Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.064
Filter
1.
J Cell Mol Med ; 28(16): e70027, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39159149

ABSTRACT

Ageing is the most prominent risk for osteoarthritis (OA) development. This study aimed to investigate the role of phosphoinositide-specific phospholipase Cγ (PLCγ) 1, previously linked to OA progression, in regulating age-related changes in articular cartilage and subchondral bone. d-galactose (d-Gal) was employed to treat chondrocytes from rats and mice or injected intraperitoneally into C57BL/6 mice. RTCA, qPCR, Western blot and immunohistochemistry assays were used to evaluate cell proliferation, matrix synthesis, senescence genes and senescence-associated secretory phenotype, along with PLCγ1 expression. Subchondral bone morphology was assessed through micro-CT. In mice with chondrocyte-specific Plcg1 deficiency (Plcg1flox/flox; Col2a1-CreERT), articular cartilage and subchondral bone were examined over different survival periods. Our results showed that d-Gal induced chondrocyte senescence, expedited articular cartilage ageing and caused subchondral bone abnormalities. In d-Gal-induced chondrocytes, diminished PLCγ1 expression was observed, and its further inhibition by U73122 exacerbated chondrocyte senescence. Plcg1flox/flox; Col2a1-CreERT mice exhibited more pronounced age-related changes in articular cartilage and subchondral bone compared to Plcg1flox/flox mice. Therefore, not only does d-Gal induce senescence in chondrocytes and age-related changes in articular cartilage and subchondral bone, as well as diminished PLCγ1 expression, but PLCγ1 deficiency in chondrocytes may also accelerate age-related changes in articular cartilage and subchondral bone. PLCγ1 may be a promising therapeutic target for mitigating age-related changes in joint tissue.


Subject(s)
Cartilage, Articular , Chondrocytes , Mice, Inbred C57BL , Phospholipase C gamma , Animals , Chondrocytes/metabolism , Phospholipase C gamma/metabolism , Phospholipase C gamma/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Mice , Aging/metabolism , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/etiology , Cellular Senescence , Rats , Estrenes/pharmacology , Galactose/metabolism , Cell Proliferation , Male , Bone and Bones/metabolism , Bone and Bones/pathology , Bone and Bones/diagnostic imaging , Pyrrolidinones/pharmacology
2.
Bone Res ; 12(1): 46, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39183236

ABSTRACT

Osteogenesis imperfecta (OI) is a disorder of low bone mass and increased fracture risk due to a range of genetic variants that prominently include mutations in genes encoding type I collagen. While it is well known that OI reflects defects in the activity of bone-forming osteoblasts, it is currently unclear whether OI also reflects defects in the many other cell types comprising bone, including defects in skeletal vascular endothelium or the skeletal stem cell populations that give rise to osteoblasts and whether correcting these broader defects could have therapeutic utility. Here, we find that numbers of skeletal stem cells (SSCs) and skeletal arterial endothelial cells (AECs) are augmented in Col1a2oim/oim mice, a well-studied animal model of moderate to severe OI, suggesting that disruption of a vascular SSC niche is a feature of OI pathogenesis. Moreover, crossing Col1a2oim/oim mice to mice lacking a negative regulator of skeletal angiogenesis and bone formation, Schnurri 3 (SHN3), not only corrected the SSC and AEC phenotypes but moreover robustly corrected the bone mass and spontaneous fracture phenotypes. As this finding suggested a strong therapeutic utility of SHN3 inhibition for the treatment of OI, a bone-targeting AAV was used to mediate Shn3 knockdown, rescuing the Col1a2oim/oim phenotype and providing therapeutic proof-of-concept for targeting SHN3 for the treatment of OI. Overall, this work both provides proof-of-concept for inhibition of the SHN3 pathway and more broadly addressing defects in the stem/osteoprogenitor niche as is a strategy to treat OI.


Subject(s)
Disease Models, Animal , Osteogenesis Imperfecta , Stem Cell Niche , Animals , Osteogenesis Imperfecta/pathology , Osteogenesis Imperfecta/genetics , Mice , Bone and Bones/pathology , Bone and Bones/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Collagen Type I/metabolism , Collagen Type I/genetics , Osteogenesis/drug effects , Mice, Inbred C57BL
3.
Biofabrication ; 16(4)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39116896

ABSTRACT

Osteoporosis is the most common bone disorder, which is a highly dangerous condition that can promote bone metastases. As the current treatment for osteoporosis involves long-term medication therapy and a cure for bone metastasis is not known, ongoing efforts are required for drug development for osteoporosis. Animal experiments, traditionally used for drug development, raise ethical concerns and are expensive and time-consuming. Organ-on-a-chip technology is being developed as a tool to supplement such animal models. In this study, we developed a bone-on-a-chip by co-culturing osteoblasts, osteocytes, and osteoclasts in an extracellular matrix environment that can represent normal bone, osteopenia, and osteoporotic conditions. We then simulated bone metastases using breast cancer cells in three different bone conditions and observed that bone metastases were most active in osteoporotic conditions. Furthermore, it was revealed that the promotion of bone metastasis in osteoporotic conditions is due to increased vascular permeability. The bone-on-a-chip developed in this study can serve as a platform to complement animal models for drug development for osteoporosis and bone metastasis.


