Skip to main content
Top
Published in: Experimental Hematology & Oncology 1/2024

Open Access 01-12-2024 | Leukemia | Research

Single-cell analysis defines highly specific leukemia-induced neutrophils and links MMP8 expression to recruitment of tumor associated neutrophils during FGFR1 driven leukemogenesis

Authors: Tianxiang Hu, Bo Cheng, Atsuko Matsunaga, Ting Zhang, Xiaocui Lu, Hui Fang, Stephanie F. Mori, Xuexiu Fang, Gavin Wang, Hongyan Xu, Huidong Shi, John K. Cowell

Published in: Experimental Hematology & Oncology | Issue 1/2024

Login to get access

Abstract

Background

Leukemias driven by activated, chimeric FGFR1 kinases typically progress to AML which have poor prognosis. Mouse models of this syndrome allow detailed analysis of cellular and molecular changes occurring during leukemogenesis. We have used these models to determine the effects of leukemia development on the immune cell composition in the leukemia microenvironment during leukemia development and progression.

Methods

Single cell RNA sequencing (scRNA-Seq) was used to characterize leukemia associated neutrophils and define gene expression changes in these cells during leukemia progression.

Results

scRNA-Seq revealed six distinct subgroups of neutrophils based on their specific differential gene expression. In response to leukemia development, there is a dramatic increase in only two of the neutrophil subgroups. These two subgroups show specific gene expression signatures consistent with neutrophil precursors which give rise to immature polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Analysis of gene expression in these precursor cells identified pathways that were specifically upregulated, the most pronounced of which involved matrix metalloproteinases Mmp8 and Mmp9, during leukemia progression. Pharmacological inhibition of MMPs using Ilomastat preferentially restricted in vitro migration of neutrophils from leukemic mice and led to a significantly improved survival in vivo, accompanied by impaired PMN-MDSC recruitment. As a result, levels of T-cells were proportionally increased. In clinically annotated TCGA databases, MMP8 was shown to act as an independent indicator for poor prognosis and correlated with higher neutrophil infiltration and poor pan-cancer prognosis.

