Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Mil Med Res ; 10(1): 54, 2023 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-37941072

RESUMO

Degenerative musculoskeletal diseases are structural and functional failures of the musculoskeletal system, including osteoarthritis, osteoporosis, intervertebral disc degeneration (IVDD), and sarcopenia. As the global population ages, degenerative musculoskeletal diseases are becoming more prevalent. However, the pathogenesis of degenerative musculoskeletal diseases is not fully understood. Previous studies have revealed that endoplasmic reticulum (ER) stress is a stress response that occurs when impairment of the protein folding capacity of the ER leads to the accumulation of misfolded or unfolded proteins in the ER, contributing to degenerative musculoskeletal diseases. By affecting cartilage degeneration, synovitis, meniscal lesion, subchondral bone remodeling of osteoarthritis, bone remodeling and angiogenesis of osteoporosis, nucleus pulposus degeneration, annulus fibrosus rupture, cartilaginous endplate degeneration of IVDD, and sarcopenia, ER stress is involved in the pathogenesis of degenerative musculoskeletal diseases. Preclinical studies have found that regulation of ER stress can delay the progression of multiple degenerative musculoskeletal diseases. These pilot studies provide foundations for further evaluation of the feasibility, efficacy, and safety of ER stress modulators in the treatment of musculoskeletal degenerative diseases in clinical trials. In this review, we have integrated up-to-date research findings of ER stress into the pathogenesis of degenerative musculoskeletal diseases. In a future perspective, we have also discussed possible directions of ER stress in the investigation of degenerative musculoskeletal disease, potential therapeutic strategies for degenerative musculoskeletal diseases using ER stress modulators, as well as underlying challenges and obstacles in bench-to-beside research.


Assuntos
Degeneração do Disco Intervertebral , Osteoartrite , Osteoporose , Sarcopenia , Humanos , Estresse do Retículo Endoplasmático/fisiologia , Degeneração do Disco Intervertebral/tratamento farmacológico , Degeneração do Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/patologia
2.
Eur J Orthod ; 45(2): 224-234, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-36576115

RESUMO

OBJECTIVE: Teriparatide (TPTD) and abaloparatide (ABL) are two osteoanabolic drugs targeting parathyroid hormone (PTH)1R signalling. This study aimed to investigate the effects of TPTD and ABL on the adolescent mandibular growth. METHOD: In total, 70 4-week-old male Sprague-Dawley rats were randomly divided into 14 groups, treated with intermittent TPDT or ABL at various doses, accompanied by mandibular advancement (MA) or not. 3D printing was used to fabricate an innovative splint for MA. After a 4-week treatment, morphological measurement, histological and immunohistochemical analysis were performed. Mandibular condylar chondrocytes (MCCs) were treated with TPTD or ABL, followed by CCK-8 assay, alcian blue staining, real time-PCR and immunofluorescent staining. RESULT: In vivo, TPTD or ABL alone increased the condylar length and cartilage thickness, with up-regulated SOX9 and COL II, whilst down-regulated COL X; however, when combined with MA, the promotive effects were attenuated. TPTD or ABL alone increased the mandibular body height and mandibular angle width, whilst increased the mandibular body length and alveolar bone width when combined with MA. In vitro, TPTD or ABL enhanced the MCC proliferation, glycosaminoglycan synthesis, COL II and SOX9 expression, whilst down-regulated COL X, Ihh and PTH1R expression. CONCLUSION: Both ABL and TPTD enhance mandibular growth in adolescent rats with site-specific and mechano-related effects, including propelling chondrogenesis at the condylar cartilage and promoting bone apposition at other mechano-responsive sites. They behave as promising drugs for mandibular growth modification, and in general ABL seems more potent than TPTD in this context.


Assuntos
Conservadores da Densidade Óssea , Teriparatida , Ratos , Masculino , Animais , Teriparatida/farmacologia , Teriparatida/uso terapêutico , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/uso terapêutico , Ratos Sprague-Dawley , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Proteína Relacionada ao Hormônio Paratireóideo/uso terapêutico
3.
Front Cell Dev Biol ; 10: 1014789, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36330332

