AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (5.4 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Neuron-to-vessel signaling is a required feature of aberrant stem cell commitment after soft tissue trauma

Qizhi Qin1,Mario Gomez-Salazar1,Masnsen Cherief1Chase A. Pagani2Seungyong Lee1Charles Hwang3Robert J. Tower2,4Sharon Onggo1Yuxiao Sun2Abhinav Piplani1Zhao Li1Sowmya Ramesh1Thomas L. Clemens4,5Benjamin Levi2( )Aaron W. James1 ( )
Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
Center for Organogenesis and Trauma, Department of Surgery, University of Texas, Southwestern, TX, USA
Department of Plastic Surgery, Harvard, Cambridge, MA 02138, USA
Department of Orthopaedics, Johns Hopkins University, Baltimore, MD 21205, USA
Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA

These authors contributed equally: Qizhi Qin, Mario Gomez-Salazar

Show Author Information

Abstract

The functional interdependence of nerves and blood vessels is a well-established concept during tissue morphogenesis, yet the role of neurovascular coupling in proper and aberrant tissue repair is an emerging field of interest. Here, we sought to define the regulatory relationship of peripheral nerves on vasculature in a severe extremity trauma model in mice, which results in aberrant cell fate and heterotopic ossification (HO). First, a high spatial degree of neurovascular congruency was observed to exist within extremity injury associated heterotopic ossification. Vascular and perivascular cells demonstrate characteristic responses to injury, as assessed by single cell RNA sequencing. This vascular response to injury was blunted in neurectomized mice, including a decrease in endothelial proliferation and type H vessel formation, and a downregulation of key transcriptional networks associated with angiogenesis. Independent mechanisms to chemically or genetically inhibit axonal ingrowth led to similar deficits in HO site angiogenesis, a reduction in type H vessels, and heterotopic bone formation. Finally, a combination of single cell transcriptomic approaches within the dorsal root ganglia identified key neural-derived angiogenic paracrine factors that may mediate neuron-to-vascular signaling in HO. These data provide further understanding of nerve-to-vessel crosstalk in traumatized soft tissues, which may reflect a key determinant of mesenchymal progenitor cell fate after injury.

References

1

Mukouyama, Y. S., Shin, D., Britsch, S., Taniguchi, M. & Anderson, D. J. Sensory nerves determine the pattern of arterial differentiation and blood vessel branching in the skin. Cell 109, 693–705 (2002).

2

Liu, L., Dana, R. & Yin, J. Sensory neurons directly promote angiogenesis in response to inflammation via substance P signaling. FASEB J. 34, 6229–6243 (2020).

3

Carmeliet, P. & Tessier-Lavigne, M. Common mechanisms of nerve and blood vessel wiring. Nature 436, 193–200 (2005).

4

Meyers, C. A. et al. A neurotrophic mechanism directs sensory nerve transit in cranial bone. Cell Rep. 31, 107696 (2020).

5

Li, Z. et al. Fracture repair requires TrkA signaling by skeletal sensory nerves. J. Clin. Investig. 129, 5137–5150 (2019).

6

Lee, S. et al. NGF-TrkA signaling dictates neural ingrowth and aberrant osteochondral differentiation after soft tissue trauma. Nat. Commun. 12, 4939 (2021).

7

Martin, P. & Lewis, J. Origins of the neurovascular bundle: interactions between developing nerves and blood vessels in embryonic chick skin. Int J. Dev. Biol. 33, 379–387 (1989).

8

Li, W. et al. Peripheral nerve-derived CXCL12 and VEGF-A regulate the patterning of arterial vessel branching in developing limb skin. Dev. Cell 24, 359–371 (2013).

9

Honig, M. G., Frase, P. A. & Camilli, S. J. The spatial relationships among cutaneous, muscle sensory and motoneuron axons during development of the chick hindlimb. Development 125, 995–1004 (1998).

10

Honig, M. G., Lance-Jones, C. & Landmesser, L. The development of sensory projection patterns in embryonic chick hindlimb under experimental conditions. Dev. Biol. 118, 532–548 (1986).

11

Bates, D., Taylor, G. I. & Newgreen, D. F. The pattern of neurovascular development in the forelimb of the quail embryo. Dev. Biol. 249, 300–320 (2002).

12
Dorfman, H. D. & Czerniak, B. Bone Tumors, (Mosby, 1998).
13

Bedi, A. et al. The incidence of heterotopic ossification after hip arthroscopy. Am. J. Sports Med. 40, 854–863 (2012).

14

Daniels, C. M. et al. Has the proportion of combat-related amputations that develop heterotopic ossification increased? J. Orthop. Trauma 32, 283–287 (2018).

15

Sandeep, K. N., Suresh, G., Gopisankar, B., Abhishek, N. & Sujiv, A. Does excision of heterotopic ossification of the elbow result in satisfactory patient-rated outcomes? Malays. Orthop. J. 11, 35–40 (2017).

16

Meyers, C. et al. Heterotopic ossification: a comprehensive review. JBMR 3, e10172 (2019).

17

Ranganathan, K. et al. Heterotopic ossification: basic-science principles and clinical correlates. J. Bone Jt. Surg. Am. 97, 1101–1111 (2015).

18

Hoch, B. & Montag, A. Reactive bone lesions mimicking neoplasms. Semin. Diagn. Pathol. 28, 102–112 (2011).

19

Rosenberg, A. E. Pseudosarcomas of soft tissue. Arch. Pathol. Lab. Med. 132, 579–586 (2008).

20

Reznik, J. E. et al. A preliminary investigation on the effect of extracorporeal shock wave therapy as a treatment for neurogenic heterotopic ossification following traumatic brain injury. Part I: Eff. Pain. Brain Inj. 31, 526–532 (2017).

21

Tran, L., Stein, N. & Miller, S. Fibrodysplasia ossificans progressiva: early diagnosis is critical yet challenging. J. Pediatr. 157, 860.e1 (2010).

22

Kaplan, F. S. et al. Fibrodysplasia ossificans progressiva. Best. Pr. Res. Clin. Rheumatol. 22, 191–205 (2008).

23

Kaplan, F. S. et al. Early diagnosis of fibrodysplasia ossificans progressiva. Pediatrics 121, e1295–e1300 (2008).

24

Salisbury, E. et al. Sensory nerve induced inflammation contributes to heterotopic ossification. J. Cell Biochem. 112, 2748–2758 (2011).

25

Salisbury, E., Sonnet, C., Heggeness, M., Davis, A. R. & Olmsted-Davis, E. Heterotopic ossification has some nerve. Crit. Rev. Eukar Gene 20, 313–324 (2010).

26

Abeynayake, N., Arthur, A. & Gronthos, S. Crosstalk between skeletal and neural tissues is critical for skeletal health. Bone 142, 115645 (2021).

27

Olmsted-Davis, E. A. et al. Progenitors in peripheral nerves launch heterotopic ossification. Stem Cells Transl. Med. 6, 1109–1119 (2017).

28

Chen, M. et al. Skeleton-vasculature chain reaction: a novel insight into the mystery of homeostasis. Bone Res. 9, 21 (2021).

29

Hwang, C. et al. Mesenchymal VEGFA induces aberrant differentiation in heterotopic ossification. Bone Res. 7, 36 (2019).

30

Agarwal, S. et al. Inhibition of Hif1 alpha prevents both trauma-induced and genetic heterotopic ossification. P Natl Acad. Sci. USA 113, E338–E347 (2016).

31

Agarwal, S. et al. Analysis of bone-cartilage-stromal progenitor populations in trauma induced and genetic models of heterotopic ossification. Stem Cells 34, 1692–1701 (2016).

32

Agarwal, S. et al. Surgical excision of heterotopic ossification leads to re-emergence of mesenchymal stem cell populations responsible for recurrence. Stem Cells Transl. Med. 6, 799–806 (2017).

33

Peterson, J. R. et al. Direct mouse trauma/burn model of heterotopic ossification. J. Vis. Exp. 102, e52880 (2015).

34

Blanco, R. & Gerhardt, H. VEGF and Notch in tip and stalk cell selection. Cold Spring Harb. Perspect. Med. 3, a006569 (2013).

35

Peng, Y., Wu, S., Li, Y. & Crane, J. L. Type H blood vessels in bone modeling and remodeling. Theranostics 10, 426–436 (2020).

36

Kusumbe, A. P., Ramasamy, S. K. & Adams, R. H. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 507, 323–328 (2014).

37

Renthal, W. et al. Transcriptional reprogramming of distinct peripheral sensory neuron subtypes after axonal injury. Neuron 108, 128–144.e9 (2020).

38

Nyberg, P., Xie, L. & Kalluri, R. Endogenous inhibitors of angiogenesis. Cancer Res. 65, 3967–3979 (2005).

39

Behr, B. et al. Fgf-18 is required for osteogenesis but not angiogenesis during long bone repair. Tissue Eng. Part A 17, 2061–2069 (2011).

40

Orlandini, M. et al. Vascular endothelial growth factor-D activates VEGFR-3 expressed in osteoblasts inducing their differentiation. J. Biol. Chem. 281, 17961–17967 (2006).

41

Dai, J. & Rabie, A. B. M. VEGF: an essential mediator of both angiogenesis and endochondral ossification. J. Dent. Res. 86, 937–950 (2007).

42

Duan, X. et al. Vegfa regulates perichondrial vascularity and osteoblast differentiation in bone development. Development 142, 1984–1991 (2015).

43

Yang, Y. Q. et al. The role of vascular endothelial growth factor in ossification. Int J. Oral. Sci. 4, 64–68 (2012).

44

Hu, K. & Olsen, B. R. Osteoblast-derived VEGF regulates osteoblast differentiation and bone formation during bone repair. J. Clin. Investig. 126, 509–526 (2016).

45

He, F. L. & Soriano, P. Dysregulated PDGFR alpha signaling alters coronal suture morphogenesis and leads to craniosynostosis through endochondral ossification. Development 144, 4026–4036 (2017).

46

Ding, H. et al. A specific requirement for PDGF-C in palate formation and PDGFR-alpha signaling. Nat. Genet 36, 1111–1116 (2004).

47

Hsieh, H. H. S. et al. Evaluation of salivary cytokines for diagnosis of both trauma-induced and genetic heterotopic ossification. Front. Endocrinol 8, 74 (2017).

48

Ranganathan, K. et al. The role of the adaptive immune system in burn-induced heterotopic ossification and mesenchymal cell osteogenic differentiation. J. Surg. Res. 206, 53–61 (2016).

49

Kan, L. X. et al. Dysregulation of local stem/progenitor cells as a common cellular mechanism for heterotopic ossification. Stem Cells 27, 150–156 (2009).

50

Genet, F. et al. Neurological heterotopic ossification following spinal cord injury is triggered by macrophage-mediated inflammation in muscle. J. Pathol. 236, 229–240 (2015).

51

Sorkin, M. et al. Regulation of heterotopic ossification by monocytes in a mouse model of aberrant wound healing. Nat. Commun. 11, 722 (2020).

52

Wang, X. et al. Inhibition of overactive TGF-beta attenuates progression of heterotopic ossification in mice. Nat. Commun. 9, 551 (2018).

53

Kraft, C. T. et al. Trauma-induced heterotopic bone formation and the role of the immune system: A review. J. Trauma Acute Care Surg. 80, 156–165 (2016).

54

Chen, Q. et al. Fate decision of mesenchymal stem cells: adipocytes or osteoblasts? Cell Death Differ. 23, 1128–1139 (2016).

55

Cao, R. H., Farnebo, J., Kurimoto, M. & Cao, Y. H. Interleukin-18 acts as an angiogenesis and tumor suppressor. Faseb J. 13, 2195–2202 (1999).

56

Shen, J. K. et al. Interleukin-18 has antipermeablity and antiangiogenic activities in the eye: reciprocal suppression with VEGF. J. Cell Physiol. 229, 974–983 (2014).

57

Dai, S. M., Nishioka, K. & Yudoh, K. Interleukin (IL) 18 stimulates osteoclast formation through synovial T cells in rheumatoid arthritis: comparison with IL1 beta and tumour necrosis factor alpha. Ann. Rheum. Dis. 63, 1379–1386 (2004).

58

Zhu, S. et al. Chondromodulin-1 in health, osteoarthritis, cancer, and heart disease. Cell Mol. Life Sci. 76, 4493–4502 (2019).

59

Nakamichi, Y. et al. Chondromodulin Ⅰ is a bone remodeling factor. Mol. Cell Biol. 23, 636–644 (2003).

60

Yukata, K. et al. Altered fracture callus formation in chondromodulin-Ⅰ deficient mice. Bone 43, 1047–1056 (2008).

61

Shukunami, C. & Hiraki, Y. Role of cartilage-derived anti-angiogenic factor, chondromodulin-Ⅰ, during endochondral bone formation. Osteoarthr. Cartil. 9, S91–S101 (2001).

62

Shore, E. M. & Kaplan, F. S. Inherited human diseases of heterotopic bone formation. Nat. Rev. Rheumatol. 6, 518–527 (2010).

63

Tower, R. J. et al. Spatial transcriptomics reveals a role for sensory nerves in preserving cranial suture patency through modulation of BMP/TGF-beta signaling. Proc. Natl. Acad. Sci. USA 118, e2103087118 (2021).

64

Chen, X. et al. A chemical-genetic approach to studying neurotrophin signaling. Neuron 46, 13–21 (2005).

65

Tomlinson, R. E. et al. NGF-TrkA signaling by sensory nerves coordinates the vascularization and ossification of developing endochondral bone. Cell Rep. 16, 2723–2735 (2016).

66

Ashraf, S., Bouhana, K. S., Pheneger, J., Andrews, S. W. & Walsh, D. A. Selective inhibition of tropomyosin-receptor-kinase A (TrkA) reduces pain and joint damage in two rat models of inflammatory arthritis. Arthritis Res. Ther. 18, 97 (2016).

67

Nwosu, L. N., Mapp, P. I., Chapman, V. & Walsh, D. A. Blocking the tropomyosin receptor kinase A (TrkA) receptor inhibits pain behaviour in two rat models of osteoarthritis. Ann. Rheum. Dis. 75, 1246–1254 (2016).

Bone Research
Article number: 43
Cite this article:
Qin Q, Gomez-Salazar M, Cherief M, et al. Neuron-to-vessel signaling is a required feature of aberrant stem cell commitment after soft tissue trauma. Bone Research, 2022, 10: 43. https://doi.org/10.1038/s41413-022-00216-x

84

Views

2

Downloads

15

Crossref

10

Web of Science

13

Scopus

Altmetrics

Received: 23 November 2021
Revised: 04 April 2022
Accepted: 29 April 2022
Published: 01 June 2022
© The Author(s) 2022

This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Return