Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 14.012
1.
Front Immunol ; 15: 1396122, 2024.
Article En | MEDLINE | ID: mdl-38817601

As the world population ages, osteoporosis, the most common disease of bone metabolism, affects more than 200 million people worldwide. The etiology is an imbalance in bone remodeling process resulting in more significant bone resorption than bone remodeling. With the advent of the osteoimmunology field, the immune system's role in skeletal pathologies is gradually being discovered. The cytokine interferon-gamma (IFN-γ), a member of the interferon family, is an important factor in the etiology and treatment of osteoporosis because it mediates bone remodeling. This review starts with bone remodeling process and includes the cellular and key signaling pathways of bone remodeling. The effects of IFN-γ on osteoblasts, osteoclasts, and bone mass are discussed separately, while the overall effects of IFN-γ on primary and secondary osteoporosis are summarized. The net effect of IFN-γ on bone appears to be highly dependent on the environment, dose, concentration, and stage of cellular differentiation. This review focuses on the mechanisms of bone remodeling and bone immunology, with a comprehensive discussion of the relationship between IFN-γ and osteoporosis. Finding the paradoxical balance of IFN-γ in bone immunology and exploring the potential of its clinical application provide new ideas for the clinical treatment of osteoporosis and drug development.


Bone Remodeling , Interferon-gamma , Osteoporosis , Humans , Bone Remodeling/drug effects , Osteoporosis/immunology , Osteoporosis/etiology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Animals , Osteoclasts/immunology , Osteoclasts/metabolism , Osteoblasts/immunology , Osteoblasts/metabolism , Signal Transduction , Bone and Bones/immunology , Bone and Bones/metabolism , Bone and Bones/pathology
2.
Sci Transl Med ; 16(745): eadi8214, 2024 May.
Article En | MEDLINE | ID: mdl-38691622

Mucopolysaccharidosis type I Hurler (MPSIH) is characterized by severe and progressive skeletal dysplasia that is not fully addressed by allogeneic hematopoietic stem cell transplantation (HSCT). Autologous hematopoietic stem progenitor cell-gene therapy (HSPC-GT) provides superior metabolic correction in patients with MPSIH compared with HSCT; however, its ability to affect skeletal manifestations is unknown. Eight patients with MPSIH (mean age at treatment: 1.9 years) received lentiviral-based HSPC-GT in a phase 1/2 clinical trial (NCT03488394). Clinical (growth, measures of kyphosis and genu velgum), functional (motor function, joint range of motion), and radiological [acetabular index (AI), migration percentage (MP) in hip x-rays and MRIs and spine MRI score] parameters of skeletal dysplasia were evaluated at baseline and multiple time points up to 4 years after treatment. Specific skeletal measures were retrospectively compared with an external cohort of HSCT-treated patients. At a median follow-up of 3.78 years after HSPC-GT, all patients treated with HSPC-GT exhibited longitudinal growth within WHO reference ranges and a median height gain greater than that observed in patients treated with HSCT after 3-year follow-up. Patients receiving HSPC-GT experienced complete and earlier normalization of joint mobility compared with patients treated with HSCT. Mean AI and MP showed progressive decreases after HSPC-GT, suggesting a reduction in acetabular dysplasia. Typical spine alterations measured through a spine MRI score stabilized after HSPC-GT. Clinical, functional, and radiological measures suggested an early beneficial effect of HSPC-GT on MPSIH-typical skeletal features. Longer follow-up is needed to draw definitive conclusions on HSPC-GT's impact on MPSIH skeletal dysplasia.


Genetic Therapy , Hematopoietic Stem Cell Transplantation , Mucopolysaccharidosis I , Humans , Mucopolysaccharidosis I/therapy , Mucopolysaccharidosis I/pathology , Mucopolysaccharidosis I/genetics , Male , Female , Child, Preschool , Infant , Treatment Outcome , Hematopoietic Stem Cells/metabolism , Child , Bone and Bones/pathology , Magnetic Resonance Imaging
3.
Curr Opin Hematol ; 31(4): 163-167, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38723188

PURPOSE OF REVIEW: The repair of bone after injury requires the participation of many different immune cell populations, which are derived from the hematopoietic lineage. The field of osteoimmunology, or the study of the interactions between bone and the immune system, is a growing field with emerging impact on both the basic science and clinical aspects of fracture healing. RECENT FINDINGS: Despite previous focus on the innate immune system in fracture healing, recent studies have revealed an important role for the adaptive immune system in bone repair. The composition of adaptive and innate immune cell populations present at the fracture site is significantly altered during aging and diet-induced obesity, which may contribute to delayed healing. Recent data also suggest a complicated relationship between fracture repair and systemic inflammation, raising the possibility that immune populations from distant sites such as the gut can impact the bone repair process. SUMMARY: These findings have important implications for the treatment of fracture patients with antibiotics or anti-inflammatory drugs. Furthermore, the effects of systemic inflammation on fracture repair in the contexts of aging or obesity should be carefully interpreted, as they may not be uniformly detrimental.


Fracture Healing , Hematopoiesis , Humans , Animals , Inflammation/metabolism , Inflammation/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Fractures, Bone/therapy , Fractures, Bone/metabolism , Fractures, Bone/pathology , Obesity/metabolism , Obesity/pathology
4.
Int J Mol Sci ; 25(9)2024 Apr 28.
Article En | MEDLINE | ID: mdl-38732046

Obesity, type 2 diabetes mellitus (T2DM) and osteoporosis are serious diseases with an ever-increasing incidence that quite often coexist, especially in the elderly. Individuals with obesity and T2DM have impaired bone quality and an elevated risk of fragility fractures, despite higher and/or unchanged bone mineral density (BMD). The effect of obesity on fracture risk is site-specific, with reduced risk for several fractures (e.g., hip, pelvis, and wrist) and increased risk for others (e.g., humerus, ankle, upper leg, elbow, vertebrae, and rib). Patients with T2DM have a greater risk of hip, upper leg, foot, humerus, and total fractures. A chronic pro-inflammatory state, increased risk of falls, secondary complications, and pharmacotherapy can contribute to the pathophysiology of aforementioned fractures. Bisphosphonates and denosumab significantly reduced the risk of vertebral fractures in patients with both obesity and T2DM. Teriparatide significantly lowered non-vertebral fracture risk in T2DM subjects. It is important to recognize elevated fracture risk and osteoporosis in obese and T2DM patients, as they are currently considered low risk and tend to be underdiagnosed and undertreated. The implementation of better diagnostic tools, including trabecular bone score, lumbar spine BMD/body mass index (BMI) ratio, and microRNAs to predict bone fragility, could improve fracture prevention in this patient group.


Bone Density , Diabetes Mellitus, Type 2 , Obesity , Osteoporosis , Humans , Diabetes Mellitus, Type 2/complications , Osteoporosis/etiology , Osteoporosis/drug therapy , Obesity/complications , Fractures, Bone/etiology , Bone and Bones/metabolism , Bone and Bones/pathology
5.
Cell Rep Med ; 5(5): 101574, 2024 May 21.
Article En | MEDLINE | ID: mdl-38776873

The existing suite of therapies for bone diseases largely act to prevent further bone loss but fail to stimulate healthy bone formation and repair. We describe an endogenous osteopeptide (PEPITEM) with anabolic osteogenic activity, regulating bone remodeling in health and disease. PEPITEM acts directly on osteoblasts through NCAM-1 signaling to promote their maturation and formation of new bone, leading to enhanced trabecular bone growth and strength. Simultaneously, PEPITEM stimulates an inhibitory paracrine loop: promoting osteoblast release of the decoy receptor osteoprotegerin, which sequesters RANKL, thereby limiting osteoclast activity and bone resorption. In disease models, PEPITEM therapy halts osteoporosis-induced bone loss and arthritis-induced bone damage in mice and stimulates new bone formation in osteoblasts derived from patient samples. Thus, PEPITEM offers an alternative therapeutic option in the management of diseases with excessive bone loss, promoting an endogenous anabolic pathway to induce bone remodeling and redress the imbalance in bone turnover.


Bone Resorption , Osteoblasts , Osteogenesis , Animals , Humans , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Mice , Bone Resorption/pathology , Bone Resorption/metabolism , Anabolic Agents/pharmacology , Anabolic Agents/therapeutic use , Bone Remodeling/drug effects , Osteoporosis/pathology , Osteoporosis/metabolism , Osteoporosis/drug therapy , RANK Ligand/metabolism , Osteoclasts/metabolism , Osteoclasts/drug effects , Bone Development/drug effects , Osteoprotegerin/metabolism , Female , Signal Transduction/drug effects , Peptides/pharmacology , Male , Mice, Inbred C57BL , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology
7.
Sci Rep ; 14(1): 8030, 2024 04 05.
Article En | MEDLINE | ID: mdl-38580668

Apical periodontitis (AP) is a condition characterized by inflammatory and infectious components in the tooth canal. AP affects periradicular tissues and has systemic repercussions. Physical exercise is a structured activity that requires cardiorespiratory function, and can modulate the inflammatory profile in pathological conditions. As a result, this study aimed to determine the effects of aerobic physical training (PT) on the alveolar bone with and without AP, and its systemic inflammatory repercussions. AP was induced in the mandibular first molars, and PT was performed on a treadmill for five consecutive days over four weeks, with progressive increases in speed and activity time. Blood samples were collected to determine serum cytokine levels using immunoassays, and alveolar bone samples were collected for histopathological evaluation, lesion volume and microarchitecture assessment using computed microtomography. Animals with AP had increased pro-inflammatory cytokines levels compared to those without AP; however, these levels were attenuated or restored by PT. Compared to the AP group, the AP + PT group had a smaller lesion volume and greater preservation of the bone trabeculae in the remaining alveolar bone surrounding the lesion. In overall, PT minimized the severity of AP proving to be a valid strategy for individuals undergoing endodontic treatment.


Cytokines , Periapical Periodontitis , Humans , Animals , Periapical Periodontitis/therapy , Periapical Periodontitis/pathology , Exercise , Bone and Bones/pathology
8.
J Nanobiotechnology ; 22(1): 153, 2024 Apr 05.
Article En | MEDLINE | ID: mdl-38580995

BACKGROUND: Osteoporosis is characterized by an imbalance in bone homeostasis, resulting in the excessive dissolution of bone minerals due to the acidified microenvironment mediated by overactive osteoclasts. Oroxylin A (ORO), a natural flavonoid, has shown potential in reversing osteoporosis by inhibiting osteoclast-mediated bone resorption. The limited water solubility and lack of targeting specificity hinder the effective accumulation of Oroxylin A within the pathological environment of osteoporosis. RESULTS: Osteoclasts' microenvironment-responsive nanoparticles are prepared by incorporating Oroxylin A with amorphous calcium carbonate (ACC) and coated with glutamic acid hexapeptide-modified phospholipids, aiming at reinforcing the drug delivery efficiency as well as therapeutic effect. The obtained smart nanoparticles, coined as OAPLG, could instantly neutralize acid and release Oroxylin A in the extracellular microenvironment of osteoclasts. The combination of Oroxylin A and ACC synergistically inhibits osteoclast formation and activity, leading to a significant reversal of systemic bone loss in the ovariectomized mice model. CONCLUSION: The work highlights an intelligent nanoplatform based on ACC for spatiotemporally controlled release of lipophilic drugs, and illustrates prominent therapeutic promise against osteoporosis.


Bone Resorption , Osteoporosis , Mice , Animals , Osteoclasts , Nanomedicine , Osteoporosis/drug therapy , Bone Resorption/drug therapy , Bone and Bones/pathology , Cell Differentiation
9.
J Neuroinflammation ; 21(1): 111, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38685040

BACKGROUND: It is well known that high-fat diet (HFD)-induced metabolic syndrome plays a crucial role in cognitive decline and brain-blood barrier (BBB) breakdown. However, whether the bone-brain axis participates in this pathological process remains unknown. Here, we report that platelet-derived growth factor-BB (PDGF-BB) secretion by preosteoclasts in the bone accelerates neuroinflammation. The expression of alkaline phosphatase (ALPL), a nonspecific transcytosis marker, was upregulated during HFD challenge. MAIN BODY: Preosteoclast-specific Pdgfb transgenic mice with high PDGF-BB concentrations in the circulation recapitulated the HFD-induced neuroinflammation and transcytosis shift. Preosteoclast-specific Pdgfb knockout mice were partially rescued from hippocampal neuroinflammation and transcytosis shifts in HFD-challenged mice. HFD-induced PDGF-BB elevation aggravated microglia-associated neuroinflammation and interleukin-1ß (IL-1ß) secretion, which increased ALPL expression and transcytosis shift through enhancing protein 1 (SP1) translocation in endothelial cells. CONCLUSION: Our findings confirm the role of bone-secreted PDGF-BB in neuroinflammation and the transcytosis shift in the hippocampal region during HFD challenge and identify a novel mechanism of microglia-endothelial crosstalk in HFD-induced metabolic syndrome.


Becaplermin , Diet, High-Fat , Endothelial Cells , Hippocampus , Metabolic Syndrome , Microglia , Transcytosis , Animals , Mice , Becaplermin/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Transcytosis/physiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Microglia/metabolism , Microglia/pathology , Diet, High-Fat/adverse effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice, Transgenic , Mice, Inbred C57BL , Mice, Knockout , Male , Bone and Bones/metabolism , Bone and Bones/pathology
10.
BMJ Case Rep ; 17(4)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38604742

This is a case of primary hyperparathyroidism in a female teenager with multiple fractures and severe bone manifestations. The histopathology revealed atypical parathyroid adenoma, an exceedingly rare form of hyperparathyroidism; its main differential diagnosis is parathyroid carcinoma, as it shares both clinical and histological characteristics with it, in addition to its still uncertain malignant potential.


Hyperparathyroidism , Parathyroid Neoplasms , Humans , Adolescent , Female , Parathyroid Neoplasms/diagnosis , Parathyroid Neoplasms/diagnostic imaging , Parathyroid Glands/diagnostic imaging , Parathyroid Glands/pathology , Bone and Bones/pathology
11.
Peptides ; 177: 171228, 2024 Jul.
Article En | MEDLINE | ID: mdl-38657908

Diabetes mellitus and obesity are rapidly growing worldwide. Aside from metabolic disturbances, these two disorders also affect bone with a higher prevalence of bone fractures. In the last decade, a growing body of evidence suggested that several gut hormones, including ghrelin, gastrin, glucose-dependent insulinotropic polypeptide (GIP), glucagon, and glucagon-like peptide-1 and 2 (GLP-1 and GLP-2, respectively) may affect bone physiology. Several gut hormone analogues have been developed for the treatment of type 2 diabetes and obesity, and could represent a new alternative in the therapeutic arsenal against bone fragility. In the present review, a summary of the physiological roles of these gut hormones and their analogues is presented at the cellular level but also in several preclinical models of bone fragility disorders including type 2 diabetes mellitus, especially on bone mineral density, microarchitecture and bone material properties. The present review also summarizes the impact of GLP-1 receptor agonists approved for the treatment of type 2 diabetes mellitus and the more recent dual or triple analogue on bone physiology and strength.


Diabetes Mellitus, Type 2 , Gastrointestinal Hormones , Obesity , Humans , Obesity/drug therapy , Obesity/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/complications , Animals , Gastrointestinal Hormones/metabolism , Bone Density/drug effects , Bone and Bones/metabolism , Bone and Bones/drug effects , Bone and Bones/pathology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/analogs & derivatives , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/therapeutic use
12.
Biomed Pharmacother ; 175: 116571, 2024 Jun.
Article En | MEDLINE | ID: mdl-38677248

Diabetes can lead to a disorder of bone-fat balance, a significant cause of osteoporosis due to changes in environmental factors. Baicalin (Bai), an active ingredient of Scutellaria baicalensis, has been confirmed to possess antioxidant, hypoglycemic, and anti-osteoporotic effects. However, a comprehensive understanding of Bai's influence on diabetic osteoporosis (DOP), including its effects and underlying mechanisms, remains elusive. This study investigated Bai's impact on the bone-fat equilibrium in rats with DOP. The results indicated that Bai alleviated bone damage in DOP by promoting osteogenesis and inhibiting adipogenesis. Concurrently, through bioinformatics analysis, it was suggested that Bai's mechanism of action might involve the P38-MAPK pathway. In vitro, Bai was found to enhance the development of bone marrow mesenchymal stem cells (BMSCs) towards osteogenic lineages while suppressing their differentiation towards adipogenic lineages. It was discovered that Bai's promotion of BMSC osteogenic differentiation depends on the P38-MAPK pathway. Additionally, the synergistic effect mediated by Bai and P38-MAPK inhibitor suppressed BMSC adipogenic differentiation. Our research indicates that the P38-MAPK pathway play a role in Bai's effects on the osteogenic-adipogenic differentiation of BMSCs, showcasing the potential for DOP treatment. This study highlights Bai's ability to regulate the equilibrium between bone and fat, presenting a novel approach to adressing DOP.


Adipogenesis , Cell Differentiation , Flavonoids , Mesenchymal Stem Cells , Osteogenesis , Osteoporosis , Rats, Sprague-Dawley , p38 Mitogen-Activated Protein Kinases , Animals , Flavonoids/pharmacology , Flavonoids/therapeutic use , p38 Mitogen-Activated Protein Kinases/metabolism , Osteoporosis/drug therapy , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Adipogenesis/drug effects , Male , Rats , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , MAP Kinase Signaling System/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Cells, Cultured
13.
Calcif Tissue Int ; 114(6): 638-649, 2024 Jun.
Article En | MEDLINE | ID: mdl-38642089

Type 2 diabetes (T2D) increases fracture incidence and fracture-related mortality rates (KK.Cg-Ay/J. The Jackson Laboratory; Available from: https://www.jax.org/strain/002468 ). While numerous mouse models for T2D exist, few effectively stimulate persistent hyperglycemia in both sexes, and even fewer are suitable for bone studies. Commonly used models like db/db and ob/ob have altered leptin pathways, confounding bone-related findings since leptin regulates bone properties (Fajardo et al. in Journal of Bone and Mineral Research 29(5): 1025-1040, 2014). The Yellow Kuo Kondo (KK/Ay) mouse, a polygenic mutation model of T2D, is able to produce a consistent diabetic state in both sexes and addresses the lack of a suitable model of T2D for bone studies. The diabetic state of KK/Ay stems from a mutation in the agouti gene, responsible for coat color in mice. This mutation induces ectopic gene expression across various tissue types, resulting in diabetic mice with yellow fur coats (Moussa and Claycombe in Obesity Research 7(5): 506-514, 1999). Male and female KK/Ay mice exhibited persistent hyperglycemia, defining them as diabetic with blood glucose (BG) levels consistently exceeding 300 mg/dL. Notably, male control mice in this study were also diabetic, presenting a significant limitation. Nevertheless, male and female KK/Ay mice showed significantly elevated BG levels, HbA1c, and serum insulin concentration when compared to the non-diabetic female control mice. Early stages of T2D are characterized by hyperglycemia and hyperinsulinemia resulting from cellular insulin resistance, whereas later stages may feature hypoinsulinemia due to ß-cell apoptosis (Banday et al. Avicenna Journal of Medicine 10(04): 174-188, 2020 and Klein et al. Cell Metabolism 34(1): 11-20, 2022). The observed hyperglycemia, hyperinsulinemia, and the absence of differences in ß-cell mass suggest that KK/Ay mice in this study are modeling the earlier stages of T2D. While compromised bone microarchitecture was observed in this study, older KK/Ay mice, representing more advanced stages of T2D, might exhibit more pronounced skeletal manifestations. Compared to the control group, the femora of KK/Ay mice had higher cortical area and cortical thickness, and improved trabecular properties which would typically be indicative of greater bone strength. However, KK/Ay mice displayed lower cortical tissue mineral density in both sexes and increased cortical porosity in females. Fracture instability toughness of the femora was lower in KK/Ay mice overall compared to controls. These findings indicate that decreased mechanical integrity noted in the femora of KK/Ay mice was likely due to overall bone quality being compromised.


Diabetes Mellitus, Type 2 , Disease Models, Animal , Mutation , Obesity , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Mice , Female , Male , Mutation/genetics , Obesity/genetics , Obesity/metabolism , Obesity/complications , Bone and Bones/metabolism , Bone and Bones/pathology , Mice, Obese , Bone Density/genetics
14.
Nano Lett ; 24(17): 5154-5164, 2024 May 01.
Article En | MEDLINE | ID: mdl-38602357

Developing novel strategies for defeating osteoporosis has become a world-wide challenge with the aging of the population. In this work, novel supramolecular nanoagonists (NAs), constructed from alkaloids and phenolic acids, emerge as a carrier-free nanotherapy for efficacious osteoporosis treatment. These precision nanoagonists are formed through the self-assembly of berberine (BER) and chlorogenic acid (CGA), utilizing noncovalent electrostatic, π-π, and hydrophobic interactions. This assembly results in a 100% drug loading capacity and stable nanostructure. Furthermore, the resulting weights and proportions of CGA and BER within the NAs are meticulously controlled with strong consistency when the CGA/BER assembly feed ratio is altered from 1:1 to 1:4. As anticipated, our NAs themselves could passively target osteoporotic bone tissues following prolonged blood circulation, modulate Wnt signaling, regulate osteogenic differentiation, and ameliorate bone loss in ovariectomy-induced osteoporotic mice. We hope this work will open a new strategy to design efficient herbal-derived Wnt NAs for dealing with intractable osteoporosis.


Berberine , Chlorogenic Acid , Osteoporosis , Osteoporosis/drug therapy , Animals , Mice , Berberine/pharmacology , Berberine/therapeutic use , Berberine/chemistry , Berberine/administration & dosage , Berberine/pharmacokinetics , Chlorogenic Acid/chemistry , Chlorogenic Acid/pharmacology , Chlorogenic Acid/therapeutic use , Chlorogenic Acid/administration & dosage , Female , Humans , Osteogenesis/drug effects , Bone and Bones/drug effects , Bone and Bones/pathology , Nanostructures/chemistry , Nanostructures/therapeutic use
15.
Front Med ; 18(2): 237-257, 2024 Apr.
Article En | MEDLINE | ID: mdl-38619691

Osteoarthritis (OA) is a degenerative bone disease associated with aging. The rising global aging population has led to a surge in OA cases, thereby imposing a significant socioeconomic burden. Researchers have been keenly investigating the mechanisms underlying OA. Previous studies have suggested that the disease starts with synovial inflammation and hyperplasia, advancing toward cartilage degradation. Ultimately, subchondral-bone collapse, sclerosis, and osteophyte formation occur. This progression is deemed as "top to bottom." However, recent research is challenging this perspective by indicating that initial changes occur in subchondral bone, precipitating cartilage breakdown. In this review, we elucidate the epidemiology of OA and present an in-depth overview of the subchondral bone's physiological state, functions, and the varied pathological shifts during OA progression. We also introduce the role of multifunctional signal pathways (including osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL)/receptor activator of nuclear factor-kappa B (RANK), and chemokine (CXC motif) ligand 12 (CXCL12)/CXC motif chemokine receptor 4 (CXCR4)) in the pathology of subchondral bone and their role in the "bottom-up" progression of OA. Using vivid pattern maps and clinical images, this review highlights the crucial role of subchondral bone in driving OA progression, illuminating its interplay with the condition.


Disease Progression , Osteoarthritis , Osteoprotegerin , Humans , Osteoarthritis/pathology , Osteoarthritis/physiopathology , Osteoarthritis/etiology , Osteoarthritis/metabolism , Osteoprotegerin/metabolism , Bone and Bones/pathology , Bone and Bones/metabolism , RANK Ligand/metabolism , Signal Transduction , Cartilage, Articular/pathology , Chemokine CXCL12/metabolism , Receptors, CXCR4/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism
16.
Bone ; 184: 117106, 2024 Jul.
Article En | MEDLINE | ID: mdl-38641232

Osteogenesis imperfecta (OI) increases fracture risk due to changes in bone quantity and quality caused by mutations in collagen and its processing proteins. Current therapeutics improve bone quantity, but do not treat the underlying quality deficiencies. Male and female G610C+/- mice, a murine model of OI, were treated with a combination of raloxifene and in vivo axial tibial compressive loading starting at 10 weeks of age and continuing for 6 weeks to improve bone quantity and quality. Bone geometry and mechanical properties were measured to determine whole bone and tissue-level material properties. A colocalized Raman/nanoindentation system was used to measure chemical composition and nanomechanical properties in newly formed bone compared to old bone to determine if bone formed during the treatment regimen differed in quality compared to bone formed prior to treatment. Lastly, lacunar geometry and osteocyte apoptosis were assessed. OI mice were able to build bone in response to the loading, but this response was less robust than in control mice. Raloxifene improved some bone material properties in female but not male OI mice. Raloxifene did not alter nanomechanical properties, but loading did. Lacunar geometry was largely unchanged with raloxifene and loading. However, osteocyte apoptosis was increased with loading in raloxifene treated female mice. Overall, combination treatment with raloxifene and loading resulted in positive but subtle changes to bone quality.


Disease Models, Animal , Osteogenesis Imperfecta , Raloxifene Hydrochloride , Animals , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Osteogenesis Imperfecta/drug therapy , Osteogenesis Imperfecta/pathology , Female , Male , Mice , Bone and Bones/drug effects , Bone and Bones/pathology , Biomechanical Phenomena/drug effects , Apoptosis/drug effects , Anabolic Agents/pharmacology , Anabolic Agents/therapeutic use , Weight-Bearing , Osteocytes/drug effects , Osteocytes/metabolism , Osteocytes/pathology
20.
Diabetes Metab ; 50(3): 101525, 2024 May.
Article En | MEDLINE | ID: mdl-38442769

OBJECTIVE: We aimed to assess the feasibility and diagnostic performance of ultrasound-guided bone biopsies at the bedside of diabetic patients admitted for suspected foot osteitis not requiring surgery. RESEARCH DESIGN AND METHODS: In this retrospective monocentric study, we compared the performance of ultrasound-guided (n = 29 consecutive patients, Dec.2020-Oct.2022) versus surgical (n = 24 consecutive patients, Jan.2018-Nov.2020) bone biopsies at confirming or ruling out diabetic foot osteitis (primary outcome). RESULTS: Patient characteristics were similar in the two intervention groups, including arteritis prevalence (62.3 %), SINBAD score, and wound location (phalanges 36 %, metatarsus 43 %, and calcaneus 21 %). However, the ultrasound-guided group was older (67 ± 11 versus 60 ± 13 years respectively, P = 0.047) and had more type 2 diabetes (97 % versus 75 %, P = 0.038). Diagnostic performance (i.e., capacity to confirm or rule out suspected osteitis) was similar for ultrasound-guided (28/29 cases: 25 confirmations, 3 invalidations) and surgical (24 confirmations/24) biopsies, P = 0.358. No biopsy-related side effect or complication was observed for either intervention, even for patients on antiaggregation and/or anticoagulation therapy. The mean (± standard deviation) time necessary to perform the biopsy was shorter in the ultrasound-guided group (2.6 ± 3.0 versus 7.2 ± 5.8 days, respectively, P < 0.001) and wound evolution at three months was more favorable (83.3 versus 41.2 %, P = 0.005) (94.4 % versus 66.7 %, respectively, patients with new surgical procedure within six months excluded; P = 0.055). Even though not statistically significant, healing rates in terms of wound and osteitis at six months were also better in the ultrasound-guided group (wound: 40.9 % versus 36.8 %; P = 0.790, and osteitis: 81.8 vs 55.6 % P = 0.071). CONCLUSION: In diabetic patients with suspected foot osteitis not requiring surgery, bedside ultrasound-guided bone biopsies may constitute a promising alternative to surgical biopsies. This intervention provided excellent tolerance and microbiological documentation, short lead-times, and more favorable wound prognosis.


Diabetic Foot , Image-Guided Biopsy , Humans , Diabetic Foot/diagnostic imaging , Male , Female , Middle Aged , Aged , Retrospective Studies , Image-Guided Biopsy/methods , Osteitis , Ultrasonography, Interventional/methods , Bone and Bones/pathology , Bone and Bones/diagnostic imaging
...