Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.130
Filtrar
1.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 256-262, 2024 Mar 20.
Artigo em Chinês | MEDLINE | ID: mdl-38645858

RESUMO

Runt-related transcription factor (RUNX1) is a transcription factor closely involved in hematopoiesis. RUNX1 gene mutation plays an essential pathogenic role in the initiation and development of hematological tumors, especially in acute myeloid leukemia. Recent studies have shown that RUNX1 is also involved in the regulation of bone development and the pathological progression of bone-related diseases. RUNX1 promotes the differentiation of mesenchymal stem cells into chondrocytes and osteoblasts and modulates the maturation and extracellular matrix formation of chondrocytes. The expression of RUNX1 in mesenchymal stem cells, chondrocytes, and osteoblasts is of great significance for maintaining normal bone development and the mass and quality of bones. RUNX1 also inhibits the differentiation and bone resorptive activities of osteoclasts, which may be influenced by sexual dimorphism. In addition, RUNX1 deficiency contributes to the pathogenesis of osteoarthritis, delayed fracture healing, and osteoporosis, which was revealed by the RUNX1 conditional knockout modeling in mice. However, the roles of RUNX1 in regulating the hypertrophic differentiation of chondrocytes, the sexual dimorphism of activities of osteoclasts, as well as bone loss in diabetes mellitus, senescence, infection, chronic inflammation, etc, are still not fully understood. This review provides a systematic summary of the research progress concerning RUNX1 in the field of bone biology, offering new ideas for using RUNX1 as a potential target for bone related diseases, especially osteoarthritis, delayed fracture healing, and osteoporosis.


Assuntos
Desenvolvimento Ósseo , Diferenciação Celular , Condrócitos , Subunidade alfa 2 de Fator de Ligação ao Core , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Humanos , Animais , Desenvolvimento Ósseo/fisiologia , Desenvolvimento Ósseo/genética , Condrócitos/metabolismo , Osteoblastos/metabolismo , Osteoblastos/citologia , Osteoclastos/metabolismo , Osteoclastos/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Doenças Ósseas/genética , Doenças Ósseas/metabolismo , Osteoporose/genética , Osteoporose/metabolismo , Osteoartrite/metabolismo , Osteoartrite/genética , Osteoartrite/etiologia
2.
Int J Mol Sci ; 25(6)2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38542453

RESUMO

Promoting the efficiency of bone regeneration in bone loss diseases is a significant clinical challenge. Traditional therapies often fail to achieve better therapeutic outcomes and shorter treatment times. However, in recent years, extracellular vesicles (EVs) have gained significant attention due to their exceptional osteogenic function in bone regeneration and superior therapeutic effects compared to traditional cell therapy. EVs have emerged as a promising therapy for tissue defect regeneration due to their various physiological functions, such as regulating the immune response and promoting tissue repair and regeneration. Moreover, EVs have good biocompatibility, low immunogenicity, and long-term stability, and can be improved through pretreatment and other methods. Studies investigating the mechanisms by which extracellular vesicles promote bone regeneration and applying EVs from different sources using various methods to animal models of bone defects have increased. Therefore, this paper reviews the types of EVs used for bone regeneration, their sources, roles, delivery pathways, scaffold biomaterials, and applications.


Assuntos
Doenças Ósseas , Vesículas Extracelulares , Animais , Regeneração Óssea/fisiologia , Osteogênese , Vesículas Extracelulares/metabolismo , Materiais Biocompatíveis/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Doenças Ósseas/terapia , Doenças Ósseas/metabolismo
3.
J Bone Miner Res ; 39(3): 326-340, 2024 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-38477820