RESUMO

Femoral head necrosis (FHN), one of the most popular joint diseases in the musculoskeletal system, is usually attributed to local ischemia of the femoral head. Thus, regenerating the vascularization capacity and restoring the local perfusion of the femoral head becomes an efficient therapeutic approach for FHN. We investigated the function of autologous lipoaspirate cells (LPCs) in regenerating circulation in FHN animal models and human subjects in this study. We also explored the mechanisms of why LPCs show a superior effect than that of the bone marrow-derived stem cells (BMSCs) in vascularization. Thirty-four FHN patients were recruited for the randomized clinical trial. Harris Hip Score (HHS) and digital subtraction arteriography (DSA) and interventional technique were used to compare the efficacy of LPCs treatment and vehicle therapy in improving femoral head circulation and hip joint function. Cellular mechanism that underlies the beneficial effect of LPCs in restoring blood supply and rescuing bone architecture was further explored using canine and mouse FHN animal models. We found that LPCs perfusion through the medial circumflex artery will promote the femoral head vascularization and bone structure significantly in both FHN patients and animal models. The HHS in LPCs treated patients was significantly improved relative to vehicle group. The levels of angiogenesis factor secreted by LPCs such as VEGF, FGF2, VEC, TGF-ß, were significantly higher than that of BMSCs. As the result, LPCs showed a better effect in promoting the tube structure formation of human vascular endothelial cells (HUVEC) than that of BMSCs. Moreover, LPCs contains a unique CD44+CD34+CD31- population. The CD44+CD34+CD31- LPCs showed significantly higher angiogenesis potential as compared to that of BMSCs. Taken together, our results show that LPCs possess a superior vascularization capacity in both autonomous and paracrine manner, indicating that autologous LPCs perfusion via the medial circumflex artery is an effective therapy for FHN.

4.
Bone Res ; 10(1): 44, 2022 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-35668080

RESUMO

The field of research on pain originating from various bone diseases is expanding rapidly, with new mechanisms and targets asserting both peripheral and central sites of action. The scope of research is broadening from bone biology to neuroscience, neuroendocrinology, and immunology. In particular, the roles of primary sensory neurons and non-neuronal cells in the peripheral tissues as important targets for bone pain treatment are under extensive investigation in both pre-clinical and clinical settings. An understanding of the peripheral mechanisms underlying pain conditions associated with various bone diseases will aid in the appropriate application and development of optimal strategies for not only managing bone pain symptoms but also improving bone repairing and remodeling, which potentially cures the underlying etiology for long-term functional recovery. In this review, we focus on advances in important preclinical studies of significant bone pain conditions in the past 5 years that indicated new peripheral neuronal and non-neuronal mechanisms, novel targets for potential clinical interventions, and future directions of research.

5.
Bone Res ; 10(1): 24, 2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35232979

RESUMO

The molecular control of osteoclast formation is still not clearly elucidated. Here, we show that a process of cell recognition mediated by Siglec15-TLR2 binding is indispensable and occurs prior to cell fusion in RANKL-mediated osteoclastogenesis. Siglec15 has been shown to regulate osteoclastic bone resorption. However, the receptor for Siglec15 has not been identified, and the signaling mechanism involving Siglec15 in osteoclast function remains unclear. We found that Siglec15 bound sialylated TLR2 as its receptor and that the binding of sialylated TLR2 to Siglec15 in macrophages committed to the osteoclast-lineage initiated cell fusion for osteoclast formation, in which sialic acid was transferred by the sialyltransferase ST3Gal1. Interestingly, the expression of Siglec15 in macrophages was activated by M-CSF, whereas ST3Gal1 expression was induced by RANKL. Both Siglec15-specific deletion in macrophages and intrafemoral injection of sialidase abrogated cell recognition and reduced subsequent cell fusion for the formation of osteoclasts, resulting in increased bone formation in mice. Thus, our results reveal that cell recognition mediated by the binding of sialylated TLR2 to Siglec15 initiates cell fusion for osteoclast formation.

6.
Bone Res ; 9(1): 47, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34719673

RESUMO

Osteoporosis (OP) is a common age-related disease characterized by a deterioration of bone mass and structure that predisposes patients to fragility fractures. Pharmaceutical therapies that promote anabolic bone formation in OP patients and OP-induced fracture are needed. We investigated whether a neutralizing antibody against Siglec-15 can simultaneously inhibit bone resorption and stimulate bone formation. We found that the multinucleation of osteoclasts was inhibited in SIGLEC-15 conditional knockout mice and mice undergoing Siglec-15 neutralizing antibody treatment. The secretion of platelet-derived growth factor-BB (PDGF-BB), the number of tartrate-resistant acid phosphatase-positive (TRAP+) mononuclear cells, and bone formation were significantly increased in the SIGLEC-15 conditional knockout mice and antibody-treated mice. The anabolic effect of the Siglec-15 neutralizing antibody on bone formation was blunted in mice with Pdgfb deleted in TRAP+ cells. These findings showed that the anabolic effect of the Siglec-15 neutralizing antibody was mediated by elevating PDGF-BB production of TRAP+ mononuclear cells. To test the therapeutic potential of the Siglec-15 neutralizing antibody, we injected the antibody in an ovariectomy-induced osteoporotic mouse model, which mimics postmenopausal osteoporosis in women, and in two fracture healing models because fracture is the most serious health consequence of osteoporosis. The Siglec-15 neutralizing antibody effectively reduced bone resorption and stimulated bone formation in estrogen deficiency-induced osteoporosis. Of note, the Siglec-15 neutralizing antibody promoted intramembranous and endochondral ossification at the damaged area of cortical bone in fracture healing mouse models. Thus, the Siglec-15 neutralizing antibody shows significant translational potential as a novel therapy for OP and bone fracture.

7.
J Rheumatol ; 48(10): 1608-1615, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34329188

RESUMO

OBJECTIVE: To investigate the metabolic syndrome (MetS) association with radiographic and symptomatic hand osteoarthritis (HOA). METHODS: Using 1:2 propensity score matching for relevant confounders, we included 2509 participants (896 MetS positive and 1613 MetS negative) from the Osteoarthritis Initiative dataset. MetS and its components, according to the International Diabetes Federation criteria, were extracted from baseline data, and included hypertension, abdominal obesity, dyslipidemia, and diabetes. We scored distinct hand joints based on the modified Kellgren-Lawrence (mKL) grade of baseline radiographs, with HOA defined as mKL ≥ 2. In the cross-sectional analysis, we investigated the association between MetS and its components with radiographic HOA and the presence of nodal and erosive HOA phenotypes using regression models. In the longitudinal analysis, we performed Cox regression analysis for hand pain incidence in follow-up visits. RESULTS: MetS was associated with higher odds of radiographic HOA, including the number of joints with OA (OR 1.32, 95% CI 1.08-1.62), the sum of joints mKLs (OR 2.42, 95% CI 1.24-4.71), mainly in distal interphalangeal joints (DIPs) and proximal interphalangeal joints (PIPs; OR 1.52, 95% CI 1.08-2.14 and OR 1.38, 95% CI 1.09-1.75, respectively), but not metacarpophalangeal (MCP) and first carpometacarpal (CMC1) joints. Hand pain incidence during follow-up was higher with MetS presence (HR 1.25, 95% CI 1.07-1.47). The erosive HOA phenotype and joints' nodal involvement were more frequent with MetS (OR 1.40, 95% CI 1.01-1.97 and OR 1.28, 95% CI 1.02-1.60, respectively). CONCLUSION: MetS, a potentially modifiable risk factor, is associated with radiographic DIP and PIP OA and longitudinal hand pain incidence while sparing MCPs and CMC1s. Nodal and erosive HOA phenotypes are associated with MetS, suggestive of possible distinct pathophysiology.


Assuntos
Articulação da Mão , Síndrome Metabólica , Osteoartrite , Estudos Transversais , Articulação da Mão/diagnóstico por imagem , Humanos , Síndrome Metabólica/complicações , Síndrome Metabólica/diagnóstico por imagem , Síndrome Metabólica/epidemiologia , Osteoartrite/diagnóstico por imagem , Osteoartrite/epidemiologia , Pontuação de Propensão
8.
Nat Commun ; 12(1): 1706, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33731712

RESUMO

Our incomplete understanding of osteoarthritis (OA) pathogenesis has significantly hindered the development of disease-modifying therapy. The functional relationship between subchondral bone (SB) and articular cartilage (AC) is unclear. Here, we found that the changes of SB architecture altered the distribution of mechanical stress on AC. Importantly, the latter is well aligned with the pattern of transforming growth factor beta (TGFß) activity in AC, which is essential in the regulation of AC homeostasis. Specifically, TGFß activity is concentrated in the areas of AC with high mechanical stress. A high level of TGFß disrupts the cartilage homeostasis and impairs the metabolic activity of chondrocytes. Mechanical stress stimulates talin-centered cytoskeletal reorganization and the consequent increase of cell contractile forces and cell stiffness of chondrocytes, which triggers αV integrin-mediated TGFß activation. Knockout of αV integrin in chondrocytes reversed the alteration of TGFß activation and subsequent metabolic abnormalities in AC and attenuated cartilage degeneration in an OA mouse model. Thus, SB structure determines the patterns of mechanical stress and the configuration of TGFß activation in AC, which subsequently regulates chondrocyte metabolism and AC homeostasis.


Assuntos
Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Estresse Mecânico , Fator de Crescimento Transformador beta/metabolismo , Animais , Osso e Ossos/patologia , Linhagem Celular , Condrócitos/metabolismo , Citoesqueleto/metabolismo , Homeostase , Humanos , Integrina alfaV/genética , Integrina alfaV/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoartrite/metabolismo , Osteoartrite/patologia , Transdução de Sinais , Talina/metabolismo
9.
Elife ; 102021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33646122

RESUMO

Osteoarthritis, a highly prevalent degenerative joint disorder, is characterized by joint pain and disability. Available treatments fail to modify osteoarthritis progression and decrease joint pain effectively. Here, we show that intermittent parathyroid hormone (iPTH) attenuates osteoarthritis pain by inhibiting subchondral sensory innervation, subchondral bone deterioration, and articular cartilage degeneration in a destabilized medial meniscus (DMM) mouse model. We found that subchondral sensory innervation for osteoarthritis pain was significantly decreased in PTH-treated DMM mice compared with vehicle-treated DMM mice. In parallel, deterioration of subchondral bone microarchitecture in DMM mice was attenuated by iPTH treatment. Increased level of prostaglandin E2 in subchondral bone of DMM mice was reduced by iPTH treatment. Furthermore, uncoupled subchondral bone remodeling caused by increased transforming growth factor ß signaling was regulated by PTH-induced endocytosis of the PTH type 1 receptor-transforming growth factor ß type 2 receptor complex. Notably, iPTH improved subchondral bone microarchitecture and decreased level of prostaglandin E2 and sensory innervation of subchondral bone in DMM mice by acting specifically through PTH type 1 receptor in Nestin+ mesenchymal stromal cells. Thus, iPTH could be a potential disease-modifying therapy for osteoarthritis.


Over time the cartilage between our bones gets worn down, and this can lead to a painful joint disorder known as osteoarthritis. Nearly 40 million people with osteoarthritis in the United States experience chronic pain. Although there are a number of drugs available for these patients, none of them provide sustained pain relief, and some have substantial side effects when ingested over a long period of time. Bone tissue is continuously broken down into minerals, such as calcium, that can be reabsorbed into the blood. In 2013, a group of researchers found that the tissue in the layer of bone below the cartilage ­ known as the subchondral bone ­ is reabsorbed and replaced incorrectly in patients with osteoarthritis. This irregular 'remodeling' stimulates nerve cells to grow into the subchondral layer, leading to increased sensitivity in the joint. A protein called parathyroid hormone, or PTH for short, plays an important role in the loss and formation of bone. A drug containing PTH is used to treat patients with another bone condition called osteoporosis, and could potentially work as a treatment for osteoarthritis pain. To investigate this, Sun et al. ­ including some of the researchers involved in the 2013 study ­ tested this drug on a mouse model that mimics the symptoms of osteoarthritis. This revealed that PTH significantly decreases the number of nerves present in the subchondral bone, which caused the mice to experience less pain. PTH also slowed down the progression of osteoarthritis, by preventing the cartilage on the subchondral layer from deteriorating as quickly. Sun et al. found that the subchondral bones of treated mice also had a more stable structure and reduced levels of a protein involved in the reabsorption of bone. The results suggest that PTH is able to correct the errors in bone remodeling caused by osteoarthritis, and that this drug could potentially alleviate patients' chronic pain. This drug has already been approved by the US Food and Drug Administration (FDA), and could be used in clinical trials to see if PTH has the same beneficial effects on patients with osteoarthritis.


Assuntos
Remodelação Óssea/efeitos dos fármacos , Osteoartrite/tratamento farmacológico , Dor/tratamento farmacológico , Hormônio Paratireóideo/farmacologia , Animais , Osso e Ossos/inervação , Osso e Ossos/patologia , Dinoprostona , Modelos Animais de Doenças , Membro Posterior , Masculino , Menisco/lesões , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Hormônio Paratireóideo/administração & dosagem
10.
Elife ; 92020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32441256

RESUMO

Pain is the most prominent symptom of osteoarthritis (OA) progression. However, the relationship between pain and OA progression remains largely unknown. Here we report osteoblast secret prostaglandin E2 (PGE2) during aberrant subchondral bone remodeling induces pain and OA progression in mice. Specific deletion of the major PGE2 producing enzyme cyclooxygenase 2 (COX2) in osteoblasts or PGE2 receptor EP4 in peripheral nerve markedly ameliorates OA symptoms. Mechanistically, PGE2 sensitizes dorsal root ganglia (DRG) neurons by modifying the voltage-gated sodium channel NaV1.8, evidenced by that genetically or pharmacologically inhibiting NaV1.8 in DRG neurons can substantially attenuate OA. Moreover, drugs targeting aberrant subchondral bone remodeling also attenuates OA through rebalancing PGE2 production and NaV1.8 modification. Thus, aberrant subchondral remodeling induced NaV1.8 neuronal modification is an important player in OA and is a potential therapeutic target in multiple skeletal degenerative diseases.


Many people will suffer from joint pain as they age, particularly in their knees. The most common cause of this pain is osteoarthritis, a disease that affects a tissue inside joints called cartilage. In a healthy knee, cartilage acts as a shock absorber. It cushions the ends of bones and enables them to move smoothly against one another. But in osteoarthritis, cartilage gradually wears away. As a result, the bones within a joint rub against each other whenever a person moves. This makes activities such as running or climbing stairs painful. But how does this pain arise? Previous work has implicated cells called osteoblasts. Osteoblasts are found in the area of the bone just below the cartilage. They produce new bone tissue throughout our lives, enabling our bones to regenerate and repair. Each time we move, forces acting on the knee joint activate osteoblasts. The cells respond by releasing a key molecule called PGE2, which is a factor in pain pathways. The joints of people with osteoarthritis produce too much PGE2. But exactly how this leads to increased pain sensation has been unclear. Zhu et al. now complete this story by working out how PGE2 triggers pain. Experiments in mice reveal that PGE2 irritates the nerve fibers that carry pain signals from the knee joint to the brain. It does this by activating a channel protein called Nav1.8, which allows sodium ions through the membranes of those nerve fibers. Zhu et al. show that, in a mouse model of osteoarthritis, Nav1.8 opens too widely in response to binding of PGE2, so the nerve cells become overactive and transmit a stronger pain sensation. This means that even small movements cause intense pain signals to travel from the joints to the brain. Building on their findings, Zhu et al. developed a drug that acts directly on bone to reduce PGE2 production, and show that this drug reduces pain in mice with osteoarthritis. At present, there are no treatments that reverse the damage that occurs during osteoarthritis, but further testing will determine whether this new drug could one day relieve joint pain in patients.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Osteoartrite/metabolismo , Osteoblastos/metabolismo , Animais , Remodelação Óssea , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Osteoartrite/genética , Osteoartrite/fisiopatologia , Ratos , Ratos Sprague-Dawley
11.
JCI Insight ; 5(8)2020 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-32208385

RESUMO

Increased subchondral bone angiogenesis with blood vessels breaching the tidemark into the avascular cartilage is a diagnostic feature of human osteoarthritis. However, the mechanisms that initiate subchondral bone angiogenesis remain unclear. We show that abnormally increased platelet-derived growth factor-BB (PDGF-BB) secretion by mononuclear preosteoclasts induces subchondral bone angiogenesis, contributing to osteoarthritis development. In mice after destabilization of the medial meniscus (DMM), aberrant joint subchondral bone angiogenesis developed during an early stage of osteoarthritis, before articular cartilage damage occurred. Mononuclear preosteoclasts in subchondral bone secrete excessive amounts of PDGF-BB, which activates platelet-derived growth factor receptor-ß (PDGFR-ß) signaling in pericytes for neo-vessel formation. Selective knockout of PDGF-BB in preosteoclasts attenuates subchondral bone angiogenesis and abrogates joint degeneration and subchondral innervation induced by DMM. Transgenic mice that express PDGF-BB in preosteoclasts recapitulate pathological subchondral bone angiogenesis and develop joint degeneration and subchondral innervation spontaneously. Our study provides the first evidence to our knowledge that PDGF-BB derived from preosteoclasts is a key driver of pathological subchondral bone angiogenesis during osteoarthritis development and offers a new avenue for developing early treatments for this disease.


Assuntos
Becaplermina/metabolismo , Neovascularização Patológica/patologia , Osteoartrite/patologia , Osteoclastos/metabolismo , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Osteoartrite/metabolismo , Osteoclastos/patologia
12.
J Clin Invest ; 130(7): 3483-3498, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32191640

RESUMO

The sensory nerve was recently identified as being involved in regulation of bone mass accrual. We previously discovered that prostaglandin E2 (PGE2) secreted by osteoblasts could activate sensory nerve EP4 receptor to promote bone formation by inhibiting sympathetic activity. However, the fundamental units of bone formation are active osteoblasts, which originate from mesenchymal stromal/stem cells (MSCs). Here, we found that after sensory denervation, knockout of the EP4 receptor in sensory nerves, or knockout of COX-2 in osteoblasts, could significantly promote adipogenesis and inhibit osteogenesis in adult mice. Furthermore, injection of SW033291 (a small molecule that locally increases the PGE2 level) or propranolol (a beta blocker) significantly promoted osteogenesis and inhibited adipogenesis. This effect of SW033291, but not propranolol, was abolished in conditional EP4-KO mice under normal conditions or in the bone repair process. We conclude that the PGE2/EP4 sensory nerve axis could regulate MSC differentiation in bone marrow of adult mice.


Assuntos
Adipogenia , Dinoprostona/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/genética , Técnicas de Inativação de Genes , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Knockout , Osteoblastos/metabolismo , Osteoblastos/patologia , Receptores de Prostaglandina E Subtipo EP4/genética , Células Receptoras Sensoriais/patologia
13.
J Bone Miner Res ; 35(6): 1188-1202, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32078184

RESUMO

In the growing skeleton, angiogenesis is intimately coupled with osteogenesis. Chronic, high doses of glucocorticoids (GCs) are associated with decreased bone vasculature and induce osteoporosis and growth failure. The mechanism of GC-suppression of angiogenesis and relationship to osteoporosis and growth retardation remains largely unknown. Type H vessels, which are regulated by preosteoclast (POC) platelet-derived growth factor-BB (PDGF-BB), are specifically coupled with bone formation and development. We determined the effect of GCs on POC synthesis of PDGF-BB in relation to type H vessel formation, bone mass, and bone growth in the distal femur of 2-week-old young mice receiving prednisolone or vehicle for 2, 4, or 6 weeks. After 2 weeks of prednisolone, the number of POCs were unchanged while POC synthesis of PDGF-BB was reduced. Longer treatment with prednisolone reduced POCs numbers and PDGF-BB. These changes were associated with a reduction in type H vessels, bone formation rate, bone mass, and bone length at each time point. In vitro, excessive concentrations of prednisolone (10-6 M) resulted in decreased PDGF-BB concentration and POC numbers. Conditioned medium from POC cultures treated with control concentration of prednisolone (10-7 M) or recombinant PDGF-BB stimulated endothelial tube formation, whereas conditioned medium from control concentration of prednisolone-treated POC cultures neutralized by PDGF-BB antibody or excessive prednisolone inhibited endothelial tube formation. Administration of excessive prednisolone attenuated the P65 subunit of nuclear factor kappa B (NF-κB) binding to the Pdgfb promoter, resulting in lower Pdgfb transcription. Co-treatment with excessive prednisolone and the glucocorticoid receptor (GR) antagonist (RU486), GR siRNA, or TNFα rescued NF-κB binding to the Pdgfb promoter and endothelial tube formation. These results indicate that PDGF-BB synthesis in POCs is suppressed by GCs through transrepression of GR/NF-κB, thus inhibiting type H vessel formation and associated osteoporosis and growth failure. © 2020 American Society for Bone and Mineral Research.


Assuntos
Glucocorticoides , Osteoporose , Proteínas Proto-Oncogênicas c-sis , Animais , Células Cultivadas , Glucocorticoides/farmacologia , Camundongos , NF-kappa B , Osteogênese , Proteínas Proto-Oncogênicas c-sis/genética
15.
Nat Commun ; 10(1): 5643, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31822662

RESUMO

Spinal pain is a major clinical problem, however, its origins and underlying mechanisms remain unclear. Here we report that in mice, osteoclasts induce sensory innervation in the porous endplates which contributes to spinal hypersensitivity in mice. Sensory innervation of the porous areas of sclerotic endplates in mice was confirmed. Lumbar spine instability (LSI), or aging, induces spinal hypersensitivity in mice. In these conditions, we show that there are elevated levels of PGE2 which activate sensory nerves, leading to sodium influx through Nav 1.8 channels. We show that knockout of PGE2 receptor 4 in sensory nerves significantly reduces spinal hypersensitivity. Inhibition of osteoclast formation by knockout Rankl in the osteocytes significantly inhibits LSI-induced porosity of endplates, sensory innervation, and spinal hypersensitivity. Knockout of Netrin-1 in osteoclasts abrogates sensory innervation into porous endplates and spinal hypersensitivity. These findings suggest that osteoclast-initiated porosity of endplates and sensory innervation are potential therapeutic targets for spinal pain.


Assuntos
Hipersensibilidade/patologia , Placa Motora/patologia , Netrina-1/metabolismo , Osteoclastos/metabolismo , Células Receptoras Sensoriais/metabolismo , Coluna Vertebral/patologia , Envelhecimento/patologia , Animais , Comportamento Animal , Dinoprostona , Modelos Animais de Doenças , Humanos , Hiperalgesia/patologia , Vértebras Lombares/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Netrina-1/deficiência , Dor/patologia , Porosidade , Transdução de Sinais
16.
Bone Res ; 7: 29, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31666999

RESUMO

Osteoarthritis (OA) causes the destruction of joints. Its pathogenesis is still under investigation, and there is no effective disease-modifying therapy. Here, we report that elevated cyclooxygenase-2 (COX-2) expression in the osteocytes of subchondral bone causes both spontaneous OA and rheumatoid arthritis (RA). The knockout of COX-2 in osteocytes or treatment with a COX-2 inhibitor effectively rescues the structure of subchondral bone and attenuates cartilage degeneration in spontaneous OA (STR/Ort) mice and tumor necrosis factor-α transgenic RA mice. Thus, elevated COX-2 expression in subchondral bone induces both OA-associated and RA-associated joint cartilage degeneration. The inhibition of COX-2 expression can potentially modify joint destruction in patients with arthritis.

17.
Nat Commun ; 10(1): 181, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30643142

RESUMO

Whether sensory nerve can sense bone density or metabolic activity to control bone homeostasis is unknown. Here we found prostaglandin E2 (PGE2) secreted by osteoblastic cells activates PGE2 receptor 4 (EP4) in sensory nerves to regulate bone formation by inhibiting sympathetic activity through the central nervous system. PGE2 secreted by osteoblasts increases when bone density decreases as demonstrated in osteoporotic animal models. Ablation of sensory nerves erodes the skeletal integrity. Specifically, knockout of the EP4 gene in the sensory nerves or cyclooxygenase-2 (COX2) in the osteoblastic cells significantly reduces bone volume in adult mice. Sympathetic tone is increased in sensory denervation models, and propranolol, a ß2-adrenergic antagonist, rescues bone loss. Furthermore, injection of SW033291, a small molecule to increase PGE2 level locally, significantly boostes bone formation, whereas the effect is obstructed in EP4 knockout mice. Thus, we show that PGE2 mediates sensory nerve to control bone homeostasis and promote regeneration.


Assuntos
Osso e Ossos/metabolismo , Dinoprostona/metabolismo , Osteoporose/patologia , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Células Receptoras Sensoriais/metabolismo , Fibras Adrenérgicas/efeitos dos fármacos , Fibras Adrenérgicas/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Animais , Densidade Óssea/efeitos dos fármacos , Regeneração Óssea/efeitos dos fármacos , Osso e Ossos/citologia , Osso e Ossos/inervação , Osso e Ossos/patologia , Células Cultivadas , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Retroalimentação Fisiológica , Feminino , Humanos , Hidroxiprostaglandina Desidrogenases/antagonistas & inibidores , Hidroxiprostaglandina Desidrogenases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoporose/etiologia , Propranolol/farmacologia , Piridinas/farmacologia , Receptores de Prostaglandina E Subtipo EP4/genética , Células Receptoras Sensoriais/efeitos dos fármacos , Tiofenos/farmacologia
18.
J Biomed Mater Res B Appl Biomater ; 107(2): 435-444, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29675920

RESUMO

Novel neural interfaces capable of reliably capturing electrical signals are crucial for the development of prostheses. Longitudinal intrafascicular electrodes (LIFEs) have been proposed as a promising technology, and their feasibility and biocompatibility need to be investigated for long-term implantation. In this study, custom-designed 95%Pt-5%Ir intrafascicular electrodes were implanted into the sciatic nerves of 14 rabbits using our novel direct microsurgical technique. The biocompatibility and their ability to record electrophysiological signals were serially investigated up to 9 months after implantation. Nerve tissues were examined using light and transmitted electron microscopy, and axon diameters were quantified, evaluated over time, and compared with sham-control (N = 4). Selective stimulation and stable recording properties of electrical signals were achieved by intrafascicular electrodes along the experimental period. While electrophysiological signal amplitude decreased by as early as 1 month after implantation (p < 0.05), the signal strength recovered to baseline levels by 3-5 months (p > 0.05). Axon diameter results showed a similar trend of initial decline (10.8% reduction, p < 0.01) followed by gradual recovery by 6 months (p > 0.05). Microstructural and ultrastructural analysis revealed modest tissue damage at the implantation site after implantation with gradual normalization over time. Intrafascicular electrodes implanted with direct microsurgical techniques demonstrated good biocompatibility and have great potential for long-term implantation and electrophysiological recordings. Though subtle tissue damage impaired ability to capture electrophysiological signals in the first 2 months, this damage gradually normalized after 3 months, and was fully normalized by 6 months. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 107B: 435-444, 2019.


Assuntos
Eletrodos Implantados , Teste de Materiais , Nervo Isquiático , Animais , Estudos de Viabilidade , Coelhos , Nervo Isquiático/metabolismo , Nervo Isquiático/cirurgia
19.
J Clin Invest ; 129(3): 1076-1093, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30530994

RESUMO

Joint pain is the defining symptom of osteoarthritis (OA) but its origin and mechanisms remain unclear. Here, we investigated an unprecedented role of osteoclast-initiated subchondral bone remodeling in sensory innervation for OA pain. We show that osteoclasts secrete netrin-1 to induce sensory nerve axonal growth in subchondral bone. Reduction of osteoclast formation by knockout of receptor activator of nuclear factor kappa-B ligand (Rankl) in osteocytes inhibited the growth of sensory nerves into subchondral bone, dorsal root ganglion neuron hyperexcitability, and behavioral measures of pain hypersensitivity in OA mice. Moreover, we demonstrated a possible role for netrin-1 secreted by osteoclasts during aberrant subchondral bone remodeling in inducing sensory innervation and OA pain through its receptor DCC (deleted in colorectal cancer). Importantly, knockout of Netrin1 in tartrate-resistant acid phosphatase-positive (TRAP-positive) osteoclasts or knockdown of Dcc reduces OA pain behavior. In particular, inhibition of osteoclast activity by alendronate modifies aberrant subchondral bone remodeling and reduces innervation and pain behavior at the early stage of OA. These results suggest that intervention of the axonal guidance molecules (e.g., netrin-1) derived from aberrant subchondral bone remodeling may have therapeutic potential for OA pain.


Assuntos
Gânglios Espinais/metabolismo , Netrina-1/metabolismo , Osteoartrite/metabolismo , Osteoclastos/metabolismo , Dor/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Remodelação Óssea/genética , Receptor DCC/genética , Receptor DCC/metabolismo , Gânglios Espinais/patologia , Masculino , Camundongos , Netrina-1/genética , Osteoartrite/genética , Osteoartrite/patologia , Osteoclastos/patologia , Dor/genética , Dor/patologia , Células Receptoras Sensoriais/patologia
20.
Ann N Y Acad Sci ; 1433(1): 29-40, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30091466

RESUMO

Camurati-Engelmann disease (CED) is a genetic bone-modeling disorder mainly caused by mutations in the gene that encodes transforming growth factor-ß1 (TGF-ß1). Symptoms of CED include bone pain, fractures, and dysplasia. Currently, effective therapies for bone fracture and dysplasia in CED are urgently needed. We have demonstrated that TGF-ß1 is a coupling factor for bone remodeling and is aberrantly activated in CED. Daily injection of TGF-ß type 1 receptor inhibitor (TßR1I) attenuated CED symptoms, but this systemic administration caused serious side effects. In this study, we created a conjugate linking TßR1I and alendronate, which delivered TßR1I specifically to bone. After weekly injection of the conjugate for 8 weeks, normal bone morphology and remodeling in CED mice was maintained with a minimum effective dose 700 times lower than TßR1I injection. Additionally, we found that the conjugate restored normal bone turnover by reducing the number of osteoblasts and osteoclasts, maintained a regular osteogenic microenvironment by regulating the formation of CD31 and Endomucin double-positive vessels, and preserved ordinary bone formation via inhibition of the migration of leptin-receptor-positive cells. Thus, targeting delivery of TßR1I to bone is a promising therapy for CED and other uncoupled bone remodeling disorders.


Assuntos
Remodelação Óssea/efeitos dos fármacos , Síndrome de Camurati-Engelmann/tratamento farmacológico , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Alendronato/administração & dosagem , Alendronato/química , Animais , Remodelação Óssea/genética , Síndrome de Camurati-Engelmann/genética , Síndrome de Camurati-Engelmann/patologia , Células Cultivadas , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Osteogênese/efeitos dos fármacos , Fator de Crescimento Transformador beta1/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...