Subject(s)
Bone Neoplasms , Lab-On-A-Chip Devices , Osteoporosis , Osteoporosis/pathology , Osteoporosis/drug therapy , Bone Neoplasms/secondary , Bone Neoplasms/pathology , Animals , Humans , Osteoblasts/metabolism , Coculture Techniques , Mice , Osteoclasts/pathology , Osteoclasts/metabolism , Osteocytes/pathology , Osteocytes/metabolism , Bone and Bones/pathology , Cell Line, Tumor , Female
4.
Cells ; 13(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39120270

ABSTRACT

Degenerative disorders like osteoarthritis (OA) might impair the ability of tissue-resident mesenchymal stem/stromal cells (MSCs) for tissue regeneration. As primary cells with MSC-like properties are exploited for patient-derived stem cell therapies, a detailed evaluation of their in vitro properties is needed. Here, we aimed to compare synovium-derived and bone-derived MSCs in early hip OA with those of patients without OA (non-OA). Tissues from three synovial sites of the hip (paralabral synovium, cotyloid fossa, inner surface of peripheral capsule) were collected along with peripheral trabecular bone from 16 patients undergoing hip arthroscopy (8 early OA and 8 non-OA patients). Primary cells isolated from tissues were compared using detailed in vitro analyses. Gene expression profiling was performed for the skeletal stem cell markers podoplanin (PDPN), CD73, CD164 and CD146 as well as for immune-related molecules to assess their immunomodulatory potential. Synovium-derived and bone-derived MSCs from early OA patients showed comparable clonogenicity, cumulative population doublings, osteogenic, adipogenic and chondrogenic potential, and immunophenotype to those of non-OA patients. High PDPN/low CD146 profile (reminiscent of skeletal stem cells) was identified mainly for non-OA MSCs, while low PDPN/high CD146 mainly defined early OA MSCs. These data suggest that MSCs from early OA patients are not affected by degenerative changes in the hip. Moreover, the synovium represents an alternative source of MSCs for patient-derived stem cell therapies, which is comparable to bone. The expression profile reminiscent of skeletal stem cells suggests the combination of low PDPN and high CD146 as potential biomarkers in early OA.


Subject(s)
Mesenchymal Stem Cells , Synovial Membrane , Humans , Mesenchymal Stem Cells/metabolism , Synovial Membrane/pathology , Synovial Membrane/metabolism , Female , Male , Middle Aged , Cell Differentiation , Aged , Osteoarthritis/pathology , Osteoarthritis/metabolism , Bone and Bones/pathology , Bone and Bones/metabolism , Adult , Biomarkers/metabolism , Chondrogenesis , Osteogenesis , Cells, Cultured
5.
Front Endocrinol (Lausanne) ; 15: 1286365, 2024.
Article in English | MEDLINE | ID: mdl-39129916

ABSTRACT

Introduction: Fibroblast growth factor 20 (Fgf20), a member of the Fgf9 subfamily, was identified as an important regulator of bone differentiation and homeostasis processes. However, the role of Fgf20 in bone physiology has not been approached yet. Here we present a comprehensive bone phenotype analysis of mice with functional ablation of Fgf20. Methods: The study conducts an extensive analysis of Fgf20 knockout mice compared to controls, incorporating microCT scanning, volumetric analysis, Fgf9 subfamily expression and stimulation experiment and histological evaluation. Results: The bone phenotype could be detected especially in the area of​ the lumbar and caudal part of the spine and in fingers. Regarding the spine, Fgf20-/- mice exhibited adhesions of the transverse process of the sixth lumbar vertebra to the pelvis as well as malformations in the distal part of their tails. Preaxial polydactyly and polysyndactyly in varying degrees of severity were also detected. High resolution microCT analysis of distal femurs and the fourth lumbar vertebra showed significant differences in structure and mineralization in both cortical and trabecular bone. These findings were histologically validated and may be associated with the expression of Fgf20 in chondrocytes and their progenitors. Moreover, histological sections demonstrated increased bone tissue formation, disruption of Fgf20-/- femur cartilage, and cellular-level alterations, particularly in osteoclasts. We also observed molar dysmorphology, including root taurodontism, and described variations in mineralization and dentin thickness. Discussion: Our analysis provides evidence that Fgf20, together with other members of the Fgf9 subfamily, plays a crucial regulatory role in skeletal development and bone homeostasis.


Subject(s)
Fibroblast Growth Factors , Mice, Knockout , Animals , Mice , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , X-Ray Microtomography , Bone and Bones/metabolism , Bone and Bones/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/abnormalities , Calcification, Physiologic , Male , Osteogenesis , Female , Mice, Inbred C57BL , Phenotype
6.
Cell Biochem Funct ; 42(6): e4107, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39154288

ABSTRACT

Despite their diverse physiologies and roles, the heart, skeletal muscles, and smooth muscles all derive from a common embryonic source as bones. Moreover, bone tissue, skeletal and smooth muscles, and the heart share conserved signaling pathways. The maintenance of skeletal health is precisely regulated by osteocytes, osteoblasts, and osteoclasts through coordinated secretion of bone-derived factors known as osteokines. Increasing evidence suggests the involvement of osteokines in regulating atherosclerotic vascular disease. Therefore, this review aims to examine the evidence for the role of osteokines in atherosclerosis development and progression comprehensively. Specifically discussed are extensively studied osteokines in atherosclerosis such as osteocalcin, osteopontin, osteoprotegerin, and fibroblast growth factor 23. Additionally, we highlighted the effects of exercise on modulating these key regulators derived from bone tissue metabolism. We believe that gaining an enhanced understanding of how osteocalcin contributes to the process of atherosclerosis will enable us to develop targeted and comprehensive therapeutic strategies against diseases associated with its progression.


Subject(s)
Atherosclerosis , Osteocalcin , Humans , Atherosclerosis/metabolism , Atherosclerosis/pathology , Animals , Osteocalcin/metabolism , Osteopontin/metabolism , Fibroblast Growth Factors/metabolism , Osteoprotegerin/metabolism , Bone and Bones/metabolism , Bone and Bones/pathology
7.
Cell Rep Med ; 5(8): 101665, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39168101

ABSTRACT

Mesenchymal stromal cell (MSC) senescence is a key factor in skeletal aging, affecting the potential of MSC applications. Identifying targets to prevent MSC and skeletal senescence is crucial. Here, we report increased miR-29 expression in bone tissues of aged mice, osteoporotic patients, and senescent MSCs. Genetic overexpression of miR-29 in Prx1-positive MSCs significantly accelerates skeletal senescence, reducing cortical bone thickness and trabecular bone mass, while increasing femur cross-sectional area, bone marrow adiposity, p53, and senescence-associated secretory phenotype (SASP) levels. Mechanistically, miR-29 promotes senescence by upregulating p53 via targeting Kindlin-2 mRNA. miR-29 knockdown in BMSCs impedes skeletal senescence, enhances bone mass, and accelerates calvarial defect regeneration, also reducing lipopolysaccharide (LPS)-induced organ injuries and mortality. Thus, our findings underscore miR-29 as a promising therapeutic target for senescence-related skeletal diseases and acute inflammation-induced organ damage.


Subject(s)
Cellular Senescence , Mesenchymal Stem Cells , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Cellular Senescence/genetics , Humans , Mice , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Mice, Inbred C57BL , Mesenchymal Stem Cell Transplantation/methods , Senescence-Associated Secretory Phenotype/genetics , Osteoporosis/genetics , Osteoporosis/pathology , Osteoporosis/therapy , Osteoporosis/metabolism , Male , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Bone and Bones/metabolism , Bone and Bones/pathology , Female , Lipopolysaccharides/pharmacology , Aging
8.
Int Immunopharmacol ; 138: 112611, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38976947

ABSTRACT

OBJECTIVE: Osteoporosis (OP) is a disease characterized by decreased bone mass, deteriorated microstructure, and increased fragility and fracture risk. The diagnosis and prevention of OP and its complications have become major public health challenges. Therefore, exploring the complex ecological connections between the immune and skeletal systems may provide new insights for clinical prevention and treatment strategies. METHODS: First, we performed single-cell RNA sequencing on human lumbar lamina tissue and conducted clustering and subgroup analysis of quality-controlled single-cell transcriptome data to identify target subgroups. Subsequently, enrichment analysis and pseudotime analysis were performed. In addition, we conducted in-depth studies on the gene regulatory network between different cell subgroups and the communication between bone immune cells. RESULTS: In this study, we identified several cell subgroups that may be involved in the progression of OP. For example, the CCL4+ NKT and CXCL8+ neutrophils subgroups promote OP progression by mediating an inflammatory environment that disrupts bone homeostasis, and the MNDA+ Mac subgroup promotes osteoclast differentiation to promote OP. Moreover, the TNFAIP6+ Obl, NR4A2+ B and HMGN2+ erythrocyte subgroups promoted the balance of bone metabolism and suppressed OP. In the cell communication network, Obl closely interacts with immune cell subgroups through the CXCR4-CXCL12, CTGF-ITGB2, and TNFSF14-TNFRSF14 axes. CONCLUSION: Our research revealed specific subgroups and intercellular interactions that play crucial roles in the pathogenesis of OP, providing potential new insights for more precise therapeutic interventions for OP.


Subject(s)
Osteoporosis , Single-Cell Analysis , Humans , Osteoporosis/immunology , Osteoporosis/genetics , Sequence Analysis, RNA , Immune System/immunology , Transcriptome , Female , Bone and Bones/metabolism , Bone and Bones/immunology , Bone and Bones/pathology , Gene Regulatory Networks , Osteoclasts/immunology , Cell Communication , Male
9.
Tissue Cell ; 89: 102422, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39003912

ABSTRACT

Tumour necrosis factor alpha (TNF-α) is a pleiotropic cytokine synthesised primarily by mononuclear cells; it has a potent pro-inflammatory effect, playing a crucial role in metabolic, immune, and inflammatory diseases. This cytokine has been studied in various biological systems. In bone tissue, TNF-α plays an integral role in skeletal disorders such as osteoporosis, fracture repair and rheumatoid arthritis through its involvement in regulating the balance between osteoblasts and osteoclasts, mediating inflammatory responses, promoting angiogenesis and exacerbating synovial proliferation. The biological effect TNF-α exerts in this context is determined by a combination of the signalling pathway it activates, the type of receptor it binds, and the concentration and duration of exposure. This review summarises the participation and pathophysiological role of TNF-α in osteoporosis, bone damage repair, chronic immunoinflammatory bone disease and spinal cord injury, and discusses its main mechanisms.


Subject(s)
Osteoporosis , Tumor Necrosis Factor-alpha , Humans , Osteoporosis/metabolism , Osteoporosis/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Inflammation/metabolism , Inflammation/pathology , Osteoblasts/metabolism , Bone Diseases/metabolism , Bone Diseases/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Signal Transduction
10.
Biomed Pharmacother ; 177: 117139, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39018871

ABSTRACT

The notion that obesity can be a protective factor for bone health is a topic of ongoing debate. Increased body weight may have a positive impact on bone health due to its mechanical effects and the production of estrogen by adipose tissue. However, recent studies have found a higher risk of bone fracture and delayed bone healing in elderly obese patients, which may be attributed to the heightened risk of bone immune regulation disruption associated with obesity. The balanced functions of bone cells such as osteoclasts, osteoblasts, and osteocytes, would be subverted by aberrant and prolonged immune responses under obese conditions. This review aims to explore the intricate relationship between obesity and bone health from the perspective of osteoimmunology, elucidate the impact of disturbances in bone immune regulation on the functioning of bone cells, including osteoclasts, osteoblasts, and osteocytes, highlighting the deleterious effects of obesity on various diseases development such as rheumatoid arthritis (RA), osteoarthritis (AS), bone fracture, periodontitis. On the one hand, weight loss may achieve significant therapeutic effects on the aforementioned diseases. On the other hand, for patients who have difficulty in losing weight, the osteoimmunological therapies could potentially serve as a viable approach in halting the progression of these disease. Additional research in the field of osteoimmunology is necessary to ascertain the optimal equilibrium between body weight and bone health.


Subject(s)
Bone and Bones , Obesity , Humans , Obesity/immunology , Obesity/complications , Animals , Bone and Bones/immunology , Bone and Bones/metabolism , Bone and Bones/pathology , Osteocytes/metabolism , Osteocytes/immunology , Osteoclasts/immunology , Osteoclasts/metabolism , Osteoblasts/immunology , Osteoblasts/metabolism , Bone Remodeling/immunology
11.
PLoS One ; 19(7): e0306819, 2024.
Article in English | MEDLINE | ID: mdl-39083447

ABSTRACT

Paleopathology, the study of diseases and injuries from the fossil record, allows for a unique view into the life of prehistoric animals. Pathologies have nowadays been described in nearly all groups of fossil vertebrates, especially dinosaurs. Despite the large number of skeletons, pathologies had never been reported in the sauropodomorph Plateosaurus trossingensis. Here we describe the first pathologies of Plateosaurus using two individuals with pathologies in the chevrons of the tail, from the Upper Triassic of Trossingen, SW Germany. The two specimens each contain three consecutive pathological chevrons. Our results show that the pathologies were caused by external trauma in one individual and potentially tendinous trauma in the other. Healing of the lesions allowed survival of both animals. Using additional pathological specimens found in other collections and from multiple localities, we observe that 14.8% of all individuals of Plateosaurus contain pathologies within their chevrons, suggesting it was a vulnerable bone.


