Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.123
Filtrer
1.
FASEB J ; 38(14): e23810, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39042586

RÉSUMÉ

Osteofibrous dysplasia (OFD) is a rare, benign, fibro-osseous lesion that occurs most commonly in the tibia of children. Tibial involvement leads to bowing and predisposes to the development of a fracture which exhibit significantly delayed healing processes, leading to prolonged morbidity. We previously identified gain-of-function mutations in the MET gene as a cause for OFD. In our present study, we test the hypothesis that gain-of-function MET mutations impair bone repair due to reduced osteoblast differentiation. A heterozygous Met exon 15 skipping (MetΔ15-HET) mouse was created to imitate the human OFD mutation. The mutation results in aberrant and dysregulation of MET-related signaling determined by RNA-seq in the murine osteoblasts extracted from the wide-type and genetic mice. Although no gross skeletal defects were identified in the mice, fracture repair was delayed in MetΔ15-HET mice, with decreased bone formation observed 2-week postfracture. Our data are consistent with a novel role for MET-mediated signaling regulating osteogenesis.


Sujet(s)
Dysplasies osseuses , Modèles animaux de maladie humaine , Dysplasie fibreuse des os , Consolidation de fracture , Ostéogenèse , Protéines proto-oncogènes c-met , Animaux , Souris , Ostéogenèse/génétique , Protéines proto-oncogènes c-met/génétique , Protéines proto-oncogènes c-met/métabolisme , Consolidation de fracture/génétique , Dysplasies osseuses/génétique , Dysplasies osseuses/anatomopathologie , Humains , Dysplasie fibreuse des os/génétique , Dysplasie fibreuse des os/anatomopathologie , Dysplasie fibreuse des os/métabolisme , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Mutation , Différenciation cellulaire , Souris de lignée C57BL , Mâle
2.
Bull Math Biol ; 86(9): 108, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39007985

RÉSUMÉ

Fibrous dysplasia (FD) is a mosaic non-inheritable genetic disorder of the skeleton in which normal bone is replaced by structurally unsound fibro-osseous tissue. There is no curative treatment for FD, partly because its pathophysiology is not yet fully known. We present a simple mathematical model of the disease incorporating its basic known biology, to gain insight on the dynamics of the involved bone-cell populations, and shed light on its pathophysiology. We develop an analytical study of the model and study its basic properties. The existence and stability of steady states are studied, an analysis of sensitivity on the model parameters is done, and different numerical simulations provide findings in agreement with the analytical results. We discuss the model dynamics match with known facts on the disease, and how some open questions could be addressed using the model.


Sujet(s)
Simulation numérique , Dysplasie fibreuse des os , Concepts mathématiques , Modèles biologiques , Mutation , Humains , Dysplasie fibreuse des os/génétique , Dysplasie fibreuse des os/anatomopathologie , Ostéoblastes/anatomopathologie
3.
J Clin Invest ; 134(15)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38885336

RÉSUMÉ

Osteogenesis imperfecta (OI) type V is the second most common form of OI, distinguished by hyperplastic callus formation and calcification of the interosseous membranes, in addition to the bone fragility. It is caused by a recurrent, dominant pathogenic variant (c.-14C>T) in interferon-induced transmembrane protein 5 (IFITM5). Here, we generated a conditional Rosa26-knockin mouse model to study the mechanistic consequences of the recurrent mutation. Expression of the mutant Ifitm5 in osteo-chondroprogenitor or chondrogenic cells resulted in low bone mass and growth retardation. Mutant limbs showed impaired endochondral ossification, cartilage overgrowth, and abnormal growth plate architecture. The cartilage phenotype correlates with the pathology reported in patients with OI type V. Surprisingly, expression of mutant Ifitm5 in mature osteoblasts caused no obvious skeletal abnormalities. In contrast, earlier expression in osteo-chondroprogenitors was associated with an increase in the skeletal progenitor cell population within the periosteum. Lineage tracing showed that chondrogenic cells expressing the mutant Ifitm5 had decreased differentiation into osteoblastic cells in diaphyseal bone. Moreover, mutant IFITM5 disrupted early skeletal homeostasis in part by activating ERK signaling and downstream SOX9 protein, and inhibition of these pathways partially rescued the phenotype in mutant animals. These data identify the contribution of a signaling defect altering osteo-chondroprogenitor differentiation as a driver in the pathogenesis of OI type V.


Sujet(s)
Différenciation cellulaire , Système de signalisation des MAP kinases , Ostéoblastes , Ostéogenèse imparfaite , Facteur de transcription SOX-9 , Animaux , Femelle , Mâle , Souris , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris transgéniques , Mutation , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Ostéogenèse/génétique , Ostéogenèse imparfaite/génétique , Ostéogenèse imparfaite/anatomopathologie , Ostéogenèse imparfaite/métabolisme , Facteur de transcription SOX-9/génétique , Facteur de transcription SOX-9/métabolisme , Cellules souches/métabolisme , Cellules souches/anatomopathologie , Extracellular Signal-Regulated MAP Kinases
4.
Pak J Pharm Sci ; 37(1): 123-128, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38741408

RÉSUMÉ

The study aimed to investigate the effects of aspirin on patients with metastatic colorectal cancer, focusing on circulating tumor DNA levels and bone tissue. Two groups (A and B) of ten patients with osteoporosis were selected for the study. Bone tissue samples were obtained from the patients and cultured under sterile conditions. The aspirin group showed a significant decrease in circulating tumor DNA levels and an increase in bone tissue density compared to the control group. Additionally, osteoblast apoptosis was reduced, while proliferation was enhanced in the aspirin group. The protein pAkt related to the PI3K/Akt signaling pathway was upregulated in the aspirin group. These results indicate that aspirin can effectively lower circulating tumor DNA levels, promote bone tissue proliferation, inhibit apoptosis, and activate the PI3K/Akt signaling pathway, thereby influencing bone cell function. These findings provide a basis for aspirin's potential application in treating metastatic colorectal cancer and encourage further research on its mechanism and clinical use.


Sujet(s)
Apoptose , Acide acétylsalicylique , ADN tumoral circulant , Tumeurs colorectales , Humains , Acide acétylsalicylique/pharmacologie , Acide acétylsalicylique/usage thérapeutique , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Mâle , Femelle , Adulte d'âge moyen , Apoptose/effets des médicaments et des substances chimiques , ADN tumoral circulant/sang , ADN tumoral circulant/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Sujet âgé , Transduction du signal/effets des médicaments et des substances chimiques , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/anatomopathologie , Ostéoblastes/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/génétique , Densité osseuse/effets des médicaments et des substances chimiques , Ostéoporose/traitement médicamenteux
5.
Cells ; 13(10)2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38786073

RÉSUMÉ

In this study of the alterations of Glypicans 1 to 6 (GPCs) and Notum in plasma, bone marrow mesenchymal stromal cells (BM-MSCs) and osteoblasts in Osteoarthritis (OA), the levels of GPCs and Notum in the plasma of 25 patients and 24 healthy subjects were measured. In addition, BM-MSCs from eight OA patients and eight healthy donors were cultured over a period of 21 days using both a culture medium and an osteogenic medium. Protein and gene expression levels of GPCs and Notum were determined using ELISA and qPCR at 0, 7, 14 and 21 days. GPC5 and Notum levels decreased in the plasma of OA patients, while the BM-MSCs of OA patients showed downexpression of GPC6 and upregulation of Notum. A decrease in GPC5 and Notum proteins and an increase in GPC3 were found. During osteogenic differentiation, elevated GPCs 2, 4, 5, 6 and Notum mRNA levels and decreased GPC3 were observed in patients with OA. Furthermore, the protein levels of GPC2, GPC5 and Notum decreased, while the levels of GPC3 increased. Glypicans and Notum were altered in BM-MSCs and during osteogenic differentiation from patients with OA. The alterations found point to GPC5 and Notum as new candidate biomarkers of OA pathology.


Sujet(s)
Glypicanes , Cellules souches mésenchymateuses , Arthrose , Ostéoblastes , Humains , Cellules souches mésenchymateuses/métabolisme , Arthrose/sang , Arthrose/anatomopathologie , Arthrose/génétique , Arthrose/métabolisme , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Mâle , Femelle , Glypicanes/métabolisme , Glypicanes/sang , Glypicanes/génétique , Adulte d'âge moyen , Différenciation cellulaire , Ostéogenèse/génétique , Sujet âgé , Études cas-témoins , Cellules cultivées , Cellules de la moelle osseuse/métabolisme
6.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167171, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38631411

RÉSUMÉ

Patients with advanced chronic kidney disease (CKD) have elevated circulating calcium × phosphate product levels and exhibit soft tissue calcification. Besides the cardiovascular system, calcification is commonly observed in the cornea in CKD patients on hemodialysis. Cardiovascular calcification is a cell-mediated, highly regulated process, and we hypothesized that a similar regulatory mechanism is implicated in corneal calcification with the involvement of corneal epithelial cells (CECs). We established a mouse model of CKD-associated corneal calcification by inducing CKD in DBA/2J mice with an adenine and high phosphate diet. CKD was associated with aorta and corneal calcification as detected by OsteoSense staining and corneal Ca measurement (1.67-fold elevation, p < 0.001). In vitro, excess phosphate and Ca induced human CEC calcification in a dose-dependent and synergistic manner, without any influence on cell viability. High phosphate and Ca-containing osteogenic medium (OM; 2.5 mmol/L excess phosphate and 0.6 mmol/L excess Ca over control) increased the protein expression of Runx2 and induced its nuclear translocation. OM increased the expression of the bone-specific Ca-binding protein osteocalcin (130-fold increase, p < 0.001). Silencing of Runx2 attenuated OM-induced CEC calcification. Immunohistology revealed upregulation of Runx2 and overlapping between the Runx2 and the Alizarin red positive areas of calcification in the cornea of CKD mice. This work sheds light on the mechanism of CKD-induced corneal calcification and provides tools to test calcification inhibitors for the prevention of this detrimental process.


Sujet(s)
Calcinose , Calcium , Sous-unité alpha 1 du facteur CBF , Ostéoblastes , Phosphates , Insuffisance rénale chronique , Animaux , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/complications , Souris , Humains , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Phosphates/métabolisme , Calcium/métabolisme , Calcinose/anatomopathologie , Calcinose/métabolisme , Épithélium antérieur de la cornée/anatomopathologie , Épithélium antérieur de la cornée/métabolisme , Mâle , Souris de lignée DBA , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Modèles animaux de maladie humaine , Phénotype
7.
J Bone Miner Res ; 39(2): 177-189, 2024 Mar 22.
Article de Anglais | MEDLINE | ID: mdl-38477760

RÉSUMÉ

Bone histomorphometry is a well-established approach to assessing skeletal pathology, providing a standard evaluation of the cellular components, architecture, mineralization, and growth of bone tissue. However, it depends in part on the subjective interpretation of cellular morphology by an expert, which introduces bias. In addition, diseases like osteogenesis imperfecta (OI) and fibrous dysplasia are accompanied by changes in the morphology and function of skeletal tissue and cells, hindering consistent evaluation of some morphometric parameters and interpretation of the results. For instance, traditional histomorphometry combined with collagen turnover markers suggested that reduced bone formation in classical OI is accompanied by increased bone resorption. In contrast, the well-documented postpubertal reduction in fractures would be easier to explain by reduced bone resorption after puberty, highlighting the need for less ambiguous measurements. Here we propose an approach to histomorphometry based on in situ mRNA hybridization, which uses Col1a1 as osteoblast and Ctsk as osteoclast markers. This approach can be fully automated and eliminates subjective identification of bone surface cells. We validate these markers based on the expression of Bglap, Ibsp, and Acp5. Comparison with traditional histological and tartrate-resistant acid phosphatase staining of the same sections suggests that mRNA-based analysis is more reliable. Unlike inconclusive traditional histomorphometry of mice with α2(I)-Gly610 to Cys substitution in the collagen triple helix, mRNA-based measurements reveal reduced osteoclastogenesis in 11-wk-old animals consistent with the postpubertal catch-up osteogenesis observed by microCT. We optimize the technique for cryosections of mineralized bone and sections of paraffin-embedded decalcified tissue, simplifying and broadening its applications. We illustrate the application of the mRNA-based approach to human samples using the example of a McCune-Albright syndrome patient. By eliminating confounding effects of altered cellular morphology and the need for subjective morphological evaluation, this approach may provide a more reproducible and accessible evaluation of bone pathology.


Sujet(s)
Os et tissu osseux , Collagène de type I , Modèles animaux de maladie humaine , Ostéogenèse imparfaite , Ostéogenèse imparfaite/anatomopathologie , Ostéogenèse imparfaite/métabolisme , Ostéogenèse imparfaite/génétique , Animaux , Souris , Os et tissu osseux/anatomopathologie , Os et tissu osseux/métabolisme , Collagène de type I/métabolisme , Collagène de type I/génétique , Chaine alpha-1 du collagène de type I , ARN messager/métabolisme , ARN messager/génétique , Ostéoclastes/métabolisme , Ostéoclastes/anatomopathologie , Puberté , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Marqueurs biologiques/métabolisme , Ostéogenèse
9.
Acta Biomater ; 178: 83-92, 2024 04 01.
Article de Anglais | MEDLINE | ID: mdl-38387748

RÉSUMÉ

Bone metastases are the most common milestone in the lethal progression of prostate cancer and prominent in a substantial portion of renal malignancies. Interactions between cancer and bone host cells have emerged as drivers of both disease progression and therapeutic resistance. To best understand these central host-epithelial cell interactions, biologically relevant preclinical models are required. To achieve this goal, we here established and characterized tissue-engineered bone mimetic environments (BME) capable of supporting the growth of patient-derived xenograft (PDX) cells, ex vivo and in vivo. The BME consisted of a polycaprolactone (PCL) scaffold colonized by human mesenchymal stem cells (hMSCs) differentiated into osteoblasts. PDX-derived cells were isolated from bone metastatic prostate or renal tumors, engineered to express GFP or luciferase and seeded onto the BMEs. BMEs supported the growth and therapy response of PDX-derived cells, ex vivo. Additionally, BMEs survived after in vivo implantation and further sustained the growth of PDX-derived cells, their serial transplant, and their application to study the response to treatment. Taken together, this demonstrates the utility of BMEs in combination with patient-derived cells, both ex vivo and in vivo. STATEMENT OF SIGNIFICANCE: Our tissue-engineered BME supported the growth of patient-derived cells and proved useful to monitor the therapy response, both ex vivo and in vivo. This approach has the potential to enable co-clinical strategies to monitor bone metastatic tumor progression and therapy response, including identification and prioritization of new targets for patient treatment.


Sujet(s)
Tumeurs osseuses , Tumeurs de la prostate , Mâle , Humains , Tests d'activité antitumorale sur modèle de xénogreffe , Os et tissu osseux/anatomopathologie , Tumeurs osseuses/thérapie , Tumeurs osseuses/secondaire , Tumeurs de la prostate/anatomopathologie , Ostéoblastes/anatomopathologie
10.
Int J Mol Sci ; 25(3)2024 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-38338785

RÉSUMÉ

Rheumatoid arthritis (RA) is an autoimmune disease that causes inflammation, pain, and ultimately, bone erosion of the joints. The causes of this disease are multifactorial, including genetic factors, such as the presence of the human leukocyte antigen (HLA)-DRB1*04 variant, alterations in the microbiota, or immune factors including increased cytotoxic T lymphocytes (CTLs), neutrophils, or elevated M1 macrophages which, taken together, produce high levels of pro-inflammatory cytokines. In this review, we focused on the function exerted by osteoclasts on osteoblasts and other osteoclasts by means of the release of exosomal microRNAs (miRNAs). Based on a thorough revision, we classified these molecules into three categories according to their function: osteoclast inhibitors (miR-23a, miR-29b, and miR-214), osteoblast inhibitors (miR-22-3p, miR-26a, miR-27a, miR-29a, miR-125b, and miR-146a), and osteoblast enhancers (miR-20a, miR-34a, miR-96, miR-106a, miR-142, miR-199a, miR-324, and miR-486b). Finally, we analyzed potential therapeutic targets of these exosomal miRNAs, such as the use of antagomiRs, blockmiRs, agomiRs and competitive endogenous RNAs (ceRNAs), which are already being tested in murine and ex vivo models of RA. These strategies might have an important role in reestablishing the regulation of osteoclast and osteoblast differentiation making progress in the development of personalized medicine.


Sujet(s)
Polyarthrite rhumatoïde , microARN , Humains , Souris , Animaux , Ostéoclastes/anatomopathologie , microARN/génétique , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/anatomopathologie , Ostéoblastes/anatomopathologie , Macrophages/anatomopathologie , Antagomirs
11.
J Clin Endocrinol Metab ; 109(7): 1803-1813, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38214665

RÉSUMÉ

OBJECTIVE: Deficiency of cartilage-associated protein (CRTAP) can cause extremely rare autosomal recessive osteogenesis imperfecta (OI) type VII. We investigated the pathogenic mechanisms of CRTAP variants through functional studies on bones of patients with OI. METHODS: Two nonconsanguineous families with CRTAP mutations were included and their phenotypes and genotypes were evaluated. Bone specimens were obtained from 1 patient with OI and a normal control during orthopedic surgery. The impacts of the novel variant on the CRTAP transcript were confirmed. The expression levels of CRTAP mRNA and CRTAP protein were analyzed. The quantification of prolyl 3-hydroxylation in the α1 chain of type I collagen was evaluated. RESULTS: Patients with OI type VII had early-onset recurrent fractures, severe osteoporosis, and bone deformities. The c.621 + 1G > A and c.1153-3C > G mutations were identified in CRTAP in the patients with OI. The c.621 + 1G > A variant was a novel mutation that could impair mRNA transcription, leading to a truncated CRTAP protein. In a patient with c.621 + 1G > A and c.1153-3C > G mutations in CRTAP, the mRNA and protein levels of CRTAP in osteoblasts were significantly decreased and the osteoid volume and osteoblast numbers were markedly reduced compared with those in the normal control individual. This was simultaneously accompanied by significantly reduced prolyl 3-hydroxylation at Pro986 in the α1 chain of type I collagen and invisible active bone formation in bone. CONCLUSION: The novel c.621 + 1G > A mutation in CRTAP expands the genotypic spectrum of type VII OI. Biallelic mutations of c.621 + 1G > A and c.1153-3C > G in CRTAP can lead to reduced CRTAP mRNA and deficient CRTAP protein in osteoblasts, which reduces 3-hydroxylation in Pro986 of the α1 chain of type I collagen and impairs bone formation, thus contributing to severe OI type VII.


Sujet(s)
Protéines de la matrice extracellulaire , Chaperons moléculaires , Ostéogenèse imparfaite , Phénotype , Humains , Ostéogenèse imparfaite/génétique , Ostéogenèse imparfaite/anatomopathologie , Protéines de la matrice extracellulaire/génétique , Protéines de la matrice extracellulaire/métabolisme , Mâle , Femelle , Chaperons moléculaires/génétique , Mutation , Enfant , Pedigree , Enfant d'âge préscolaire , Collagène de type I/génétique , Collagène de type I/métabolisme , Génotype , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Chaine alpha-1 du collagène de type I , Adulte , Adolescent
12.
Stem Cells ; 42(4): 360-373, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38153253

RÉSUMÉ

Recent investigations have shown that the necroptosis of tissue cells in joints is important in the development of osteoarthritis (OA). This study aimed to investigate the potential effects of exogenous skeletal stem cells (SSCs) on the necroptosis of subchondral osteoblasts in OA. Human SSCs and subchondral osteoblasts isolated from human tibia plateaus were used for Western blotting, real-time PCR, RNA sequencing, gene editing, and necroptosis detection assays. In addition, the rat anterior cruciate ligament transection OA model was used to evaluate the effects of SSCs on osteoblast necroptosis in vivo. The micro-CT and pathological data showed that intra-articular injections of SSCs significantly improved the microarchitecture of subchondral trabecular bones in OA rats. Additionally, SSCs inhibited the necroptosis of subchondral osteoblasts in OA rats and necroptotic cell models. The results of bulk RNA sequencing of SSCs stimulated or not by tumor necrosis factor α suggested a correlation of SSCs-derived tumor necrosis factor α-induced protein 3 (TNFAIP3) and cell necroptosis. Furthermore, TNFAIP3-derived from SSCs contributed to the inhibition of the subchondral osteoblast necroptosis in vivo and in vitro. Moreover, the intra-articular injections of TNFAIP3-overexpressing SSCs further improved the subchondral trabecular bone remodeling of OA rats. Thus, we report that TNFAIP3 from SSCs contributed to the suppression of the subchondral osteoblast necroptosis, which suggests that necroptotic subchondral osteoblasts in joints may be possible targets to treat OA by stem cell therapy.


Sujet(s)
Arthrose , Protéine-3 induite par le facteur de nécrose tumorale alpha , Animaux , Humains , Rats , Nécroptose , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/thérapie , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Cellules souches/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/métabolisme , Protéine-3 induite par le facteur de nécrose tumorale alpha/pharmacologie
13.
J Exp Clin Cancer Res ; 42(1): 264, 2023 Oct 11.
Article de Anglais | MEDLINE | ID: mdl-37821954

RÉSUMÉ

BACKGROUND: Disseminated tumor cells (DTCs) can enter a dormant state and cause no symptoms in cancer patients. On the other hand, the dormant DTCs can reactivate and cause metastases progression and lethal relapses. In prostate cancer (PCa), relapse can happen after curative treatments such as primary tumor removal. The impact of surgical removal on PCa dissemination and dormancy remains elusive. Furthermore, as dormant DTCs are asymptomatic, dormancy-induction can be an operational cure for preventing metastases and relapse of PCa patients. METHODS: We used a PCa subcutaneous xenograft model and species-specific PCR to survey the DTCs in various organs at different time points of tumor growth and in response to tumor removal. We developed in vitro 2D and 3D co-culture models to recapitulate the dormant DTCs in the bone microenvironment. Proliferation assays, fluorescent cell cycle reporter, qRT-PCR, and Western Blot were used to characterize the dormancy phenotype. We performed RNA sequencing to determine the dormancy signature of PCa. A drug repurposing algorithm was applied to predict dormancy-inducing drugs and a top candidate was validated for the efficacy and the mechanism of dormancy induction. RESULTS: We found DTCs in almost all mouse organs examined, including bones, at week 2 post-tumor cell injections. Surgical removal of the primary tumor reduced the overall DTC abundance, but the DTCs were enriched only in the bones. We found that osteoblasts, but not other cells of the bones, induced PCa cell dormancy. RNA-Seq revealed the suppression of mitochondrial-related biological processes in osteoblast-induced dormant PCa cells. Importantly, the mitochondrial-related biological processes were found up-regulated in both circulating tumor cells and bone metastases from PCa patients' data. We predicted and validated the dormancy-mimicking effect of PF-562,271 (PF-271), an inhibitor of focal adhesion kinase (FAK) in vitro. Decreased FAK phosphorylation and increased nuclear translocation were found in both co-cultured and PF-271-treated C4-2B cells, suggesting that FAK plays a key role in osteoblast-induced PCa dormancy. CONCLUSIONS: Our study provides the first insights into how primary tumor removal enriches PCa cell dissemination in the bones, defines a unique osteoblast-induced PCa dormancy signature, and identifies FAK as a PCa cell dormancy gatekeeper.


Sujet(s)
Récidive tumorale locale , Tumeurs de la prostate , Mâle , Humains , Animaux , Souris , Focal adhesion protein-tyrosine kinases/métabolisme , Récidive tumorale locale/métabolisme , Tumeurs de la prostate/anatomopathologie , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Récidive , Lignée cellulaire tumorale , Microenvironnement tumoral
14.
Nature ; 621(7979): 602-609, 2023 Sep.
Article de Anglais | MEDLINE | ID: mdl-37704733

RÉSUMÉ

Vertebral bone is subject to a distinct set of disease processes from long bones, including a much higher rate of solid tumour metastases1-4. The basis for this distinct biology of vertebral bone has so far remained unknown. Here we identify a vertebral skeletal stem cell (vSSC) that co-expresses ZIC1 and PAX1 together with additional cell surface markers. vSSCs display formal evidence of stemness, including self-renewal, label retention and sitting at the apex of their differentiation hierarchy. vSSCs are physiologic mediators of vertebral bone formation, as genetic blockade of the ability of vSSCs to generate osteoblasts results in defects in the vertebral neural arch and body. Human counterparts of vSSCs can be identified in vertebral endplate specimens and display a conserved differentiation hierarchy and stemness features. Multiple lines of evidence indicate that vSSCs contribute to the high rates of vertebral metastatic tropism observed in breast cancer, owing in part to increased secretion of the novel metastatic trophic factor MFGE8. Together, our results indicate that vSSCs are distinct from other skeletal stem cells and mediate the unique physiology and pathology of vertebrae, including contributing to the high rate of vertebral metastasis.


Sujet(s)
Tumeurs du sein , Lignage cellulaire , Métastase tumorale , Rachis , Cellules souches , Humains , Tumeurs du sein/anatomopathologie , Différenciation cellulaire , Auto-renouvellement cellulaire , Métastase tumorale/anatomopathologie , Ostéoblastes/cytologie , Ostéoblastes/anatomopathologie , Rachis/cytologie , Rachis/anatomopathologie , Cellules souches/cytologie , Cellules souches/métabolisme , Cellules souches/anatomopathologie , Marqueurs biologiques
15.
AIDS Rev ; 25(2): 67-76, 2023.
Article de Anglais | MEDLINE | ID: mdl-37585363

RÉSUMÉ

HIV infection has been reported to cause bone loss and a higher risk of fracture. Under normal conditions, bone metabolism is regulated by mesenchymal cells, osteoclasts differentiated from mononuclear macrophages, osteoblasts, and their expression of regulatory factors, such as receptor activator of nuclear factor-kappa B ligand (RANKL), M-SCF, and transforming growth factor-beta. The balance between bone resorption and osteogenesis depends on the balance between osteoclasts and osteoblasts. In addition, some immune cells, such as B-cells, T-cells, and other non-immune cells expressing RANKL, can contribute to osteoporosis under inflammatory conditions. HIV proteins consist of three types: regulatory proteins, accessory proteins, and structural proteins, which contribute to HIV-mediated bone loss partly by upregulating NF-κB expression, tumor necrosis factor alpha content, and release of inflammatory cytokines. Even worse, although antiretroviral therapy has reduced HIV infection mortality and successfully transformed acquired immunodeficiency syndrome into a chronic disease, its impact on bone loss should not be overlooked, especially when the drug contains tenofovir. This review analyzes some reports focusing on the overall osteolytic situation due to imbalances in osteogenesis and bone resorption due to HIV infection and antiviral therapy. The intrinsic mechanism of bone loss provides a reference for researchers to analyze the risk factors for HIV patients complicated with bone loss and helps clinicians to provide ideas for the intervention and prevention of bone loss during clinical treatment and chronic disease management of HIV patients.


Sujet(s)
Résorption osseuse , Infections à VIH , Humains , Infections à VIH/complications , Infections à VIH/traitement médicamenteux , Infections à VIH/métabolisme , Ostéoclastes/métabolisme , Ostéoclastes/anatomopathologie , Résorption osseuse/métabolisme , Résorption osseuse/anatomopathologie , Résorption osseuse/prévention et contrôle , Ostéogenèse , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Ligand de RANK/métabolisme
16.
Front Immunol ; 14: 1222129, 2023.
Article de Anglais | MEDLINE | ID: mdl-37475866

RÉSUMÉ

The complicated connections and cross talk between the skeletal system and the immune system are attracting more attention, which is developing into the field of Osteoimmunology. In this field, cytokines that are among osteoblasts and osteoclasts play a critical role in bone remodeling, which is a pathological process in the pathogenesis and development of osteoporosis. Those cytokines include the tumor necrosis factor (TNF) family, the interleukin (IL) family, interferon (IFN), chemokines, and so on, most of which influence the bone microenvironment, osteoblasts, and osteoclasts. This review summarizes the effect of cytokines on osteoblasts and osteoclasts in bone remodeling in osteoporosis, aiming to providing the latest reference to the role of immunology in osteoporosis.


Sujet(s)
Ostéoclastes , Ostéoporose , Humains , Cytokines , Ostéoblastes/anatomopathologie , Remodelage osseux
17.
Int J Mol Sci ; 24(12)2023 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-37373043

RÉSUMÉ

Irisin is a peptide secreted by skeletal muscle that plays a major role in bone metabolism. Experiments in mouse models have shown that administration of recombinant irisin prevents disuse-induced bone loss. In this study, we aimed to evaluate the effects of irisin treatment for the prevention of bone loss in the ovariectomized (Ovx) mouse, the animal model commonly used to investigate osteoporosis caused by estrogen deficiency. Micro-Ct analysis conducted on Sham mice (Sham-veh) and Ovx mice treated with vehicle (Ovx-veh) or recombinant irisin (Ovx-irisn) showed bone volume fraction (BV/TV) decreases in femurs (Ovx-veh 1.39± 0.71 vs. Sham-veh 2.84 ± 1.23; p = 0.02) and tibia at both proximal condyles (Ovx-veh 1.97 ± 0.68 vs. Sham-veh 3.48 ± 1.26; p = 0.03) and the subchondral plate (Ovx-veh 6.33 ± 0.36 vs. Sham-veh 8.18 ± 0.41; p = 0.01), which were prevented by treatment with a weekly dose of irisin for 4 weeks. Moreover, histological analysis of trabecular bone showed that irisin increased the number of active osteoblasts per bone perimeter (Ovx-irisin 32.3 ± 3.9 vs. Ovx-veh 23.5 ± 3.6; p = 0.01), while decreasing osteoclasts (Ovx-irisin 7.6 ± 2.4 vs. Ovx-veh 12.9 ± 3.04; p = 0.05). The possible mechanism by which irisin enhances osteoblast activity in Ovx mice is upregulation of the transcription factor Atf4, one of the key markers of osteoblast differentiation, and osteoprotegerin, thereby inhibiting osteoclast formation.


Sujet(s)
Maladies osseuses métaboliques , Ostéoporose , Souris , Animaux , Femelle , Humains , Fibronectines/pharmacologie , Os spongieux/anatomopathologie , Ostéoporose/anatomopathologie , Modèles animaux de maladie humaine , Ostéoblastes/anatomopathologie , Ovariectomie/effets indésirables , Densité osseuse
18.
Environ Pollut ; 332: 121931, 2023 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-37268221

RÉSUMÉ

Fluoride is a widespread pollutant in the environment. There is a high risk of developing skeletal fluorosis from excessive fluoride exposure. Skeletal fluorosis has different phenotypes (including osteosclerotic, osteoporotic and osteomalacic) under the same fluoride exposure and depends on dietary nutrition. However, the existing mechanistic hypothesis of skeletal fluorosis cannot well explain the condition's different pathological manifestations and their logical relation with nutritional factors. Recent studies have shown that DNA methylation is involved in the occurrence and development of skeletal fluorosis. DNA methylation is dynamic throughout life and may be affected by nutrition and environmental factors. We speculated that fluoride exposure leads to the abnormal methylation of genes related to bone homeostasis under different nutritional statuses, resulting in different skeletal fluorosis phenotypes. The mRNA-Seq and target bisulfite sequencing (TBS) result showed differentially methylated genes in rats with different skeletal fluorosis types. The role of the differentially methylated gene Cthrc1 in the formation of different skeletal fluorosis types was explored in vivo and in vitro. Under normal nutritional conditions, fluoride exposure led to hypomethylation and high expression of Cthrc1 in osteoblasts through TET2 demethylase, which promoted osteoblast differentiation by activating Wnt3a/ß-catenin signalling pathway, and participated in the occurrence of osteosclerotic skeletal fluorosis. Meanwhile, the high CTHRC1 protein expression also inhibited osteoclast differentiation. Under poor dietary conditions, fluoride exposure led to hypermethylation and low expression of Cthrc1 in osteoblasts through DNMT1 methyltransferase, and increased the RANKL/OPG ratio, which promoted the osteoclast differentiation and participated in the occurrence of osteoporotic/osteomalacic skeletal fluorosis. Our study expands the understanding of the role of DNA methylation in regulating the formation of different skeletal fluorosis types and provides insights into new prevention and treatment strategies for patients with skeletal fluorosis.


Sujet(s)
Méthylation de l'ADN , Fluorures , Rats , Animaux , Fluorures/toxicité , Ostéogenèse , Ostéoblastes/anatomopathologie , Maturation post-traductionnelle des protéines , Glycoprotéines/génétique
19.
J Pathol ; 260(2): 137-147, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-36811349

RÉSUMÉ

Wnt signaling is a positive regulator of bone formation through the induction of osteoblast differentiation and down-regulation of osteoclast differentiation. We previously reported that muramyl dipeptide (MDP) increases bone volume by increasing osteoblast activity and attenuating osteoclast activity in receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoporotic model mice. In this study, we investigated whether MDP could alleviate post-menopausal osteoporosis through Wnt signaling regulation in an ovariectomy (OVX)-induced mouse osteoporosis model. MDP-administered OVX mice exhibited higher bone volume and bone mineral density than mice of the control group. MDP significantly increased P1NP in the serum of OVX mice, implying increased bone formation. The expression of pGSK3ß and ß-catenin in the distal femur of OVX mice was lower than that in the distal femur of sham-operated mice. Yet, the expression of pGSK3ß and ß-catenin was increased in MDP-administered OVX mice compared with OVX mice. In addition, MDP increased the expression and transcriptional activity of ß-catenin in osteoblasts. MDP inhibited the proteasomal degradation of ß-catenin via the down-regulation of its ubiquitination by GSK3ß inactivation. When osteoblasts were pretreated with Wnt signaling inhibitors, DKK1 or IWP-2, the induction of pAKT, pGSK3ß, and ß-catenin was not observed. In addition, nucleotide oligomerization domain-containing protein 2-deficient osteoblasts were not sensitive to MDP. MDP-administered OVX mice exhibited fewer tartrate-resistant acid phosphatase (TRAP)-positive cells than did OVX mice, attributed to a decrease in the RANKL/OPG ratio. In conclusion, MDP alleviates estrogen deficiency-induced osteoporosis through canonical Wnt signaling and could be an effective therapeutic for the treatment of post-menopausal bone loss. © 2023 The Pathological Society of Great Britain and Ireland.


Sujet(s)
Ostéoporose post-ménopausique , Ostéoporose , Humains , Femelle , Souris , Animaux , Voie de signalisation Wnt , Acétylmuramyl alanyl isoglutamine/métabolisme , Acétylmuramyl alanyl isoglutamine/pharmacologie , Acétylmuramyl alanyl isoglutamine/usage thérapeutique , Ostéoporose/traitement médicamenteux , Ostéoporose/étiologie , Ostéoporose/prévention et contrôle , Densité osseuse , Ostéoporose post-ménopausique/traitement médicamenteux , Ostéoporose post-ménopausique/prévention et contrôle , Ostéoporose post-ménopausique/métabolisme , Différenciation cellulaire , Ostéoclastes/métabolisme , Ostéoblastes/anatomopathologie , Oestrogènes/métabolisme
20.
J Exp Clin Cancer Res ; 41(1): 316, 2022 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-36307871

RÉSUMÉ

Bone metastasis occurs when tumour cells dissociate from primary tumours, enter the circulation (circulating tumour cells, CTCs), and colonize sites in bone (disseminated tumour cells, DTCs). The bone marrow seems to be a particularly dormancy-inducing environment for DTCs, yet the mechanisms of dormancy initiation, reactivation, and interaction within the bone marrow have to be elucidated. Intriguingly, some evidence has suggested that dormancy is a reversible state that is switched 'on' or 'off' depending on the presence of various bone marrow resident cells, particularly osteoclasts and osteoblasts. It has become clear that these two cells contribute to regulating dormant tumour cells in bone both directly (interaction) and indirectly (secreted factors). The involved mechanisms include TGFß signalling, the Wnt signalling axis, the Notch2 pathway, etc. There is no detailed review that specifically focuses on ascertaining the dynamic interactions between tumour cell dormancy and bone remodelling. In addition, we highlighted the roles of inflammatory cytokines during this 'cell-to-cell' communication. We also discussed the potential clinical relevance of remodelling the bone marrow niche in controlling dormant tumour cells. Understanding the unique role of osteoclasts and osteoblasts in regulating tumour dormancy in bone marrow will provide new insight into preventing and treating tumour bone metastasis.


Sujet(s)
Tumeurs osseuses , Cellules tumorales circulantes , Humains , Ostéoclastes/métabolisme , Ostéoclastes/anatomopathologie , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Tumeurs osseuses/anatomopathologie , Cellules tumorales circulantes/métabolisme , Moelle osseuse/métabolisme , Métastase tumorale/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE