Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
BMJ Open ; 13(12): e075502, 2023 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-38110382

RESUMO

INTRODUCTION: Exaggerated inflammatory response is one of the main mechanisms underlying heterotopic ossification (HO). It has been suggested that the antifibrinolytic drug tranexamic acid (TXA) can exert a significant anti-inflammatory effect during orthopaedic surgery. However, no prospective studies have yet investigated the effects of TXA on HO recurrence in patients following open elbow arthrolysis (OEA). METHODS AND ANALYSIS: Here, we present a protocol for a single-centre, randomised, double-blind, placebo-controlled trial to investigate the effectiveness of TXA on HO recurrence after OEA in a single hospital. A minimum sample size of 138 eligible and consenting participants randomised into treatment and control groups in a 1:1 manner will be included. Patients will receive 2 g of intravenous TXA (experimental group) or placebo (normal saline, control group) administered before skin incision. The primary outcome is HO recurrence rate within 12 months after surgery. The secondary outcomes are the serum immune-inflammatory cytokines including erythrocyte sedimentation rate, C reactive protein, interleukin (IL)-6, IL-1ß, IL-13 at the first and third day postoperatively, and elbow range of motion and functional score at 1.5, 6, 9 and 12 months after surgery. After completion of the trial, the results will be reported in accordance with the extensions of the Consolidated Standards of Reporting Trials Statement for trials. The results of this study should determine whether TXA can reduce the rates of HO occurrence after OEA. ETHICS AND DISSEMINATION: Ethical approval has been granted by the Medical Ethics Committee of the Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (reference number 2022-123-(1)). The results of this study will be disseminated through presentations at academic conferences and publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER: ChiCTR2300068106.


Assuntos
Antifibrinolíticos , Artropatias , Procedimentos Ortopédicos , Ossificação Heterotópica , Ácido Tranexâmico , Humanos , Ácido Tranexâmico/uso terapêutico , Cotovelo/cirurgia , China , Antifibrinolíticos/uso terapêutico , Método Duplo-Cego , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/etiologia , Ossificação Heterotópica/cirurgia , Resultado do Tratamento , Ensaios Clínicos Controlados Aleatórios como Assunto
2.
Semin Arthritis Rheum ; 63: 152306, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37976811

RESUMO

INTRODUCTION: Ectopic calcifications (ECs) and heterotopic ossifications (HOs) form in non-mineralized tissues, most often in subcutaneous and muscular areas. Local and systemic complications can cause severe disability. Systemic administration of sodium thiosulfate (STS) gives promising results but is difficult to use in clinical practice. OBJECTIVE: Evaluation of the efficacy and safety of topical STS in ECs and HOs. METHODS: Retrospective analysis of the CATSS-O registry that included patients receiving topical STS 25 % prepared by the pharmacy of Limoges hospital during 2014-2020. The efficacy of STS was assessed by imaging (radiography or CT) after at least 6 months' treatment. RESULTS: Among 126 patients who received STS 25 %, 35 had complete clinical and radiographic data for analysis (28 with ECs and 7 with HOs; 18 children [mean age 8.9 years, range 1.5-16], 17 adults [mean age 52.4 years, range 24-90]). Calcifications or ossifications were due to dermatomyositis (8 children, 6 adults), systemic scleroderma (6 adults) or pseudo-hypoparathyroidism 1A (7 children). They were single (37.1 %) or multiple (62.9 %). Treated regions were in the lower limbs (31.4 %), upper limbs (37.1 %) or both (28.6 %) and the axial region (2.9 %). Topical STS was clinically effective in 9/28 (32.1 %) patients with ECs and 2/7 (28.6 %) children with HOs. Three patients experienced complete disappearance of their calcifications. Response for ECs was better in children than adults (54.5% vs 17.6 %, p = 0.035). Topical STS was well tolerated. CONCLUSION: Local STS seems effective for ossifications, particularly pediatric calcifications or ossifications. Randomized and experimental studies are needed to confirm this observation and to identify the underlying mechanisms.


