osteoporosis and developing effective prevention and treatment methods are of great significance to health worldwide. LncRNA is a non-coding RNA peptide chain with more than 200 nucleotides. Researchers have identified many lncRNAs implicated in the development of diseases and lncRNA H19 is an example. RESULTS: A large amount of evidence supports the fact that long non-coding RNA (lncRNA) genes, such as H19, have multiple, far-reaching effects on various biological functions. It has been found that lncRNA H19 has a role in the regulation of different types of cells in the body including the osteoblasts, osteocytes, and osteoclasts found in bones. Therefore, it can be postulated that lncRNA H19 affects the incidence and development of osteoporosis. CONCLUSION: The prospect of targeting lncRNA H19 in the treatment of osteoporosis is promising because of the effects that lncRNA H19 has on the process of osteogenic differentiation. In this review, we summarize the molecular pathways and mechanisms of lncRNA H19 in the pathogenesis of osteoporosis and summarize the research progress of targeting H19 as a treatment option. Research is emerging that explores more effective treatment possibilities for bone metabolism diseases using molecular targets.


Assuntos
Diferenciação Celular/genética , Proliferação de Células/genética , Regulação da Expressão Gênica , Osteogênese/genética , Osteoporose/genética , RNA Longo não Codificante/genética , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Humanos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteócitos/citologia , Osteócitos/metabolismo , Transdução de Sinais/genética
14.
J Mater Sci Mater Med ; 32(9): 124, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34524552

RESUMO

The extracellular matrix regulates cell survival, proliferation, and differentiation. In vitro two-dimensional cell experiments are typically performed on a plastic plate or a substrate of a single extracellular matrix constituent such as collagen or calcium phosphate. As these approaches do not include extracellular matrix proteins or growth factors, they fail to mimic a complex cell microenvironment. The cell-derived matrix is an alternative platform for better representing the in vivo microenvironment in vitro. Standard decellularization of a cell-derived matrix is achieved by combining chemical and physical methods. In this study, we compared the decellularization efficacy of several methods: ammonium hydroxide, sodium dodecyl sulfate (SDS), or Triton X-100 with cold or heat treatment on a matrix of Saos-2 cells. We found that the protocols containing SDS were cytotoxic during recellularization. Heat treatment at 47 °C was not cytotoxic, removed cellular constituents, inactivated alkaline phosphatase activity, and maintained the levels of calcium deposition. Subsequently, we investigated the differentiation efficiency of a direct bone coculture system in the established decellularized Saos-2 matrix, an inorganic matrix of calcium phosphate, and a plastic plate as a control. We found that the decellularized Saos-2 cell matrix obtained by heat treatment at 47 °C enhanced osteoclast differentiation and matrix mineralization better than the inorganic matrix and the control. This simple and low-cost method allows us to create a Saos-2 decellularized matrix that can be used as an in vivo-like support for the growth and differentiation of bone cells.


Assuntos
Matriz Extracelular Descelularizada/síntese química , Osteoblastos/citologia , Osteoblastos/fisiologia , Engenharia Tecidual/métodos , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiologia , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Matriz Extracelular Descelularizada/química , Matriz Extracelular Descelularizada/farmacologia , Humanos , Osteoblastos/efeitos dos fármacos , Osteócitos/citologia , Osteócitos/efeitos dos fármacos , Osteócitos/fisiologia , Células THP-1 , Tecidos Suporte/química
15.
Tissue Cell ; 73: 101628, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34479072

RESUMO

Pulmonary mesenchymal stem cells (PMSCs) have great potential in lung tissue repair and regeneration, which have been isolated from some mammalian species, including mice, bovine and pig. However, the isolation, characteristics and differentiation potential of rat PMSCs have not been reported. In this study, we successfully isolated PMSCs from Sprague-Dawley rat fetal lung tissue in vitro for the first time and attempted to evaluate its multilineage differentiation potentials. The cultured PMSCs showed typical spindle-shaped morphology and high proliferative potential, and could be passaged for at least 13 passages and maintained high hereditary stability with more than 93.6 % of cells were diploid (2n = 42) by G-banding analysis. Furthermore, the PMSCs could express mesenchymal markers Sca-1, CD29, CD44, CD73 and CD90, but not hematopoietic markers CD34 and CD45. Besides, the expression of cell markers of AT2 (SFTPC), AT1 (PDPN) and macrophage (CD11b) were also negative. Cell cycle examination revealed majority of the PMSCs were in G0/G1 phase, which are similar with previously reported pig PMSCs. In addition, the PMSCs were multipotent and could differentiated into osteocytes, adipocytes, hepatocytes and neurons in vitro. Together, the present study demonstrated the stemness and multi-differentiation potentials of rat PMSCs, which conferred a potential regenerative cell resource for cell regenerative therapy of lung injury.


Assuntos
Feto/citologia , Pulmão/citologia , Pulmão/embriologia , Células-Tronco Mesenquimais/citologia , Adipócitos/citologia , Animais , Biomarcadores/metabolismo , Ciclo Celular , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Forma Celular , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Hepatócitos/citologia , Cariótipo , Neurônios/citologia , Osteócitos/citologia , Ratos Sprague-Dawley
16.
Ann N Y Acad Sci ; 1503(1): 5-22, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34403146

RESUMO

Different cellular mechanisms contribute to osteocyte development. And while critical roles for members of the zinc finger protein SNAI family (SNAIs) have been discussed in cancer-related models, there are few reviews summarizing their importance for chondrocyte-to-osteocyte development. To help fill this gap, we review the roles of SNAIs in the development of mature osteocytes from chondrocytes, including the regulation of chondro- and osteogenesis through different signaling pathways and in programmed cell death. We also discuss how epigenetic factors-including DNA methylation, histone methylation and acetylation, and noncoding RNAs-contribute differently to both chondrocyte and osteocyte development. To better grasp the important roles of SNAIs in bone development, we also review genotype-phenotype correlations in different animal models. We end with comments about the possible importance of the SNAI family in cartilage/bone development and the potential applications for therapeutic goals.


Assuntos
Diferenciação Celular , Condrócitos/citologia , Condrócitos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Osteócitos/citologia , Osteócitos/metabolismo , Fatores de Transcrição da Família Snail/genética , Animais , Desenvolvimento Ósseo/genética , Diferenciação Celular/genética , Condrogênese/genética , Epigênese Genética , Humanos , Osteogênese/genética , Transdução de Sinais , Fatores de Transcrição da Família Snail/metabolismo
17.
Appl Physiol Nutr Metab ; 46(12): 1525-1534, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34370961

RESUMO

The effects of treadmill interval training (IT) and free-fall exercise were evaluated on bone parameters including osteocyte related characteristics. Thirty-eight 4-month-old male Wistar rats were randomly divided into a control (C) group and exercise groups: IT, 10 free-fall impacts/day with a 10-s (FF10) or 20-s interval between drops (FF20), 5 days/week, for 9 weeks. We assessed bone mineral density (BMD); microarchitecture by µCT; mechanical strength by a 3-point bending test; density and occupancy of the osteocyte lacunae by toluidine blue staining; osteocalcin and NTx systemic levels by ELISA; and bone tissue Sost messenger RNA (mRNA) expression by RT-PCR. NTx levels were significantly lower in exercise groups as compared with the C group. In exercise groups the Sost mRNA expression was significantly lower than in C. Tb.N was significantly higher for IT and FF20 compared with the C group. Tb.Sp was significantly lower in FF10 compared with the C group. Both IT and FF20 were associated with higher tibial lacunar density as compared with FF10. compared with FF10, IT fat mass was lower, while tibial osteocyte lacunae occupancy and systemic osteocalcin level were higher. All exercise modes were efficient in reducing bone resorption. Both IT and free-fall impact with appropriate recovery periods, which may be beneficial for bone health and osteocyte-related characteristics. Novelty: Interval training is beneficial for bone mineral density. Exercises decreased both bone resorption and inhibition of bone formation (Sost mRNA). Longer interval recovery time favors osteocyte lacunae density.


Assuntos
Densidade Óssea , Proteínas Morfogenéticas Ósseas/genética , Osso Esponjoso/citologia , Marcadores Genéticos/genética , Osteocalcina/sangue , Osteócitos/fisiologia , Condicionamento Físico Animal/métodos , Condicionamento Físico Animal/fisiologia , Animais , Fenômenos Biomecânicos , Composição Corporal , Reabsorção Óssea , Osso Esponjoso/anatomia & histologia , Contagem de Células , Colágeno Tipo I/análise , Expressão Gênica , Masculino , Osteócitos/citologia , Osteogênese/fisiologia , Peptídeos/análise , Exercício Pliométrico , RNA Mensageiro/genética , Distribuição Aleatória , Ratos Wistar , Corrida/fisiologia , Resistência à Tração
18.
Int J Mol Sci ; 22(16)2021 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-34445472

RESUMO

Background: Platelet-Rich Plasma (PRP) induces bone regeneration; however, there is low evidence supporting its efficacy in bone healing. The lack of a standardized protocol of administration represents the main obstacle to its use in the clinical routine for bone defects' treatment. The purpose of this study was to characterize PRP and elucidate its osteogenic potential. Methods: Platelet count, fibrinogen levels, and growth factors concentration were measured in PRP obtained by four apheresis procedures. HOB-01-C1, a pre-osteocytic cell line, was used to examine the effects of different PRP dilutions (from 1% to 50%) on cell viability, growth, and differentiation. Gene expression of RUNX2, PHEX, COL1A1, and OCN was also assayed. Results: PRP showed a mean 4.6-fold increase of platelets amount compared to whole blood. Among the 36 proteins evaluated, we found the highest concentrations for PDGF isoforms, EGF, TGF-ß and VEGF-D. PDGF-AA positively correlated with platelet counts. In three of the four tested units, 25% PRP induced a growth rate comparable to the positive control (10% FBS); whereas, for all the tested units, 10% PRP treatment sustained differentiation. Conclusions: This study showed that PRP from apheresis stimulates proliferation and differentiation of pre-osteocyte cells through the release of growth factors from platelets.


Assuntos
Remoção de Componentes Sanguíneos/métodos , Regulação da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Osteócitos/citologia , Osteogênese , Plasma Rico em Plaquetas/metabolismo , Medicina Regenerativa , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Perfilação da Expressão Gênica , Humanos , Técnicas In Vitro , Osteócitos/metabolismo
19.
Biomolecules ; 11(8)2021 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-34439795

RESUMO

The cranial bones constitute the protective structures of the skull, which surround and protect the brain. Due to the limited repair capacity, the reconstruction and regeneration of skull defects are considered as an unmet clinical need and challenge. Previously, it has been proposed that the periosteum and dura mater provide reparative progenitors for cranial bones homeostasis and injury repair. In addition, it has also been speculated that the cranial mesenchymal stem cells reside in the perivascular niche of the diploe, namely, the soft spongy cancellous bone between the interior and exterior layers of cortical bone of the skull, which resembles the skeletal stem cells' distribution pattern of the long bone within the bone marrow. Not until recent years have several studies unraveled and validated that the major mesenchymal stem cell population of the cranial region is primarily located within the suture mesenchyme of the skull, and hence, they are termed suture mesenchymal stem cells (SuSCs). Here, we summarized the characteristics of SuSCs, this newly discovered stem cell population of cranial bones, including the temporospatial distribution pattern, self-renewal, and multipotent properties, contribution to injury repair, as well as the signaling pathways and molecular mechanisms associated with the regulation of SuSCs.


Assuntos
Regeneração Óssea/genética , Suturas Cranianas/citologia , Células-Tronco Mesenquimais/citologia , Osteócitos/citologia , Fraturas Cranianas/genética , Animais , Proteína Axina/genética , Proteína Axina/metabolismo , Catepsina K/genética , Catepsina K/metabolismo , Diferenciação Celular , Proliferação de Células , Suturas Cranianas/crescimento & desenvolvimento , Suturas Cranianas/lesões , Suturas Cranianas/metabolismo , Craniossinostoses/genética , Craniossinostoses/metabolismo , Craniossinostoses/patologia , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteócitos/metabolismo , Transdução de Sinais , Fraturas Cranianas/metabolismo , Fraturas Cranianas/patologia , Proteína GLI1 em Dedos de Zinco/genética , Proteína GLI1 em Dedos de Zinco/metabolismo
20.
Int J Mol Sci ; 22(15)2021 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-34361085

RESUMO

A novel aptamer-based competitive drug screening platform for osteoporosis was devised in which fluorescence-labeled, sclerostin-specific aptamers compete with compounds from selected chemical libraries for the binding of immobilized recombinant human sclerostin to achieve high-throughput screening for potential small-molecule sclerostin inhibitors and to facilitate drug repurposing and drug discovery. Of the 96 selected inhibitors and FDA-approved drugs, six were shown to result in a significant decrease in the fluorescence intensity of the aptamer, suggesting a higher affinity toward sclerostin compared with that of the aptamer. The targets of these potential sclerostin inhibitors were correlated to lipid or bone metabolism, and several of the compounds have already been shown to be potential osteogenic activators, indicating that the aptamer-based competitive drug screening assay offered a potentially reliable strategy for the discovery of target-specific new drugs. The six potential sclerostin inhibitors suppressed the level of both intracellular and/or extracellular sclerostin in mouse osteocyte IDG-SW3 and increased alkaline phosphatase activity in IDG-SW3 cells, human bone marrow-derived mesenchymal stem cells and human fetal osteoblasts hFOB1.19. Potential small-molecule drug candidates obtained in this study are expected to provide new therapeutics for osteoporosis as well as insights into the structure-activity relationship of sclerostin inhibitors for rational drug design.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Aptâmeros de Nucleotídeos/química , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteócitos/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Aptâmeros de Nucleotídeos/isolamento & purificação , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteócitos/citologia , Osteócitos/metabolismo , Osteoporose/metabolismo , Osteoporose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...