Subject(s)
Dinosaurs , Fossils , Animals , Dinosaurs/anatomy & histology , Germany , Paleopathology , Bone and Bones/pathology , Bone and Bones/anatomy & histology
12.
Ann Afr Med ; 23(2): 149-153, 2024 Apr 01.
Article in French, English | MEDLINE | ID: mdl-39028162

ABSTRACT

CONTEXT: A major indication for referrals for bone scans (BS) to establish or exclude skeletal metastases. Few patients are referred with clinically indeterminate diagnosis or cancer of unknown primary (CUP), to search for bony metastases or primary tumor. AIMS: This study aimed to assess the usefulness or otherwise for BS in such instances. SETTINGS AND DESIGN: A retrospective cross-sectional study of BS performed for CUP and indeterminate diagnosis from 2012 to 2016 in the nuclear medicine unit of a tertiary teaching hospital. SUBJECTS AND METHODS: The study involved reviews of technetium-99m diphosphonate BS for patients with CUP. BSs were reported by nuclear physicians as normal, normal variants, suspicious for metastases or as malignant, and with solitary or multiple skeletal metastases. STATISTICAL ANALYSIS USED: Data were analyzed using SPSS version 21 for descriptive analysis. Continuous data were displayed as means along with their standard deviation; categorical data were tabulated as frequencies and percentages. RESULTS: Of 2156 BS, 42 (0.02%) were eligible. Patients were aged 27-86 years, mainly in the sixth and seventh decades of life. Bone metastases were identified in 14 (33%) of these patients, whereas 17 BS (40%) were normal, BS appeared equivocal in 10 (23.8%). CONCLUSIONS: Single-photon emission computed tomography/computed tomography availability would resolve the clinical dilemma in patients with equivocal and apparently normal BS.


Résumé Contexte:Une indication majeure pour les références à des scintigraphies osseuses (BS) pour établir ou exclure des métastases squelettiques. Peu de patients sont référés avec diagnostic cliniquement indéterminé ou cancer primitif inconnu (CUP), pour rechercher des métastases osseuses ou une tumeur primitive.Objectifs:Cette étude visait à évaluer l'utilité ou non de la BS dans de tels cas.Paramètres et conception:Une étude transversale rétrospective sur le BS réalisée pour CUP et diagnostic indéterminé de 2012 à 2016 dans l'unité de médecine nucléaire d'un hôpital universitaire tertiaire.Sujets et Méthodes:L'étude comprenait des examens du diphosphonate de technétium-99m BS pour les patients atteints de CUP. Des BS ont été signalés par des médecins nucléaires comme normales, variantes normales, suspectes de métastases ou malignes, et avec métastases squelettiques solitaires ou multiples.Analyses statistiques utilisé:Les données ont été analysées à l'aide de SPSS version 21 pour une analyse descriptive. Les données continues ont été affichées comme moyennes avec leur l'ecarts t; les données catégorielles ont été présentées sous forme de fréquences et de pourcentages.Résultats:Sur 2156 BS, 42 (0,02%) étaient éligibles. Les patients étaient âgés 27 à 86 ans, principalement dans les sixième et septième décennies de la vie. Des métastases osseuses ont été identifiées chez 14 (33 %) de ces patients, alors que 17 BS (40 %) étaient normaux, la BS semblait équivoque dans 10 cas (23,8 %).Conclusions:Tomodensitométrie par émission de photons uniques/tomodensitométrie la disponibilité résoudrait le dilemme clinique chez les patients atteints de BS équivoque et apparemment normale.


Subject(s)
Bone Neoplasms , Humans , Female , Male , Bone Neoplasms/secondary , Bone Neoplasms/diagnostic imaging , Middle Aged , Cross-Sectional Studies , Retrospective Studies , Aged , Adult , Aged, 80 and over , Neoplasms, Unknown Primary/diagnostic imaging , Neoplasms, Unknown Primary/pathology , Radionuclide Imaging , Radiopharmaceuticals , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Technetium Tc 99m Medronate
13.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000517

ABSTRACT

Advancing age is associated with several age-related diseases (ARDs), with musculoskeletal conditions impacting millions of elderly people worldwide. With orthopedic conditions contributing towards considerable number of patients, a deeper understanding of bone aging is the need of the hour. One of the underlying factors of bone aging is cellular senescence and its associated senescence associated secretory phenotype (SASP). SASP comprises of pro-inflammatory markers, cytokines and chemokines that arrest cell growth and development. The accumulation of SASP over several years leads to chronic low-grade inflammation with advancing age, also known as inflammaging. The pathways and molecular mechanisms focused on bone senescence and inflammaging are currently limited but are increasingly being explored. Most of the genes, pathways and mechanisms involved in senescence and inflammaging coincide with those associated with cancer and other ARDs like osteoarthritis (OA). Thus, exploring these pathways using techniques like sequencing, identifying these factors and combatting them with the most suitable approach are crucial for healthy aging and the early detection of ARDs. Several approaches can be used to aid regeneration and reduce senescence in the bone. These may be pharmacological, non-pharmacological and lifestyle interventions. With increasing evidence towards the intricate relationship between aging, senescence, inflammation and ARDs, these approaches may also be used as anti-aging strategies for the aging bone marrow (BM).


Subject(s)
Aging , Bone and Bones , Cellular Senescence , Inflammation , Humans , Cellular Senescence/genetics , Inflammation/genetics , Inflammation/metabolism , Aging/genetics , Bone and Bones/metabolism , Bone and Bones/pathology , Animals , Senescence-Associated Secretory Phenotype/genetics , Signal Transduction
14.
Am J Biol Anthropol ; 185(1): e24994, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38963678

ABSTRACT

OBJECTIVE: Here we investigate infectious diseases that potentially contribute to osteological lesions in individuals from the early medieval necropolis of La Olmeda (6th-11th c. CE) in North Iberia. MATERIALS AND METHODS: We studied a minimum number of 268 individuals (33 adult females; 38 adult males, 77 unknown/indeterminate sex; and 120 non-adults), including articulated and commingled remains. Individuals with differential diagnoses suggesting chronic systemic infectious diseases were sampled and bioinformatically screened for ancient pathogen DNA. RESULTS: Five non-adults (and no adults) presented skeletal evidence of chronic systemic infectious disease (1.87% of the population; 4.67% of non-adults). The preferred diagnoses for these individuals included tuberculosis, brucellosis, and malaria. Ancient DNA fragments assigned to the malaria-causing pathogen, Plasmodium spp., were identified in three of the five individuals. Observed pathology includes lesions generally consistent with malaria; however, additional lesions in two of the individuals may represent hitherto unknown variation in the skeletal manifestation of this disease or co-infection with tuberculosis or brucellosis. Additionally, spondylolysis was observed in one individual with skeletal lesions suggestive of infectious disease. CONCLUSIONS: This study sheds light on the pathological landscape in Iberia during a time of great social, demographic, and environmental change. Genetic evidence challenges the hypothesis that malaria was absent from early medieval Iberia and demonstrates the value of combining osteological and archaeogenetic methods. Additionally, all of the preferred infectious diagnoses for the individuals included in this study (malaria, tuberculosis, and brucellosis) could have contributed to the febrile cases described in historical sources from this time.


Subject(s)
Malaria , Humans , Male , History, Medieval , Spain , Female , Adult , Middle Aged , Malaria/history , Young Adult , Adolescent , Child , DNA, Ancient/analysis , Child, Preschool , Infant , Bone and Bones/pathology , Bone and Bones/microbiology , Communicable Diseases/history , Paleopathology , Brucellosis/history , Tuberculosis/history
15.
Curr Osteoporos Rep ; 22(4): 396-415, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38980532

ABSTRACT

PURPOSE OF REVIEW: Quantification of the morphology of osteocyte lacunae has become a powerful tool to investigate bone metabolism, pathologies and aging. This review will provide a brief overview of 2D and 3D imaging methods for the determination of lacunar shape, orientation, density, and volume. Deviations between 2D-based and 3D-based lacunar volume estimations are often not sufficiently addressed and may give rise to contradictory findings. Thus, the systematic error arising from 2D-based estimations of lacunar volume will be discussed, and an alternative calculation proposed. Further, standardized morphological parameters and best practices for sampling and segmentation are suggested. RECENT FINDINGS: We quantified the errors in reported estimation methods of lacunar volume based on 2D cross-sections, which increase with variations in lacunar orientation and histological cutting plane. The estimations of lacunar volume based on common practice in 2D imaging methods resulted in an underestimation of lacunar volume of up to 85% compared to actual lacunar volume in an artificial dataset. For a representative estimation of lacunar size and morphology based on 2D images, at least 400 lacunae should be assessed per sample.


Subject(s)
Imaging, Three-Dimensional , Osteocytes , Humans , Osteocytes/cytology , Imaging, Three-Dimensional/methods , Bone Density , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Image Processing, Computer-Assisted/methods
16.
Bone ; 187: 117190, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38960297

ABSTRACT

This study investigates the biomechanics of type 2 diabetic bone fragility through a multiscale experimental strategy that considers structural, mechanical, and compositional components of ex vivo human trabecular and cortical bone. Human tissue samples were obtained from the femoral heads of patients undergoing total hip replacement. Mechanical testing was carried out on isolated trabecular cores using monotonic and cyclic compression loading and nanoindentation experiments, with bone microdamage analysed using micro-computed tomography (CT) imaging. Bone composition was evaluated using Raman spectroscopy, high-performance liquid chromatography, and fluorometric spectroscopy. It was found that human type 2 diabetic bone had altered mechanical, compositional, and morphological properties compared to non-type 2 diabetic bone. High-resolution micro-CT imaging showed that cores taken from the central trabecular region of the femoral head had higher bone mineral density (BMD), bone volume, trabecular thickness, and reduced trabecular separation. Type 2 diabetic bone also had enhanced macro-mechanical compressive properties under mechanical loading compared to non-diabetic controls, with significantly higher apparent modulus, yield stress, and pre-yield toughness evident, even when properties were normalised against the bone volume. Using nanoindentation, there were no significant differences in the tissue-level mechanical properties of cortical or trabecular bone in type 2 diabetic samples compared to controls. Through compositional analysis, higher levels of furosine were found in type 2 diabetic trabecular bone, and an increase in both furosine and carboxymethyl-lysine (an advanced glycation end-product) was found in cortical bone. Raman spectroscopy showed that type 2 diabetic bone had a higher mineral-to-matrix ratio, carbonate substitution, and reduced crystallinity compared to the controls. Together, this study shows that type 2 diabetes leads to distinct changes in both organic and mineral phases of the bone tissue matrix, but these changes did not coincide with any reduction in the micro- or macro-mechanical properties of the tissue under monotonic or cyclic loading.


Subject(s)
Diabetes Mellitus, Type 2 , X-Ray Microtomography , Humans , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Biomechanical Phenomena , Aged , Female , Bone and Bones/pathology , Bone and Bones/physiopathology , Bone and Bones/diagnostic imaging , Male , Spectrum Analysis, Raman , Bone Density/physiology , Cancellous Bone/pathology , Cancellous Bone/diagnostic imaging , Cancellous Bone/physiopathology , Middle Aged , Stress, Mechanical
17.
Bone ; 187: 117198, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39002837

ABSTRACT

Early B cell factor 1 (EBF1) is a transcription factor expressed by multiple lineages of stromal cells within the bone marrow. While cultures of Ebf1-deficient cells have been demonstrated to have impaired differentiation into either the osteoblast or adipogenic lineage in vitro by several groups, in vivo there has been a nominal consequence of the loss of EBF1 on skeletal development. In this study we used Prx-cre driven deletion of Ebf1 to eliminate EBF1 from the entire mesenchymal lineage of the skeleton and resolve this discrepancy. We report here that EBF1 is expressed primarily in the Mesenchymal Stem and Progenitor Cell (MSPC)-Adipo, MSPC-Osteo, and the Early Mesenchymal Progenitors, and that loss of EBF1 has a plethora of consequences to maintenance of the skeleton throughout adulthood. Stroma from the Prx-cre;Ebf1fl/fl bones had impaired osteogenic differentiation, an age-dependent loss of CFU-F, and elevated senescence accompanying Ebf1-deletion. New bone formation was reduced after 3 months, and resulted in a quiescent bone environment with fewer osteoblasts and an accompanied reduction in osteoclast-mediated remodeling. Consequently, bones were less ductile at a younger age, and deletion of EBF1 dramatically impaired fracture repair. Disruption of EBF1 in perivascular populations also rearranged the vascular network within these bones and disrupted cytokine signaling from key hematopoietic niches resulting in anemia, reductions in B cells, and myeloid skewing of marrow hematopoietic lineages. Mechanistically we observed disrupted BMP signaling within Ebf1-deficient progenitors with reduced SMAD1-phosphorylation, and elevated secretion of the soluble BMP-inhibitor Gremlin from the MSPC-Adipo cells. Ebf1-deficient progenitors also exhibited posttranslational suppression of glucocorticoid receptor expression. Together, these results suggest that EBF1 signaling is required for mesenchymal progenitor mobilization to maintain the adult skeleton, and that the primary action of EBF1 in the early mesenchymal lineage is to promote proliferation, and differentiation of these perivascular cells to sustain a healthy tissue.


Subject(s)
Cell Differentiation , Homeostasis , Osteogenesis , Trans-Activators , Animals , Trans-Activators/metabolism , Trans-Activators/genetics , Osteogenesis/genetics , Bone and Bones/metabolism , Bone and Bones/pathology , Mice , Mesenchymal Stem Cells/metabolism , Gene Deletion , Stromal Cells/metabolism , Bone Marrow/metabolism , Osteoblasts/metabolism , Mice, Inbred C57BL , Cellular Senescence/physiology , Aging/metabolism , Cellular Microenvironment
18.
Bone ; 187: 117206, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39029608

ABSTRACT

Children with type 1 diabetes (T1D) experience an increased risk of fracture, which may be related to altered bone development. We aimed to assess differences in bone, muscle and physical activity (PA), and explore if better muscle and PA measures would mitigate bone differences between children and adolescents with T1D and typically developing peers (TDP). We matched 56 children and adolescents with T1D (mean age 11.9 yrs) and 56 TDP (11.5 yrs) by sex and maturity from 171 participants with T1D and 66 TDP (6-17 yrs). We assessed the distal radius and tibia with high-resolution peripheral quantitative computed tomography (HR-pQCT), and the radius and tibia shaft bone and muscle with pQCT. We also measured muscle function from force-related measures in neuromuscular performance tests (push-up, grip test, countermovement and long jump). We compared PA based on questionnaire scores and accelerometers between groups. Bone, muscle, and neuromuscular performance measures were compared using MANOVA. We used mediation to explore the role of PA and muscle in bone differences. Children and adolescents with T1D had 6-10 % lower trabecular density, bone volume fraction, thickness and number at both distal radius and tibia, and 11 % higher trabecular separation at the distal radius than TDP. They also had 3-16 % higher cortical and tissue mineral density, and cortical thickness at the distal radius, 5-7 % higher cortical density and 1-3 % higher muscle density at both shaft sites compared to TDP. PA mediated the between-group difference in trabecular number (indirect effect -0.04) at the distal radius. Children and adolescents with T1D had lower trabecular bone density and deficits in trabecular micro-architecture, but higher cortical bone density and thickness at the radius and tibia compared to TDP. They engaged in less PA but had comparable muscle measures to those of TDP. PA participation may assist in mitigating deficit in trabecular number observed in children and adolescents with T1D.


Subject(s)
Bone Density , Bone and Bones , Diabetes Mellitus, Type 1 , Exercise , Humans , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/diagnostic imaging , Adolescent , Child , Male , Female , Exercise/physiology , Bone and Bones/physiopathology , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Bone Density/physiology , Muscle, Skeletal/physiopathology , Muscle, Skeletal/diagnostic imaging , Tomography, X-Ray Computed , Muscles/physiopathology , Muscles/pathology , Radius/diagnostic imaging , Radius/physiopathology , Radius/pathology
19.
J Theor Biol ; 593: 111894, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38992463

ABSTRACT

In this paper, we explore the effects of biological (pathological) and mechanical damage on bone tissue within a benchmark model. Using the Finite Element Methodology, we analyze and numerically test the model's components, capabilities, and performance under physiologically and pathologically relevant conditions. Our findings demonstrate the model's effectiveness in simulating bone remodeling processes and self-repair mechanisms for micro-damage induced by biological internal conditions and mechanical external ones within bone tissue. This article is the second part of a series, where the first part presented the mathematical model and the biological and physical significance of the terms used in a simplified benchmark model. It explored the bone remodeling model's application, implementation, and results under physiological conditions.


Subject(s)
Bone Remodeling , Models, Biological , Bone Remodeling/physiology , Humans , Biomechanical Phenomena , Finite Element Analysis , Bone and Bones/physiology , Bone and Bones/pathology , Animals , Stress, Mechanical , Computer Simulation
20.
Calcif Tissue Int ; 115(3): 298-314, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39012489

ABSTRACT

Obesity and type 2 diabetes (T2D) are risk factors for fragility fractures. It is unknown whether this elevated risk is due to a diet favoring obesity or the diabetes that often occurs with obesity. Therefore, we hypothesized that the fracture resistance of bone is lower in mice fed with a high fat diet (45% kcal; HFD) than in mice that fed on a similar, control diet (10% kcal; LFD), regardless of whether the mice developed overt T2D. Sixteen-week-old, male NON/ShiLtJ mice (resistant to T2D) and age-matched, male NONcNZO10/LtJ (prone to T2D) received a control LFD or HFD for 21 weeks. HFD increased the bodyweight to a greater extent in the ShiLtJ mice compared to the NZO10 mice, while blood glucose levels were significantly higher in NZO10 than in ShiLtJ mice. As such, the glycated hemoglobin A1c (HbA1c) levels exceeded 10% in NZO10 mice, but it remained below 6% in ShiLtJ mice. Diet did not affect HbA1c. HFD lowered trabecular number and bone volume fraction of the distal femur metaphysis (micro-computed tomography or µCT) in both strains. For the femur mid-diaphysis, HFD significantly reduced the yield moment (mechanical testing by three-point bending) in both strains but did not affect cross-sectional bone area, cortical thickness, nor cortical tissue mineral density (µCT). Furthermore, the effect of diet on yield moment was independent of the structural resistance of the femur mid-diaphysis suggesting a negative effect of HFD on characteristics of the bone matrix. However, neither Raman spectroscopy nor assays of advanced glycation end-products identified how HFD affected the matrix. HFD also lowered the resistance of cortical bone to crack growth in only the diabetic NZO10 mice (fracture toughness testing of other femur), while HFD reduced the ultimate force of the L6 vertebra in both strains (compression testing). In conclusion, the HFD-related decrease in bone strength can occur in mice resistant and prone to diabetes indicating that a diet high in fat deleteriously affects bone without necessarily causing hyperglycemia.


Subject(s)
Bone Density , Diabetes Mellitus, Type 2 , Diet, High-Fat , Obesity , Animals , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Obesity/metabolism , Male , Mice , Bone Density/physiology , Fractures, Bone/etiology , Bone and Bones/metabolism , Bone and Bones/pathology
SELECTION OF CITATIONS
SEARCH DETAIL