Assuntos
Calcinose , Ossificação Heterotópica , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Humanos , Lactente , Pessoa de Meia-Idade , Adulto Jovem , Calcinose/diagnóstico por imagem , Calcinose/tratamento farmacológico , Calcinose/etiologia , Ossificação Heterotópica/diagnóstico por imagem , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/complicações , Osteogênese , Estudos Retrospectivos
3.
Inflammation ; 46(4): 1414-1429, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37115368

RESUMO

Heterotopic ossification (HO) is a pathological condition that occurs in soft tissues following severe trauma. The exact pathogenesis of HO remains unclear. Studies have shown that inflammation predisposes patients to the development of HO and triggers ectopic bone formation. Macrophages are crucial mediators of inflammation and are involved in HO development. The present study investigated the inhibitory effect and underlying mechanism of metformin on macrophage infiltration and traumatic HO in mice. Our results found that abundant levels of macrophages were recruited to the injury site during early HO progression and that early administration of metformin prevented traumatic HO in mice. Furthermore, we found that metformin attenuated macrophage infiltration and the NF-κB signaling pathway in injured tissue. The monocyte-to-macrophage transition in vitro was suppressed by metformin and this event was mediated by AMPK. Finally, we showed that inflammatory mediator's regulation by macrophages targeted preosteoblasts, leading to elevated BMP signaling, and osteogenic differentiation and driving HO formation, and this effect was blocked after the activation of AMPK in macrophages. Collectively, our study suggests that metformin prevents traumatic HO by inhibiting of NF-κB signaling in macrophages and subsequently attenuating BMP signaling and osteogenic differentiation in preosteoblasts. Therefore, metformin may serve as a therapeutic drug for traumatic HO by targeting NF-κB signaling in macrophages.


Assuntos
Metformina , Ossificação Heterotópica , Camundongos , Animais , NF-kappa B/metabolismo , Metformina/farmacologia , Metformina/uso terapêutico , Proteínas Quinases Ativadas por AMP/metabolismo , Osteogênese , Macrófagos/metabolismo , Transdução de Sinais , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/etiologia , Ossificação Heterotópica/prevenção & controle
4.
Adv Sci (Weinh) ; 10(15): e2207224, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36970815

RESUMO

Heterotopic ossification (HO) is a double-edged sword. Pathological HO presents as an undesired clinical complication, whereas controlled heterotopic bone formation by synthetic osteoinductive materials shows promising therapeutic potentials for bone regeneration. However, the mechanism of material-induced heterotopic bone formation remains largely unknown. Early acquired HO being usually accompanied by severe tissue hypoxia prompts the hypothesis that hypoxia caused by the implantation coordinates serial cellular events and ultimately induces heterotopic bone formation in osteoinductive materials. The data presented herein shows a link between hypoxia, macrophage polarization to M2, osteoclastogenesis, and material-induced bone formation. Hypoxia inducible factor-1α (HIF-1α), a crucial mediator of cellular responses to hypoxia, is highly expressed in an osteoinductive calcium phosphate ceramic (CaP) during the early phase of implantation, while pharmacological inhibition of HIF-1α significantly inhibits M2 macrophage, subsequent osteoclast, and material-induced bone formation. Similarly, in vitro, hypoxia enhances M2 macrophage and osteoclast formation. Osteoclast-conditioned medium enhances osteogenic differentiation of mesenchymal stem cells, such enhancement disappears with the presence of HIF-1α inhibitor. Furthermore, metabolomics analysis reveals that hypoxia enhances osteoclastogenesis via the axis of M2/lipid-loaded macrophages. The current findings shed new light on the mechanism of HO and favor the design of more potent osteoinductive materials for bone regeneration.


Assuntos
Substitutos Ósseos , Ossificação Heterotópica , Humanos , Osteogênese , Substitutos Ósseos/uso terapêutico , Macrófagos , Hipóxia , Ossificação Heterotópica/tratamento farmacológico , Lipídeos/uso terapêutico
5.
J Shoulder Elbow Surg ; 31(10): 2157-2163, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35872167