Conclusion

We have defined specific leukemia responsive neutrophil subgroups based on their unique gene expression profile, which appear to be the precursors of neutrophils specifically associated with leukemia progression. An important event during development of these neutrophils is upregulation MMP genes which facilitated mobilization of these precursors from the BM in response to cancer progression, suggesting a possible therapeutic approach to suppress the development of immune tolerance.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jackson CC, Medeiros LJ, Miranda RN. 8p11 myeloproliferative syndrome: a review. Hum Pathol. 2010;41:461–76.PubMedCrossRef Jackson CC, Medeiros LJ, Miranda RN. 8p11 myeloproliferative syndrome: a review. Hum Pathol. 2010;41:461–76.PubMedCrossRef
2.
go back to reference Li T, Zhang G, Zhang X, et al. The 8p11 myeloproliferative syndrome: genotypic and phenotypic classification and targeted therapy. Front Oncol. 2022;12:1015792.PubMedPubMedCentralCrossRef Li T, Zhang G, Zhang X, et al. The 8p11 myeloproliferative syndrome: genotypic and phenotypic classification and targeted therapy. Front Oncol. 2022;12:1015792.PubMedPubMedCentralCrossRef
3.
go back to reference Baumann H, Kunapuli P, Tracey E, et al. The oncogenic fusion protein-tyrosine kinase ZNF198/fibroblast growth factor receptor-1 has signaling function comparable with interleukin-6 cytokine receptors. J Biol Chem. 2003;278:16198–208.PubMedCrossRef Baumann H, Kunapuli P, Tracey E, et al. The oncogenic fusion protein-tyrosine kinase ZNF198/fibroblast growth factor receptor-1 has signaling function comparable with interleukin-6 cytokine receptors. J Biol Chem. 2003;278:16198–208.PubMedCrossRef
4.
go back to reference Ren M, Li X, Cowell JK. Genetic fingerprinting of the development and progression of T-cell lymphoma in a murine model of atypical myeloproliferative disorder initiated by the ZNF198-fibroblast growth factor receptor-1 chimeric tyrosine kinase. Blood. 2009;114:1576–84.PubMedPubMedCentralCrossRef Ren M, Li X, Cowell JK. Genetic fingerprinting of the development and progression of T-cell lymphoma in a murine model of atypical myeloproliferative disorder initiated by the ZNF198-fibroblast growth factor receptor-1 chimeric tyrosine kinase. Blood. 2009;114:1576–84.PubMedPubMedCentralCrossRef
5.
go back to reference Ren M, Tidwell JA, Sharma S, et al. Acute progression of BCR-FGFR1 induced murine B-lympho/myeloproliferative disorder suggests involvement of lineages at the pro-B cell stage. PLoS ONE. 2012;7:e38265.PubMedPubMedCentralCrossRef Ren M, Tidwell JA, Sharma S, et al. Acute progression of BCR-FGFR1 induced murine B-lympho/myeloproliferative disorder suggests involvement of lineages at the pro-B cell stage. PLoS ONE. 2012;7:e38265.PubMedPubMedCentralCrossRef
6.
go back to reference Ren M, Qin H, Kitamura E, et al. Dysregulated signaling pathways in the development of CNTRL-FGFR1-induced myeloid and lymphoid malignancies associated with FGFR1 in human and mouse models. Blood. 2013;122:1007–16.PubMedPubMedCentralCrossRef Ren M, Qin H, Kitamura E, et al. Dysregulated signaling pathways in the development of CNTRL-FGFR1-induced myeloid and lymphoid malignancies associated with FGFR1 in human and mouse models. Blood. 2013;122:1007–16.PubMedPubMedCentralCrossRef
7.
go back to reference Cowell JK, Qin H, Chang C-S, et al. A model of BCR-FGFR1 driven human AML in immunocompromised mice. Br J Cancer. 2016;175:542–45. Cowell JK, Qin H, Chang C-S, et al. A model of BCR-FGFR1 driven human AML in immunocompromised mice. Br J Cancer. 2016;175:542–45.
8.
9.
go back to reference Ren M, Qin H, Ren R, et al. Ponatinib suppresses the development of myeloid and lymphoid malignancies associated with FGFR1 abnormalities. Leukemia. 2013;27:32–40.PubMedCrossRef Ren M, Qin H, Ren R, et al. Ponatinib suppresses the development of myeloid and lymphoid malignancies associated with FGFR1 abnormalities. Leukemia. 2013;27:32–40.PubMedCrossRef
10.
11.
go back to reference Cowell JK, Qin H, Hu T et al. Mutation in the FGFR1 tyrosine kinase domain or inactivation of PTEN is associated with acquired resistance to FGFR inhibitors in FGFR1-driven leukemia/lymphomas. Int J Cancer. 12017; 41: 1822-29. Cowell JK, Qin H, Hu T et al. Mutation in the FGFR1 tyrosine kinase domain or inactivation of PTEN is associated with acquired resistance to FGFR inhibitors in FGFR1-driven leukemia/lymphomas. Int J Cancer. 12017; 41: 1822-29.
13.
go back to reference Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10.PubMedCrossRef Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39:1–10.PubMedCrossRef
14.
go back to reference Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–30.PubMedCrossRef Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541:321–30.PubMedCrossRef
16.
go back to reference Dunn GP, Bruce AT, Ikeda H, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–8.PubMedCrossRef Dunn GP, Bruce AT, Ikeda H, et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol. 2002;3:991–8.PubMedCrossRef
17.
go back to reference Vinay DS, Ryan EP, Pawelec G et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol. 2015: 35 Suppl: S185-98. Vinay DS, Ryan EP, Pawelec G et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol. 2015: 35 Suppl: S185-98.
20.
go back to reference Bronte V, Brandau S, Chen SH, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:12150.PubMedPubMedCentralCrossRef Bronte V, Brandau S, Chen SH, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun. 2016;7:12150.PubMedPubMedCentralCrossRef
22.
23.
go back to reference Cai B, Liu Y, Chong Y, et al. IRAK1-regulated IFN-γ signaling induces MDSC to facilitate immune evasion in FGFR1-driven hematological malignancies. Mol Cancer. 2021;20:165.PubMedPubMedCentralCrossRef Cai B, Liu Y, Chong Y, et al. IRAK1-regulated IFN-γ signaling induces MDSC to facilitate immune evasion in FGFR1-driven hematological malignancies. Mol Cancer. 2021;20:165.PubMedPubMedCentralCrossRef
26.
go back to reference Peranzoni E, Zilio S, Marigo I, et al. Myeloid-derived suppressor cell heterogeneity and subset definition. Curr Opin Immunol. 2010;22:238–44.PubMedCrossRef Peranzoni E, Zilio S, Marigo I, et al. Myeloid-derived suppressor cell heterogeneity and subset definition. Curr Opin Immunol. 2010;22:238–44.PubMedCrossRef
27.
go back to reference Yang Y, Li C, Liu T, et al. Myeloid-derived suppressor cells in tumors: from mechanisms to Antigen specificity and Microenvironmental Regulation. Front Immunol. 2020;11:1371.PubMedPubMedCentralCrossRef Yang Y, Li C, Liu T, et al. Myeloid-derived suppressor cells in tumors: from mechanisms to Antigen specificity and Microenvironmental Regulation. Front Immunol. 2020;11:1371.PubMedPubMedCentralCrossRef
28.
go back to reference Li K, Shi H, Zhang B, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther. 2021;6:362.PubMedPubMedCentralCrossRef Li K, Shi H, Zhang B, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther. 2021;6:362.PubMedPubMedCentralCrossRef
29.
go back to reference Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C, Stuart JM. The Cancer Genome Atlas Pan-cancer analysis project. Nat Genet. 2013;45:1113–20.PubMedCentralCrossRef Cancer Genome Atlas Research Network, Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, Ellrott K, Shmulevich I, Sander C, Stuart JM. The Cancer Genome Atlas Pan-cancer analysis project. Nat Genet. 2013;45:1113–20.PubMedCentralCrossRef
30.
go back to reference Bottomly D, Long N, Schultz AR et al. Integrative analysis of drug response and clinical outcome in acute myeloid leukemia. Cancer Cell. 2022; 40:850 – 64.e9. Bottomly D, Long N, Schultz AR et al. Integrative analysis of drug response and clinical outcome in acute myeloid leukemia. Cancer Cell. 2022; 40:850 – 64.e9.
31.
go back to reference Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.PubMedCrossRef Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–4.PubMedCrossRef
32.
go back to reference Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:11.CrossRef Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:11.CrossRef
33.
go back to reference Butler A, Hoffman P, Smibert P, et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36:411–20.PubMedPubMedCentralCrossRef Butler A, Hoffman P, Smibert P, et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36:411–20.PubMedPubMedCentralCrossRef
34.
go back to reference Wu T, Hu E, Xu S, et al. ClusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innov (Camb). 2021;2:100141. Wu T, Hu E, Xu S, et al. ClusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innov (Camb). 2021;2:100141.
35.
37.
go back to reference Zhang H, Cai B, Liu Y, et al. RHOA-regulated IGFBP2 promotes invasion and drives progression of BCR-ABL1 chronic myeloid leukemia. Haematologica. 2023;108:122–34.PubMedCrossRef Zhang H, Cai B, Liu Y, et al. RHOA-regulated IGFBP2 promotes invasion and drives progression of BCR-ABL1 chronic myeloid leukemia. Haematologica. 2023;108:122–34.PubMedCrossRef
38.
go back to reference Li C, Cantor WJ, Nili N, et al. Arterial repair after stenting and the effects of GM6001, a matrix metalloproteinase inhibitor. J Am Coll Cardiol. 2002;39:1852–8.PubMedCrossRef Li C, Cantor WJ, Nili N, et al. Arterial repair after stenting and the effects of GM6001, a matrix metalloproteinase inhibitor. J Am Coll Cardiol. 2002;39:1852–8.PubMedCrossRef
39.
go back to reference Bissinger S, Hage C, Wagner V, et al. Macrophage depletion induces edema through release of matrix-degrading proteases and proteoglycan deposition. Sci Transl Med. 2021;13:eabd4550.PubMedCrossRef Bissinger S, Hage C, Wagner V, et al. Macrophage depletion induces edema through release of matrix-degrading proteases and proteoglycan deposition. Sci Transl Med. 2021;13:eabd4550.PubMedCrossRef
40.
go back to reference Li Z, Herold T, He C, et al. Identification of a 24-gene prognostic signature that improves the European LeukemiaNet risk classification of acute myeloid leukemia: an international collaborative study. J Clin Oncol. 2013;31:1172–81.PubMedPubMedCentralCrossRef Li Z, Herold T, He C, et al. Identification of a 24-gene prognostic signature that improves the European LeukemiaNet risk classification of acute myeloid leukemia: an international collaborative study. J Clin Oncol. 2013;31:1172–81.PubMedPubMedCentralCrossRef
43.
go back to reference Silva J, Chang C-S, Hu T, et al. Distinct Signaling Programs associated with progression of FGFR1 driven leukemia in a mouse model of Stem Cell Leukemia Lymphoma Syndrome. Genomics. 2019;111:1566–73.PubMedCrossRef Silva J, Chang C-S, Hu T, et al. Distinct Signaling Programs associated with progression of FGFR1 driven leukemia in a mouse model of Stem Cell Leukemia Lymphoma Syndrome. Genomics. 2019;111:1566–73.PubMedCrossRef
45.
go back to reference Veglia F, Hashimoto A, Dweep H, et al. Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor- bearing mice. J Exp Med. 2021;218:e20201803.PubMedPubMedCentralCrossRef Veglia F, Hashimoto A, Dweep H, et al. Analysis of classical neutrophils and polymorphonuclear myeloid-derived suppressor cells in cancer patients and tumor- bearing mice. J Exp Med. 2021;218:e20201803.PubMedPubMedCentralCrossRef
46.
go back to reference Grieshaber-Bouyer R, Radtke FA, et al. The neutrotime transcriptional signature defines a single continuum of neutrophils across biological compartments. Nat Commun. 2021;17:12. Grieshaber-Bouyer R, Radtke FA, et al. The neutrotime transcriptional signature defines a single continuum of neutrophils across biological compartments. Nat Commun. 2021;17:12.
47.
