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 (3.3 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

Mobilization of monocytic myeloid-derived suppressor cells is regulated by PTH1R activation in bone marrow stromal cells

Eun Jung Lee1,2,Kyoung Jin Lee1,2,Seungpil Jung3Kyong Hwa Park4Serk In Park1,2,5 ( )
Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
The BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Anam Hospital, Seoul, Republic of Korea
Division of Oncology and Hematology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, Republic of Korea
Vanderbilt Center for Bone Biology, Vanderbilt University School of Medicine, Nashville, TN, USA

These authors contributed equally: Eun Jung Lee, Kyoung Jin Lee.

Show Author Information

Abstract

Myeloid-derived suppressor cells (MDSCs) are bone marrow (BM)-derived immunosuppressive cells in the tumor microenvironment, but the mechanism of MDSC mobilization from the BM remains unclear. We investigated how BM stromal cell activation by PTH1R contributes to MDSC mobilization. PTH1R activation by parathyroid hormone (PTH) or PTH-related peptide (PTHrP), a tumor-derived counterpart, mobilized monocytic (M-) MDSCs from murine BM without increasing immunosuppressive activity. In vitro cell-binding assays demonstrated that α4β1 integrin and vascular cell adhesion molecule (VCAM)-1, expressed on M-MDSCs and osteoblasts, respectively, are key to M-MDSC binding to osteoblasts. Upon PTH1R activation, osteoblasts express VEGF-A and IL6, leading to Src family kinase phosphorylation in M-MDSCs. Src inhibitors suppressed PTHrP-induced MDSC mobilization, and Src activation in M-MDSCs upregulated two proteases, ADAM-17 and MMP7, leading to VCAM1 shedding and subsequent disruption of M-MDSC tethering to osteoblasts. Collectively, our data provide the molecular mechanism of M-MDSC mobilization in the bones of tumor hosts.

References

1

Jeong, H. M., Cho, S. W. & Park, S. I. Osteoblasts are the centerpiece of the metastatic bone microenvironment. Endocrinol. Metab. (Seoul.) 31, 485–492 (2016).

2

Buenrostro, D., Park, S. I. & Sterling, J. A. Dissecting the role of bone marrow stromal cells on bone metastases. Biomed. Res. Int. 2014, 875305 (2014).

3

Park, S. I., Soki, F. N. & McCauley, L. K. Roles of bone marrow cells in skeletal metastases: no longer bystanders. Cancer Microenviron. 4, 237–246 (2011).

4

Gabrilovich, D. I. Myeloid-derived suppressor cells. Cancer Immunol. Res. 5, 3–8 (2017).

5

Gros, A. et al. Myeloid cells obtained from the blood but not from the tumor can suppress T-cell proliferation in patients with melanoma. Clin. Cancer Res. 18, 5212–5223 (2012).

6

Weide, B. et al. Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: comparison with regulatory T cells and NY-ESO-1- or Melan-A-Specific T Cells. Clin. Cancer Res. 20, 1601–1609 (2014).

7

Lim, J.-Y. et al. Ex vivo generated human cord blood myeloid-derived suppressor cells attenuate murine chronic graft-versus-host diseases. Biol. Blood Marrow Transpl. 24, 2381–2396 (2018).

8

Marvel, D. & Gabrilovich, D. I. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J. Clin. Invest. 125, 3356–3364 (2015).

9

Bronte, V. et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 7, 12150 (2016).

10

Veglia, F., Perego, M. & Gabrilovich, D. Myeloid-derived suppressor cells coming of age. Nat. Immunol. 19, 108–119 (2018).

11

Dolcetti, L. et al. Hierarchy of immunosuppressive strength among myeloid-derived suppressor cell subsets is determined by GM-CSF. Eur. J. Immunol. 40, 22–35 (2010).

12

Marigo, I. et al. Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity 32, 790–802 (2010).

13

Luyckx, A. et al. G-CSF stem cell mobilization in human donors induces polymorphonuclear and mononuclear myeloid-derived suppressor cells. Clin. Immunol. 143, 83–87 (2012).

14

Lee, C.-R., Lee, W., Cho, S. K. & Park, S.-G. Characterization of multiple cytokine combinations and TGF-β on differentiation and functions of myeloid-derived suppressor cells. Int. J. Mol. Sci. 19, 869 (2018).

15

Tu, S. et al. Overexpression of interleukin-1beta induces gastric inflammation and cancer and mobilizes myeloid-derived suppressor cells in mice. Cancer Cell 14, 408–419 (2008).

16

Svoronos, N. et al. Tumor cell-independent estrogen signaling drives disease progression through mobilization of myeloid-derived suppressor cells. Cancer Discov. 7, 72–85 (2017).

17

Hegde, V. L., Nagarkatti, M. & Nagarkatti, P. S. Cannabinoid receptor activation leads to massive mobilization of myeloid-derived suppressor cells with potent immunosuppressive properties. Eur. J. Immunol. 40, 3358–3371 (2010).

18

Liang, H. et al. Host STING-dependent MDSC mobilization drives extrinsic radiation resistance. Nat. Commun. 8, 1736–10 (2017).

19

Park, S. I. et al. Parathyroid hormone-related protein drives a CD11b+Gr1+ cell-mediated positive feedback loop to support prostate cancer growth. Cancer Res. 73, 6574–6583 (2013).

20

Rettig, M. P., Ansstas, G. & DiPersio, J. F. Mobilization of hematopoietic stem and progenitor cells using inhibitors of CXCR4 and VLA-4. Leukemia 26, 34–53 (2012).

21

Shiozawa, Y., Pienta, K. J. & Taichman, R. S. Hematopoietic stem cell niche is a potential therapeutic target for bone metastatic tumors. Clin. Cancer Res. 17, 5553–5558 (2011).

22

Schmid, M. C. et al. PI3-kinase γ promotes Rap1a-mediated activation of myeloid cell integrin α4β1, leading to tumor inflammation and growth. PLoS One 8, e60226 (2013).

23

Schmid, M. C. et al. Combined blockade of integrin-α4β1 plus cytokines SDF-1α or IL-1β potently inhibits tumor inflammation and growth. Cancer Res. 71, 6965–6975 (2011).

24

Jin, H., Su, J., Garmy-Susini, B., Kleeman, J. & Varner, J. Integrin alpha4beta1 promotes monocyte trafficking and angiogenesis in tumors. Cancer Res. 66, 2146–2152 (2006).

25

Foubert, P., Kaneda, M. M. & Varner, J. A. PI3Kγ activates integrin α4 and promotes immune suppressive myeloid cell polarization during tumor progression. Cancer Immunol. Res. 5, 957–968 (2017).

26

Xu, J. et al. Tpl2 protects against fulminant hepatitis through mobilization of myeloid-derived suppressor cells. Front. Immunol. 10, 1980 (2019).

27

Hawila, E. et al. CCR5 directs the mobilization of CD11b+Gr1+Ly6Clow polymorphonuclear myeloid cells from the bone marrow to the blood to support tumor development. Cell Rep. 21, 2212–2222 (2017).

28

Park, S.-M. & Youn, J.-I. Role of myeloid-derived suppressor cells in immune checkpoint inhibitor therapy in cancer. Arch. Pharm. Res. 42, 560–566 (2019).

29

Liu, Y. et al. Targeting myeloid-derived suppressor cells for cancer immunotherapy. Cancer Immunol. Immunother. 67, 1181–1195 (2018).

30

McCauley, L. K. & Martin, T. J. Twenty-five years of PTHrP progress: from cancer hormone to multifunctional cytokine. J. Bone Miner. Res. 27, 1231–1239 (2012).

31

Li, J. et al. PTHrP drives breast tumor initiation, progression, and metastasis in mice and is a potential therapy target. J. Clin. Invest. 121, 4655–4669 (2011).

32

Sato, K. et al. Passive immunization with anti-parathyroid hormone-related protein monoclonal antibody markedly prolongs survival time of hypercalcemic nude mice bearing transplanted human PTHrP-producing tumors. J. Bone Miner. Res. 8, 849–860 (1993).

33

Sato, K., Onuma, E., Yocum, R. C. & Ogata, E. Treatment of malignancy-associated hypercalcemia and cachexia with humanized anti-parathyroid hormone-related protein antibody. Semin. Oncol. 30, 167–173 (2003).

34

Iguchi, H., Aramaki, Y., Maruta, S. & Takiguchi, S. Effects of anti-parathyroid hormone-related protein monoclonal antibody and osteoprotegerin on PTHrP-producing tumor-induced cachexia in nude mice. J. Bone Min. Metab. 24, 16–19 (2006).

35

Camirand, A., Fadhil, I., Luco, A.-L., Ochietti, B. & Kremer, R. B. Enhancement of taxol, doxorubicin and zoledronate anti-proliferation action on triple-negative breast cancer cells by a PTHrP blocking monoclonal antibody. Am. J. Cancer Res. 3, 500–508 (2013).

36

Liang, W. et al. Antitumor activity of targeting SRC kinases in endothelial and myeloid cell compartments of the tumor microenvironment. Clin. Cancer Res. 16, 924–935 (2010).

37

Mao, L. et al. Inhibition of SRC family kinases reduces myeloid-derived suppressor cells in head and neck cancer. Int. J. Cancer 140, 1173–1185 (2017).

38

Yu, G.-T. et al. Inhibition of SRC family kinases facilitates anti-CTLA4 immunotherapy in head and neck squamous cell carcinoma. Cell. Mol. Life Sci. 75, 4223–4234 (2018).

39

Garton, K. J. et al. Stimulated shedding of vascular cell adhesion molecule 1 (VCAM-1) is mediated by tumor necrosis factor-alpha-converting enzyme (ADAM 17). J. Biol. Chem. 278, 37459–37464 (2003).

40

Lévesque, J. P., Takamatsu, Y., Nilsson, S. K., Haylock, D. N. & Simmons, P. J. Vascular cell adhesion molecule-1 (CD106) is cleaved by neutrophil proteases in the bone marrow following hematopoietic progenitor cell mobilization by granulocyte colony-stimulating factor. Blood 98, 1289–1297 (2001).

41

Lévesque, J.-P. et al. Mobilization by either cyclophosphamide or granulocyte colony-stimulating factor transforms the bone marrow into a highly proteolytic environment. Exp. Hematol. 30, 440–449 (2002).

42

Datta, N. S., Chen, C., Berry, J. E. & McCauley, L. K. PTHrP signaling targets cyclin D1 and induces osteoblastic cell growth arrest. J. Bone Miner. Res. 20, 1051–1064 (2005).

43

Koh, A. J., Beecher, C. A., Rosol, T. J. & McCauley, L. K. 3‘,5’-Cyclic adenosine monophosphate activation in osteoblastic cells: effects on parathyroid hormone-1 receptors and osteoblastic differentiation in vitro. Endocrinology 140, 3154–3162 (1999).

44

Lee, C. et al. Dual targeting c-met and VEGFR2 in osteoblasts suppresses growth and osteolysis of prostate cancer bone metastasis. Cancer Lett. 414, 205–213 (2018).

45

Jung, Y. et al. Annexin Ⅱ expressed by osteoblasts and endothelial cells regulates stem cell adhesion, homing, and engraftment following transplantation. Blood 110, 82–90 (2007).

46

Shiozawa, Y. et al. Annexin Ⅱ/annexin Ⅱ receptor axis regulates adhesion, migration, homing, and growth of prostate cancer. J. Cell. Biochem. 105, 370–380 (2008).

Bone Research
Article number: 22
Cite this article:
Lee EJ, Lee KJ, Jung S, et al. Mobilization of monocytic myeloid-derived suppressor cells is regulated by PTH1R activation in bone marrow stromal cells. Bone Research, 2023, 11: 22. https://doi.org/10.1038/s41413-023-00255-y

140

Views

2

Downloads

3

Crossref

2

Web of Science

2

Scopus

Altmetrics

Received: 21 June 2022
Revised: 23 January 2023
Accepted: 01 March 2023
Published: 21 April 2023
© The Author(s) 2023

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