RESUMO

BACKGROUND: The aim of this study was to assess the efficacy of 3 weeks of indomethacin, a nonselective nonsteroidal anti-inflammatory drug, in comparison to 1 week of meloxicam as prophylaxis for heterotopic ossifications (HOs) after distal biceps tendon repair. METHODS: A single-center retrospective study was performed on 78 patients undergoing distal biceps tendon repair between 2008 and 2019. From 2008 to 2016, patients received meloxicam 15 mg daily for the period of 1 week as usual care. From 2016 onward, the standard protocol was changed to indomethacin 25 mg 3 times daily for 3 weeks. All patients underwent a single-incision repair with a cortical button technique. The postoperative rehabilitation protocol was similar for all patients. The postoperative radiographs at 8-week follow-up were assessed blindly by 7 independent assessors. If HOs were present, it was classified according to the Ilahi-Gabel classification for size and according to the Gärtner-Heyer classification for density. Statistical analysis was performed to analyze the difference in HO between the patients who were treated with indomethacin and with meloxicam. RESULTS: Seventy-eight patients, with a mean age of 48.8 years (range 30-72) were included. The mean follow-up after surgery was 12 months (range 2-45). Indomethacin (21 days, 25 mg 3 times per day) was prescribed to 26 (33%) patients. The 52 other patients (67%) were prescribed meloxicam 15 mg daily for 7 days. HOs were seen in 19 patients 8 weeks postoperatively. Five of 26 patients treated with indomethacin developed HO, and 14 of 52 patients treated with meloxicam developed HO (P = .5). Two patients had symptomatic HO with minor restrictions in movement; neither patient was treated with indomethacin. Significantly more HOs were seen in patients with a longer time from injury to surgery (P = .01) The intraclass correlation score for reliability between assessors for HO scoring on postoperative radiographs was good to excellent for both classifications. CONCLUSION: In this study, HOs were seen in 24% of postoperative radiographs. Three weeks of indomethacin was not superior to meloxicam for 1 week for the prevention of HO after single-incision distal biceps tendon repair.


Assuntos
Ossificação Heterotópica , Traumatismos dos Tendões , Adulto , Idoso , Anti-Inflamatórios não Esteroides/uso terapêutico , Humanos , Indometacina/uso terapêutico , Meloxicam/uso terapêutico , Pessoa de Meia-Idade , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/etiologia , Ossificação Heterotópica/prevenção & controle , Reprodutibilidade dos Testes , Estudos Retrospectivos , Ruptura/cirurgia , Traumatismos dos Tendões/cirurgia , Tendões
6.
Eur J Pharmacol ; 928: 175109, 2022 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-35738451

RESUMO

Heterotopic ossification (HO) is the formation of bony tissues in the extraskeletal system. To date, no effective therapy has been developed for the treatment of HO, although increasing evidences have shown that inhibition of TGF-ß signaling has potential as a new therapeutic approach for attenuating HO progression. Results from previous clinical trials have demonstrated that patients with malignant tumors exhibit excellent tolerability to Galunisertib, a TGF-ß receptor I kinase inhibitor. However, its therapeutic potential in preventing HO and inhibitory effect on osteogenesis remain unclear. In this study, we demonstrated that intragastrical administration of Galunisertib, at a concentration as low as 10 mg/kg, was not only fairly effective in preventing HO development in a dose-dependent manner, but also generated a non-toxic response in a novel Achilles tendon puncture-induced traumatic HO model in mice. Moreover, Galunisertib treatment in the early phases of HO development, including the inflammatory and chondrogenic period, resulted in better therapeutic effects instead of eliminating already formed bony tissues. Mechanistically, Galunisertib suppressed the osteogenic differentiation capacity of tendon-derived stem cells (TDSCs) by interfering with the Smad2/3 signaling pathway, blocking the phosphorylation of Smad2/3 translocated from cytoplasm into the nucleus to regulate the expression of both osteogenesis-related transcription factors and related proteins. Results from in vivo experiments further validated Galunisertib's effect on HO attenuation, by intercepting the TGF-ß/Smad2/3 signaling pathway. In conclusion, our findings demonstrated Galunisertib's potential as a prophylactic drug for the treatment of traumatic HO or other related diseases triggered by over-expressed TGF-ß.


Assuntos
Ossificação Heterotópica , Osteogênese , Animais , Camundongos , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/patologia , Pirazóis , Quinolinas , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/farmacologia
7.
J Oral Maxillofac Surg ; 80(5): 869-888, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35032438

RESUMO

PURPOSE: The purpose of this study was to create a treatment protocol for cases of heterotopic ossification (HO) of the temporomandibular joint (TMJ), particularly those refractory to current TMJ HO protocols. In addition, we demonstrate the success of this protocol on a unique case of recurrent HO that failed multiple TMJ HO protocols in the setting of an improvised explosive device (IED) blast in a wounded warrior. METHODS: An electronic literature review was conducted via PubMed and Web of Science. Twenty-five studies were identified to provide supporting evidence for a proposed, up-to-date protocol for the treatment of refractory TMJ HO. The authors present a case report of a wounded warrior with HO ankylosis of bilateral TMJs in the setting of IED blast and demonstrate successful use of our surgical and pharmacotherapeutic protocol. RESULTS: Based on the literature review, our proposed protocol consists of pharmacotherapy with celecoxib and etidronate, with weekly forced dilation (brisement) and home physical therapy with the TheraBite Jaw Motion Rehab System. Surgically, the TMJ should be treated with two-stage reconstruction using initial polymethyl methacrylate spacers and subsequent total joint replacement with custom prostheses, fat grafting, and 3-dimensional-navigated total resection of HO. This protocol was successfully utilized in our patient's refractory HO ankylosed TMJ secondary to IED blast, and the patient's maximal incisal opening was regained and has remained stable 2 years after surgery without recurrent HO. CONCLUSIONS: Our method for treatment in this case deviated from the standard TMJ Concepts HO protocol in that it included multimodal pharmacotherapy with celecoxib and etidronate. Based on our literature review and experience, we advise that clinicians utilize our protocol for the management of all craniofacial HO cases, particularly in cases of recurrent HO that fail conventional therapies and/or involving high-order blast trauma.


Assuntos
Ossificação Heterotópica , Transtornos da Articulação Temporomandibular , Celecoxib/uso terapêutico , Terapia Combinada , Ácido Etidrônico/uso terapêutico , Humanos , Prótese Articular , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/cirurgia , Transtornos da Articulação Temporomandibular/tratamento farmacológico , Transtornos da Articulação Temporomandibular/cirurgia
8.
Biochem Biophys Res Commun ; 573: 171-178, 2021 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-34419763

RESUMO

Rapamycin (RAPA), which was first described as an anti-fungal agent, is a potent immunosuppressant that suppresses tumors and inhibits the mTOR signaling pathway. Heterotopic ossification (HO) is abnormal bone formation outside the skeletal system (e.g., in muscles, tendons, articular capsules and other soft tissues), often due to trauma or injury. There are currently no drugs available to treat traumatic HO, largely due to limited understanding of the disease. In this study, we focused on the role of oxidative stress (OS) in the early stage of traumatic HO, and explored the underlying mechanism of traumatic HO by using RAPA to specifically inhibit the mTOR pathway, which is known to play a role in the pathogenesis of HO. To assess the effects of RAPA in traumatic HO, we used an NSE-BMP4 transgenic mouse model that develops ossification in response to traumatic injury and intramuscular injection of cardiotoxin to initiate injury. These mice were then treated with RAPA or vehicle intraperitoneally every other day for 2 weeks. Our results demonstrate that RAPA can inhibit HO through a number of different mechanisms. We show that OS and a strong inflammatory response contribute to the hypoxia associated with the early stages of HO, and that RAPA inhibits these responses. Furthermore, RAPA reduces the vascularization triggered by mTOR signaling that leads to HO formation. Therefore, we believe that RAPA could be an effective treatment for the early stages of HO.


Assuntos
Imunossupressores/farmacologia , Ossificação Heterotópica/tratamento farmacológico , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/patologia , Estresse Oxidativo/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
9.
Front Immunol ; 12: 649285, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093537

RESUMO

Heterotopic ossification (HO) is one of the most intractable disorders following musculoskeletal injury and is characterized by the ectopic presence of bone tissue in the soft tissue leading to severe loss of function in the extremities. Recent studies have indicated that immune cell infiltration and inflammation are involved in aberrant bone formation. In this study, we found increased monocyte/macrophage and mast cell accumulation during early HO progression. Macrophage depletion by clodronate liposomes and mast cell stabilization by cromolyn sodium significantly impeded HO formation. Therefore, we proposed that the dietary phytochemical quercetin could also suppress immune cell recruitment and related inflammatory responses to prevent HO. As expected, quercetin inhibited the monocyte-to-macrophage transition, macrophage polarization, and mast cell activation in vitro in a dose-dependent manner. Using a murine burn/tenotomy model, we also demonstrated that quercetin attenuated inflammatory responses and HO in vivo. Furthermore, elevated SIRT1 and decreased acetylated NFκB p65 expression were responsible for the mechanism of quercetin, and the beneficial effects of quercetin were reversed by the SIRT1 antagonist EX527 and mimicked by the SIRT agonist SRT1720. The findings in this study suggest that targeting monocyte/macrophage and mast cell activities may represent an attractive approach for therapeutic intervention of HO and that quercetin may serve as a promising therapeutic candidate for the treatment of trauma-induced HO by modulating SIRT1/NFκB signaling.


Assuntos
Queimaduras/complicações , Ossificação Heterotópica/tratamento farmacológico , Quercetina/administração & dosagem , Traumatismos dos Tendões/complicações , Animais , Queimaduras/imunologia , Carbazóis/administração & dosagem , Células Cultivadas , Modelos Animais de Doenças , Compostos Heterocíclicos de 4 ou mais Anéis/administração & dosagem , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , Ossificação Heterotópica/diagnóstico , Ossificação Heterotópica/imunologia , Ossificação Heterotópica/patologia , Cultura Primária de Células , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/metabolismo , Células THP-1 , Traumatismos dos Tendões/imunologia , Tendões/patologia , Tenotomia/efeitos adversos , Fator de Transcrição RelA/metabolismo , Microtomografia por Raio-X
10.
J Pharmacol Sci ; 145(1): 23-28, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33357776

RESUMO

Ossification of the posterior longitudinal ligament (OPLL) within the spinal canal sometimes leads to severe myelopathy. Teriparatide (TPD) is a recombinant human parathyroid hormone (PTH) (1-34), which promotes osteogenesis of mesenchymal stem cells (MSCs) via PTH 1 receptor (PTH1R). Although ligamentum flavum (LF)-MSCs from patients with OPLL have a high osteogenic potency, the effect of TPD on them remains unknown. In this study, we determined PTH1R expression in LF-MSCs from patients with OPLL and investigated whether TPD promotes osteogenic differentiation in them. First, LF-MSCs were isolated from patients with OPLL and cervical spondylotic myelopathy (CSM) (controls). Cultured LF-MSCs were treated with different concentrations of TPD on days 0, 7, and 14. On day 21, osteogenic gene expression was quantified. Mineralization was measured based on optical density after Alizarin Red S staining. LF-MSCs from both groups expressed PTH1R at the same level. TPD did not enhance osteogenic gene expression and mineralization in LF-MSCs from both groups. TPD did not promote the osteogenic differentiation of LF-MSCs from patients with OPLL. Thus, it may be safe for patients with OPLL. However, further confirmation of our results with in vivo studies is necessary.


Assuntos
Expressão Gênica/efeitos dos fármacos , Ligamento Amarelo/citologia , Ligamentos Longitudinais/patologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Ossificação Heterotópica/patologia , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Receptor Tipo 1 de Hormônio Paratireóideo/genética , Teriparatida/farmacologia , Idoso , Calcificação Fisiológica/efeitos dos fármacos , Calcificação Fisiológica/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ossificação Heterotópica/tratamento farmacológico , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Teriparatida/uso terapêutico
11.
Bone ; 139: 115517, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32622875

RESUMO

Heterotopic ossification (HO) is defined as ectopic bone formation around joints and in soft tissues following trauma, particularly blast-related extremity injuries, thermal injuries, central nerve injuries, or orthopaedic surgeries, leading to increased pain and diminished quality of life. Current treatment options include pharmacotherapy with non-steroidal anti-inflammatory drugs, radiotherapy, and surgical excision, but these treatments have limited efficacy and have associated complication profiles. In contrast, small molecule inhibitors have been shown to have higher specificity and less systemic cytotoxicity. Previous studies have shown that bone morphogenetic protein (BMP) signaling and downstream non-canonical (SMAD-independent) BMP signaling mediated induction of TGF-ß activated kinase-1 (TAK1) contributes to HO. In the current study, small molecule inhibition of TAK1, NG-25, was evaluated for its efficacy in limiting ectopic bone formation following a rat blast-associated lower limb trauma and a murine burn tenotomy injury model. A significant decrease in total HO volume in the rat blast injury model was observed by microCT imaging with no systemic complications following NG-25 therapy. Furthermore, tissue-resident mesenchymal progenitor cells (MPCs) harvested from rats treated with NG-25 demonstrated decreased proliferation, limited osteogenic differentiation capacity, and reduced gene expression of Tac1, Col10a1, Ibsp, Smad3, and Sox2 (P < 0.05). Single cell RNA-sequencing of murine cells harvested from the injury site in a burn tenotomy injury model showed increased expression of these genes in MPCs during stages of chondrogenic differentiation. Additional in vitro cell cultures of murine tissue-resident MPCs and osteochondrogenic progenitors (OCPs) treated with NG-25 demonstrated reduced chondrogenic differentiation by 10.2-fold (P < 0.001) and 133.3-fold (P < 0.001), respectively, as well as associated reduction in chondrogenic gene expression. Induction of HO in Tak1 knockout mice demonstrated a 7.1-fold (P < 0.001) and 2.7-fold reduction (P < 0.001) in chondrogenic differentiation of murine MPCs and OCPs, respectively, with reduced chondrogenic gene expression. Together, our in vivo models and in vitro cell culture studies demonstrate the importance of TAK1 signaling in chondrogenic differentiation and HO formation and suggest that small molecule inhibition of TAK1 is a promising therapy to limit the formation and progression of HO.


Assuntos
Ossificação Heterotópica , Osteogênese , Animais , Condrogênese , Extremidade Inferior , Camundongos , Ossificação Heterotópica/tratamento farmacológico , Qualidade de Vida , Ratos
14.
Rev. chil. reumatol ; 36(4): 125-133, 2020. ilus, tab
Artigo em Espanhol | LILACS | ID: biblio-1282557

RESUMO

Introducción: La Fibrodisplasia osificante progresiva es una enfermedad congénita autosómica dominante poco frecuente, caracterizada por malformaciones esqueléticas y osificación heterotópica progresiva e invalidante. Caso clínico: Niño de 11 años consulta por múltiples lesiones osificadas en tronco y región cervical con importante limitación en su movilidad. En el examen físico destaca un ortejo mayor corto. Estudio genético muestra mutación del gen ACVR1. Recibe tratamiento con periodos cortos de corticosteroides posterior a traumas y previo a procedimientos, asociado a un manejo multidisciplinario. Revisión de la literatura: A la fecha el principal tratamiento es la prevención de los brotes de osificación y el uso de corticosteroides o antiinflamatorios cuando los brotes ya se iniciaron. Están en curso ensayos clínicos con bifosfonatos y anticuerpos anti-activina A. Conclusión: En la actualidad no existe un tratamiento específico, sin embargo, un diagnóstico precoz, la prevención de brotes y nuevas terapias podrían mejorar el pronóstico de los pacientes.


ntroduction: Fibrodysplasia ossificans progressiva is a rare autosomal dominant congenital disease characterized by skeletal malformations and progressive disabling heterotopic ossification. Clinical case: An 11-year-old boy consulted with multiple ossified lesions in the trunk and cervical regions associated with significant limitation in mobility. On physical examination, the big toe is short. Genetic study shows ACVR1 gene mutation. He received treatment with short corticosteroid periods after traumas and prior to clinical procedures, as well as a multidisciplinary management.Literature review: To date the main treatment is the prevention of ossification flare-ups and the use of corticosteroids or anti-inflammatories when they have already started. Clinical trials are ongoing with bisphosphonates and anti-activin A antibodies.Conclusion: There is currently no specific treatment, however, early diagnosis, prevention of flare-ups and new therapies could improve the prognosis of patients.


Assuntos
Humanos , Masculino , Criança , Ossificação Heterotópica/tratamento farmacológico , Miosite Ossificante/diagnóstico , Miosite Ossificante/terapia , Ossificação Heterotópica/diagnóstico , Corticosteroides/uso terapêutico
15.
Curr Osteoporos Rep ; 17(6): 387-394, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31721068

RESUMO

PURPOSE OF REVIEW: Heterotopic ossification (HO) is associated with inflammation. The goal of this review is to examine recent findings on the roles of inflammation and the immune system in HO. We examine how inflammation changes in fibrodysplasia ossificans progressiva, in traumatic HO, and in other clinical conditions of HO. We also discuss how inflammation may be a target for treating HO. RECENT FINDINGS: Both genetic and acquired forms of HO show similarities in their inflammatory cell types and signaling pathways. These include macrophages, mast cells, and adaptive immune cells, along with hypoxia signaling pathways, mesenchymal stem cell differentiation signaling pathways, vascular signaling pathways, and inflammatory cytokines. Because there are common inflammatory mediators across various types of HO, these mediators may serve as common targets for blocking HO. Future research may focus on identifying new inflammatory targets and testing combinatorial therapies based on these results.


Assuntos
Inflamação/imunologia , Miosite Ossificante/imunologia , Ossificação Heterotópica/imunologia , Ferimentos e Lesões/imunologia , Imunidade Adaptativa/imunologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Artroplastia de Quadril , Traumatismos por Explosões/imunologia , Lesões Encefálicas Traumáticas/imunologia , Queimaduras/imunologia , Diferenciação Celular/imunologia , Citocinas/imunologia , Humanos , Hipóxia/imunologia , Imunossupressores/uso terapêutico , Inflamação/tratamento farmacológico , Inibidores de Janus Quinases/uso terapêutico , Macrófagos/imunologia , Mastócitos/imunologia , Células-Tronco Mesenquimais , Miosite Ossificante/tratamento farmacológico , Ossificação Heterotópica/tratamento farmacológico , Complicações Pós-Operatórias/imunologia , Pirazóis/uso terapêutico , Receptores do Ácido Retinoico/agonistas , Transdução de Sinais , Sirolimo/uso terapêutico , Traumatismos da Medula Espinal/imunologia , Estilbenos/uso terapêutico , Receptor gama de Ácido Retinoico
16.
J Shoulder Elbow Surg ; 28(12): 2379-2385, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31757369

