4.6 Article

Depletion of Mast Cells and Macrophages Impairs Heterotopic Ossification in an Acvr1(R206H) Mouse Model of Fibrodysplasia Ossificans Progressiva

期刊

JOURNAL OF BONE AND MINERAL RESEARCH
卷 33, 期 2, 页码 269-282

出版社

WILEY
DOI: 10.1002/jbmr.3304

关键词

FIBRODYSPLASIA OSSIFICANS PROGRESSIVA; FOP; HETEROTOPIC OSSIFICATION; TISSUE INJURY; CHRONIC INFLAMMATION; BONE MORPHOGENETIC PROTEIN SIGNALING; BMP; ACVR1

资金

  1. International Fibrodysplasia Ossificans Progressiva Association (IFOPA)
  2. Whitney Weldon Endowment for FOP Research
  3. Center for Research in FOP and Related Disorders
  4. Ian Cali Endowment for FOP Research
  5. Ashley Martucci FOP Research Fund
  6. Isaac and Rose Nassau Professorship of Orthopaedic Molecular Medicine
  7. Cali-Weldon Professorship of FOP Research
  8. US National Institutes of Health [R01-AR041916]
  9. Penn Center for Musculoskeletal Diseases [NIH P30-AR050950]

向作者/读者索取更多资源

Heterotopic ossification (HO) is a clinical condition that often reduces mobility and diminishes quality of life for affected individuals. The most severe form of progressive HO occurs in those with fibrodysplasia ossificans progressiva (FOP; OMIM #135100), a genetic disorder caused by a recurrent heterozygous gain-of-function mutation (R206H) in the bone morphogenetic protein (BMP) type I receptor ACVR1/ALK2. In individuals with FOP, episodes of HO frequently follow injury. The first sign of active disease is commonly an inflammatory flare-up that precedes connective tissue degradation, progenitor cell recruitment, and endochondral HO. We used a conditional-on global knock-in mouse model expressing Acvr1(R206H) (referred to as Acvr1(cR206H/+)) to investigate the cellular and molecular inflammatory response in FOP lesions following injury. We found that the Acvr1 R206H mutation caused increased BMP signaling in posttraumatic FOP lesions and early divergence from the normal skeletal muscle repair program with elevated and prolonged immune cell infiltration. The proinflammatory cytokine response of TNF, IL-1, and IL-6 was elevated and prolonged in Acvr1(cR206H/+) lesions and in Acvr1(cR206H/+) mast cells. Importantly, depletion of mast cells and macrophages significantly impaired injury-induced HO in Acvr1(cR206H/+) mice, reducing injury-induced HO volume by approximate to 50% with depletion of each cell population independently, and approximate to 75% with combined depletion of both cell populations. Together, our data show that the immune system contributes to the initiation and development of HO in FOP. Further, the expression of Acvr1(R206H) in immune cells alters cytokine expression and cellular response to injury and unveils novel therapeutic targets for treatment of FOP and nongenetic forms of HO. (c) 2017 American Society for Bone and Mineral Research.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据