Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.249
Filter
1.
ACS Biomater Sci Eng ; 10(5): 2659-2679, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38697939

ABSTRACT

Connective tissue attaches to bone across an insertion with spatial gradients in components, microstructure, and biomechanics. Due to regional stress concentrations between two mechanically dissimilar materials, the insertion is vulnerable to mechanical damage during joint movements and difficult to repair completely, which remains a significant clinical challenge. Despite interface stress concentrations, the native insertion physiologically functions as the effective load-transfer device between soft tissue and bone. This review summarizes tendon, ligament, and meniscus insertions cross-sectionally, which is novel in this field. Herein, the similarities and differences between the three kinds of insertions in terms of components, microstructure, and biomechanics are compared in great detail. This review begins with describing the basic components existing in the four zones (original soft tissue, uncalcified fibrocartilage, calcified fibrocartilage, and bone) of each kind of insertion, respectively. It then discusses the microstructure constructed from collagen, glycosaminoglycans (GAGs), minerals and others, which provides key support for the biomechanical properties and affects its physiological functions. Finally, the review continues by describing variations in mechanical properties at the millimeter, micrometer, and nanometer scale, which minimize stress concentrations and control stretch at the insertion. In summary, investigating the contrasts between the three has enlightening significance for future directions of repair strategies of insertion diseases and for bioinspired approaches to effective soft-hard interfaces and other tough and robust materials in medicine and engineering.


Subject(s)
Tendons , Humans , Biomechanical Phenomena/physiology , Tendons/physiology , Tendons/anatomy & histology , Animals , Bone and Bones/physiology , Ligaments/physiology , Fibrocartilage/physiology , Fibrocartilage/chemistry , Fibrocartilage/metabolism , Collagen/chemistry , Collagen/metabolism , Stress, Mechanical
2.
Biofabrication ; 16(3)2024 May 13.
Article in English | MEDLINE | ID: mdl-38688259

ABSTRACT

Bone scaffolds are widely employed for treating various bone disorders, including defects, fractures, and accidents. Gradient bone scaffolds present a promising approach by incorporating gradients in shape, porosity, density, and other properties, mimicking the natural human body structure. This design offers several advantages over traditional scaffolds. A key advantage is the enhanced matching of human tissue properties, facilitating cell adhesion and migration. Furthermore, the gradient structure fosters a smooth transition between scaffold and surrounding tissue, minimizing the risk of inflammation or rejection. Mechanical stability is also improved, providing better support for bone regeneration. Additionally, gradient bone scaffolds can integrate drug delivery systems, enabling controlled release of drugs or growth factors to promote specific cellular activities during the healing process. This comprehensive review examines the design aspects of gradient bone scaffolds, encompassing structure and drug delivery capabilities. By optimizing the scaffold's inherent advantages through gradient design, bone regeneration outcomes can be improved. The insights presented in this article contribute to the academic understanding of gradient bone scaffolds and their applications in bone tissue engineering.


Subject(s)
Bone Diseases , Bone Regeneration , Tissue Engineering , Tissue Scaffolds , Humans , Tissue Scaffolds/chemistry , Bone Regeneration/drug effects , Bone Diseases/therapy , Animals , Bone and Bones/physiology , Drug Delivery Systems
3.
Poult Sci ; 103(5): 103597, 2024 May.
Article in English | MEDLINE | ID: mdl-38471225

ABSTRACT

Laying hens, selectively bred for high egg production, often suffer from bone fragility and fractures, impacting their welfare and causing economic losses. Additionally, gut health and muscle quality are crucial for overall health and productivity. This study aimed to evaluate the effects of ß-Hydroxy-ß-methylbutyrate (HMB) supplementation on performance, bone metabolism, intestinal morphology, and muscle quality in laying hens. Forty-eight Bovans Brown hens were divided into a control group and an HMB-supplemented group (0.02% HMB in diet). The study spanned from the 31st to the 60th wk of age. Assessments included bone mechanical testing, serum hormonal analysis, histological analysis of bone and intestine, and muscle quality analysis. The HMB supplementation led to decreased feed intake without affecting body weight or laying rate in laying hens. It caused an increase in both mean daily and total egg weight, indicating improved feed utilization, without influencing the feed intake to egg weight ratio. Enhanced bone formation markers and altered intestinal morphometric parameters were observed, along with improved trabecular bone structure. However, no changes in measured other bone quality indices, including geometric, densitometric, or mechanical properties were observed. Muscle analysis revealed no significant changes in overall meat quality, except for a decrease in cholesterol content and alterations in the fatty acid profile, notably a reduction in total n-3 polyunsaturated and total polyunsaturated fatty acids (PUFA). In conclusion, although not all effects of HMB supplementation were unequivocally beneficial, the positive changes in performance data and trabecular bone microarchitecture support further research into various doses and durations of supplementation. Such studies are necessary to fully understand and optimize the benefits of HMB for enhancing the health and productivity of laying hens.


Subject(s)
Animal Feed , Chickens , Diet , Dietary Supplements , Intestines , Valerates , Animals , Chickens/physiology , Valerates/administration & dosage , Valerates/pharmacology , Dietary Supplements/analysis , Female , Animal Feed/analysis , Diet/veterinary , Intestines/drug effects , Intestines/physiology , Intestines/anatomy & histology , Bone and Bones/drug effects , Bone and Bones/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Random Allocation , Animal Nutritional Physiological Phenomena/drug effects
4.
J Bone Miner Res ; 39(2): 106-115, 2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38477743

ABSTRACT

Artificial intelligence (AI) chatbots utilizing large language models (LLMs) have recently garnered significant interest due to their ability to generate humanlike responses to user inquiries in an interactive dialog format. While these models are being increasingly utilized to obtain medical information by patients, scientific and medical providers, and trainees to address biomedical questions, their performance may vary from field to field. The opportunities and risks these chatbots pose to the widespread understanding of skeletal health and science are unknown. Here we assess the performance of 3 high-profile LLM chatbots, Chat Generative Pre-Trained Transformer (ChatGPT) 4.0, BingAI, and Bard, to address 30 questions in 3 categories: basic and translational skeletal biology, clinical practitioner management of skeletal disorders, and patient queries to assess the accuracy and quality of the responses. Thirty questions in each of these categories were posed, and responses were independently graded for their degree of accuracy by four reviewers. While each of the chatbots was often able to provide relevant information about skeletal disorders, the quality and relevance of these responses varied widely, and ChatGPT 4.0 had the highest overall median score in each of the categories. Each of these chatbots displayed distinct limitations that included inconsistent, incomplete, or irrelevant responses, inappropriate utilization of lay sources in a professional context, a failure to take patient demographics or clinical context into account when providing recommendations, and an inability to consistently identify areas of uncertainty in the relevant literature. Careful consideration of both the opportunities and risks of current AI chatbots is needed to formulate guidelines for best practices for their use as source of information about skeletal health and biology.


Artificial intelligence chatbots are increasingly used as a source of information in health care and research settings due to their accessibility and ability to summarize complex topics using conversational language. However, it is still unclear whether they can provide accurate information for questions related to the medicine and biology of the skeleton. Here, we tested the performance of three prominent chatbots­ChatGPT, Bard, and BingAI­by tasking them with a series of prompts based on well-established skeletal biology concepts, realistic physician­patient scenarios, and potential patient questions. Despite their similarities in function, differences in the accuracy of responses were observed across the three different chatbot services. While in some contexts, chatbots performed well, and in other cases, strong limitations were observed, including inconsistent consideration of clinical context and patient demographics, occasionally providing incorrect or out-of-date information, and citation of inappropriate sources. With careful consideration of their current weaknesses, artificial intelligence chatbots offer the potential to transform education on skeletal health and science.


Subject(s)
Artificial Intelligence , Bone and Bones , Humans , Bone and Bones/physiology , Bone Diseases/therapy
5.
Acta Biomater ; 179: 164-179, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38513725

ABSTRACT

Failure-resistant designs are particularly crucial for bones subjected to rapid loading, as is the case for the ambush-hunting northern pike (Esox lucius). These fish have slim and low-density osteocyte-lacking bones. As part of the swallowing mechanism, the cleithrum bone opens and closes the jaw. The cleithrum needs sufficient strength and damage tolerance, to withstand years of repetitive rapid gape-and-suck cycles of feeding. The thin wing-shaped bone comprises anisotropic layers of mineralized collagen fibers that exhibit periodic variations in mineral density on the mm and micrometer length scales. Wavy collagen fibrils interconnect these layers yielding a highly anisotropic structure. Hydrated cleithra exhibit Young's moduli spanning 3-9 GPa where the yield stress of ∼40 MPa increases markedly to exceed ∼180 MPa upon drying. This 5x observation of increased strength corresponds to a change to brittle fracture patterns. It matches the emergence of compressive residual strains of ∼0.15% within the mineral crystals due to forces from shrinking collagen layers. Compressive stresses on the nanoscale, combined with the layered anisotropic microstructure on the mm length scale, jointly confer structural stability in the slender and lightweight bones. By employing a range of X-ray, electron and optical imaging and mechanical characterization techniques, we reveal the structure and properties that make the cleithra impressively damage resistant composites. STATEMENT OF SIGNIFICANCE: By combining structural and mechanical characterization techniques spanning the mm to the sub-nanometer length scales, this work provides insights into the structural organization and properties of a resilient bone found in pike fish. Our observations show how the anosteocytic bone within the pectoral gridle of these fish, lacking any biological (remodeling) repair mechanisms, is adapted to sustain natural repeated loading cycles of abrupt jaw-gaping and swallowing. We find residual strains within the mineral apatite nanocrystals that contribute to forming a remarkably resilient composite material. Such information gleaned from bony structures that are different from the usual bones of mammals showcases how nature incorporates smart features that induce damage tolerance in bone material, an adaptation acquired through natural evolutionary processes.


Subject(s)
Esocidae , Animals , Esocidae/physiology , Bone and Bones/physiology , Stress, Mechanical , Nanoparticles/chemistry , Compressive Strength , Biological Evolution , Elastic Modulus , Collagen/chemistry
6.
Curr Osteoporos Rep ; 22(2): 290-298, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38358401

ABSTRACT

PURPOSE OF REVIEW: Interfacial tissue exists throughout the body at cartilage-to-bone (osteochondral interface) and tendon-to-bone (enthesis) interfaces. Healing of interfacial tissues is a current challenge in regenerative approaches because the interface plays a critical role in stabilizing and distributing the mechanical stress between soft tissues (e.g., cartilage and tendon) and bone. The purpose of this review is to identify new directions in the field of interfacial tissue development and physiology that can guide future regenerative strategies for improving post-injury healing. RECENT FINDINGS: Cues from interfacial tissue development may guide regeneration including biological cues such as cell phenotype and growth factor signaling; structural cues such as extracellular matrix (ECM) deposition, ECM, and cell alignment; and mechanical cues such as compression, tension, shear, and the stiffness of the cellular microenvironment. In this review, we explore new discoveries in the field of interfacial biology related to ECM remodeling, cellular metabolism, and fate. Based on emergent findings across multiple disciplines, we lay out a framework for future innovations in the design of engineered strategies for interface regeneration. Many of the key mechanisms essential for interfacial tissue development and adaptation have high potential for improving outcomes in the clinic.


Subject(s)
Bone Regeneration , Extracellular Matrix , Humans , Extracellular Matrix/physiology , Bone Regeneration/physiology , Bone and Bones/physiology , Tendons/physiology , Tissue Engineering/methods , Cartilage/physiology , Regeneration/physiology , Wound Healing/physiology
7.
Bone ; 179: 116985, 2024 02.
Article in English | MEDLINE | ID: mdl-38052372

ABSTRACT

Sclerostin is an extracellular inhibitor of canonical Wnt signaling that inhibits bone formation and stimulates bone resorption. Anti-sclerostin antibodies (Scl-Ab) have been developed as bone-building agents. DKK1, another extracellular inhibitor of the pathway, is upregulated in osteocytes in response to sclerostin inhibition. To further enhance bone-forming effects, a bispecific antibody inhibiting both sclerostin and DKK1 was created (AMG 147). In nonclinical safety studies, AMG 147 resulted in novel skull findings. In the rat, there was increased thickness of skull bones of neural crest origin due to increased subperiosteal compact lamellar and intramembranous woven bone. Externally, subperiosteal fibroblastic/osteoblastic stromal cell proliferation with woven bone and hemorrhage was also observed. Scl-Ab alone resulted in increased skull thickness in the rat, like AMG 147, but without the stromal cell proliferation/woven bone formation. In contrast to embryonic flat bone development, intramembranous bone formed similar to plexiform bone. In the monkey, AMG 147 resulted in macroscopic skull thickening due to a diffuse increase in appositional lamellar bone and increased intramembranous bone on both periosteal surfaces of all skull bones. These data demonstrate that dual inhibition of sclerostin and DDK1 results in unique effects on the skull not observed with sclerostin inhibition alone.


Subject(s)
Adaptor Proteins, Signal Transducing , Antibodies , Bone and Bones , Intercellular Signaling Peptides and Proteins , Animals , Rats , Antibodies/pharmacology , Osteogenesis , Primates , Skull , Intercellular Signaling Peptides and Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Bone and Bones/drug effects , Bone and Bones/physiology
8.
J Ethnopharmacol ; 311: 116399, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-36997131

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Tiger bone, which had long been used in traditional Chinese medicine, had the action of removing wind and alleviating pain, strengthening the sinews and bones, and often used to treat bone impediment, and atrophic debility of bones in TCM clinical practice. As a substitute of natural bone tiger, artificial tiger bone Jintiange (JTG), has been approved by the State Food and Drug Administration of China for relief the symptom of osteoporosis, such as lumbago and back pain, lassitude in loin and legs, flaccidity and weakness legs, and walk with difficulty based on TCM theory. JTG has similar chemical profile to natural tiger bone, and contains mineral substance, peptides and proteins, and has been shown to protect bone loss in ovariectomized mice and exert the regulatory effects on osteoblast and osteoclast activities. But how the peptides and proteins in JTG modulate bone formation remains unclear. AIM: To investigate the stimulating effects of JTG proteins on osteogenesis and explore the possible underlying mechanisms. MATERIALS AND METHODS: JTG proteins were prepared from JTG Capsules by extracting calcium, phosphorus and other inorganic elements using SEP-PaktC18 desalting column. MC3T3-E1 cells were treated with JTG proteins to evaluate their effects and explore the underlying mechanisms. Osteoblast proliferation was detected by CCK-8 method. ALP activity was detected using a relevant assay kit, and bone mineralized nodules were stained with alizarin red-Tris-HCl solution. Cell apoptosis was analyzed by flow cytometry. Autophagy was observed by MDC staining, and autophagosomes were observed by TEM. Nuclear translocations of LC3 and CHOP were detected by immunofluorescence and observed under a laser confocal microscope. The expression of key proteins related to osteogenesis, apoptosis, autophagy and PI3K/AKT and ER stress pathways was analyzed by Western Blot analysis. RESULTS: JTG proteins improved osteogenesis as evidenced by the alteration of proliferation, differentiation and mineralization of MC3T3-E1 osteoblasts, inhibited their apoptosis, and enhanced autophagosome formation and autophagy. They also regulated the expression of key proteins of PI3K/AKT and ER stress pathways. In addition, PI3K/AKT and ER stress pathway inhibitors could reverse the regulatory effects of JTG proteins on osteogenesis, apoptosis, autophagy and PI3K/AKT and ER stress pathways. CONCLUSION: JTG proteins increased the osteogenesis and inhibited osteoblast apoptosis by enhancing autophagy via PI3K/AKT and ER stress signaling pathways.


Subject(s)
Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Ethnopharmacology , Osteoblasts , Osteogenesis , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects , Apoptosis/drug effects , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tigers , Bone and Bones/chemistry , Bone and Bones/drug effects , Bone and Bones/physiology , Cell Line , Metabolic Networks and Pathways/drug effects , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Animals , Mice , Ovariectomy , Female
9.
Calcif Tissue Int ; 112(1): 13-23, 2023 01.
Article in English | MEDLINE | ID: mdl-36261652

ABSTRACT

Achondroplasia (ACH) is a skeletal disorder caused by fibroblast growth factor receptor 3 (FGFR3) variants. Volumetric bone mineral density (vBMD), bone microarchitecture, and strength have not been evaluated in these patients previously. This study aims to evaluate vBMD, bone microarchitecture, and strength in ACH patients. Seventeen patients underwent clinical and biochemical evaluations, and genetic testing. High-resolution peripheral quantitative computed tomography was performed in 10 ACH patients and 21 age- and sex-matched healthy subjects. All individuals had the hotspot mutation of c.1138G > A in FGFR3. Linear growth retardation, disproportionate short stature, and genu varum are the most common manifestations. The mean height was 108.82 ± 24.08 cm (Z score: - 5.72 ± 0.96). Total vBMD in the ACH and the control groups was 427.08 ± 49.29 mg HA/cm3 versus 300.35 ± 69.92 mg HA/cm3 (p < 0.001) at the radius and 336.90 ± 79.33 mg HA/cm3 versus 292.20 ± 62.35 mg HA/cm3 (p = 0.098) at the tibia; both at the radius and tibia, vBMD of trabecular bones was significantly lower in the ACH group than in the control group, but vBMD of cortical bones was slightly higher in the ACH group. Trabecular separation and cortical thickness in the ACH group were significantly higher than those in the control group, but trabecular number was significantly decreased in the ACH group. Stiffness and failure load were only better at the radius in the ACH group. ACH patients have higher total and cortical vBMD, lower trabecular vBMD, worse trabecular bone microarchitecture, thicker cortical bone thickness, and better estimated bone strength.


Subject(s)
Achondroplasia , Bone Density , Receptor, Fibroblast Growth Factor, Type 3 , Humans , Absorptiometry, Photon , Achondroplasia/genetics , Achondroplasia/metabolism , Bone Density/genetics , Cross-Sectional Studies , Mutation , Radius , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Tibia , Bone and Bones/anatomy & histology , Bone and Bones/physiology
10.
J Bone Miner Res ; 37(9): 1733-1749, 2022 09.
Article in English | MEDLINE | ID: mdl-35773783

ABSTRACT

Biallelic ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) deficiency induces vascular/soft tissue calcifications in generalized arterial calcification of infancy (GACI), and low bone mass with phosphate-wasting rickets in GACI survivors (autosomal hypophosphatemic rickets type-2). ENPP1 haploinsufficiency induces early-onset osteoporosis and mild phosphate wasting in adults. Both conditions demonstrate the unusual combination of reduced accrual of skeletal mineral, yet excess and progressive heterotopic mineralization. ENPP1 is the only enzyme that generates extracellular pyrophosphate (PPi), a potent inhibitor of both bone and heterotopic mineralization. Life-threatening vascular calcification in ENPP1 deficiency is due to decreased plasma PPi; however, the mechanism by which osteopenia results is not apparent from an understanding of the enzyme's catalytic activity. To probe for catalysis-independent ENPP1 pathways regulating bone, we developed a murine model uncoupling ENPP1 protein signaling from ENPP1 catalysis, Enpp1T238A mice. In contrast to Enpp1asj mice, which lack ENPP1, Enpp1T238A mice have normal trabecular bone microarchitecture and favorable biomechanical properties. However, both models demonstrate low plasma Pi and PPi, increased fibroblast growth factor 23 (FGF23), and by 23 weeks, osteomalacia demonstrating equivalent phosphate wasting in both models. Reflecting findings in whole bone, calvarial cell cultures from Enpp1asj mice demonstrated markedly decreased calcification, elevated transcription of Sfrp1, and decreased nuclear ß-catenin signaling compared to wild-type (WT) and Enpp1T238A cultures. Finally, the decreased calcification and nuclear ß-catenin signaling observed in Enpp1asj cultures was restored to WT levels by knockout of Sfrp1. Collectively, our findings demonstrate that catalysis-independent ENPP1 signaling pathways regulate bone mass via the expression of soluble Wnt inhibitors such as secreted frizzled-related protein 1 (SFRP1), whereas catalysis dependent pathways regulate phosphate homeostasis through the regulation of plasma FGF23. © 2022 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Bone and Bones/physiology , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/metabolism , Animals , Catalysis , Familial Hypophosphatemic Rickets , Fibroblast Growth Factors , Mammals/metabolism , Mice , Phosphates/metabolism , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/genetics , Vascular Calcification , beta Catenin
11.
ACS Biomater Sci Eng ; 8(8): 3199-3219, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35816626

ABSTRACT

As bone grafts become more commonly needed by patients and as donors become scarcer, acellularized bone grafts (ABGs) are becoming more popular for restorative purposes. While autogeneic grafts are reliable as a gold standard, allogeneic and xenogeneic ABGs have been shown to be of particular interest due to the limited availability of autogeneic resources and reduced patient well-being in long-term surgeries. Because of the complete similarity of their structures with native bone, excellent mechanical properties, high biocompatibility, and similarities of biological behaviors (osteoinductive and osteoconductive) with local bones, successful outcomes of allogeneic and xenogeneic ABGs in both in vitro and in vivo research have raised hopes of repairing patients' bone injuries in clinical applications. However, clinical trials have been delayed due to a lack of standardized protocols pertaining to acellularization, cell seeding, maintenance, and diversity of ABG evaluation criteria. This study sought to uncover these factors by exploring the bone structures, ossification properties of ABGs, sources, benefits, and challenges of acellularization approaches (physical, chemical, and enzymatic), cell loading, and type of cells used and effects of each of the above items on the regenerative technologies. To gain a perspective on the repair and commercialization of products before implementing new research activities, this study describes the differences between ABGs created by various techniques and methods applied to them. With a comprehensive understanding of ABG behavior, future research focused on treating bone defects could provide a better way to combine the treatment approaches needed to treat bone defects.


Subject(s)
Bone Regeneration , Bone Transplantation/methods , Bone and Bones/pathology , Transplantation, Heterologous/standards , Transplantation, Homologous/standards , Bone Transplantation/standards , Bone and Bones/physiology , Bone and Bones/surgery , Humans , Osteogenesis , Transplantation, Heterologous/methods , Transplantation, Homologous/methods
12.
Int J Mol Sci ; 23(7)2022 Mar 22.
Article in English | MEDLINE | ID: mdl-35408784

ABSTRACT

Lysophosphatidic acid (LPA) is a natural bioactive phospholipid with pleiotropic activities affecting multiple tissues, including bone. LPA exerts its biological functions by binding to G-protein coupled LPA receptors (LPA1-6) to stimulate cell migration, proliferation, and survival. It is largely produced by autotaxin (ATX), a secreted enzyme with lysophospholipase D activity that converts lysophosphatidylcholine (LPC) into active LPA. Beyond its enzymatic activity, ATX serves as a docking molecule facilitating the efficient delivery of LPA to its specific cell surface receptors. Thus, LPA effects are the result of local production by ATX in a given tissue or cell type. As a consequence, the ATX/LPA axis should be considered as an entity to better understand their roles in physiology and pathophysiology and to propose novel therapeutic strategies. Herein, we provide not only an extensive overview of the relevance of the ATX/LPA axis in bone cell commitment and differentiation, skeletal development, and bone disorders, but also discuss new working hypotheses emerging from the interplay of ATX/LPA with well-established signaling pathways regulating bone mass.


Subject(s)
Bone Diseases , Bone and Bones , Lysophospholipids , Phosphoric Diester Hydrolases , Bone and Bones/physiology , Humans , Lysophospholipids/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Lysophosphatidic Acid/metabolism
13.
Am J Hum Biol ; 34(8): e23756, 2022 08.
Article in English | MEDLINE | ID: mdl-35481615

ABSTRACT

OBJECTIVES: Ethnic groups differ in prevalence of calcium-related diseases. Differences in the physiology and the endogenous circadian rhythm (CR) of calcium and bone homeostasis may play a role. Thus, we aimed to investigate details of CR pattern in calcium and bone homeostasis in East African Maasai. METHODS: Ten clinically healthy adult Maasai men and women from Tanzania were examined. Blood samples were collected every 2nd hour for 24 h. Serum levels of total calcium, albumin, parathyroid hormone (PTH), 25(OH)D, creatinine, C-terminal telopeptide (CTX), bone-specific alkaline phosphatase (BSAP), procollagen type 1 N-terminal propeptide (P1NP), and osteocalcin were measured. Circadian patterns were derived from graphic curves of medians, and rhythmicity was assessed with Fourier analysis. RESULTS: PTH-levels varied over the 24 h exhibiting a bimodal pattern. Nadir level corresponded to 65% of total 24-h mean. CTX and P1NP showed 24-h variations with a morning nadir and nocturnal peak with nadir levels corresponding to 23% and 79% of the 24-h mean, respectively. Albumin-corrected calcium level was held in a narrow range and alterations were corresponding to alterations in PTH. There was no distinct pattern in 24-h variations of 25(OH)D, creatinine, osteocalcin, or BSAP. CONCLUSIONS: All participants showed pronounced 24-h variations in PTH and bone turnover markers CTX and P1NP. These findings support that Maasai participants included in this study have typical patterns of CR in calcium and bone homeostasis consistent with findings from other ethnic populations.


Subject(s)
Bone and Bones , Calcium , Circadian Rhythm , Adult , Albumins , Biomarkers , Bone and Bones/physiology , Calcium/physiology , Circadian Rhythm/physiology , Creatinine , Ethnicity , Female , Homeostasis , Humans , Male , Osteocalcin , Parathyroid Hormone/physiology , Tanzania
14.
Life Sci ; 295: 120406, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35182555

ABSTRACT

AIMS: To investigate the effects of hyperbaric oxygen therapy (HBOT) on metabolic disturbance, aging and bone remodeling in D-galactose-induced aging rats with and without obesity by determining the metabolic parameters, aging and oxidative stress markers, bone turnover markers, bone microarchitecture, and bone biomechanical strength. MATERIALS AND METHODS: Male Wistar rats were fed either a normal diet (ND; n = 18) or a HFD (n = 12) for 22 weeks. At week 13, vehicle (0.9% NaCl) was injected into ND-fed rats (NDV; n = 6), while 150 mg/kg/day of D-galactose was injected into 12 ND-fed rats (NDD) and 12 HFD-fed rats (HFDD) for 10 weeks. At week 21, rats were treated with either sham (NDVS, NDDS, or HFDDS; n = 6/ group) or HBOT (NDDH, or HFDDH; n = 6/group) for 14 days. Rats were then euthanized. Blood samples, femora, and tibiae were collected. KEY FINDINGS: Both NDD and HFDD groups developed aging as indicated by increased AGE level, increased inflammation and oxidative stress as shown by raised serum TNF-α and MDA levels, impaired bone remodeling as indicated by an increase in levels of CTX-1, TRACP-5b, and impaired bone structure/strength, when compared with those of the NDVS group. HFD aggravated these indicators of bone dyshomeostasis in D-galactose-treated rats. HBOT restored bone remodeling and bone structure/strength in the NDD group, however HBOT ameliorated bone dyshomeostasis in the HFDD group. SIGNIFICANCE: HBOT is a potential intervention to decrease the risk of osteoporosis and bone fracture in aging with or without obesity.


Subject(s)
Aging/physiology , Bone and Bones/metabolism , Hyperbaric Oxygenation/methods , Age Factors , Animals , Bone Remodeling/physiology , Bone and Bones/physiology , Diet, High-Fat , Galactose/adverse effects , Galactose/pharmacology , Homeostasis , Inflammation/metabolism , Insulin Resistance , Male , Obesity/metabolism , Obesity/physiopathology , Osteoporosis/metabolism , Oxidative Stress , Rats , Rats, Wistar
15.
Sci Rep ; 12(1): 2492, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169187

ABSTRACT

Bone fractures commonly repair by forming a bridging structure called callus, which begins as soft tissue and gradually ossifies to restore rigidity to the bone. Virtual mechanical testing is a promising technique for image-based assessment of structural bone healing in both preclinical and clinical settings, but its accuracy depends on the validity of the material model used to assign tissue mechanical properties. The goal of this study was to develop a constitutive model for callus that captures the heterogeneity and biomechanical duality of the callus, which contains both soft tissue and woven bone. To achieve this, a large-scale optimization analysis was performed on 2363 variations of 3D finite element models derived from computed tomography (CT) scans of 33 osteotomized sheep under normal and delayed healing conditions. A piecewise material model was identified that produced high absolute agreement between virtual and physical tests by differentiating between soft and hard callus based on radiodensity. The results showed that the structural integrity of a healing long bone is conferred by an internal architecture of mineralized hard callus that is supported by interstitial soft tissue. These findings suggest that with appropriate material modeling, virtual mechanical testing is a reliable surrogate for physical biomechanical testing.


Subject(s)
Bone and Bones/physiology , Fracture Healing/physiology , Fractures, Bone/physiopathology , Mechanical Tests/methods , Osteogenesis/physiology , Animals , Biomechanical Phenomena , Bone and Bones/diagnostic imaging , Connective Tissue/diagnostic imaging , Connective Tissue/physiology , Finite Element Analysis , Sheep , Tomography, X-Ray Computed/methods
16.
Elife ; 112022 02 08.
Article in English | MEDLINE | ID: mdl-35132953

ABSTRACT

Mechanical stimulation, such as physical exercise, is essential for bone formation and health. Here, we demonstrate the critical role of osteocytic Cx43 hemichannels in anabolic function of bone in response to mechanical loading. Two transgenic mouse models, R76W and Δ130-136, expressing dominant-negative Cx43 mutants in osteocytes were adopted. Mechanical loading of tibial bone increased cortical bone mass and mechanical properties in wild-type and gap junction-impaired R76W mice through increased PGE2, endosteal osteoblast activity, and decreased sclerostin. These anabolic responses were impeded in gap junction/hemichannel-impaired Δ130-136 mice and accompanied by increased endosteal osteoclast activity. Specific inhibition of Cx43 hemichannels by Cx43(M1) antibody suppressed PGE2 secretion and impeded loading-induced endosteal osteoblast activity, bone formation and anabolic gene expression. PGE2 administration rescued the osteogenic response to mechanical loading impeded by impaired hemichannels. Together, osteocytic Cx43 hemichannels could be a potential new therapeutic target for treating bone loss and osteoporosis.


Subject(s)
Bone Remodeling , Bone and Bones/physiology , Connexin 43/metabolism , Prostaglandins/metabolism , Animals , Biomechanical Phenomena , Connexin 43/genetics , Dinoprostone/metabolism , Gap Junctions/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Osteocytes/metabolism , Stress, Mechanical , Weight-Bearing
17.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35163219

ABSTRACT

Prostate cancer (PCa) is the most frequent malignancy in older men with a high propensity for bone metastases. Characteristically, PCa causes osteosclerotic lesions as a result of disrupted bone remodeling. Extracellular vesicles (EVs) participate in PCa progression by conditioning the pre-metastatic niche. However, how EVs mediate the cross-talk between PCa cells and osteoprogenitors in the bone microenvironment remains poorly understood. We found that EVs derived from murine PCa cell line RM1-BM increased metabolic activity, vitality, and cell proliferation of osteoblast precursors by >60%, while significantly impairing mineral deposition (-37%). The latter was further confirmed in two complementary in vivo models of ossification. Accordingly, gene and protein set enrichments of osteoprogenitors exposed to EVs displayed significant downregulation of osteogenic markers and upregulation of proinflammatory factors. Additionally, transcriptomic profiling of PCa-EVs revealed the abundance of three microRNAs, miR-26a-5p, miR-27a-3p, and miR-30e-5p involved in the suppression of BMP-2-induced osteogenesis in vivo, suggesting the critical role of these EV-derived miRNAs in PCa-mediated suppression of osteoblast activity. Taken together, our results indicate the importance of EV cargo in cancer-bone cross-talk in vitro and in vivo and suggest that exosomal miRNAs may contribute to the onset of osteosclerotic bone lesions in PCa.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/genetics , Osteoblasts/physiology , Prostatic Neoplasms/genetics , Animals , Bone and Bones/metabolism , Bone and Bones/physiology , Cell Communication , Cell Line, Tumor , Cell Proliferation , Exosome Multienzyme Ribonuclease Complex/metabolism , Exosomes/genetics , Extracellular Vesicles/metabolism , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Male , Mesenchymal Stem Cells , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Osteogenesis , Transcriptome/genetics , Tumor Microenvironment
18.
Int J Mol Sci ; 23(3)2022 Feb 03.
Article in English | MEDLINE | ID: mdl-35163687

ABSTRACT

It has been observed that bone fractures carry a risk of high mortality and morbidity. The deployment of a proper bone healing method is essential to achieve the desired success. Over the years, bone tissue engineering (BTE) has appeared to be a very promising approach aimed at restoring bone defects. The main role of the BTE is to apply new, efficient, and functional bone regeneration therapy via a combination of bone scaffolds with cells and/or healing promotive factors (e.g., growth factors and bioactive agents). The modern approach involves also the production of living bone grafts in vitro by long-term culture of cell-seeded biomaterials, often with the use of bioreactors. This review presents the most recent findings concerning biomaterials, cells, and techniques used for the production of living bone grafts under in vitro conditions. Particular attention has been given to features of known bioreactor systems currently used in BTE: perfusion bioreactors, rotating bioreactors, and spinner flask bioreactors. Although bioreactor systems are still characterized by some limitations, they are excellent platforms to form bioengineered living bone grafts in vitro for bone fracture regeneration. Moreover, the review article also describes the types of biomaterials and sources of cells that can be used in BTE as well as the role of three-dimensional bioprinting and pulsed electromagnetic fields in both bone healing and BTE.


Subject(s)
Bioengineering , Bioreactors , Bone Transplantation , Tissue Engineering , Animals , Bone and Bones/physiology , Humans , Printing, Three-Dimensional
19.
Int J Mol Sci ; 23(4)2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35216233

ABSTRACT

The primary cilium is a hair-like immotile organelle with specific membrane receptors, including the receptor of Hedgehog signaling, smoothened. The cilium organized in preosteoblasts promotes differentiation of the cells into osteoblasts (osteoblast differentiation) by mediating Hedgehog signaling to achieve bone formation. Notably, 4.1G is a plasma membrane-associated cytoskeletal protein that plays essential roles in various tissues, including the peripheral nervous system, testis, and retina. However, its function in the bone remains unexplored. In this study, we identified 4.1G expression in the bone. We found that, in the 4.1G-knockout mice, calcium deposits and primary cilium formation were suppressed in the trabecular bone, which is preosteoblast-rich region of the newborn tibia, indicating that 4.1G is a prerequisite for osteoblast differentiation by organizing the primary cilia in preosteoblasts. Next, we found that the primary cilium was elongated in the differentiating mouse preosteoblast cell line MC3T3-E1, whereas the knockdown of 4.1G suppressed its elongation. Moreover, 4.1G-knockdown suppressed the induction of the cilia-mediated Hedgehog signaling and subsequent osteoblast differentiation. These results demonstrate a new regulatory mechanism of 4.1G in bone formation that promotes the primary ciliogenesis in the differentiating preosteoblasts and induction of cilia-mediated osteoblast differentiation, resulting in bone formation at the newborn stage.


Subject(s)
Cell Differentiation/physiology , Cilia/metabolism , Cilia/physiology , Microfilament Proteins/metabolism , Osteoblasts/metabolism , Osteoblasts/physiology , Osteogenesis/physiology , 3T3 Cells , Animals , Bone and Bones/metabolism , Bone and Bones/physiology , Calcification, Physiologic/physiology , Cell Line , Mice , Mice, Knockout , Signal Transduction/physiology
20.
Genes (Basel) ; 13(1)2022 01 13.
Article in English | MEDLINE | ID: mdl-35052478

ABSTRACT

The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.


Subject(s)
Bone Diseases/pathology , Bone and Bones/physiology , Homeostasis , Osteogenesis , Wnt Signaling Pathway , Animals , Bone Diseases/metabolism , Bone and Bones/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...