Cargando…

A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach

Breast cancer (BC) is one of the most common cancers, accounting for 2.3 million cases worldwide. BC can be molecularly subclassified into luminal A, luminal B HER2-, luminal B HER2+, HER2+, and triple-negative breast cancer (TNBC). These molecular subtypes differ in their prognosis and treatment st...

Descripción completa

Detalles Bibliográficos
Autores principales: Amer, Hoda T., Eissa, Reda A., El Tayebi, Hend M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9649622/
https://www.ncbi.nlm.nih.gov/pubmed/36387279
http://dx.doi.org/10.3389/fmolb.2022.1032517
_version_ 1784827836884844544
author Amer, Hoda T.
Eissa, Reda A.
El Tayebi, Hend M.
author_facet Amer, Hoda T.
Eissa, Reda A.
El Tayebi, Hend M.
author_sort Amer, Hoda T.
collection PubMed
description Breast cancer (BC) is one of the most common cancers, accounting for 2.3 million cases worldwide. BC can be molecularly subclassified into luminal A, luminal B HER2-, luminal B HER2+, HER2+, and triple-negative breast cancer (TNBC). These molecular subtypes differ in their prognosis and treatment strategies; thus, understanding the tumor microenvironment (TME) of BC could lead to new potential treatment strategies. The TME hosts a population of cells that act as antitumorigenic such as tumor-associated eosinophils or pro-tumorigenic such as cancer-associated fibroblasts (CAFs), tumor-associated neutrophils (TANs), monocytic-derived populations such as MDSCs, or most importantly “tumor-associated macrophages (TAMs),” which are derived from CD14(+) monocytes. TAMs are reported to have the pro-inflammatory phenotype M1, which is found only in the very early stages of tumor and is not correlated with progression; however, the M2 phenotype is anti-inflammatory that is correlated with tumor progression and metastasis. The current study focused on controlling the anti-inflammatory activity in TAMs of hormonal, HER2+, and TNBC by epigenetic fine-tuning of two immunomodulatory proteins, namely, CD80 and mesothelin (MSLN), which are known to be overexpressed in BC with pro-tumorigenic activity. Long non-coding RNAs are crucial key players in tumor progression whether acting as oncogenic or tumor suppressors. We focused on the regulatory role of MALAT1 and HOTAIR lncRNAs and their role in controlling the tumorigenic activity of TAMs. This study observed the impact of manipulation of MALAT1 and HOTAIR on the expression of both CD80 and MSLN in TAMs of BC. Moreover, we analyzed the interlinkage between HOTAIR and MALAT1 as regulators to one another in TAMs of BC. The current study reported an upstream regulatory effect of HOTAIR on MALAT1. Moreover, our results showed a promising use of MALAT1 and HOTAIR in regulating oncogenic immune-modulatory proteins MSLN and CD80 in TAMs of HER2+ and TNBC. The downregulation of MALAT1 and HOTAIR resulted in the upregulation of CD80 and MSLN, which indicates that they might have a cell-specific activity in TAMs. These data shed light on novel key players affecting the anti-inflammatory activity of TAMs as a possible therapeutic target in HER2+ and TNBC.
format Online
Article
Text
id pubmed-9649622
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-96496222022-11-15 A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach Amer, Hoda T. Eissa, Reda A. El Tayebi, Hend M. Front Mol Biosci Molecular Biosciences Breast cancer (BC) is one of the most common cancers, accounting for 2.3 million cases worldwide. BC can be molecularly subclassified into luminal A, luminal B HER2-, luminal B HER2+, HER2+, and triple-negative breast cancer (TNBC). These molecular subtypes differ in their prognosis and treatment strategies; thus, understanding the tumor microenvironment (TME) of BC could lead to new potential treatment strategies. The TME hosts a population of cells that act as antitumorigenic such as tumor-associated eosinophils or pro-tumorigenic such as cancer-associated fibroblasts (CAFs), tumor-associated neutrophils (TANs), monocytic-derived populations such as MDSCs, or most importantly “tumor-associated macrophages (TAMs),” which are derived from CD14(+) monocytes. TAMs are reported to have the pro-inflammatory phenotype M1, which is found only in the very early stages of tumor and is not correlated with progression; however, the M2 phenotype is anti-inflammatory that is correlated with tumor progression and metastasis. The current study focused on controlling the anti-inflammatory activity in TAMs of hormonal, HER2+, and TNBC by epigenetic fine-tuning of two immunomodulatory proteins, namely, CD80 and mesothelin (MSLN), which are known to be overexpressed in BC with pro-tumorigenic activity. Long non-coding RNAs are crucial key players in tumor progression whether acting as oncogenic or tumor suppressors. We focused on the regulatory role of MALAT1 and HOTAIR lncRNAs and their role in controlling the tumorigenic activity of TAMs. This study observed the impact of manipulation of MALAT1 and HOTAIR on the expression of both CD80 and MSLN in TAMs of BC. Moreover, we analyzed the interlinkage between HOTAIR and MALAT1 as regulators to one another in TAMs of BC. The current study reported an upstream regulatory effect of HOTAIR on MALAT1. Moreover, our results showed a promising use of MALAT1 and HOTAIR in regulating oncogenic immune-modulatory proteins MSLN and CD80 in TAMs of HER2+ and TNBC. The downregulation of MALAT1 and HOTAIR resulted in the upregulation of CD80 and MSLN, which indicates that they might have a cell-specific activity in TAMs. These data shed light on novel key players affecting the anti-inflammatory activity of TAMs as a possible therapeutic target in HER2+ and TNBC. Frontiers Media S.A. 2022-10-28 /pmc/articles/PMC9649622/ /pubmed/36387279 http://dx.doi.org/10.3389/fmolb.2022.1032517 Text en Copyright © 2022 Amer, Eissa and El Tayebi. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Molecular Biosciences
Amer, Hoda T.
Eissa, Reda A.
El Tayebi, Hend M.
A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach
title A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach
title_full A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach
title_fullStr A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach
title_full_unstemmed A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach
title_short A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach
title_sort cutting-edge immunomodulatory interlinkage between hotair and malat1 in tumor-associated macrophages in breast cancer: a personalized immunotherapeutic approach
topic Molecular Biosciences
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9649622/
https://www.ncbi.nlm.nih.gov/pubmed/36387279
http://dx.doi.org/10.3389/fmolb.2022.1032517
work_keys_str_mv AT amerhodat acuttingedgeimmunomodulatoryinterlinkagebetweenhotairandmalat1intumorassociatedmacrophagesinbreastcancerapersonalizedimmunotherapeuticapproach
AT eissaredaa acuttingedgeimmunomodulatoryinterlinkagebetweenhotairandmalat1intumorassociatedmacrophagesinbreastcancerapersonalizedimmunotherapeuticapproach
AT eltayebihendm acuttingedgeimmunomodulatoryinterlinkagebetweenhotairandmalat1intumorassociatedmacrophagesinbreastcancerapersonalizedimmunotherapeuticapproach
AT amerhodat cuttingedgeimmunomodulatoryinterlinkagebetweenhotairandmalat1intumorassociatedmacrophagesinbreastcancerapersonalizedimmunotherapeuticapproach
AT eissaredaa cuttingedgeimmunomodulatoryinterlinkagebetweenhotairandmalat1intumorassociatedmacrophagesinbreastcancerapersonalizedimmunotherapeuticapproach
AT eltayebihendm cuttingedgeimmunomodulatoryinterlinkagebetweenhotairandmalat1intumorassociatedmacrophagesinbreastcancerapersonalizedimmunotherapeuticapproach