RESUMO

BACKGROUND AND HYPOTHESIS: Heterotopic ossification (HO) is a recognized sequela after trauma and arthroplasty. The purpose of this study was to evaluate the therapeutic effect of celecoxib on HO. We hypothesized that celecoxib may inhibit the progression of initiated HO. METHODS: We performed a retrospective review of 37 patients who underwent elbow joint surgery between January 2014 and June 2018. Seventeen patients were prescribed orally administered celecoxib (200 mg/dose, twice daily) for 2 months after the diagnosis of HO, whereas the remaining 20 patients were administered celecoxib for 1 month starting immediately after surgery. HO progression was evaluated by plain radiographs. By use of an Achilles tendon puncture-induced HO mouse model, the curative effect of celecoxib was illustrated at different HO progression stages. The mice were assigned to 1 of 4 groups: sham group, vehicle group, group receiving celecoxib on day 1, and group receiving celecoxib in week 6. Achilles tendons were analyzed by micro-computed tomography and histochemistry after 12 weeks. RESULTS: Celecoxib did not inhibit the progression of initiated HO in the patients in whom HO was diagnosed, whereas those who received celecoxib after surgery had lower morbidity. Achilles tendon puncture effectively induced typical HO in mice. The ectopic bone volume was significantly reduced in the day 1 celecoxib group compared with the vehicle group; however, the difference was not statistically significant in the week 6 celecoxib group. CONCLUSIONS: Administration of celecoxib starting immediately after surgery can significantly inhibit the formation of HO. Once HO is visible on plain radiographs or micro-computed tomography, celecoxib cannot effectively attenuate further progression of HO in humans and mice.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Celecoxib/uso terapêutico , Progressão da Doença , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/prevenção & controle , Complicações Pós-Operatórias/tratamento farmacológico , Tendão do Calcâneo/diagnóstico por imagem , Tendão do Calcâneo/lesões , Tendão do Calcâneo/patologia , Adulto , Idoso , Animais , Modelos Animais de Doenças , Articulação do Cotovelo/diagnóstico por imagem , Articulação do Cotovelo/cirurgia , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Ossificação Heterotópica/diagnóstico por imagem , Ossificação Heterotópica/etiologia , Complicações Pós-Operatórias/diagnóstico por imagem , Complicações Pós-Operatórias/etiologia , Estudos Retrospectivos , Microtomografia por Raio-X , Adulto Jovem
17.
Med Sci Monit ; 25: 7872-7881, 2019 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-31631887

RESUMO

BACKGROUND Heterotopic ossification (HO) is a kind of abnormal mineralized bone which usually occurs in muscle, tendon, or ligament. There are currently no effective drugs for the treatment and prevention of HO. Developing effective drugs that can inhibit HO is of profound significance and would provide new strategies for clinical treatment of this disease. The present investigation evaluated the inhibitory effect of tamoxifen against HO. MATERIAL AND METHODS Using an Achilles tendon trauma-induced HO female mice model, we screened different doses of tamoxifen (1, 3, and 9 mg/kg) in mice to determine the optimal dosage on the inhibition of the HO formation. The curative effect of tamoxifen was also illustrated at different HO progression stages including inflammation, chondrogenesis, osteogenesis, and HO maturation. RESULTS Heterotopic bone was formed with typical endochondral ossification in Achilles tendons 6 weeks after surgery and continued to enlarge up to 12 weeks. The formation of HO was significantly inhibited with the treatment of tamoxifen at the dosage of 9 mg/kg, whereas 1 mg/kg and 3 mg/kg did not reduce HO bone volume remarkably. The progression of HO was both attenuated by tamoxifen from Day 1 and Week 4 post-surgery, whereas no inhibitory effect was shown at the osteogenesis and maturation stages treated with tamoxifen. CONCLUSIONS Tamoxifen exerts an inhibitory effect on the heterotopic bone progression at inflammation and chondrogenesis stages, with the TGF-ß signaling pathway suppressed following the increase in estrogen receptor alpha activity.


Assuntos
Tendão do Calcâneo/efeitos dos fármacos , Ossificação Heterotópica/tratamento farmacológico , Tamoxifeno/farmacologia , Animais , Osso e Ossos/metabolismo , China , Condrogênese/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Músculos/metabolismo , Ossificação Heterotópica/metabolismo , Osteogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/metabolismo , Traumatismos dos Tendões/tratamento farmacológico
19.
Mol Med Rep ; 20(4): 2979-2989, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31432174

RESUMO

Heterotopic ossification (HO) refers to the appearance of osteoblasts in soft tissues under pathological conditions, such as trauma or infection. HO arises in an unpredictable way without any recognizable initiation. Activin receptor­like kinase­2 (ALK2) is a type I cell surface receptor for bone morphogenetic proteins (BMPs). The dysregulation of ALK2 signaling is associated with a variety of diseases, including cancer and HO. At present, the prevention and treatment of HO in the clinic predominantly includes nonsteroidal anti­inflammatory drugs (NSAIDs), bisphosphonates and other drug treatments, low­dose local radiation therapy and surgical resection, rehabilitation treatment and physical therapy. However, most of these therapies have adverse effects. These methods do not prevent the occurrence of HO. The pathogenesis of HO is not being specifically targeted; the current treatment strategies target the symptoms, not the disease. These treatments also cannot solve the fundamental problem of the occurrence of HO. Therefore, scholars have been working to develop targeted therapies based on the pathogenesis of HO. The present review focuses on advances in the understanding of the underlying mechanisms of HO, and possible options for the prevention and treatment of HO. In addition, the role of ALK2 in the process of HO is introduced and the progress made towards the targeted inhibition of ALK2 is discussed. The present study aims to offer a platform for further research on possible targets for the prevention and treatment of HO.


Assuntos
Receptores de Ativinas Tipo I , Sistemas de Liberação de Medicamentos , Ossificação Heterotópica , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais , Receptores de Ativinas Tipo I/antagonistas & inibidores , Receptores de Ativinas Tipo I/metabolismo , Humanos , Ossificação Heterotópica/tratamento farmacológico , Ossificação Heterotópica/enzimologia
20.
Front Immunol ; 10: 377, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30899259

RESUMO

Neurogenic heterotopic ossifications (NHO) are very incapacitating complications of traumatic brain and spinal cord injuries (SCI) which manifest as abnormal formation of bone tissue in periarticular muscles. NHO are debilitating as they cause pain, partial or total joint ankylosis and vascular and nerve compression. NHO pathogenesis is unknown and the only effective treatment remains surgical resection, however once resected, NHO can re-occur. To further understand NHO pathogenesis, we developed the first animal model of NHO following SCI in genetically unmodified mice, which mimics most clinical features of NHO in patients. We have previously shown that the combination of (1) a central nervous system lesion (SCI) and (2) muscular damage (via an intramuscular injection of cardiotoxin) is required for NHO development. Furthermore, macrophages within the injured muscle play a critical role in driving NHO pathogenesis. More recently we demonstrated that macrophage-derived oncostatin M (OSM) is a key mediator of both human and mouse NHO. We now report that inflammatory monocytes infiltrate the injured muscles of SCI mice developing NHO at significantly higher levels compared to mice without SCI. Muscle infiltrating monocytes and neutrophils expressed OSM whereas mouse muscle satellite and interstitial cell expressed the OSM receptor (OSMR). In vitro recombinant mouse OSM induced tyrosine phosphorylation of the transcription factor STAT3, a downstream target of OSMR:gp130 signaling in muscle progenitor cells. As STAT3 is tyrosine phosphorylated by JAK1/2 tyrosine kinases downstream of OSMR:gp130, we demonstrated that the JAK1/2 tyrosine kinase inhibitor ruxolitinib blocked OSM driven STAT3 tyrosine phosphorylation in mouse muscle progenitor cells. We further demonstrated in vivo that STAT3 tyrosine phosphorylation was not only significantly higher but persisted for a longer duration in injured muscles of SCI mice developing NHO compared to mice with muscle injury without SCI. Finally, administration of ruxolitinib for 7 days post-surgery significantly reduced STAT3 phosphorylation in injured muscles in vivo as well as NHO volume at all analyzed time-points up to 3 weeks post-surgery. Our results identify the JAK/STAT3 signaling pathway as a potential therapeutic target to reduce NHO development following SCI.


Assuntos
Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Inibidores de Janus Quinases/farmacologia , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/patologia , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Imuno-Histoquímica , Janus Quinase 1/metabolismo , Janus Quinase 2/metabolismo , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/metabolismo , Células Musculares , Ossificação Heterotópica/tratamento farmacológico , Fosforilação , Fator de Transcrição STAT3/metabolismo , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/etiologia , Microtomografia por Raio-X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA