Cargando…

BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution

Rationale: In the bone marrow microenvironment (BMME), mesenchymal stem/stromal cells (MSCs) control the self-renewal of both healthy and cancerous hematopoietic stem/progenitor cells (HSPCs). We previously showed that in vivo leukemia-derived MSCs change neighbor MSCs into leukemia-permissive state...

Descripción completa

Detalles Bibliográficos
Autores principales: Chai, Chengyan, Sui, Ke, Tang, Jun, Yu, Hao, Yang, Chao, Zhang, Hongyang, Li, Shengwen Calvin, Zhong, Jiang F., Wang, Zheng, Zhang, Xi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10405834/
https://www.ncbi.nlm.nih.gov/pubmed/37554265
http://dx.doi.org/10.7150/thno.83178
_version_ 1785085620668858368
author Chai, Chengyan
Sui, Ke
Tang, Jun
Yu, Hao
Yang, Chao
Zhang, Hongyang
Li, Shengwen Calvin
Zhong, Jiang F.
Wang, Zheng
Zhang, Xi
author_facet Chai, Chengyan
Sui, Ke
Tang, Jun
Yu, Hao
Yang, Chao
Zhang, Hongyang
Li, Shengwen Calvin
Zhong, Jiang F.
Wang, Zheng
Zhang, Xi
author_sort Chai, Chengyan
collection PubMed
description Rationale: In the bone marrow microenvironment (BMME), mesenchymal stem/stromal cells (MSCs) control the self-renewal of both healthy and cancerous hematopoietic stem/progenitor cells (HSPCs). We previously showed that in vivo leukemia-derived MSCs change neighbor MSCs into leukemia-permissive states and boost leukemia cell proliferation, survival, and chemotherapy resistance. But the mechanisms behind how the state changes are still not fully understood. Methods: Here, we took a reverse engineering approach to determine BCR-ABL1+ leukemia cells activated transcriptional factor C/EBPβ, resulting in miR130a/b-3p production. Then, we back-tracked from clinical specimen transcriptome sequencing to cell co-culture, molecular and cellular assays, flow cytometry, single-cell transcriptome, and transcriptional regulation to determine the molecular mechanisms of BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications. Results: BCR-ABL1-driven exosome-miR130a/b-3p mediated gap-junction Cx43 (a.k.a., GJA1) BMSC intercellular communications for subclonal evolution in leukemic microenvironment by targeting BMSCs-expressed HLAs, thereby potentially maintaining BMSCs with self-renewal properties and reduced BMSC immunogenicity. The Cx43(low) and miR-130a/b(high) subclonal MSCs subsets of differentiation state could be reversed to Cx43(high) and miR-130a/b(low) subclones of the higher stemness state in Cx43-overexpressed subclonal MSCs. Both miR-130a and miR-130b might only inhibit Cx43 translation or degrade Cx43 proteins and did not affect Cx43 mRNA stability. The subclonal evolution was further confirmed by single-cell transcriptome profiling of MSCs, which suggested that Cx43 regulated their stemness and played normal roles in immunomodulation antigen processing. Thus, upregulated miR-130a/b promoted osteogenesis and adipogenesis from BMSCs, thereby decreasing cancer progression. Our clinical data validated that the expression of many genes in human major histocompatibility was negatively associated with the stemness of MSCs, and several immune checkpoint proteins contributing to immune escape in tumors were overexpressed after either miR-130a or miR-130b overexpression, such as CD274, LAG3, PDCD1, and TNFRSF4. Not only did immune response-related cytokine-cytokine receptor interactions and PI3K-AKT pathways, including EGR3, TNFRSF1B, but also NDRG2 leukemic-associated inflammatory factors, such as IFNB1, CXCL1, CXCL10, and CCL7 manifest upon miR-130a/b overexpression. Either BCR siRNAs or ABL1 siRNAs assay showed significantly decreased miR-130a and miR-130b expression, and chromatin immunoprecipitation sequencing confirmed that the regulation of miR-130a and miR-130b expression is BCR-ABL1-dependent. BCR-ABL1 induces miR-130a/b expression through the upregulation of transcriptional factor C/EBPβ. C/EBPβ could bind directly to the promoter region of miR-130b-3p, not miR-130a-3p. BCR-ABL1-driven exosome-miR130a-3p could interact with Cx43, and further impact GJIC in TME. Conclusion: Our findings shed light on how leukemia BCR-ABL1-driven exosome-miR130b-3p could interact with gap-junction Cx43, and further impact GJIC in TME, implications for leukemic therapies of subclonal evolution.
format Online
Article
Text
id pubmed-10405834
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-104058342023-08-08 BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution Chai, Chengyan Sui, Ke Tang, Jun Yu, Hao Yang, Chao Zhang, Hongyang Li, Shengwen Calvin Zhong, Jiang F. Wang, Zheng Zhang, Xi Theranostics Research Paper Rationale: In the bone marrow microenvironment (BMME), mesenchymal stem/stromal cells (MSCs) control the self-renewal of both healthy and cancerous hematopoietic stem/progenitor cells (HSPCs). We previously showed that in vivo leukemia-derived MSCs change neighbor MSCs into leukemia-permissive states and boost leukemia cell proliferation, survival, and chemotherapy resistance. But the mechanisms behind how the state changes are still not fully understood. Methods: Here, we took a reverse engineering approach to determine BCR-ABL1+ leukemia cells activated transcriptional factor C/EBPβ, resulting in miR130a/b-3p production. Then, we back-tracked from clinical specimen transcriptome sequencing to cell co-culture, molecular and cellular assays, flow cytometry, single-cell transcriptome, and transcriptional regulation to determine the molecular mechanisms of BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications. Results: BCR-ABL1-driven exosome-miR130a/b-3p mediated gap-junction Cx43 (a.k.a., GJA1) BMSC intercellular communications for subclonal evolution in leukemic microenvironment by targeting BMSCs-expressed HLAs, thereby potentially maintaining BMSCs with self-renewal properties and reduced BMSC immunogenicity. The Cx43(low) and miR-130a/b(high) subclonal MSCs subsets of differentiation state could be reversed to Cx43(high) and miR-130a/b(low) subclones of the higher stemness state in Cx43-overexpressed subclonal MSCs. Both miR-130a and miR-130b might only inhibit Cx43 translation or degrade Cx43 proteins and did not affect Cx43 mRNA stability. The subclonal evolution was further confirmed by single-cell transcriptome profiling of MSCs, which suggested that Cx43 regulated their stemness and played normal roles in immunomodulation antigen processing. Thus, upregulated miR-130a/b promoted osteogenesis and adipogenesis from BMSCs, thereby decreasing cancer progression. Our clinical data validated that the expression of many genes in human major histocompatibility was negatively associated with the stemness of MSCs, and several immune checkpoint proteins contributing to immune escape in tumors were overexpressed after either miR-130a or miR-130b overexpression, such as CD274, LAG3, PDCD1, and TNFRSF4. Not only did immune response-related cytokine-cytokine receptor interactions and PI3K-AKT pathways, including EGR3, TNFRSF1B, but also NDRG2 leukemic-associated inflammatory factors, such as IFNB1, CXCL1, CXCL10, and CCL7 manifest upon miR-130a/b overexpression. Either BCR siRNAs or ABL1 siRNAs assay showed significantly decreased miR-130a and miR-130b expression, and chromatin immunoprecipitation sequencing confirmed that the regulation of miR-130a and miR-130b expression is BCR-ABL1-dependent. BCR-ABL1 induces miR-130a/b expression through the upregulation of transcriptional factor C/EBPβ. C/EBPβ could bind directly to the promoter region of miR-130b-3p, not miR-130a-3p. BCR-ABL1-driven exosome-miR130a-3p could interact with Cx43, and further impact GJIC in TME. Conclusion: Our findings shed light on how leukemia BCR-ABL1-driven exosome-miR130b-3p could interact with gap-junction Cx43, and further impact GJIC in TME, implications for leukemic therapies of subclonal evolution. Ivyspring International Publisher 2023-07-03 /pmc/articles/PMC10405834/ /pubmed/37554265 http://dx.doi.org/10.7150/thno.83178 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Chai, Chengyan
Sui, Ke
Tang, Jun
Yu, Hao
Yang, Chao
Zhang, Hongyang
Li, Shengwen Calvin
Zhong, Jiang F.
Wang, Zheng
Zhang, Xi
BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution
title BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution
title_full BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution
title_fullStr BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution
title_full_unstemmed BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution
title_short BCR-ABL1-driven exosome-miR130b-3p-mediated gap-junction Cx43 MSC intercellular communications imply therapies of leukemic subclonal evolution
title_sort bcr-abl1-driven exosome-mir130b-3p-mediated gap-junction cx43 msc intercellular communications imply therapies of leukemic subclonal evolution
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10405834/
https://www.ncbi.nlm.nih.gov/pubmed/37554265
http://dx.doi.org/10.7150/thno.83178
work_keys_str_mv AT chaichengyan bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT suike bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT tangjun bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT yuhao bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT yangchao bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT zhanghongyang bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT lishengwencalvin bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT zhongjiangf bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT wangzheng bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution
AT zhangxi bcrabl1drivenexosomemir130b3pmediatedgapjunctioncx43mscintercellularcommunicationsimplytherapiesofleukemicsubclonalevolution