RESUMO

Proteasome activator subunit 3 (PA28γ) is a member of the proteasome activator family, which mainly regulates the degradation and stability of proteins. Studies have shown that it plays crucial roles in lipid formation, stemness maintenance, and blood vessel formation. However, few studies have clarified the association between PA28γ and bone diseases. Herein, we identified PA28γ as a previously unknown regulator of bone homeostasis that coordinates bone formation and lipid accumulation. PA28γ-knockout mice presented with the characteristics of low bone mass and accumulation of lipids. Suppressed expression of PA28γ restrained the osteogenic differentiation and enhanced the adipogenic differentiation of bone marrow stromal cells (BMSCs). Overexpression of PA28γ promoted osteogenic differentiation and inhibited adipogenic differentiation of BMSCs. Mechanistically, PA28γ interacted with Wnt5α, and the two interactors appeared to be positively correlated. PA28γ mainly activated the downstream Wnt/ß-catenin signaling pathway, which affects BMSCs differentiation homeostasis. Deletion of Wnt5α significantly delayed the promotion of osteogenic differentiation and partially alleviated the inhibitory effect of adipogenic differentiation of BMSCs in the PA28γ-overexpressing group. Furthermore, we demonstrated that PA28γ-knockout mice had an inhibited rate of bone healing in a drill-hole femoral bone defect model in vivo. Therefore, our results confirm the effects of PA28γ on bone formation and bone defect repair, indicating that PA28γ mainly interacts with Wnt5α to activate the Wnt/ß-catenin signaling pathway regulating BMSCs differentiation homeostasis. Our results reveal the function of PA28γ in bone diseases and provide a new theoretical basis for expanding the treatment of bone diseases.


Assuntos
Autoantígenos , Doenças Ósseas , Células-Tronco Mesenquimais , Camundongos , Animais , Complexo de Endopeptidases do Proteassoma/metabolismo , Complexo de Endopeptidases do Proteassoma/farmacologia , Osteogênese , beta Catenina/metabolismo , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Via de Sinalização Wnt/fisiologia , Doenças Ósseas/metabolismo , Células da Medula Óssea/metabolismo , Células Cultivadas , Camundongos Knockout , Lipídeos
4.
Int Immunopharmacol ; 128: 111453, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38241841

RESUMO

BACKGROUND: Tumor necrosis factor-α (TNF-α) is involved in inflammatory responses and promotes cell death and the inhibition of osteogenic differentiation. MicroRNA (miRNA) plays a crucial role in the infected bone diseases, however, the biological role of miRNAs in inflammation-induced impaired osteogenic differentiation remains unclear. This study aimed to explore the role of miRNA-18a-5p (miR-18a) in regulating PANoptosis and osteogenic differentiation in an inflammatory environment via hypoxia-inducible factor-1α (HIF1-α). METHODS: The expression of miR-18a in MC3T3-E1 cells was analyzed using quantitative reverse transcription-polymerase chain reaction in an inflammatory environment induced by TNF-α. The expression of HIF1-α and NLRP3 in LV-miR-18a or sh-miR-18a cells was analyzed using western blotting. Fluorescence imaging for cell death, flow cytometry, and alkaline phosphatase activity analysis were used to analyze the role of miR-18a in TNF-α-induced PANoptosis and the inhibition of osteogenic differentiation. An animal model of infectious bone defect was established to validate the regulatory role of miR-18a in an inflammatory environment. RESULTS: The expression of miRNA-18a in the MC3T3-E1 cell line was significantly lower under TNF-α stimulation than in the normal environment. miR-18a significantly inhibited the expression of HIF1-α and NLRP3, and inhibition of HIF1-α expression further inhibited NLRP3 expression. Furthermore, inhibition of miR-18a expression promoted the TNF-α-induced PANoptosis and inhibition of osteogenic differentiation, whereas miR-18a overexpression and the inhibition of both HIF1-α and NLRP3 reduced the effects of TNF-α. These findings are consistent with those of the animal experiments. CONCLUSION: miRNA-18a negatively affects HIF1-α/NLRP3 expression, inhibits inflammation-induced PANoptosis, and impairs osteogenic differentiation. Thus, it is a potential therapeutic candidate for developing anti-inflammatory strategies for infected bone diseases.


Assuntos
Doenças Ósseas , MicroRNAs , Animais , Apoptose , Doenças Ósseas/metabolismo , Diferenciação Celular , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/metabolismo , MicroRNAs/genética , Necroptose , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Osteoblastos/metabolismo , Osteogênese , Piroptose , Fator de Necrose Tumoral alfa/metabolismo , Camundongos
5.
Bioessays ; 46(3): e2300173, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38161246

RESUMO

Endosteal stem cells are a subclass of bone marrow skeletal stem cell populations that are particularly important for rapid bone formation occurring in growth and regeneration. These stem cells are strategically located near the bone surface in a specialized microenvironment of the endosteal niche. These stem cells are abundant in young stages but eventually depleted and replaced by other stem cell types residing in a non-endosteal perisinusoidal niche. Single-cell molecular profiling and in vivo cell lineage analyses play key roles in discovering endosteal stem cells. Importantly, endosteal stem cells can transform into bone tumor-making cells when deleterious mutations occur in tumor suppressor genes. The emerging hypothesis is that osteoblast-chondrocyte transitional identities confer a special subset of endosteal stromal cells with stem cell-like properties, which may make them susceptible for tumorigenic transformation. Endosteal stem cells are likely to represent an important therapeutic target of bone diseases caused by aberrant bone formation.


Assuntos
Doenças Ósseas , Medula Óssea , Humanos , Medula Óssea/metabolismo , Osteogênese , Osteoblastos/metabolismo , Doenças Ósseas/metabolismo , Doenças Ósseas/patologia , Células-Tronco , Células da Medula Óssea/metabolismo
6.
Cells ; 12(23)2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-38067111

RESUMO

Sex-related differences are a current topic in contemporary science. In addition to hormonal regulation, cell-autonomous mechanisms are important in bone homeostasis and regeneration. In this study, human skeletal stem cells (SSCs) from female and male adults were cultured and analyzed with immunological assays and osteogenic differentiation assessments. Female SSCs exhibited a mean doubling time of 100.6 h, whereas male SSCs displayed a mean doubling time of 168.0 h. Immunophenotyping revealed the expression of the stem cell markers Nestin, CD133, and CD164, accompanied by the neural-crest marker SOX9. Furthermore, multiparameter flow cytometric analyses revealed a substantial population of multipotent SSCs, comprising up to 80% in both sexes. An analysis of the osteogenic differentiation potential demonstrated a strong mineralization in both male and female SSCs under physiological conditions. Recognizing the prevailing association of bone diseases with inflammatory processes, we also analyzed the osteogenic potential of SSCs from both sexes under pro-inflammatory conditions. Upon TNF-α and IL-1ß treatment, we observed no sexual dimorphism on osteogenesis. In summary, we demonstrated the successful isolation and characterization of SSCs capable of rapid osteogenic differentiation. Taken together, in vitro cultured SSCs might be a suitable model to study sexual dimorphisms and develop drugs for degenerative bone diseases.


Assuntos
Doenças Ósseas , Células-Tronco Mesenquimais , Humanos , Masculino , Feminino , Osteogênese , Caracteres Sexuais , Células-Tronco Mesenquimais/metabolismo , Células-Tronco , Doenças Ósseas/metabolismo
7.
J Orthop Surg Res ; 18(1): 975, 2023 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-38114998

RESUMO

BACKGROUND: The traditional Chinese kidney-tonifying granules, known as Bushen Zhongyao Keli (BSZYKL), have been found to stimulate calcium salt deposition, enhance bone formation, and foster bone growth within the bone matrix at sites of bone defects. On the other hand, platelet-rich plasma (PRP) is enriched with various growth factors capable of facilitating the repair of bone defects and enhancing bone strength following fractures. This study is dedicated to investigating the combined efficacy of BSZYKL and PRP gel (PRP-G) in the treatment of bone defects. METHODS: We established a femur defect model in male Sprague-Dawley (SD) rats and filled the defect areas with autologous coccygeal bone and PRP-G. For 8 consecutive weeks, those rats were given with intragastric administration of BSZYKL. Biomechanical characteristics of the femur were assessed 28 days after intramuscular administration. On day 56, bone formation was examined using X-ray, micro-CT, and transmission electron microscopy. Additionally, we analyzed the expression of bone formation markers, Runx2 and Osterix, in femur tissues through qPCR, Western blotting, and immunohistochemistry. RESULTS: Rats receiving the combined treatment of BSZYKL and PRP-G exhibited drastically enhanced femoral peak torsion, failure angle, energy absorption capacity, and torsional stiffness as compared to control group. This combination therapy also led to marked improvements in bone volume, mass, and microarchitecture, accompanied by elevated expressions of Runx2 and Osterix when compared to control group. Notably, the synergistic effects of BSZYKL and PRP-G in treating bone defects surpassed the effects of either treatment alone. CONCLUSIONS: These findings revealed the potential of BSZYKL in combination with PRP-G in improving bone defects.


Assuntos
Doenças Ósseas , Plasma Rico em Plaquetas , Ratos , Masculino , Animais , Ratos Sprague-Dawley , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Fêmur , Doenças Ósseas/metabolismo , Géis , Plasma Rico em Plaquetas/metabolismo , Rim , China , Regeneração Óssea
8.
Curr Osteoporos Rep ; 21(6): 787-805, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37897675

RESUMO

PURPOSE OF REVIEW: Metabolic and genetic bone disorders affect not only bone mass but often also the bone material, including degree of mineralization, matrix organization, and lacunar porosity. The quality of juvenile bone is moreover highly influenced by skeletal growth. This review aims to provide a compact summary of the present knowledge on the complex interplay between bone modeling and remodeling during skeletal growth and to alert the reader to the complexity of bone tissue characteristics in children with bone disorders. RECENT FINDINGS: We describe cellular events together with the characteristics of the different tissues and organic matrix organization (cartilage, woven and lamellar bone) occurring during linear growth. Subsequently, we present typical alterations thereof in disorders leading to over-mineralized bone matrix compared to those associated with low or normal mineral content based on bone biopsy studies. Growth spurts or growth retardation might amplify or mask disease-related alterations in bone material, which makes the interpretation of bone tissue findings in children complex and challenging.


Assuntos
Doenças Ósseas , Calcinose , Criança , Humanos , Osso e Ossos , Doenças Ósseas/metabolismo , Matriz Óssea/metabolismo , Densidade Óssea , Calcinose/metabolismo
9.
Molecules ; 28(13)2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37446790

RESUMO

Neurofibromatosis type 1 is a rare autosomal dominant genetic disorder, with up to 50% of patients clinically displaying skeletal defects. Currently, the pathogenesis of bone disorders in NF1 patients is unclear, and there are no effective preventive and treatment measures. In this study, we found that knockout of the NF1 gene reduced cAMP levels and osteogenic differentiation in an osteoblast model, and icariin activated the cAMP/PKA/CREB pathway to promote osteoblast differentiation of the NF1 gene knockout cell model by increasing intracellular cAMP levels. The PKA selective inhibitor H89 significantly impaired the stimulatory effect of icariin on osteogenesis in the NF1 cell model. In this study, an osteoblast model of NF1 was successfully constructed, and icariin was applied to the cell model for the first time. The results will help to elucidate the molecular mechanism of NF1 bone disease and provide new ideas for the clinical prevention and treatment of NF1 bone disease and drug development in the future.


Assuntos
Doenças Ósseas , Neurofibromatose 1 , Humanos , Osteogênese/genética , Neurofibromatose 1/tratamento farmacológico , Neurofibromatose 1/genética , Neurofibromatose 1/metabolismo , Genes da Neurofibromatose 1 , Técnicas de Inativação de Genes , Diferenciação Celular/genética , Doenças Ósseas/metabolismo , Osteoblastos
10.
Nanoscale ; 15(31): 12840-12852, 2023 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-37482769

RESUMO

Bone loss is prevalent in clinical pathological phenomena such as osteoporosis, which is characterized by decreased osteoblast function and number, increased osteoclast activity, and imbalanced bone homeostasis. However, current treatment strategies for bone diseases are limited. Regulated cell death (RCD) is a programmed cell death pattern activated by the expression of specific genes in response to environmental changes. Various studies have shown that RCD is closely associated with bone diseases, and manipulating the death fate of osteoblasts could contribute to effective bone treatment. Recently, microRNA-targeting therapy drugs have emerged as a potential solution because of their precise targeting, powerful curative effect, and limited side effects. Nevertheless, their clinical application is limited by their inherent instability, easy enzymatic degradation, and poor membrane penetrability. To address this challenge, a self-assembling tetrahedral DNA nanostructure (TDN)-based microRNA (Tmi) delivery system has been proposed. TDN features excellent biocompatibility, cell membrane penetrability, serum stability, and modification versatility, making it an ideal nucleic acid carrier for miRNA protection and intracellular transport. Once inside cells, Tmi can dissociate and release miRNAs to manipulate key molecules in the RCD signaling pathway, thereby regulating bone homeostasis and curing diseases caused by abnormal RCD activation. In this paper, we discuss the impact of the miRNA network on the initiation and termination of four critical RCD programs in bone tissues: apoptosis, autophagy, pyroptosis, and ferroptosis. Furthermore, we present the Tmi delivery system as a miRNA drug vector. This provides insight into the clinical translation of miRNA nucleic acid drugs and the application of miRNA drugs in bone diseases.


Assuntos
Doenças Ósseas , MicroRNAs , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Preparações Farmacêuticas , Osteoclastos/metabolismo , Osso e Ossos , Doenças Ósseas/metabolismo
11.
Front Endocrinol (Lausanne) ; 14: 1150068, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37415664

RESUMO

Diabetes mellitus (DM) has been shown to be a clinical risk factor for bone diseases including osteoporosis and fragility. Bone metabolism is a complicated process that requires coordinated differentiation and proliferation of bone marrow mesenchymal stem cells (BMSCs). Owing to the regenerative properties, BMSCs have laid a robust foundation for their clinical application in various diseases. However, mounting evidence indicates that the osteogenic capability of BMSCs is impaired under high glucose conditions, which is responsible for diabetic bone diseases and greatly reduces the therapeutic efficiency of BMSCs. With the rapidly increasing incidence of DM, a better understanding of the impacts of hyperglycemia on BMSCs osteogenesis and the underlying mechanisms is needed. In this review, we aim to summarize the current knowledge of the osteogenesis of BMSCs in hyperglycemia, the underlying mechanisms, and the strategies to rescue the impaired BMSCs osteogenesis.


Assuntos
Doenças Ósseas , Hiperglicemia , Células-Tronco Mesenquimais , Humanos , Osteogênese , Diferenciação Celular , Doenças Ósseas/metabolismo , Hiperglicemia/metabolismo
12.
J Cell Physiol ; 238(7): 1431-1464, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37183350

RESUMO

Osteoclasts, the bone-resorbing cells, are essential for the bone remodeling process and are involved in the pathophysiology of several bone-related diseases. The extensive corpus of in vitro research and crucial mouse model studies in the 1990s demonstrated the key roles of monocyte/macrophage colony-stimulating factor, receptor activator of nuclear factor kappa B ligand (RANKL) and integrin αvß3 in osteoclast biology. Our knowledge of the molecular mechanisms by which these variables control osteoclast differentiation and function has significantly advanced in the first decade of this century. Recent developments have revealed a number of novel insights into the fundamental mechanisms governing the differentiation and functional activity of osteoclasts; however, these mechanisms have not yet been adequately documented. Thus, in the present review, we discuss various regulatory factors including local and hormonal factors, innate as well as adaptive immune cells, noncoding RNAs (ncRNAs), etc., in the molecular regulation of the intricate and tightly regulated process of osteoclastogenesis. ncRNAs have a critical role as epigenetic controllers of osteoclast physiologic activities, including differentiation and bone resorption. The primary ncRNAs, which include micro-RNAs, circular RNAs, and long noncoding RNAs, form a complex network that affects gene transcription activities associated with osteoclast biological activity. Greater knowledge of the involvement of ncRNAs in osteoclast biological activities will contribute to the treatment and management of several skeletal diseases such as osteoporosis, osteoarthritis, rheumatoid arthritis, etc. Moreover, we further outline potential therapies targeting these regulatory pathways of osteoclastogenesis in distinct bone pathologies.


Assuntos
Doenças Ósseas , Reabsorção Óssea , Animais , Camundongos , Osteogênese/genética , Osteoclastos/metabolismo , Reabsorção Óssea/patologia , Diferenciação Celular/genética , Osteoblastos/metabolismo , Doenças Ósseas/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo
13.
J Cell Physiol ; 238(6): 1368-1380, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37021796

RESUMO

Human mesenchymal stem cells (hMSCs) are the cornerstone of regenerative medicine; large quantities of hMSCs are required via in vitro expansion to meet therapeutic purposes. However, hMSCs quickly lose their osteogenic differentiation potential during in vitro expansion, which is a major roadblock to their clinical applications. In this study, we found that the osteogenic differentiation potential of human bone marrow stem cells (hBMSCs), dental pulp stem cells (hDPSCs), and adipose stem cells (hASCs) was severely impaired after in vitro expansion. To clarify the molecular mechanism underlying this in vitro expansion-related loss of osteogenic capacity in hMSCs, the transcriptome changes following in vitro expansion of these hMSCs were compared. Cysteine-rich secretory protein LCCL domain-containing 2 (CRISPLD2) was identified as the most downregulated gene shared by late passage hBMSCs, hDPSCs, and hASCs. Both the secreted and non-secreted CRISPLD2 proteins progressively declined in hMSCs during in vitro expansion when the cells gradually lost their osteogenic potential. We thus hypothesized that the expression of CRISPLD2 is critical for hMSCs to maintain their osteogenic differentiation potential during in vitro expansion. Our studies showed that the knockdown of CRISPLD2 in early passage hBMSCs inhibited the cells' osteogenic differentiation in a siRNA dose-dependent manner. Transcriptome analysis and immunoblotting indicated that the CRISPLD2 knockdown-induced osteogenesis suppression might be attributed to the downregulation of matrix metallopeptidase 1 (MMP1) and forkhead box Q1 (FOXQ1). Furthermore, adeno-associated virus (AAV)-mediated CRISPLD2 overexpression could somewhat rescue the impaired osteogenic differentiation of hBMSCs during in vitro expansion. These results revealed that the downregulation of CRISPLD2 contributes to the impaired osteogenic differentiation of hMSCs during in vitro expansion. Our findings shed light on understanding the loss of osteogenic differentiation in hMSCs and provide a potential therapeutic target gene for bone-related diseases.


Assuntos
Doenças Ósseas , Células-Tronco Mesenquimais , Humanos , Osteogênese/genética , Células-Tronco Mesenquimais/metabolismo , Diferenciação Celular/genética , RNA Interferente Pequeno/metabolismo , Doenças Ósseas/metabolismo , Células Cultivadas , Fatores de Transcrição Forkhead/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Fatores Reguladores de Interferon/metabolismo
14.
Blood Cancer J ; 13(1): 63, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-37105956

RESUMO

Multiple myeloma (MM) bone disease is a significant cause of morbidity but there is a paucity of data on the impact of malignant plasma cells on adjacent trabecular bone within the BM. Here, we characterize the proteome of trabecular bone tissue from BM biopsies of 56 patients with monoclonal gammopathy of undetermined significance (MGUS), smoldering (SMM), newly diagnosed (NDMM), relapsed MM (RMM), and normal controls. Proteins involved in extracellular matrix (ECM) formation and immunity pathways were decreased in SMM and active MM. Among the proteins most decreased were immunoglobulins, type IV collagen, and TIMP3, suggesting increased immunoparesis and decreased ECM remodelling within trabecular bone. Proteins most increased in SMM/MM were APP (enhances osteoclast activity), ENPP1 (enhances bone mineralization), and MZB1 (required for normal plasmablast differentiation). Pathway analyses showed that proteins involved in gamma -carboxylation, a pathway implicated in osteocalcin function, osteoblast differentiation, and normal hematopoiesis, were also overexpressed in SMM/MM. This study is the first comprehensive proteomic atlas of the BM bone proteome in dysproteinemias. We identify new key proteins and pathways for MM bone disease and potentially impaired hematopoiesis, and show for the first time that gamma -carboxylation pathways are increased in the bone tissue of SMM/MM.


Assuntos
Doenças Ósseas , Gamopatia Monoclonal de Significância Indeterminada , Mieloma Múltiplo , Humanos , Medula Óssea/patologia , Mieloma Múltiplo/patologia , Proteoma/metabolismo , Proteômica , Gamopatia Monoclonal de Significância Indeterminada/diagnóstico , Osso e Ossos/metabolismo , Doenças Ósseas/metabolismo , Progressão da Doença
15.
Physiol Rep ; 11(6): e15650, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36967231

RESUMO

Loss of the mediator Of cell motility 1 (Memo1) in mice caused kidney disease and a bone disease with diminished osteoblast and osteoclast biomarkers in serum, resembling alterations occurring in adynamic bone disease in humans with chronic kidney disease or in Klotho-deficient mice. Here, we investigated whether Memo1 expression in osteoblasts is required for normal bone structure and FGF23 expression. We deleted Memo1 in the osteoblast-osteocyte lineage in Memo fl/fl mice using a Cre under Col1a1 promotor to obtain osteoblast-specific knockout (obKO) mice. We studied organs by micro-computed tomography, qPCR, and western blot. We challenged mice with folic acid for acute kidney injury (AKI) and analyzed organs. Memo obKO were viable without changes in gross anatomy, serum electrolytes, or circulating FGF23 concentrations compared to controls. Memo1 expression was blunted in bones of Memo obKO, whereas it remained unchanged in other organs. Micro-CT revealed no differences between genotypes in bone structure of vertebrae, femur, and tibia. During AKI, Fgf23 expression in calvaria, and renal transcriptional changes were comparable between genotypes. However, renal injury marker expression, circulating FGF23, and parathyroid hormone revealed a sex difference with more severely affected females than males of either genotype. The present data imply that Memo1 in osteoblasts is dispensable for bone structure and expression of Fgf23. Moreover, we found evidence of potential sex differences in murine folic acid nephropathy similar to other experimental models of renal injury that are important to consider when using this experimental model of renal injury.


Assuntos
Injúria Renal Aguda , Doenças Ósseas , Peptídeos e Proteínas de Sinalização Intracelular , Animais , Feminino , Humanos , Masculino , Camundongos , Injúria Renal Aguda/metabolismo , Doenças Ósseas/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Ácido Fólico/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Rim/metabolismo , Osteoblastos/metabolismo , Microtomografia por Raio-X
16.
Int J Mol Sci ; 24(5)2023 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-36902326

RESUMO

The study of osteoblast (OB) metabolism has recently received increased attention due to the considerable amount of energy used during the bone remodeling process. In addition to glucose, the main nutrient for the osteoblast lineages, recent data highlight the importance of amino acid and fatty acid metabolism in providing the fuel necessary for the proper functioning of OBs. Among the amino acids, it has been reported that OBs are largely dependent on glutamine (Gln) for their differentiation and activity. In this review, we describe the main metabolic pathways governing OBs' fate and functions, both in physiological and pathological malignant conditions. In particular, we focus on multiple myeloma (MM) bone disease, which is characterized by a severe imbalance in OB differentiation due to the presence of malignant plasma cells into the bone microenvironment. Here, we describe the most important metabolic alterations involved in the inhibition of OB formation and activity in MM patients.


Assuntos
Doenças Ósseas , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/patologia , Osteoblastos/metabolismo , Osso e Ossos/metabolismo , Doenças Ósseas/metabolismo , Diferenciação Celular/fisiologia , Microambiente Tumoral
17.
Biomater Sci ; 11(7): 2581-2589, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36794531

RESUMO

CrkII, a member of the adaptor protein family, is known to participate in bone homeostasis via the regulation of osteoclasts and osteoblasts. Therefore, silencing CrkII would beneficially impact the bone microenvironment. In this study, CrkII siRNA encapsulated by a bone-targeting peptide (AspSerSer)6-liposome was evaluated for its therapeutic applications using a receptor activator of nuclear factor kappa-B ligand (RANKL)-induced bone loss model. (AspSerSer)6-liposome-siCrkII maintained its gene-silencing ability in both osteoclasts and osteoblasts in vitro and significantly reduced osteoclast formation while increasing osteoblast differentiation in vitro. Fluorescence image analyses showed that the (AspSerSer)6-liposome-siCrkII was present largely in bone, where it remained present for up to 24 hours and was cleared by 48 hours, even when systemically administrated. Importantly, microcomputed-tomography revealed that bone loss induced by RANKL administration was recovered by systemic administration of (AspSerSer)6-liposome-siCrkII. Collectively, the findings of this study suggest that (AspSerSer)6-liposome-siCrkII is a promising therapeutic strategy for the development of treatments for bone diseases, as it overcomes the adverse effects derived from ubiquitous expression via bone-specific delivery of siRNA.


Assuntos
Doenças Ósseas , Reabsorção Óssea , Humanos , Osteogênese , RNA Interferente Pequeno/metabolismo , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Lipossomos/metabolismo , Osteoclastos , Osteoblastos , Doenças Ósseas/metabolismo , Diferenciação Celular
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 31(1): 170-178, 2023 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-36765495

RESUMO

OBJECTIVE: To investigate the expression of pyruvate kinase M2 (PKM2) in bone marrow mesenchymal stem cells (BMSCs) in myeloma bone disease (MBD) and its effect on osteogenic and adipogenic differentiation of BMSCs. METHODS: BMSCs were isolated from bone marrow of five patients with multiple myeloma (MM) (MM group) and five with iron deficiency anemia (control group) for culture and identification. The expression of PKM2 protein were compared between the two groups. The differences between osteogenic and adipogenic differentiation of BMSCs were assessed by using alkaline phosphatase (ALP) and oil red O staining, and detecting marker genes of osteogenesis and adipogenesis. The effect of MM cell line (RPMI-8226) and BMSCs co-culture on the expression of PKM2 was explored. Functional analysis was performed to investigate the correlations of PKM2 expression of MM-derived BMSCs with osteogenic and adipogenic differentiation by employing PKM2 activator and inhibitor. The role of orlistat was explored in regulating PKM2 expression, osteogenic and adipogenic differentiation of MM-derived BMSCs. RESULTS: Compared with control, MM-originated BMSCs possessed the ability of increased adipogenic and decreased osteogenic differentiation, and higher level of PKM2 protein. Co-culture of MM cells with BMSCs markedly up-regulated the expression of PKM2 of BMSCs. Up-regulation of PKM2 expression could promote adipogenic differentiation and inhibit osteogenic differentiation of MM-derived BMSCs, while down-regulation of PKM2 showed opposite effect. Orlistat significantly promoted osteogenic differentiation in MM-derived BMSCs via inhibiting the expression of PKM2. CONCLUSION: The overexpression of PKM2 can induce the inhibition of osteogenic differentiation of BMSCs in MBD. Orlistat can promote the osteogenic differentiation of BMSCs via inhibiting the expression of PKM2, indicating a potential novel agent of anti-MBD therapy.


Assuntos
Doenças Ósseas , Células-Tronco Mesenquimais , Mieloma Múltiplo , Humanos , Adipogenia , Doenças Ósseas/metabolismo , Células da Medula Óssea , Diferenciação Celular , Células Cultivadas , Células-Tronco Mesenquimais/fisiologia , Mieloma Múltiplo/metabolismo , Orlistate/metabolismo , Orlistate/farmacologia , Osteogênese/genética
19.
Acta Histochem ; 125(1): 151998, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36630753

RESUMO

MicroRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) are involved in the regulation of bone metabolism. The BMP/Smad pathway is a key signaling pathway for classical regulation of osteogenic differentiation. Non-coding RNAs (ncRNAs) and the BMP/Smad pathway both have important roles for osteogenic differentiation of stem cells, bone regeneration, and development of bone diseases. There is increasing evidence that ncRNAs interact with the BMP/Smad pathway to regulate not only osteogenic differentiation of stem cells but also progression of bone diseases, such as osteoporosis (OP), myeloma, and osteonecrosis of the femoral head (ONFH), by controlling the expression of bone disease-related genes. Therefore, ncRNAs that interact with BMP/Smad pathway molecules are potential targets for bone regeneration as well as bone disease diagnosis, prevention, and treatment. However, despite extensive studies on ncRNAs associated with the BMP/Smad pathway and osteogenic differentiation of stem cells, there is a lack of comparability. Moreover, some bone disease-associated ncRNAs with low abundance can be difficult to detect and there is a lack of mature delivery systems for their stable translocation to target sites, thus limiting their application. In this review, we summarize the research progress on interactions between ncRNAs and the BMP/Smad pathway during osteogenic differentiation of various stem cells and in the regulation of bone regeneration and bone diseases.


Assuntos
Doenças Ósseas , Osteogênese , Humanos , Doenças Ósseas/metabolismo , Diferenciação Celular/genética , Osteogênese/genética , Transdução de Sinais/fisiologia , Células-Tronco , RNA Longo não Codificante/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Smad/metabolismo
20.
IEEE Trans Nanobioscience ; 22(1): 11-18, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-34928800

RESUMO

Magnetic iron oxide nanoparticles (MNPs) coated with citric acid (MG@CA) are proposed as raw materials for the treatment of bone diseases. Citric acid (CA) was selected as coating due to its role in the stabilization of apatite nanocrystals and as a signaling agent for osteoblast activation. Raloxifene (Ral), curcumine (Cur) and methylene blue (MB) were employed as model drugs as therapeutic agents for bone diseases. Characterization of raw and drug loaded nanosystems was conducted in order to elucidate the mechanisms governing interactions between therapeutics and the magnetic platform. Biocompatibility studies were performed on red blood cells (RBCs) from peripheral human blood. Cytotoxicity was evaluated on endothelial cells (ECs); and viability was studied for bone cells exposed at concentrations of 1, 10 and 100 [Formula: see text]/mL of the magnetic nano-platform. MG@CA exhibited proper physicochemical properties for the applications intended within this work. It presented satisfactory biocompatibility on peripheral red blood cells. Only doses of 100 [Formula: see text]/mL induced a decrease in metabolic activity of ECs and MC3T3-E1 cells. Drug adsorption efficiency was estimated as 62.0, 15.0 and 54.0 % for Ral, Cur and MB and drug loading capability of 12.0, 20.0 and 13.6%, respectively.


Assuntos
Doenças Ósseas , Nanopartículas de Magnetita , Humanos , Células Endoteliais/metabolismo , Sistemas de Liberação de Medicamentos , Cloridrato de Raloxifeno/metabolismo , Doenças Ósseas/metabolismo , Ácido Cítrico/química , Ácido Cítrico/metabolismo , Fenômenos Magnéticos , Nanopartículas de Magnetita/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...