1
|
Li J, Liu L, Fan R. The PKM2/HIF-1α Axis is Involved in the Pathogenesis of Endometriosis via TGF-β1 under Endometrial Polyps. FRONT BIOSCI-LANDMRK 2024; 29:417. [PMID: 39735997 DOI: 10.31083/j.fbl2912417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Endometriosis patients exhibit a cancer-like glycolytic phenotype. The pyruvate kinase M2 (PKM2)/hypoxia-inducible factor-1 alpha (HIF-1α) axis plays important roles in glycolysis-related diseases, but its role in patients with endometrial polyps (EPs) combined with endometriosis has not been validated. METHODS EP samples were collected from patients with and without endometriosis. PKM2, HIF-1α, and transforming growth factor-beta 1 (TGF-β1) levels were detected by immunohistochemistry (IHC), quantitative polymerase chain reaction, western blotting, and/or immunofluorescence. Primary endometrial stromal cells (ESCs) and non-endometriotic patient-derived ESCs (NESCs) were isolated from patients with EP with or without endometriosis. PKM2 loss-of-function assays in ESCs and gain-of-function assays in NESCs were performed to assess the function of PKM2. The effects of PKM2 and TGF-β1 on the promoter activity of HIF-1α were determined by dual-luciferase reporter assay. RESULTS PKM2 was overexpressed in ESCs compared to NESCs. Furthermore, PKM2 knockdown repressed viability, decreased migration and invasion, and restrained glycolysis of ESCs, accompanied by reduced HIF-1α levels and weakened promoter activity of HIF-1α. In addition, PKM2 overexpression had the opposite effect on these indicators in NESCs. Of note, an anti-TGF-β1 Ab reversed the PKM2-overexpression-mediated effects on cell viability, migration, and invasion, but not glycolysis or HIF-1α promoter activity, in NESCs. Additionally, PKM2, HIF-1α, and TGF-β1 levels were higher in EP samples with endometriosis than in EP samples without endometriosis, and there were positive correlations between PKM2, HIF-1α, and TGF-β1 IHC scores in all EP samples. CONCLUSIONS PKM2/HIF-1α-axis-dependent glycolysis participates in the pathogenesis of EP combined with endometriosis by mediating TGF-β1 signaling.
Collapse
Affiliation(s)
- Jianjuan Li
- Department of Reproductive Medicine, Dongying People's Hospital, 257091 Dongying, Shandong, China
| | - Li Liu
- Department of Obstetrics, Dongying People's Hospital, 257091 Dongying, Shandong, China
| | - Ruiqi Fan
- Department of Reproductive Medicine, Dongying People's Hospital, 257091 Dongying, Shandong, China
| |
Collapse
|
2
|
Parpex G, Chassaing B, Bourdon M, Santulli P, Doridot L, Thomas M, Batteux F, Chouzenoux S, Chapron C, Nicco C, Marcellin L. Western diet promotes endometriotic lesion growth in mice and induces depletion of Akkermansia muciniphila in intestinal microbiota. BMC Med 2024; 22:513. [PMID: 39501247 PMCID: PMC11539706 DOI: 10.1186/s12916-024-03738-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Endometriosis, affecting 10% of women in their reproductive years, remains poorly understood. Both individual and environmental unexplained factors are implicated in this heterogenous condition. This study aims to examine the influence of a Western diet on endometriosis lesion development in mice and to uncover the mechanisms involved. METHODS Mice were fed either a control diet or a Western diet (high in fatty acids and low in fiber) for 4 weeks. Endometriosis was then surgically induced, and lesion development was monitored by ultrasound. After 7 weeks, the mice were sacrificed for analysis of lesion characteristics through RT-qPCR, immunohistochemistry, and flow cytometry. Additionally, the intestinal microbiota was assessed using 16S rRNA gene sequencing. RESULTS Mice on the Western diet developed lesions that were significantly twice as large compared to those on the control diet. These lesions exhibited greater fibrosis and proliferation, alongside enhanced macrophage activity and leptin pathway expression. Changes in the intestinal microbiota were significantly noted after endometriosis induction, regardless of diet. Notably, mice on the Western diet with the most substantial lesions showed a loss of Akkermansia Muciniphila in their intestinal microbiota. CONCLUSIONS A Western diet significantly exacerbates lesion size in a mouse model of endometriosis, accompanied by metabolic and immune alterations. The onset of endometriosis also leads to substantial shifts in intestinal microbiota, suggesting a potential link between diet, intestinal health, and endometriosis development.
Collapse
Affiliation(s)
- Guillaume Parpex
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France.
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France.
| | - Benoît Chassaing
- Institut Pasteur, Université Paris Cité, Microbiome-Host Interaction Group, INSERM U1306, Paris, France
| | - Mathilde Bourdon
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Pietro Santulli
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Ludivine Doridot
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Marine Thomas
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Frédéric Batteux
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | | | - Charles Chapron
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Carole Nicco
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| | - Louis Marcellin
- Department of Gynecology Obstetrics II and Reproductive Medicine (Professor Chapron), Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, 123 boulevard de Port-Royal, Paris, 75014, France
- Université Paris Cité, CNRS, Institut Cochin, Paris, Inserm, France
| |
Collapse
|
3
|
Yang R, Yang F, Wei Y, Huang B, Cao T, Tan H, Liu D, Zou Q, Wen J, Wen L, Lu X, Yu C, Cai H, Xie X, Jiang S, Yao S, Liang Y. Hypoxia-induced Semaphorin 3A promotes the development of endometriosis through regulating macrophage polarization. Int Immunopharmacol 2024; 138:112559. [PMID: 38955028 DOI: 10.1016/j.intimp.2024.112559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Semaphorin 3A (Sema3A) is a member of neural guidance factor family well-known for inducing the collapse of nerve cell growth cone and regulating nerve redistribution. It also has been characterized as an immunoregulatory and tumor promoting factor. Our previous study showed that Sema3A was involved in the regulation of sympathetic innervation and neuropathic pain of endometriosis. Nevertheless, the role of Sema3A in the development of endometriosis and its potential upstreaming factor are still not clear. METHODS Histology experiments were carried to detect the expression of Sema3A, hypoxia -inducible factor 1α (HIF-1α) and the distribution of macrophages. Cell experiments were used to explore the effect of Sema3A on the proliferation and migration of endometrial stromal cells (ESCs) and to confirm the regulatory action of HIF-1α on Sema3A. In vivo experiments were carried out to explore the role of Sema3A on the development of endometriosis. RESULTS Sema3A was highly expressed in endometriotic lesions and could enhanced the proliferation and migration abilities of ESCs. Aberrant macrophage distribution was found in endometriotic lesions. Sema3A also promoted the differentiation of monocytes into anti-inflammatory macrophages, so indirectly mediating the proliferation and migration of ESCs. Hypoxic microenvironment induced Sema3A mRNA and protein expression in ESCs via HIF-1α. Administration of Sema3A promoted the development of endometriosis in a mouse model. CONCLUSIONS Sema3A, which is regulated by HIF-1α, is a promoting factor for the development of endometriosis. Targeting Sema3A may be a potential treatment strategy to control endometriotic lesions.
Collapse
Affiliation(s)
- Ruyu Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China
| | - Fan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China
| | - Yajing Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China
| | - Biqi Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China
| | - Tiefeng Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China
| | - Hao Tan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China
| | - Duo Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China
| | - Qiuyu Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China
| | - Jinjuan Wen
- Department of Obstetrics and Gynecology, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-sen University), Jieyang 522081, Guangdong, China
| | - Lei Wen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Xi Lu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Changyang Yu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| | - Heng Cai
- Department of Obstetrics and Gynecology, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-sen University), Jieyang 522081, Guangdong, China
| | - Xiaofei Xie
- Department of Obstetrics and Gynecology, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-sen University), Jieyang 522081, Guangdong, China
| | - Shaoru Jiang
- Department of Obstetrics and Gynecology, Jieyang People's Hospital (Jieyang Affiliated Hospital, Sun Yat-sen University), Jieyang 522081, Guangdong, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China.
| | - Yanchun Liang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China; Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, Guangdong, China; Department of Obstetrics and Gynecology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning 530022, Guangxi, China.
| |
Collapse
|
4
|
Sarsenova M, Lawarde A, Pathare ADS, Saare M, Modhukur V, Soplepmann P, Terasmaa A, Käämbre T, Gemzell-Danielsson K, Lalitkumar PGL, Salumets A, Peters M. Endometriotic lesions exhibit distinct metabolic signature compared to paired eutopic endometrium at the single-cell level. Commun Biol 2024; 7:1026. [PMID: 39169201 PMCID: PMC11339455 DOI: 10.1038/s42003-024-06713-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Current therapeutics of endometriosis focus on hormonal disruption of endometriotic lesions (ectopic endometrium, EcE). Recent findings show higher glycolysis utilization in EcE, suggesting non-hormonal strategy for disease treatment that addresses cellular metabolism. Identifying metabolically altered cell types in EcE is important for targeted metabolic drug therapy without affecting eutopic endometrium (EuE). Here, using single-cell RNA-sequencing, we examine twelve metabolic pathways in paired samples of EuE and EcE from women with confirmed endometriosis. We detect nine major cell types in both EuE and EcE. Metabolic pathways are most differentially regulated in perivascular, stromal, and endothelial cells, with the highest changes in AMPK signaling, HIF-1 signaling, glutathione metabolism, oxidative phosphorylation, and glycolysis. We identify transcriptomic co-activation of glycolytic and oxidative metabolism in perivascular and stromal cells of EcE, indicating a critical role of metabolic reprogramming in maintaining endometriotic lesion growth. Perivascular cells, involved in endometrial stroma repair and angiogenesis, may be potential targets for non-hormonal treatment of endometriosis.
Collapse
Affiliation(s)
- Meruert Sarsenova
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Division of Neonatology, Obstetrics and Gynecology, and Reproductive Health, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- WHO Collaborating Centre, Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Ankita Lawarde
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Amruta D S Pathare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Vijayachitra Modhukur
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | | | - Anton Terasmaa
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Tuuli Käämbre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Tallinn, Estonia
| | - Kristina Gemzell-Danielsson
- Division of Neonatology, Obstetrics and Gynecology, and Reproductive Health, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- WHO Collaborating Centre, Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Parameswaran Grace Luther Lalitkumar
- Division of Neonatology, Obstetrics and Gynecology, and Reproductive Health, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- WHO Collaborating Centre, Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.
- Competence Centre on Health Technologies, Tartu, Estonia.
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden.
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Maire Peters
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| |
Collapse
|
5
|
Wen X, Zhang J, Xu Z, Li M, Dong X, Du Y, Xu Z, Yan L. Highly expressed lncRNA H19 in endometriosis promotes aerobic glycolysis and histone lactylation. Reproduction 2024; 168:e240018. [PMID: 38744310 PMCID: PMC11301422 DOI: 10.1530/rep-24-0018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/13/2024] [Indexed: 05/16/2024]
Abstract
In brief Abnormal glucose metabolism may be involved in the pathogenesis of endometriosis. The present study identifies that highly expressed H19 leads to increased aerobic glycolysis and histone lactylation levels in endometriosis. Abstract Previous studies from our group and others have shown increased IncRNA H19 expression in both the eutopic endometrium and the ectopic endometriosis tissue during endometriosis. In this study, we use immunofluorescence, immunohistochemistry, and protein quantification to determine that levels of aerobic glycolysis and histone lactylation are increased in endometriosis tissues. In human endometrial stromal cells, we found that high H19 expression resulted in abnormal glucose metabolism by examining the levels of glucose, lactate, and ATP and measuring protein levels of enzymes that participate in glycolysis. At the same time, immunofluorescence and western blotting demonstrated increased histone lactylation in H19 overexpressing cells. Altering aerobic glycolysis and histone lactylation levels through the addition of sodium lactate and 2-deoxy-d-glucose demonstrated that increased aerobic glycolysis and histone lactylation levels resulted in enhanced cell proliferation and cell migration, contributing to endometriosis. To validate these findings in vivo, we constructed an endometriosis mouse model, demonstrating similar changes in endometriosis tissues in vivo. Both aerobic glycolysis and histone lactylation levels were elevated in endometriotic lesions. Taken together, these data demonstrate elevated expression levels of H19 in endometriosis patients promote abnormal glucose metabolism and elevated histone lactylation levels in vivo, enhancing cell proliferation and migration and promoting the progression of endometriosis. Our study provides a functional link between H19 expression and histone lactylation and glucose metabolism in endometriosis, providing new insights into disease mechanisms that could result in novel therapeutic approaches.
Collapse
Affiliation(s)
- Xiaoyang Wen
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Reproductive Hospital Affiliated to Shandong University, Jinan, Shandong, People’s Republic of China
| | - Jingyang Zhang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Reproductive Hospital Affiliated to Shandong University, Jinan, Shandong, People’s Republic of China
| | - Zihan Xu
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Reproductive Hospital Affiliated to Shandong University, Jinan, Shandong, People’s Republic of China
| | - Muzi Li
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Reproductive Hospital Affiliated to Shandong University, Jinan, Shandong, People’s Republic of China
| | - Xiaotong Dong
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Reproductive Hospital Affiliated to Shandong University, Jinan, Shandong, People’s Republic of China
| | - Yanbo Du
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Reproductive Hospital Affiliated to Shandong University, Jinan, Shandong, People’s Republic of China
| | - Zhen Xu
- Center for Medical Genetics and Prenatal Diagnosis, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, Shandong, China
- Shandong Medicine and Health Key Laboratory of Birth Defect Prevention and Genetic Medicine, Jinan, Shandong, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Jinan, Shandong, China
| | - Lei Yan
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Reproductive Medicine, Jinan, Shandong, People’s Republic of China
- Reproductive Hospital Affiliated to Shandong University, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
6
|
Dai W, Guo R, Na X, Jiang S, Liang J, Guo C, Fang Y, Na Z, Li D. Hypoxia and the endometrium: An indispensable role for HIF-1α as therapeutic strategies. Redox Biol 2024; 73:103205. [PMID: 38815332 PMCID: PMC11167393 DOI: 10.1016/j.redox.2024.103205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024] Open
Abstract
Hypoxia-inducible factor 1 alpha (HIF-1α) is a major molecular mediator of the hypoxic response. In the endometrium, local hypoxic conditions induced by hormonal fluctuations and endometrial vascular remodeling contribute to the production of HIF-1α, which plays an indispensable role in a series of physiological activities, such as menstruation and metamorphosis. The sensitive regulation of HIF-1α maintains the cellular viability and regenerative capacity of the endometrium against cellular stresses induced by hypoxia and excess reactive oxygen species. In contrast, abnormal HIF-1α levels exacerbate the development of various endometrial pathologies. This knowledge opens important possibilities for the development of promising HIF-1α-centered strategies to ameliorate endometrial disease. Nonetheless, additional efforts are required to elucidate the regulatory network of endometrial HIF-1α and promote the applications of HIF-1α-centered strategies in the human endometrium. Here, we summarize the role of the HIF-1α-mediated pathway in endometrial physiology and pathology, highlight the latest HIF-1α-centered strategies for treating endometrial diseases, and improve endometrial receptivity.
Collapse
Affiliation(s)
- Wanlin Dai
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Renhao Guo
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinni Na
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuyi Jiang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Junzhi Liang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cuishan Guo
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Fang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China; NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
| | - Zhijing Na
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China; NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
| | - Da Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China; NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China; Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China.
| |
Collapse
|
7
|
Kobayashi H, Matsubara S, Yoshimoto C, Shigetomi H, Imanaka S. The role of mitochondrial dynamics in the pathophysiology of endometriosis. J Obstet Gynaecol Res 2023; 49:2783-2791. [PMID: 37681703 DOI: 10.1111/jog.15791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Abstract
AIM Endometriosis is a chronic disease of reproductive age, associated with pelvic pain and infertility. Endometriotic cells adapt to changing environments such as oxidative stress and hypoxia in order to survive. However, the underlying mechanisms remain to be fully elucidated. In this review, we summarize our current understanding of the pathogenesis of endometriosis, focusing primarily on the molecular basis of energy metabolism, redox homeostasis, and mitochondrial function, and discuss perspectives on future research directions. METHODS Papers published up to March 31, 2023 in the PubMed and Google Scholar databases were included in this narrative literature review. RESULTS Mitochondria serve as a central hub sensing a multitude of physiological processes, including energy production and cellular redox homeostasis. Under hypoxia, endometriotic cells favor glycolysis and actively produce pyruvate, nicotinamide adenine dinucleotide phosphate (NADPH), and other metabolites for cell proliferation. Mitochondrial fission and fusion dynamics may regulate the phenotypic plasticity of cellular energy metabolism, that is, aerobic glycolysis or OXPHOS. Endometriotic cells have been reported to have reduced mitochondrial numbers, increased lamellar cristae, improved energy efficiency, and enhanced cell proliferation and survival. Increased mitochondrial fission and fusion turnover by hypoxic and normoxic conditions suggests an activation of mitochondrial quality control mechanisms. Recently, candidate molecules that influence mitochondrial dynamics have begun to be identified. CONCLUSION This review suggests that unique energy metabolism and redox homeostasis driven by mitochondrial dynamics may be linked to the pathophysiology of endometriosis. However, further studies are needed to elucidate the regulatory mechanisms of mitochondrial dynamics in endometriosis.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Medicine, Kei Oushin Clinic, Nishinomiya, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, Nara, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, Nara, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| |
Collapse
|
8
|
Nothnick WB, Arachchige SP, Minchella P, Stephens EB, Graham A. Targeting c-MYC: a potential non-hormonal therapeutic approach for endometriosis treatment. Front Cell Dev Biol 2023; 11:1225055. [PMID: 38078012 PMCID: PMC10702580 DOI: 10.3389/fcell.2023.1225055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/01/2023] [Indexed: 01/23/2025] Open
Abstract
Endometriosis is a benign gynecological disease in which eutopic endometrial tissue composed of glands and stroma grow within the pelvic cavity. The disease affects females of reproductive age and is characterized by pelvic pain, infertility and reduced quality of life. The majority of pharmacologic treatment modalities for endometriosis focus on suppression of estradiol production and/or action; an approach associated with adverse side effects. c-MYC is elevated in eutopic endometrium and endometriotic lesion tissue in patients with endometriosis and the disease shares many similar pathological characteristics with that of endometrial carcinoma. While targeting of c-MYC with Omomyc has recently gained substantial interest in the field of cancer research, there has been no recent attempt to evaluate the potential utility in targeting c-MYC for endometriosis treatment. The following perspective article compares the similarities between endometriosis and endometrial cancer and presents preliminary data suggesting that targeting c-MYC with Omomyc reduces endometriotic cell proliferation and viability in vitro. Future application of targeting c-MYC in endometriosis treatment and potential pros and cons are then discussed.
Collapse
Affiliation(s)
- Warren B. Nothnick
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Center for Reproductive Sciences, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sachith Polpitiya Arachchige
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Paige Minchella
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Edward B. Stephens
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Amanda Graham
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
9
|
Chen J, Qin P, Sun Y, Han S. Histone lactylation promotes cell proliferation, migration and invasion through targeting HMGB1 in endometriosis. J Biomed Res 2023; 37:470-478. [PMID: 37945340 PMCID: PMC10687535 DOI: 10.7555/jbr.37.20230095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 11/12/2023] Open
Abstract
Endometriosis is defined as a condition with endometrium-like tissues migrating outside of the pelvic cavity. However, the mechanism of endometriosis is still unclear. Lactate can be covalently modified to lysine residues of histones and other proteins, which is called lactylation. The results showed that the higher level of lactate and lactate dehydrogenase A enhanced the histone H3 lysine 18 lactylation (H3K18lac) in ectopic endometrial tissues and ectopic endometrial stromal cells than that in normal endometrial tissues and normal endometrial stromal cells. Lactate promoted cell proliferation, migration, and invasion in endometriosis. Mechanistically, lactate induced H3K18lac to promote the expression of high-mobility group box 1 (HMGB1) in endometriosis, and HMGB1 knockdown significantly reduced the cell proliferation, migration, and invasion of the lactate-treated cells through the phosphorylation of AKT. In conclusion, lactate could induce histone lactylation to promote endometriosis progression by upregulating the expression of HMGB1, which may provide a novel target for the prevention and treatment of endometriosis.
Collapse
Affiliation(s)
- Jie Chen
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, China
| | - Pengfei Qin
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, China
| | - Yanli Sun
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, China
| | - Suping Han
- Department of Gynecology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210004, China
| |
Collapse
|
10
|
Chang LY, Shan J, Hou XX, Li DJ, Wang XQ. Synergy between Th1 and Th2 responses during endometriosis: A review of current understanding. J Reprod Immunol 2023; 158:103975. [PMID: 37331087 DOI: 10.1016/j.jri.2023.103975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/27/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
Endometriosis is widely perceived as an estrogen-dependent chronic disorder with infertility and pelvic pain. Although the etiology of endometriosis has remained elusive, many studies have proclaimed the relevance of immune system disorders with endometriosis. With the discovery that the dysregulation of multiple biological functions in endometriosis is caused by the aberrant differentiation of T helper cells, a shift towards Th2 immune response may account for the disease progression. This review attempts to present mechanisms of cytokines, chemokines, signal pathways, transcription factors and some other factors related with the derivation of Th1/Th2 immune response involved in the development of endometriosis. The current understanding of treatment approaches and potential therapeutic targets will also be outlined with brief discussion.
Collapse
Affiliation(s)
- Ling-Yu Chang
- Hospital of Obstetrics and Gynecology, Fudan University, Shanghai Medical College, Shanghai 200011, China
| | - Jing Shan
- Hospital of Obstetrics and Gynecology, Fudan University, Shanghai Medical College, Shanghai 200011, China
| | - Xin-Xin Hou
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Da-Jin Li
- Hospital of Obstetrics and Gynecology, Fudan University, Shanghai Medical College, Shanghai 200011, China.
| | - Xiao-Qiu Wang
- Hospital of Obstetrics and Gynecology, Fudan University, Shanghai Medical College, Shanghai 200011, China.
| |
Collapse
|
11
|
Crookenden MA, Burke CR, Mitchell MD, Phyn CVC, Roche JR, Heiser A. Effect of nonsteroidal anti-inflammatory drugs on the inflammatory response of bovine endometrial epithelial cells in vitro. J Dairy Sci 2023; 106:2651-2666. [PMID: 36653292 DOI: 10.3168/jds.2021-21742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 09/30/2022] [Indexed: 01/19/2023]
Abstract
Chronic postpartum uterine infection detrimentally affects subsequent fertility. Nonsteroidal anti-inflammatory drugs (NSAID) are used to alleviate pain and treat inflammatory conditions in transition dairy cows with varying success. To screen the efficacy of NSAID in the absence of animal experiments, we have established an in vitro model to study uterine inflammation. Inflammation was induced in cultured bovine endometrial epithelial cells by challenging cells with an inflammation cocktail: lipopolysaccharide and proinflammatory cytokines, interleukin-1β (IL1β) and tumor necrosis factor α (TNFα). Release of the inflammation markers, serum amyloid A (SAA) and α-1-acid glycoprotein (αAGP), was measured by ELISA. Concentration of these markers was used to indicate the effectiveness in dampening inflammation of 5 NSAID: meloxicam, flunixin meglumine, aspirin, ketoprofen, and tolfenamic acid. Three NSAID, meloxicam, flunixin meglumine, and tolfenamic acid, were successful at dampening the release of SAA and αAGP into cell-culture supernatant, and the corresponding treated cells were selected for down-stream mRNA expression analysis. Expression of 192 genes involved in regulation of inflammatory pathways were investigated using Nanostring. Of the genes investigated, 81 were above the mRNA expression-analysis threshold criteria and were included in expression analysis. All SAA genes investigated (SAA2, SAA3, M-SAA3.2) were upregulated in response to the inflammation cocktail, relative to mRNA expression in control cells; however, AGP mRNA expression was below the expression analysis threshold and was, therefore, excluded from analysis. Treatment with NSAID downregulated genes involved in regulating chemokine signaling (e.g., CXCL2, CXCR4, CXCL5, and CXCL16) and genes that regulate the eicosanoid pathway (e.g., LTA4H, PTGS2, PLA2G4A, and PTGDS). Of the 5 NSAID investigated, meloxicam, flunixin meglumine, and tolfenamic acid are recommended for further investigation into treatment of postpartum uterine inflammation. The results from this study confirm the immunomodulatory properties of the endometrial epithelium in response to inflammatory stimuli and suggest that NSAID may be beneficial in alleviating uterine inflammation.
Collapse
Affiliation(s)
- M A Crookenden
- Hopkirk Research Institute, AgResearch, Palmerston North 4442, New Zealand.
| | - C R Burke
- DairyNZ Ltd., Private Bag 3221, Hamilton 3240, New Zealand
| | - M D Mitchell
- Institute of Health and Biomedical Innovation - Centre for Children's Health Research, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, 4101, Australia
| | - C V C Phyn
- DairyNZ Ltd., Private Bag 3221, Hamilton 3240, New Zealand
| | - J R Roche
- School of Biological Sciences, University of Auckland, Auckland 1010, New Zealand
| | - A Heiser
- Hopkirk Research Institute, AgResearch, Palmerston North 4442, New Zealand
| |
Collapse
|
12
|
Teworte S, Luciani P. Endometriosis, the Silent Disease: Molecular Targets, Active Principles, and Drug Delivery Systems. Helv Chim Acta 2022. [DOI: 10.1002/hlca.202200132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Sarah Teworte
- Department of Chemistry Biochemistry and Pharmaceutical Sciences University of Bern Freiestrasse 3 CH-3012 Bern Switzerland
| | - Paola Luciani
- Department of Chemistry Biochemistry and Pharmaceutical Sciences University of Bern Freiestrasse 3 CH-3012 Bern Switzerland
| |
Collapse
|
13
|
Li L, Sun B, Sun Y. Identification of functional TF-miRNA-hub gene regulatory network associated with ovarian endometriosis. Front Genet 2022; 13:998417. [PMID: 36212136 PMCID: PMC9540245 DOI: 10.3389/fgene.2022.998417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Endometriosis (EMs), one of the most common gynecological diseases, seriously affects the health and wellness of women; however, the underlying pathogenesis remains unclear. This study focused on dysregulated genes and their predicted transcription factors (TFs) and miRNAs, which may provide ideas for further mechanistic research. The microarray expression dataset GSE58178, which included six ovarian endometriosis (OE) samples and six control samples, was downloaded from the Gene Expression Omnibus (GEO) to identify differentially expressed genes (DEGs). Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to study the cellular and organism-level functions of DEGs. The protein-protein interaction (PPI) network was built and visualized using Cytoscape, and modules and hub genes were explored using various algorithms. Furthermore, we predicted miRNAs and TFs of hub genes using online databases, and constructed the TF-miRNA-hub gene network. There were 124 upregulated genes and 66 downregulated genes in EMs tissues. GO enrichment analysis showed that DEGs were concentrated in reproductive structure development and collagen-containing extracellular matrix, while KEGG pathway analysis showed that glycolysis/gluconeogenesis and central carbon metabolism in cancer require further exploration. Subsequently, HIF1A, LDHA, PGK1, TFRC, and CD9 were identified as hub genes, 22 miRNAs and 34 TFs were predicted to be upstream regulators of hub genes, and these molecules were pooled together. In addition, we found three key feedback loops in the network, MYC-miR-34a-5p-LDHA, YY1-miR-155-5p-HIF1A, and RELA-miR-93-5p-HIF1A, which may be closely related to OE development. Taken together, our study structured a TF-miRNA-hub gene network to decipher the molecular mechanism of OE, which may provide novel insights for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Lu Li
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bo Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Yingpu Sun,
| |
Collapse
|
14
|
Li B, Liu Y, Sun S. Pump proton inhibitors display anti-tumour potential in glioma. Cell Prolif 2022:e13321. [PMID: 35961680 DOI: 10.1111/cpr.13321] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/28/2022] [Accepted: 07/14/2022] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVES Glioma is one of the most aggressive brain tumours with poor overall survival despite advanced technology in surgical resection, chemotherapy and radiation. Progression and recurrence are the hinge causes of low survival. Our aim is to explain the concrete mechanism in the proliferation and progression of tumours based on tumour microenvironment (TME). The main purpose is to illustrate the mechanism of proton pump inhibitors (PPIs) in affecting acidity, hypoxia, oxidative stress, inflammatory response and autophagy based on the TME to induce apoptosis and enhance the sensitivity of chemoradiotherapy. FINDINGS TME is the main medium for tumour growth and progression. Acidity, hypoxia, inflammatory response, autophagy, angiogenesis and so on are the main causes of tumour progress. PPIs, as a common clinical drug to inhibit gastric acid secretion, have the advantages of fast onset, long action time and small adverse reactions. Nowadays, several kinds of literature highlight the potential of PPIs in inhibiting tumour progression. However, long-term use of PPIs alone also has obvious side effects. Therefore, till now, how to apply PPIs to promote the effect of radio-chemotherapy and find the concrete dose and concentration of combined use are novel challenges. CONCLUSIONS PPIs display the potential in enhancing the sensitivity of chemoradiotherapy to defend against glioma based on TME. In the clinic, it is also necessary to explore specific concentrations and dosages in synthetic applications.
Collapse
Affiliation(s)
- Bihan Li
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Ying Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| | - Shilong Sun
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
15
|
Assaf L, Eid AA, Nassif J. Role of AMPK/mTOR, mitochondria, and ROS in the pathogenesis of endometriosis. Life Sci 2022; 306:120805. [PMID: 35850246 DOI: 10.1016/j.lfs.2022.120805] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/27/2022] [Accepted: 07/10/2022] [Indexed: 12/19/2022]
Abstract
Endometriosis is the presence of endometrial tissue outside the uterine cavity usually in the ovaries, fallopian tube, and pelvic cavity. It's a chronic enigmatic gynecological condition associated with dysmenorrhea, dyspareunia, pelvic pain, and infertility. Endometriosis lesions exist in a unique microenvironment characterized by increased concentrations of hormones, inflammation, and oxidative stress. This environment promotes cell survival through the binding of membrane receptors and subsequent cascading activation of intracellular kinases that stimulate a cellular response. In endometriosis, well-established signaling pathways, mTOR and AMPK, are altered via steroid hormones and other factors to promote cell growth, migration, and proliferation. This is accompanied by dysfunction in the mitochondria that increase energy production to sustain proliferation demands consequently leading to reactive oxygen species overproduction. This review aims to summarize the role of altered mTOR/AMPK signaling pathway, mitochondrial dysfunction, and reactive oxygen species overproduction along with providing therapeutic and diagnostic approaches. Highlighting these factors would provide a better understanding to reach a coherent theory for the pathogenesis of endometriosis.
Collapse
Affiliation(s)
- Lama Assaf
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon; AUB Diabetes, American University of Beirut, Beirut, Lebanon.
| | - Joseph Nassif
- Division of Minimally Invasive Gynecology, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
16
|
Silvestro S, Diomede F, Chiricosta L, Zingale VD, Marconi GD, Pizzicannella J, Valeri A, Avanzini MA, Calcaterra V, Pelizzo G, Mazzon E. The Role of Hypoxia in Improving the Therapeutic Potential of Mesenchymal Stromal Cells. A Comparative Study From Healthy Lung and Congenital Pulmonary Airway Malformations in Infants. Front Bioeng Biotechnol 2022; 10:868486. [PMID: 35774062 PMCID: PMC9237219 DOI: 10.3389/fbioe.2022.868486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/20/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) play an important role in the field of regenerative medicine thanks to their immunomodulatory properties and their ability to secrete paracrine factors. The use of MSCs has also been tested in children with congenital lung diseases inducing fibrosis and a decrease in lung function. Congenital malformations of the pulmonary airways (CPAM) are the most frequently encountered lung lesion that results from defects in early development of airways. Despite the beneficial properties of MSCs, interventions aimed at improving the outcome of cell therapy are needed. Hypoxia may be an approach aimed to ameliorate the therapeutic potential of MSCs. In this regard, we evaluated the transcriptomic profile of MSCs collected from pediatric patients with CPAM, analyzing similarities and differences between healthy tissue (MSCs-lung) and cystic tissue (MSCs-CPAM) both in normoxia and in cells preconditioned with hypoxia (0.2%) for 24 h. Study results showed that hypoxia induces cell cycle activation, increasing in such a way the cell proliferation ability, and enhancing cell anaerobic metabolism in both MSCs-lung and MSCs-CPAM-lung. Additionally, hypoxia downregulated several pro-apoptotic genes preserving MSCs from apoptosis and, at the same time, improving their viability in both comparisons. Finally, data obtained indicates that hypoxia leads to a greater expression of genes involved in the regulation of the cytoskeleton in MSCs-lung than MSCs-CPAM.
Collapse
Affiliation(s)
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | | | | | - Guya Diletta Marconi
- Department of Medical, Oral and Biotechnological Sciences, University “G. D’Annunzio” Chieti-Pescara, Chieti, Italy
| | | | - Andrea Valeri
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Messina, Italy
| | - Maria Antonietta Avanzini
- Cell Factory, Pediatric Hematology Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Valeria Calcaterra
- Pediatrics and Adolescentology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy
- Pediatric Department, Children’s Hospital “Vittore Buzzi”, Milano, Italy
| | - Gloria Pelizzo
- Pediatric Surgery Department, Children’s Hospital “Vittore Buzzi”, Milano, Italy
- Department of Biomedical and Clinical Sciences-L. Sacco, University of Milan, Milan, Italy
| | | |
Collapse
|
17
|
Optimal Nutrition Parameters for Neonates and Infants with Congenital Heart Disease. Nutrients 2022; 14:nu14081671. [PMID: 35458233 PMCID: PMC9029500 DOI: 10.3390/nu14081671] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/04/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
Congenital heart defects are known causes of malnutrition. Optimal nutritional management is paramount in improving short and long-term prognosis for neonates and infants with congenital heart malformations, as current strategies target preoperative and postoperative feeding requirements. Standardized enteral and/or parenteral feeding protocols, depending on the systemic implications of the cardiac defect, include the following common practices: diagnosing and managing feeding intolerance, choosing the right formula, and implementing a monitoring protocol. The latest guidelines from the American Society for Parenteral and Enteral Nutrition and the European Society of Paediatric and Neonatal Intensive Care, as well as a significant number of recent scientific studies, offer precious indications for establishing the best feeding parameters for neonates and infants with heart defects.
Collapse
|
18
|
Cho MK, Jin L, Han JH, Jin JS, Cheon SY, Shin S, Bae SJ, Park JK, Ha KT. Water-Extracted Prunella vulgaris Alleviates Endometriosis by Reducing Aerobic Glycolysis. Front Pharmacol 2022; 13:872810. [PMID: 35444541 PMCID: PMC9014096 DOI: 10.3389/fphar.2022.872810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/15/2022] [Indexed: 12/31/2022] Open
Abstract
Endometriosis is a chronic inflammatory disorder caused by abnormal adhesion of endometrial tissue to the outside of the uterus. The combination of surgery, non-steroidal anti-inflammatory drugs, and hormone treatment is well established therapy for endometriosis, however, case reports have showed that high rates of relapse and unpleasant side effect. For these reasons, recently, the studies have been focused on the Warburg-like metabolic shift of endometriosis. Prunella vulgaris is one of traditionally used herbal medicine for inflammatory disease and the anti-estrogenic effects of P. vulgaris is well-established. Therefore, in this work, we evaluated water-extracted P. vulgaris (PV) as a potential treatment for endometriosis. To this, we artificially induced endometriosis in ovarectomized mice by intra-peritoneal inoculation of uterus extracts. PV was orally administered, and PV significantly alleviated endometriosis, particularly the growth of ectopic endometrial lesions in artificially endometriosis-induced mice. For the mechanism study of anti-endometriosis by PV, we designed an in vitro study using human normal endometrial stromal cells (T-HESCs) and human endometrial cell (12Z) obtained from patients with endometriosis. PV strongly induced the apoptosis of 12Z cells rather than T-HESCs by control the activity or expression of aerobic glycolysis enzymes, such as lactate dehydrogenase A (LDHA), pyruvate dehydrogenase A, and pyruvate dehydrogenase kinase 1/3. In addition, lactate production was enhanced, and oxygen consumption rate was suppressed in 12Z cells upon PV treatment. These changes in aerobic glycolysis eventually caused mitochondrial damage following decreased mitochondrial membrane potential and excessive mitochondrial ROS production. Especially, ulsolic acid (UA), one of the compounds in PV considerably led 12Z cell apoptosis with inhibition of LDHA activity. Therefore, UA could be a major active substance of PV in terms of endometriosis inhibitors. In conclusion, this study provides the evidence that the beneficial efficacy of PV for the prevention/treatment of endometriosis.
Collapse
Affiliation(s)
- Min Kyoung Cho
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, South Korea
| | - Ling Jin
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, South Korea,Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, South Korea
| | - Jung Ho Han
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, South Korea,Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, South Korea
| | - Jung-Suk Jin
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, South Korea
| | - Se-Yun Cheon
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, South Korea
| | - Su Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, South Korea
| | - Sung-Jin Bae
- Department of Anatomy, Kosin University College of Medicine, Busan, South Korea
| | - Jang-Kyung Park
- Department of Korean Obstetrics and Gynecology, Pusan National University Korean Medicine Hospital, Yangsan, South Korea,*Correspondence: Jang-Kyung Park, ; Ki-Tae Ha,
| | - Ki-Tae Ha
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, South Korea,Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, South Korea,*Correspondence: Jang-Kyung Park, ; Ki-Tae Ha,
| |
Collapse
|
19
|
Kobayashi H, Imanaka S. Understanding the molecular mechanisms of macrophage polarization and metabolic reprogramming in endometriosis: A narrative review. Reprod Med Biol 2022; 21:e12488. [PMID: 36310658 PMCID: PMC9596393 DOI: 10.1002/rmb2.12488] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/15/2022] [Accepted: 09/29/2022] [Indexed: 12/03/2022] Open
Abstract
Background Endometriosis is an estrogen-dependent disease and causes pelvic pain and infertility. The limits of current pharmacotherapy in women who desire to become pregnant prompt the development of various targeted molecules for more effective treatment. A review article focused on the unique aspect of cellular metabolic reprogramming of endometriotic cells has been reported. The cellular metabolic pathways are reprogrammed to adapt to a variety of environmental stresses (e.g., nutrient starvation or glucose deprivation, hypoxic stress, excessive reactive oxygen species generation, and other environmental factors). This review aims to summarize macrophage polarization and metabolic reprogramming in endometriosis. Methods A literature search was performed between January 2000 and March 2022 in the PubMed and Google Scholar databases using a combination of specific terms. Results Macrophage cellular metabolism has a marked influence on its phenotype and function. Preclinical studies showed that metabolic conversion toward glycolysis or oxidative phosphorylation drives macrophage polarization to M1 or M2 phenotype, respectively. Such cellular metabolic rewiring can offer new therapeutic opportunities. Conclusion A better understanding of metabolic reprogramming biology in endometriosis-associated macrophages is essential in considering novel therapeutic approach for endometriosis. However, there are currently no detailed studies on therapeutic strategies targeting the cellular metabolic properties of endometriosis-associated macrophages.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of GynecologyMs.Clinic MayOneKashihara, NaraJapan
- Department of Obstetrics and GynecologyNara Medical UniversityKashihara, NaraJapan
| | - Shogo Imanaka
- Department of GynecologyMs.Clinic MayOneKashihara, NaraJapan
- Department of Obstetrics and GynecologyNara Medical UniversityKashihara, NaraJapan
| |
Collapse
|
20
|
Mortezaee K, Majidpoor J. The impact of hypoxia on immune state in cancer. Life Sci 2021; 286:120057. [PMID: 34662552 DOI: 10.1016/j.lfs.2021.120057] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022]
Abstract
Hypoxia is a known feature of solid tumors and a critical promoter of tumor hallmarks. Hypoxia influences tumor immunity in a way favoring immune evasion and resistance. Extreme hypoxia and aberrant hypoxia-inducible factor-1 (HIF-1) activity in tumor microenvironment (TME) is a drawback for effective immunotherapy. Infiltration and activity of CD8+ T cells is reduced in such condition, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) show high activities. Highly hypoxic TME also impairs maturation and activity of dendritic cell (DCs) and natural killer (NK) cells. In addition, the hypoxic TME positively is linked positively with metabolic changes in cells of immune system. These alterations are indicative of a need for hypoxia modulation as a complementary targeting strategy to go with immune checkpoint inhibitor (ICI) therapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|