go back to reference Ding ZC, Aboelella NS, Bryan L, et al. The monocytes that repopulate in mice after cyclophosphamide treatment acquire a Nnutrophil precursor gene ssgnature and immunosuppressive activity. Front Immunol. 2021;1:594540.CrossRef Ding ZC, Aboelella NS, Bryan L, et al. The monocytes that repopulate in mice after cyclophosphamide treatment acquire a Nnutrophil precursor gene ssgnature and immunosuppressive activity. Front Immunol. 2021;1:594540.CrossRef
48.
go back to reference Metzger P, Kirchleitnar SV, Kluge M et al. Immunostimulatory RNA leads to functional reprogramming of myeloid-derived suppressor cells in pancreatic cancer. J Immunother Cancer. 2019;7:288. Erratum in: J Immunother Cancer. 2019;7:349. Metzger P, Kirchleitnar SV, Kluge M et al. Immunostimulatory RNA leads to functional reprogramming of myeloid-derived suppressor cells in pancreatic cancer. J Immunother Cancer. 2019;7:288. Erratum in: J Immunother Cancer. 2019;7:349.
49.
go back to reference Acuff HB, Carter KJ, Fingleton B, et al. Matrix metalloproteinase-9 from bone marrow-derived cells contributes to survival but not growth of tumor cells in the lung microenvironment. Cancer Res. 2006;66:259–66.PubMedPubMedCentralCrossRef Acuff HB, Carter KJ, Fingleton B, et al. Matrix metalloproteinase-9 from bone marrow-derived cells contributes to survival but not growth of tumor cells in the lung microenvironment. Cancer Res. 2006;66:259–66.PubMedPubMedCentralCrossRef
50.
go back to reference Kawai K, Xue F, Takahara T, et al. Matrix metalloproteinase-9 contributes to the mobilization of bone marrow cells in the injured liver. Cell Transpl. 2012;21:453–64.CrossRef Kawai K, Xue F, Takahara T, et al. Matrix metalloproteinase-9 contributes to the mobilization of bone marrow cells in the injured liver. Cell Transpl. 2012;21:453–64.CrossRef
51.
go back to reference Steinl C, Essl M, Schreiber TD, et al. Release of matrix metalloproteinase-8 duringphysiological trafficking and induced mobilization of human hematopoietic stem cells. Stem Cells Dev. 2013;22:1307–18.PubMedCrossRef Steinl C, Essl M, Schreiber TD, et al. Release of matrix metalloproteinase-8 duringphysiological trafficking and induced mobilization of human hematopoietic stem cells. Stem Cells Dev. 2013;22:1307–18.PubMedCrossRef
52.
go back to reference Klein G, Schmal O, Aicher WK. Matrix metalloproteinases in stem cell mobilization. Matrix Biol. 2015; 44–46: 175 – 83. Klein G, Schmal O, Aicher WK. Matrix metalloproteinases in stem cell mobilization. Matrix Biol. 2015; 44–46: 175 – 83.
53.
go back to reference Ding ZC, Shi H, Aboelelle NS, et al. Persistent STAT5 activation reprograms the epigenetic landscape in CD4(+) T cells to drive polyfunctionality and antitumor immunity. Sci Immunol. 2020;5:eaba5962.PubMedPubMedCentralCrossRef Ding ZC, Shi H, Aboelelle NS, et al. Persistent STAT5 activation reprograms the epigenetic landscape in CD4(+) T cells to drive polyfunctionality and antitumor immunity. Sci Immunol. 2020;5:eaba5962.PubMedPubMedCentralCrossRef
54.
go back to reference Trapnell C, Cacchiarelli D, Grimsby J, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014;32:381–6.PubMedPubMedCentralCrossRef Trapnell C, Cacchiarelli D, Grimsby J, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014;32:381–6.PubMedPubMedCentralCrossRef
55.
go back to reference Zhang Q, He Y, Luo N et al. Landscape and Dynamics of Single Immune Cells in Hepatocellular Carcinoma. Cell. 2019;179:829 – 45.e20. Zhang Q, He Y, Luo N et al. Landscape and Dynamics of Single Immune Cells in Hepatocellular Carcinoma. Cell. 2019;179:829 – 45.e20.
56.
57.
go back to reference Alshetaiwi H, Pervolarakis N, McIntyre LL, et al. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci Immunol. 2020;5:eaay6017.PubMedPubMedCentralCrossRef Alshetaiwi H, Pervolarakis N, McIntyre LL, et al. Defining the emergence of myeloid-derived suppressor cells in breast cancer using single-cell transcriptomics. Sci Immunol. 2020;5:eaay6017.PubMedPubMedCentralCrossRef
