Obesity Contributes to Transformation of Myometrial Stem-Cell Niche to Leiomyoma via Inducing Oxidative Stress, DNA Damage, Proliferation, and Extracellular Matrix Deposition

Leiomyomas (fibroids) are monoclonal tumors in which myometrial stem cells (MSCs) turn tumorigenic after mutation, abnormal methylation, or aberrant signaling. Several factors contribute to metabolic dysfunction in obesity, including abnormal cellular proliferation, oxidative stress, and DNA damage....

Full description

Bibliographic Details
Main Authors: Sadia Afrin, Gregory W. Kirschen, Mostafa A. Borahay
Format: Article
Language:English
Published: MDPI AG 2023-08-01
Series:Genes
Subjects:
Online Access:https://www.mdpi.com/2073-4425/14/8/1625
_version_ 1797584635935326208
author Sadia Afrin
Gregory W. Kirschen
Mostafa A. Borahay
author_facet Sadia Afrin
Gregory W. Kirschen
Mostafa A. Borahay
author_sort Sadia Afrin
collection DOAJ
description Leiomyomas (fibroids) are monoclonal tumors in which myometrial stem cells (MSCs) turn tumorigenic after mutation, abnormal methylation, or aberrant signaling. Several factors contribute to metabolic dysfunction in obesity, including abnormal cellular proliferation, oxidative stress, and DNA damage. The present study aims to determine how adipocytes and adipocyte-secreted factors affect changes in MSCs in a manner that promotes the growth of uterine leiomyomas. Myometrial stem cells were isolated from the uteri of patients by fluorescence-activated cell sorting (FACS) using CD44/Stro1 antibodies. Enzyme-linked immunosorbent assay (ELISA), Western blot, and immunocytochemistry assays were performed on human adipocytes (SW872) co-cultured with MSCs and treated with leptin or adiponectin to examine the effects of proliferation, extracellular matrix (ECM) deposition, oxidative damage, and DNA damage. Co-culture with SW872 increased MSC proliferation compared to MSC culture alone, according to 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) results. The expressions of PCNA and COL1A increased significantly with SW872 co-culture. In addition, the expression of these markers was increased after leptin treatment and decreased after adiponectin treatment in MSCs. The Wnt/β-catenin and TGF-β/SMAD signaling pathways promote proliferation and ECM deposition in uterine leiomyomas. The expression of Wnt4, β-catenin, TGFβ3, and pSMAD2/3 of MSCs was increased when co-cultured with adipocytes. We found that the co-culture of MSCs with adipocytes resulted in increased NOX4 expression, reactive oxygen species production, and γ-H2AX expression. Leptin acts by binding to its receptor (LEP-R), leading to signal transduction, resulting in the transcription of genes involved in cellular proliferation, angiogenesis, and glycolysis. In MSCs, co-culture with adipocytes increased the expression of LEP-R, pSTAT3/STAT3, and pERK1/2/ERK/12. Based on the above results, we suggest that obesity may mediate MSC initiation of tumorigenesis, resulting in leiomyomas.
first_indexed 2024-03-10T23:54:27Z
format Article
id doaj.art-888caca44e134133ad68c6d6a71f7af0
institution Directory Open Access Journal
issn 2073-4425
language English
last_indexed 2024-03-10T23:54:27Z
publishDate 2023-08-01
publisher MDPI AG
record_format Article
series Genes
spelling doaj.art-888caca44e134133ad68c6d6a71f7af02023-11-19T01:16:07ZengMDPI AGGenes2073-44252023-08-01148162510.3390/genes14081625Obesity Contributes to Transformation of Myometrial Stem-Cell Niche to Leiomyoma via Inducing Oxidative Stress, DNA Damage, Proliferation, and Extracellular Matrix DepositionSadia Afrin0Gregory W. Kirschen1Mostafa A. Borahay2Department of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD 21287, USADepartment of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD 21287, USADepartment of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD 21287, USALeiomyomas (fibroids) are monoclonal tumors in which myometrial stem cells (MSCs) turn tumorigenic after mutation, abnormal methylation, or aberrant signaling. Several factors contribute to metabolic dysfunction in obesity, including abnormal cellular proliferation, oxidative stress, and DNA damage. The present study aims to determine how adipocytes and adipocyte-secreted factors affect changes in MSCs in a manner that promotes the growth of uterine leiomyomas. Myometrial stem cells were isolated from the uteri of patients by fluorescence-activated cell sorting (FACS) using CD44/Stro1 antibodies. Enzyme-linked immunosorbent assay (ELISA), Western blot, and immunocytochemistry assays were performed on human adipocytes (SW872) co-cultured with MSCs and treated with leptin or adiponectin to examine the effects of proliferation, extracellular matrix (ECM) deposition, oxidative damage, and DNA damage. Co-culture with SW872 increased MSC proliferation compared to MSC culture alone, according to 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) results. The expressions of PCNA and COL1A increased significantly with SW872 co-culture. In addition, the expression of these markers was increased after leptin treatment and decreased after adiponectin treatment in MSCs. The Wnt/β-catenin and TGF-β/SMAD signaling pathways promote proliferation and ECM deposition in uterine leiomyomas. The expression of Wnt4, β-catenin, TGFβ3, and pSMAD2/3 of MSCs was increased when co-cultured with adipocytes. We found that the co-culture of MSCs with adipocytes resulted in increased NOX4 expression, reactive oxygen species production, and γ-H2AX expression. Leptin acts by binding to its receptor (LEP-R), leading to signal transduction, resulting in the transcription of genes involved in cellular proliferation, angiogenesis, and glycolysis. In MSCs, co-culture with adipocytes increased the expression of LEP-R, pSTAT3/STAT3, and pERK1/2/ERK/12. Based on the above results, we suggest that obesity may mediate MSC initiation of tumorigenesis, resulting in leiomyomas.https://www.mdpi.com/2073-4425/14/8/1625adipocytesadipokinesleptinfibroidcollagen
spellingShingle Sadia Afrin
Gregory W. Kirschen
Mostafa A. Borahay
Obesity Contributes to Transformation of Myometrial Stem-Cell Niche to Leiomyoma via Inducing Oxidative Stress, DNA Damage, Proliferation, and Extracellular Matrix Deposition
Genes
adipocytes
adipokines
leptin
fibroid
collagen
title Obesity Contributes to Transformation of Myometrial Stem-Cell Niche to Leiomyoma via Inducing Oxidative Stress, DNA Damage, Proliferation, and Extracellular Matrix Deposition
title_full Obesity Contributes to Transformation of Myometrial Stem-Cell Niche to Leiomyoma via Inducing Oxidative Stress, DNA Damage, Proliferation, and Extracellular Matrix Deposition
title_fullStr Obesity Contributes to Transformation of Myometrial Stem-Cell Niche to Leiomyoma via Inducing Oxidative Stress, DNA Damage, Proliferation, and Extracellular Matrix Deposition
title_full_unstemmed Obesity Contributes to Transformation of Myometrial Stem-Cell Niche to Leiomyoma via Inducing Oxidative Stress, DNA Damage, Proliferation, and Extracellular Matrix Deposition
title_short Obesity Contributes to Transformation of Myometrial Stem-Cell Niche to Leiomyoma via Inducing Oxidative Stress, DNA Damage, Proliferation, and Extracellular Matrix Deposition
title_sort obesity contributes to transformation of myometrial stem cell niche to leiomyoma via inducing oxidative stress dna damage proliferation and extracellular matrix deposition
topic adipocytes
adipokines
leptin
fibroid
collagen
url https://www.mdpi.com/2073-4425/14/8/1625
work_keys_str_mv AT sadiaafrin obesitycontributestotransformationofmyometrialstemcellnichetoleiomyomaviainducingoxidativestressdnadamageproliferationandextracellularmatrixdeposition
AT gregorywkirschen obesitycontributestotransformationofmyometrialstemcellnichetoleiomyomaviainducingoxidativestressdnadamageproliferationandextracellularmatrixdeposition
AT mostafaaborahay obesitycontributestotransformationofmyometrialstemcellnichetoleiomyomaviainducingoxidativestressdnadamageproliferationandextracellularmatrixdeposition