58.
go back to reference Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21:485–98.PubMedPubMedCentralCrossRef Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol. 2021;21:485–98.PubMedPubMedCentralCrossRef
60.
go back to reference Wang D, Yu Y, Haarberg K, et al. Dynamic change and impact of myeloid-derived suppressor cells in allogeneic bone marrow transplantation in mice. Biol Blood Marrow Transpl. 2013;19:692–702.CrossRef Wang D, Yu Y, Haarberg K, et al. Dynamic change and impact of myeloid-derived suppressor cells in allogeneic bone marrow transplantation in mice. Biol Blood Marrow Transpl. 2013;19:692–702.CrossRef
61.
go back to reference Kotsakis A, Harasymczuk M, Schilling B, et al. Myeloid-derived suppressor cell measurements in fresh and cryopreserved blood samples. J Immunol Methods. 2012;381:14–22.PubMedPubMedCentralCrossRef Kotsakis A, Harasymczuk M, Schilling B, et al. Myeloid-derived suppressor cell measurements in fresh and cryopreserved blood samples. J Immunol Methods. 2012;381:14–22.PubMedPubMedCentralCrossRef
62.
go back to reference Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood. 2010;116:354–65.PubMedCrossRef Grimwade D, Hills RK, Moorman AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood. 2010;116:354–65.PubMedCrossRef
63.
go back to reference Pei D. Leukolysin MMP25/MT6-MMP: a novel matrix metalloproteinase specifically expressed in the leukocyte lineage. Cell Res. 1999;9:291–303.PubMedCrossRef Pei D. Leukolysin MMP25/MT6-MMP: a novel matrix metalloproteinase specifically expressed in the leukocyte lineage. Cell Res. 1999;9:291–303.PubMedCrossRef
64.
go back to reference Kang T, Yi J, Guo A, et al. Subcellular distribution and cytokine- and chemokine- regulated secretion of leukolysin/MT6-MMP/MMP-25 in neutrophils. J Biol Chem. 2001;276:21960–8.PubMedCrossRef Kang T, Yi J, Guo A, et al. Subcellular distribution and cytokine- and chemokine- regulated secretion of leukolysin/MT6-MMP/MMP-25 in neutrophils. J Biol Chem. 2001;276:21960–8.PubMedCrossRef
65.
go back to reference Fortin CF, Sohail A, Sun Q, et al. MT6-MMP is present in lipid rafts and faces inward in living human PMNs but translocates to the cell surface during neutrophil apoptosis. Int Immunol. 2010;22:637–49.PubMedPubMedCentralCrossRef Fortin CF, Sohail A, Sun Q, et al. MT6-MMP is present in lipid rafts and faces inward in living human PMNs but translocates to the cell surface during neutrophil apoptosis. Int Immunol. 2010;22:637–49.PubMedPubMedCentralCrossRef
66.
go back to reference English WR, Velasco G, Stracke JO, et al. Catalytic activities of membrane-type 6 matrix metalloproteinase (MMP25). FEBS Lett. 2001;491:137–42.PubMedCrossRef English WR, Velasco G, Stracke JO, et al. Catalytic activities of membrane-type 6 matrix metalloproteinase (MMP25). FEBS Lett. 2001;491:137–42.PubMedCrossRef
67.
go back to reference Ardi VC, Kupriyanova TA, Deryugina EI, et al. Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis. Proc Natl Acad Sci. 2007;104:20262–7.PubMedPubMedCentralCrossRef Ardi VC, Kupriyanova TA, Deryugina EI, et al. Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis. Proc Natl Acad Sci. 2007;104:20262–7.PubMedPubMedCentralCrossRef
Metadata
Title
Single-cell analysis defines highly specific leukemia-induced neutrophils and links MMP8 expression to recruitment of tumor associated neutrophils during FGFR1 driven leukemogenesis
Authors
Tianxiang Hu
Bo Cheng
Atsuko Matsunaga
Ting Zhang
Xiaocui Lu
Hui Fang
Stephanie F. Mori
Xuexiu Fang
Gavin Wang
Hongyan Xu
Huidong Shi
John K. Cowell
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Experimental Hematology & Oncology / Issue 1/2024
Electronic ISSN: 2162-3619
DOI
https://doi.org/10.1186/s40164-024-00514-6

Other articles of this Issue 1/2024

Experimental Hematology & Oncology 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine