1
|
Wang Z, Li X, Liu X, Yang Y, Yan Y, Cui D, Meng C, Ali MI, Zhang J, Yao Z, Long Y, Yang R. Mechanistic insights into the anti-fibrotic effects of estrogen via the PI3K-Akt pathway in frozen shoulder. J Steroid Biochem Mol Biol 2025; 249:106701. [PMID: 39947440 DOI: 10.1016/j.jsbmb.2025.106701] [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: 11/07/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
The development of frozen shoulder (FS) is primarily characterized by pathological fibrosis, yet clinical treatment options remain limited. Recent studies have identified estrogen depletion during perimenopause as a significant contributor to the onset of FS and fibrosis. This study investigates the role of estradiol (E2) and the estrogen-related receptor (GPER) in fibrotic processes associated with FS to elucidate the underlying mechanisms. The functional relationship between E2, GPER, and FS progression was examined using a rat immobilization model and synovial-derived fibroblasts (SFs) from FS patients. E2's effects on GPER expression, fibroblast activation, and tissue fibrosis were evaluated through Western blotting, immunofluorescence staining, collagen contraction assays, wound healing assays, and histological staining. RNA sequencing identified signaling pathways and key regulators involved in E2 treatment. Both E2 and the GPER activator G1 exhibited antifibrotic effects, improving shoulder mobility, reducing extracellular matrix (ECM) deposition in the periarticular capsule, and decreasing the expression of fibrosis-related genes, including fibronectin, α-SMA, and COL3. In contrast, the GPER inhibitor G15 reversed these effects, suggesting that E2 mediates its antifibrotic action through GPER activation. Mechanistically, KEGG pathway analysis revealed that E2 suppresses the PI3K/AKT signaling pathway by inhibiting PI3K and AKT phosphorylation, thereby preventing fibroblast activation and reversing FS-associated fibrosis. These findings provide mechanistic insights into the previously unrecognized role of GPER in FS progression and may open new avenues for research to optimize future clinical therapies.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xinhao Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaoshan Liu
- Pediatric Oncology, Children's Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Yitao Yang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yan Yan
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Dedong Cui
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chenyang Meng
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Maslah Idiris Ali
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jinming Zhang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zeyu Yao
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yi Long
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Rui Yang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
2
|
Yan J, Meng Q, Hao F, Xu M. cFos-mediated β-Arrestin1 in the RVLM alleviates sympathetic hyperactivity induced by ovariectomy. Mol Cell Endocrinol 2025; 601:112520. [PMID: 40096880 DOI: 10.1016/j.mce.2025.112520] [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: 12/12/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Sympathetic hyperactivity is a key feature of cardiovascular dysfunction in postmenopausal women and is closely linked to the onset, progression, and outcomes of cardiovascular events. However, the mechanisms underlying sympathetic nerve hyperactivity due to menopause remain unclear. β-arrestin is a versatile class of intracellular proteins that were initially discovered for their ability to disrupt the G protein-coupled receptors (GPCRs) signaling by binding to activated receptors. A notable reduction in the expression of β-arrestin1 in the rostral ventrolateral medulla (RVLM) associated with increased sympathetic activity and elevated blood pressure (BP) in spontaneously hypertensive rats. It has been reported that the cellular oncogene fos (cFos), as a transcription factor, plays a crucial role in BP regulation. This study aimed to investigate whether β-arrestin1, regulated by cFos in the RVLM, contributes to sympathetic hyperactivity induced by menopause. Bilateral ovariectomy (OVX) was performed to establish a postmenopausal rat model. We found that the expression of β-arrestin1 in the RVLM of OVX rats was reduced, whereas estrogen supplementation increased the expression of β-arrestin1. Furthermore, overexpression of β-arrestin1 in the RVLM of OVX rats attenuated the sympathetic hyperactivity. Conversely, reducing β-arrestin1 expression in the RVLM compromised the cardioprotective effects of estrogen in OVX rats. Additionally, inhibiting the expression of the transcription factor cFos in the RVLM of OVX rats diminished the estrogen-induced increase in the expression of β-arrestin1. These findings suggest that estrogen enhances the expression of β-arrestin1 mediated by cFos in the RVLM of OVX rats, thereby alleviating sympathetic nerve hyperactivity and hypertension.
Collapse
Affiliation(s)
- Jiuqiong Yan
- Department of Obstetrics and Gynecology, Changhai Hospital, Naval Medical University, 200433, PR China
| | - Qi Meng
- Department of Obstetrics and Gynecology, Changhai Hospital, Naval Medical University, 200433, PR China; Marine Corps Hospital, PLA, 521011, PR China
| | - Fan Hao
- Department of Obstetrics and Gynecology, Changhai Hospital, Naval Medical University, 200433, PR China; Reproductive Medical Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, 200092, PR China
| | - Mingjuan Xu
- Department of Obstetrics and Gynecology, Changhai Hospital, Naval Medical University, 200433, PR China.
| |
Collapse
|
3
|
Li J, Chen Y, Yu Q, Li S, Zhang X, Cheng Y, Fu X, Li J, Zhu L. Estrogen receptor β alleviates colitis in intestinal epithelial cells and activates HIF-1a and ATG-9a-mediated autophagy. Exp Cell Res 2025; 447:114520. [PMID: 40107441 DOI: 10.1016/j.yexcr.2025.114520] [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: 10/22/2024] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Estrogen receptor β (ERβ) plays a pivotal role in regulating intestinal epithelial function and inflammation. Its involvement in inflammatory bowel diseases (IBD), particularly in ulcerative colitis (UC), remains poorly understood, despite emerging evidence pointing to its anti-inflammatory properties. This study investigated ERβ expression in UC patients using quantitative PCR, Western blot, and immunofluorescence. To investigate the functional role of ERβ, a DSS-induced colitis mouse model and LPS-treated HT-29 cells were used. Autophagy activity was evaluated through Western blot, transmission electron microscopy (TEM), and autophagy inhibitors. Co-immunoprecipitation (Co-IP) and dual luciferase reporter assays were employed to explore the interaction between ERβ and hypoxia-inducible factor-1α (HIF-1α), as well as the regulation of ATG-9a expression. The results demonstrated that ERβ expression was significantly downregulated in the inflammatory colons of UC patients. In vivo, ERβ activation by ERB041 alleviated DSS-induced colitis in mice, reducing weight loss, histopathological damage, and inflammatory cytokine levels. In vitro, ERB041 enhanced autophagy in LPS-treated HT-29 cells, accompanied by a reduction in pro-inflammatory cytokines. Furthermore, ERβ activation promoted the expression of tight junction proteins and preserved epithelial barrier integrity. Co-IP and dual luciferase assays revealed that ERβ interacted with HIF-1α and modulated ATG-9a-mediated autophagy. These results indicate that ERβ alleviates intestinal inflammation and activates HIF-1a and ATG-9a-mediated autophagy, providing new insights into the therapeutic potential of targeting ERβ in UC and highlighting its role in maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Junrong Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Division of Gastroenterology, Chongqing Hospital Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Chongqing, China
| | - Yidong Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Yu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuang Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaopeng Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyu Fu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiamin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangru Zhu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Bacchetti A, LeBoff M, Kaplan TB. Osteoporosis and multiple sclerosis: Exploring the complex intersection between bone and brain, a clarion call for change. Mult Scler Relat Disord 2025; 98:106420. [PMID: 40203605 DOI: 10.1016/j.msard.2025.106420] [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: 10/30/2024] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Osteoporosis is a common but often underrecognized condition in people with multiple sclerosis (PwMS), occurring more frequently and at an earlier age compared to the general population. It leads to an increased risk of fractures due to reduced bone mineral density (BMD). This review identifies key risk factors contributing to osteoporosis in PwMS, including age, vitamin D deficiency, reduced mobility, and long-term corticosteroid use. In addition to these factors, shared inflammatory mechanisms between MS and osteoporosis, such as elevated pro-inflammatory cytokines, further exacerbate bone loss. Strategies for early identification and proactive management are also discussed, highlighting their importance in reducing fracture risk and improving outcomes. The ultimate aim is to promote tailored pharmacological treatments, effective interventions, and comprehensive management of osteoporosis in PwMS.
Collapse
Affiliation(s)
- Anna Bacchetti
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, United States.
| | - Meryl LeBoff
- Division of Endocrinology, Diabetes, and Hypertension, Mass General Brigham Hospital, Harvard Medical School, Boston, MA, 02115, United States.
| | - Tamara B Kaplan
- Department of Neurology, Mass General Brigham Hospital, Harvard Medical School, Boston, MA, 02115, United States.
| |
Collapse
|
5
|
Ou M, Xu S, Huang Z, Xu X. In silico toxicology investigation of μ-conotoxin KIIIA on human Na + channel Na v1.2. Int J Biol Macromol 2025; 298:140092. [PMID: 39832599 DOI: 10.1016/j.ijbiomac.2025.140092] [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: 10/15/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
Conotoxins(CTXs) can specifically act on multiple ion channels, which are crucial for the development of neurobiology and novel targeted drug development. At present, >10,000 kinds of CTXs have been sequenced, it would be extremely laborious to conduct experiments for each. μ-CTX KIIIA is a type of substance that can selectively recognize voltage-gated sodium ion channels. This article constructs four derivatives of KIIIA and predicts their 3D structures; afterwards, their molecular orbital arrangements and physicochemical properties were calculated using DFT; then, predicted their toxicokinetic parameters such as absorption, distribution, metabolism, excretion (ADME) and toxicity (T) through Machine Learning (ML); finally, molecular docking and molecular dynamics are used to investigate the interaction modes and binding affinity. The results indicate that the toxicity of KIIIA and its derivatives (KIIIA-1 -KIIIA-4) to the human body is mainly concentrated in the liver and respiratory tract. Among four derivatives, KIIIA-2 (5 Ser → Arg) has better toxicokinetics properties and its binding energy to Nav1.2 is -65.32 kcal/mol, which is higher than that of wild type(-32.13 kcal/mol). This study indicate that computational toxicology can facilitate the druggability research of CTXs, and KIIIA-2 can be developed as a potential antiepileptic drug.
Collapse
Affiliation(s)
- Minrui Ou
- College of Chemistry, Fuzhou University, Fuzhou 350116, China.
| | - Suyan Xu
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Zhixuan Huang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Xiaoping Xu
- College of Chemistry, Fuzhou University, Fuzhou 350116, China
| |
Collapse
|
6
|
Canzian J, Conforti F, Jacobs F, Benvenuti C, Gaudio M, Gerosa R, De Sanctis R, Zambelli A. Sex-Related Differences in Immunotherapy Toxicities: Insights into Dimorphic Responses. Cancers (Basel) 2025; 17:1054. [PMID: 40227458 PMCID: PMC11987764 DOI: 10.3390/cancers17071054] [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: 02/11/2025] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Significant sex-based differences exist in the immune system and antitumor immune responses, potentially leading to variations in both the efficacy and toxicity of anticancer immunotherapies. Women generally mount stronger innate and adaptive immune responses than men, which can result in more severe immune-related adverse events (irAEs) during treatments with immune checkpoint inhibitors (ICIs). However, the importance of sex dimorphism in the safety of cancer immunotherapy remains underexplored in clinical oncology, despite its profound implications for treatment outcomes. Our review highlights the critical influence of biological sex on pharmacokinetics, pharmacodynamics, and immune responses, shaping ICI efficacy and the prevalence, type, and severity of irAEs. Integrating sex as a critical variable in cancer treatment and clinical trial design is essential for personalizing therapeutic strategies, bridging existing knowledge gaps, and enhancing survival rates and quality of life for patients across genders.
Collapse
Affiliation(s)
- Jacopo Canzian
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy; (J.C.); (C.B.); (M.G.); (R.G.)
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Fabio Conforti
- Division of Medical Oncology, Humanitas Gavazzeni, 24125 Bergamo, Italy;
| | - Flavia Jacobs
- Division of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Chiara Benvenuti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy; (J.C.); (C.B.); (M.G.); (R.G.)
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Mariangela Gaudio
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy; (J.C.); (C.B.); (M.G.); (R.G.)
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Riccardo Gerosa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy; (J.C.); (C.B.); (M.G.); (R.G.)
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Rita De Sanctis
- Oncology Unit, ASST Papa Giovanni XXIII Hospital, 24127 Bergamo, Italy;
| | - Alberto Zambelli
- Oncology Unit, ASST Papa Giovanni XXIII Hospital, 24127 Bergamo, Italy;
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
7
|
Motta F, Di Simone N, Selmi C. The Impact of Menopause on Autoimmune and Rheumatic Diseases. Clin Rev Allergy Immunol 2025; 68:32. [PMID: 40117049 PMCID: PMC11928423 DOI: 10.1007/s12016-025-09031-8] [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] [Accepted: 02/05/2025] [Indexed: 03/23/2025]
Abstract
The prevalence of autoimmune and rheumatological diseases is significantly higher in women, likely due to the effect of sex hormones influencing the development and function of the immune system, a phenomenon observed particularly during pregnancy. Oestrogens, in particular, appear to be a major factor in modulating the immune response, as their receptors are present in nearly all immune cells, where they regulate the expression of genes involved in inflammation. However, there is limited data on how menopause impacts autoimmune diseases, despite evidence suggesting that the menopausal perturbation of hormone levels may lead to the development of autoimmune conditions or alter the course of an already established disease. This review focuses on rheumatic conditions, aiming to provide a comprehensive understanding of how menopause influences the onset, progression, and clinical features of autoimmune diseases. The best evidence is available for rheumatoid arthritis and systemic lupus erythematosus, two paradigmatic autoimmune diseases in which menopause elicits opposite outcomes. Despite these data, there is a notable lack of evidence and research on the impact of menopause in other inflammatory arthritis and connective tissue diseases. This gap highlights a crucial area for future research and unmet needs to be addressed. Understanding how menopausal changes impact autoimmunity and rheumatic diseases will be crucial for improving the management of autoimmune and rheumatological diseases in women.
Collapse
Affiliation(s)
- Francesca Motta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas S. Pio X Hospital, Milan, Italy
| | - Carlo Selmi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan, Italy.
| |
Collapse
|
8
|
Brandão ML, Hermsdorff HHM, Leal ACG, Bressan J, Pimenta AM. Vaccination and food consumption: association with Post-Acute COVID-19 Syndrome in Brazilian adults (CUME Study). Front Nutr 2025; 12:1549747. [PMID: 40161300 PMCID: PMC11950691 DOI: 10.3389/fnut.2025.1549747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025] Open
Abstract
Background Post-Acute COVID-19 Syndrome (PACS) is an important sequalae of COVID-19. Then, our objective was to analyze the risk and protective factors for PACS in Brazilian adults participating in the Cohort of Universities of Minas Gerais (CUME Study), with emphasis on COVID-19 vaccination and food consumption. Methods In this sub-study, we included 2,065 participants of CUME Study who answered the baseline questionnaire in 2016 or 2018 or 2020 or 2022, and the follow-up COVID-19/PACS-specific questionnaire in 2023. PACS diagnosis was based on self-reporting of continuation or development of new symptoms 3 months after the initial SARS-CoV-2 infection, with these symptoms lasting for at least 2 months with no other explanation. To estimate the risk and protective factors for PACS, hierarchical multivariate statistical analysis was conducted using the Cox regression technique, producing two models: (1) focusing on consumption of macro and micronutrients; (2) focusing on consumption of food groups. Results After a median of 5.5 years of follow-up, 54.4% of the participants reported PACS. When we analyzed the consumption of macro and micronutrients, higher intake of proteins (HR: 1.36; 95% CI: 1.06-1.74-4th quartile) and lipids (HR: 1.23; 95% CI: 1.02-1.48-4th quartile) were risk factors for PACS. On the other hand, higher intake of vitamin C (HR: 0.78; 95% CI: 0.64-0.94-4th quartile), vitamin D (HR: 0.81; 95% CI: 0.67-0.99-4th quartile), and zinc (HR: 0.66; 95% CI: 0.52-0.83-4th quartile) were protective factors for the outcome (model 1). When we analyzed the consumption of food groups, higher intake of eggs (HR: 1.59; 95% CI: 1.34-1.89-4th quartile) increased the risk of PACS, whereas, respectively, higher and intermediate consumption of white meat (HR: 0.84; 95% CI: 0.71-1.00-4th quartile) and vegetables (HR: 0.81; 95% CI: 0.67-0.99-2nd quartile; HR: 0.81; 95% CI: 0.67-0.99-3rd quartile) decreased the risk of the outcome (model 2). In both models, pre-infection COVID-19 vaccination was a protective factor for PACS. Conclusion A healthy diet, with higher consumption of white meat, vegetables and specific micronutrients (vitamin C, vitamin D, zinc), in parallel with pre-infection COVID-19 vaccination, is essential to reduce the risk of PACS.
Collapse
Affiliation(s)
- Marlise Lima Brandão
- Posgraduate Program in Nursing, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | | | | | - Josefina Bressan
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | |
Collapse
|
9
|
Miao M, Liu X, Zhang H, Dai H. Immuno-inflammatory mechanisms in cardio-oncology: new hopes for immunotargeted therapies. Front Oncol 2025; 15:1516977. [PMID: 40182041 PMCID: PMC11966441 DOI: 10.3389/fonc.2025.1516977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
Cardio-oncology is an emerging interdisciplinary field concerned with cancer treatment-related cardiovascular toxicities (CTR-CVT) and concomitant cardiovascular diseases (CVD) in patients with cancer. Inflammation and immune system dysregulation are common features of tumors and cardiovascular disease (CVD). In addition to the mutual exacerbating effect through inflammation, tumor treatments, including immunotherapy, chemotherapy, radiation therapy, and targeted therapy, may induce immune inflammatory reactions leading to cardiovascular damage. Cancer immunotherapy is currently a new method of cancer treatment. Immunotherapeutic agents, such as immune checkpoint inhibitors (ICIs), chimeric antigen receptor T cell immunotherapy (CAR-T), mRNA vaccines, etc., can induce anti-tumor effects by enhancing the host immune response to eliminate tumor cells. They have achieved remarkable therapeutic efficacy in clinical settings but lead to many immune-related adverse events (irAEs), especially CTR-CVT. Establishing specific evaluation, diagnostic, and monitoring criteria (e.g., inflammatory biomarkers) for both immunotherapy and anti-inflammatory therapy-related cardiovascular toxicity is vital to guide clinical practice. This article explores the role of immune response and inflammation in tumor cardiology, unravels the underlying mechanisms, and provides improved methods for monitoring and treating in CTR-CVT in the field of cardio-oncology.
Collapse
Affiliation(s)
- Meiqi Miao
- Department of Cardiology, Kunshan Hospital of Chinese Medicine, Kunshan, China
| | - Xinxin Liu
- Postdoctoral Mobile Station, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- The Innovation Base, Mudanjiang Collaborative Innovation Center for the Development and Application of Northern Medicinal Resources, Mudanjiang, China
| | - Han Zhang
- Department of Cardiology, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hailong Dai
- Department of Cardiology, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
10
|
Tseng YT, Chen II, Wang CH. Association of Hormone Replacement Therapy with Inflammatory Bowel Disease Risk in Women with Menopausal Disorders: A Population-Based Retrospective Cohort Study. Healthcare (Basel) 2025; 13:578. [PMID: 40077140 PMCID: PMC11899444 DOI: 10.3390/healthcare13050578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background: The long-term effects of hormone replacement therapy (HRT) on inflammatory bowel disease (IBD) remain unclear, necessitating further investigations of the association between HRT and the development of ulcerative colitis and Crohn's disease in postmenopausal women. Methods: This retrospective cohort study utilized Taiwan's National Health Insurance claims (2001-2018) to identify postmenopausal women aged ≥ 50 years with HRT use. A one-year washout period was applied before the index date to ensure new HRT users. To address the immortal time bias, follow-up for HRT users began at HRT initiation. The non-HRT group was selected by 1:1 propensity score matching. Cox proportional hazards models with adjustments for comorbidities and medications were used to estimate hazard ratios. Results: A total of 10,126 postmenopausal women (5063 per group) were included. During a mean follow-up of 11.1 years, the incidence rates of ulcerative colitis were 0.14 and 0.11 per 1000 person-years in the HRT and non-HRT groups, respectively. The adjusted hazard ratios were 1.33 (95% CI, 0.46-3.83; p = 0.600) for ulcerative colitis and 0.72 (95% CI, 0.45-1.16; p = 0.177) for Crohn's disease. Conclusions: This longitudinal study suggests that HRT use is not significantly associated with the risk of IBD among postmenopausal women. These findings indicate that IBD risk may not need to be a primary concern when considering HRT in this population.
Collapse
Affiliation(s)
- Yuan-Tsung Tseng
- Department of Public Health, College of Medicine, National Cheng Kung University, Tainan City 701, Taiwan;
- Department of Medical Research, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), Tainan City 701, Taiwan
| | - I-I Chen
- Department of Hepatogastroenterology, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), No. 670, Chongde Road, East District, Tainan City 701, Taiwan;
| | - Chun-Hsiang Wang
- Department of Hepatogastroenterology, Tainan Municipal Hospital (Managed by Show Chwan Medical Care Corporation), No. 670, Chongde Road, East District, Tainan City 701, Taiwan;
| |
Collapse
|
11
|
Chen HN, Hu YN, Ran LL, Wang M, Zhang Z. Sexual dimorphism in aortic aneurysm: A review of the contributions of sex hormones and sex chromosomes. Vascul Pharmacol 2025; 158:107460. [PMID: 39716526 DOI: 10.1016/j.vph.2024.107460] [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: 10/07/2024] [Revised: 11/23/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Aortic aneurysm is a common cardiovascular disease. Over time, the disease damages the structural and functional integrity of the aorta, causing it to abnormally expand and potentially rupture, which can be fatal. Sex differences are evident in the disease, with men experiencing an earlier onset and higher incidence. However, women may face a worse prognosis and a higher risk of rupture. While there are some studies on the cellular and molecular mechanisms of aneurysm formation, it remains unclear how sex factors contribute to sexual dimorphism. Therefore, this review aims to summarize the role of sex in the occurrence of aortic aneurysms, offering valuable insights for disease prevention and the development of appropriate treatment options.
Collapse
Affiliation(s)
- Hao-Nan Chen
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Yan-Ni Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Li-Ling Ran
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China
| | - Mi Wang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha 410013, Hunan, China.
| |
Collapse
|
12
|
Guerra G, Nakase T, Kachuri L, McCoy L, Hansen HM, Rice T, Wiemels JL, Wiencke JK, Molinaro AM, Wrensch M, Francis SS. Association of immunoglobulin E levels with glioma risk and survival. J Natl Cancer Inst 2025; 117:545-553. [PMID: 39447063 PMCID: PMC11884848 DOI: 10.1093/jnci/djae265] [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: 04/16/2024] [Revised: 08/20/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Previous epidemiological studies have reported an association of serum immunoglobulin E (IgE) levels with reduced glioma risk, but the association between IgE and glioma prognosis has not been characterized. This study aimed to examine how sex, tumor subtype, and IgE class modulate the association of serum IgE levels with glioma risk and survival. METHODS We conducted a case-control study using participants from the University of California San Francisco Adult Glioma Study (1997-2010). Serum IgE levels for total, respiratory, and food allergy were measured in adults diagnosed with glioma (n = 1319) and cancer-free control individuals (n = 1139) matched based on age, sex, and race and ethnicity. Logistic regression was adjusted for patient demographics to assess the association between IgE levels and glioma risk. Multivariable Cox regression adjusted for patient-specific and tumor-specific factors compared survival between the elevated and normal IgE groups. All statistical tests were 2-sided. RESULTS Elevated total IgE was associated with reduced risk of IDH wild-type glioma (risk ratio [RR] = 0.78, 95% CI = 0.71 to 0.86) and IDH-mutant glioma (RR = 0.73, 95% CI = 0.63 to 0.85). In multivariable Cox regression, positive respiratory IgE was associated with improved survival for IDH wild-type glioma (RR = 0.79, 95% CI = 0.67 to 0.93). The reduction in mortality risk was statistically significant in female individuals only (RR = 0.75, 95% CI = 0.57 to 0.98), with an improvement in median survival of 6.9 months (P < .001). CONCLUSION Elevated serum IgE was associated with improved prognosis for IDH wild-type glioma, with a more pronounced protective effect in female than male individuals, which has implications for the future study of IgE-based immunotherapies for glioma.
Collapse
Affiliation(s)
- Geno Guerra
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Taishi Nakase
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, United States
| | - Linda Kachuri
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Lucie McCoy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Helen M Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Terri Rice
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Joseph L Wiemels
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, United States
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Stephen S Francis
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, United States
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, United States
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
13
|
Xie Y, Chen J, Xu S, Cheng O. Estrogen deficiency promotes neurodegeneration in female hemi-parkinsonian mice: The role of regulatory T cells. Int Immunopharmacol 2025; 148:114104. [PMID: 39862633 DOI: 10.1016/j.intimp.2025.114104] [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: 05/14/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Circulating levels of the female hormone estrogen has been associated with the development of Parkinson's disease (PD), although the underlying mechanism remains unclear. Immune homeostasis mediated by peripheral regulatory T cells (Treg) is a crucial factor in PD. The aim of this study was to explore the effects of estrogen deficiency on neuroinflammation and neurodegeneration in a rodent model of PD, with particular reference to Treg. METHODS Estrogen deficiency was established in a mouse model by bilateral ovariectomy (OVX). PD was modeled by the injection of LPS into the striatum. Motor performance was assessed in each experimental group. Dopaminergic degeneration was evaluated using tyrosine-hydroxylase (Th) immunohistochemical staining of the substantia nigra (SN) and striatum. Dopamine and dopamine metabolite levels in the striatum were also evaluated, together with the infiltration of CD4 T cells into the SN. Neuroinflammation was assessed by evaluating the mRNA level of microglial and M1/M2 phenotype markers, as well as the abundance of pro-inflammatory cytokines in the midbrain. The frequency of peripheral Treg cells was evaluated using flow cytometry. RESULTS OVX prior to LPS injection markedly aggravated neurodegeneration, neuroinflammation, motor performance, and CD4 T cell infiltration compared to the LPS-only group. Estradiol treatment or activation of the G protein-coupled estrogen receptor (GPER) in OVX mice prior to LPS injection induced functional improvement and reduced the levels of neurodegeneration, neuroinflammation, and CD4 T cell infiltration. OVX prior to LPS injection also led to a decreased frequency of Treg compared to the LPS-only group. Moreover, estradiol treatment or GPER activation of OVX mice prior to LPS injection significantly increased the Treg frequency. Antibody-mediated depletion of Treg after GPER activation counteracted the ability of GPER to alleviate neurodegeneration, CD4 T cell infiltration, the release of pro-inflammatory cytokines, and motor performance in the PD model. CONCLUSION Estrogen deficiency can disrupt Treg-mediated immune homeostasis and thus further aggravate microglial inflammation and dopaminergic degeneration in PD model. The effects of estrogen on Treg may be partially mediated by GPER signaling, and thus GPER is a promising target for PD, especially in estrogen-deficient women.
Collapse
Affiliation(s)
- Yangzhi Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiacheng Chen
- Department of Intensive Care Unit, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China
| | - Sicong Xu
- Department of Neurology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
14
|
He C, He P, Ou Y, Tang X, Wei H, Xu Y, Bai S, Guo Z, Hu R, Xiong K, Du G, Sun X. Rectifying the Crosstalk between the Skeletal and Immune Systems Improves Osteoporosis Treatment by Core-Shell Nanocapsules. ACS NANO 2025; 19:5549-5567. [PMID: 39879106 DOI: 10.1021/acsnano.4c14728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Contemporary osteoporosis treatment often neglects the intricate interactions among immune cells, signaling proteins, and cytokines within the osteoporotic microenvironment. Here, we developed core-shell nanocapsules composed of a cationized lactoferrin core and an alendronate polymer shell. By tuning the size of these nanocapsules and leveraging the alendronate shell, we enabled precise delivery of small interfering RNA targeting the Semaphorin 4D gene (siSema4D) to specific bone sites. This strategy integrates the antiresorptive drug alendronate with siSema4D, efficiently inhibiting osteoclast (OC) differentiation and bone resorption, while promoting osteogenesis to restore the balance between osteoblasts (OBs) and OCs. Moreover, encapsulating siSema4D within the nanocapsules helps to mitigate immunological cascades, thereby reversing the inflammatory microenvironment and restoring immune homeostasis and providing insights into the immunomodulatory effects of Sema4D in osteoporosis therapy. In both ovariectomized and senile osteoporotic mouse models, local intramuscular administration of core-shell nanocapsules effectively rectified the imbalance between the skeletal and immune systems, significantly enhancing the overall efficacy of osteoporosis treatment. Our findings underscore the therapeutic promise of addressing the multifaceted osteoporotic microenvironment through targeted interventions.
Collapse
Affiliation(s)
- Chunting He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Penghui He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yangsen Ou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xue Tang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hongjiao Wei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yanhua Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Shuting Bai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhaofei Guo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rui Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Kun Xiong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Das GM, Oturkar CC, Menon V. Interaction between Estrogen Receptors and p53: A Broader Role for Tamoxifen? Endocrinology 2025; 166:bqaf020. [PMID: 39891710 PMCID: PMC11837209 DOI: 10.1210/endocr/bqaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/20/2025] [Accepted: 01/30/2025] [Indexed: 02/03/2025]
Abstract
Tamoxifen is one of the most widely used anticancer drugs in the world. It is a safe drug with generally well-tolerated side effects and has been prescribed for the treatment of early-stage and advanced-stage or metastatic estrogen receptor α (ERα/ESR1)-positive breast cancer. Tamoxifen therapy also provides a 38% reduction of the risk of developing breast cancer in women at high risk. With the advent of newer medications targeting ERα-positive breast cancer, tamoxifen is now mainly used as adjuvant therapy for lower-risk premenopausal breast cancer and cancer prevention. It is widely accepted that tamoxifen as a selective estrogen receptor modulator exerts its therapeutic effect by competitively binding to ERα, leading to the recruitment of corepressors and inhibition of transcription of genes involved in the proliferation of breast cancer epithelium. As such, expression of ERα in breast tumors has been considered necessary for tumors to be responsive to tamoxifen therapy. However, ERα-independent effects of tamoxifen in various in vitro and in vivo contexts have been reported over the years. Importantly, the recent discovery that ERα and estrogen receptor β (ERβ/ESR2) can bind tumor suppressor protein p53 with functional consequences has provided new insights into the mechanisms underlying response to tamoxifen therapy and resistance. Furthermore, these findings have paved the way for broadening the use of tamoxifen by potentially repurposing it to treat triple negative (negative for ERα, human epidermal growth factor receptor 2, and progesterone receptor) breast cancer. Herein, we summarize these developments and discuss their mechanistic underpinnings and clinical implications.
Collapse
Affiliation(s)
- Gokul M Das
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Chetan C Oturkar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Vishnu Menon
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
16
|
Wu Y, Chen Y, Liu F, Li K. The Immunomodulatory Role of Estrogen in Malaria: A Review of Sex Differences and Therapeutic Implications. Immun Inflamm Dis 2025; 13:e70148. [PMID: 39898752 PMCID: PMC11789271 DOI: 10.1002/iid3.70148] [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: 08/07/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Malaria remains a significant global health challenge, with substantial mortality rates, particularly in tropical and subtropical regions. A notable sexual dimorphism exists in malaria, with males often experiencing higher infection and mortality rates compared to females. OBJECTIVE This review explores the role of estrogen in modulating immune responses to malaria, potentially explaining the observed sex differences. Estrogen, through its receptors, influences immune cell activation and cytokine production, which are critical in the immune response to malaria. RESULTS Utilizing data from the Global Burden of Disease (GBD) study, we analyzed sex differences in malaria burden in Central Sub-Saharan Africa from 2000 to 2021, revealing a significantly lower mortality burden for females compared to males. Epidemiological data and animal model results support the notion that estrogen plays a significant role in modulating immune responses to malaria. Estrogen receptors are widely expressed in immune cells, and estrogen can influence the activation, proliferation, and differentiation of these cells, thereby affecting cytokine production and immune response type. Additionally, selective estrogen receptor modulators (SERMs) show potential as therapeutic agents, with some studies demonstrating their efficacy in reducing parasitemia and improving malaria outcomes. CONCLUSION Understanding the sex differences in the pathogenesis of malaria is crucial for its prevention, treatment, and vaccine development. Estrogen's role in immune regulation highlights the need for sex-specific approaches in disease management.
Collapse
Affiliation(s)
- Ye Wu
- Department of Pharmacy, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Ying‐Chun Chen
- Department of Laboratory Medicine, The Sixth Hospital of WuhanAffiliated Hospital of Jianghan UniversityWuhanPeople's Republic of China
| | - Fang‐Fang Liu
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Ke Li
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| |
Collapse
|
17
|
Miao HT, Wang J, Shao JJ, Song RX, Li WG, Sun JK, Jia SY, Zhang DX, Li XM, Zhao JY, Zhang LM. Astrocytic NLRP3 cKO mitigates depression-like behaviors induced by mild TBI in mice. Neurobiol Dis 2025; 205:106785. [PMID: 39793767 DOI: 10.1016/j.nbd.2024.106785] [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: 08/13/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Reports indicate that depression is a common mental health issue following traumatic brain injury (TBI). Our prior research suggests that Nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-related neuroinflammation, modulated by glial cells such as astrocytes, is likely to play a crucial role in the progression of anxiety and cognitive dysfunction. However, there is limited understanding of the potential of astrocytic NLRP3 in treating depression under mild TBI condition. This study aimed to determine whether astrocytic NLRP3 knockout (KO) could mitigate depressive-like behaviors following mild TBI and explore potential variations in such behaviors between genders post-mild TBI. METHODS Mild TBI was induced in mice using Feeney's weight-drop method. Behavioral assessments included neurological severity scores (NSS), social interaction test (SI), tail suspension test (TST), and forced swimming test (FST). Pathological changes were evaluated through immunofluorescence and local field potential (LFP) recordings at various time points post-injury. RESULTS Our findings indicated that astrocyte-specific NLRP3 KO decreased cleaved caspase-1 colocalized with astrocytes, decreased pathogenic astrocytes and increased Postsynaptic density protein 95 (PSD95) intensity, and significantly alleviated mild TBI-induced depression-like behaviors. It also led to the upregulation of protective astrocytes and apoptosis-associated factors, including cleaved caspase-3 post-mild TBI. Additionally, astrocyte-specific NLRP3 deletion resulting in improved θ and γ power and θ-γ phase coupling in the social interaction test (SI). Notably, under mild TBI conditions, astrocyte-specific NLRP3 exhibited greater neuroprotective effects in female knockout mice compared to males. CONCLUSION Astrocyte NLRP3 knockout demonstrated a protective mechanism in mice subjected to mild TBI, possibly attributed to the inhibition of pyroptosis through the NLRP3 signaling pathway in astrocytes.
Collapse
Affiliation(s)
- Hui-Tao Miao
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China,; Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China; Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Jun Wang
- Department of Orthopaedics, Tianjin Hospital, Tianjin University, Tianjin, China
| | - Jing-Jing Shao
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong-Xin Song
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China
| | - Wen-Guang Li
- Graduated School, Hebei Medical University, Shijiazhuang, China
| | - Jian-Kai Sun
- Graduated School, Hebei Medical University, Shijiazhuang, China
| | - Shi-Yan Jia
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China
| | - Dong-Xue Zhang
- Department of Gerontology, Cangzhou Central Hospital, Cangzhou, China
| | - Xiao-Ming Li
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Jian-Yong Zhao
- Hebei Key Laboratory of Integrated Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, China
| | - Li-Min Zhang
- Department of Anesthesiology, Hebei Province, Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, China,.
| |
Collapse
|
18
|
Singh P, Mohanty B. Neurotensin receptor agonist PD149163 modulates LPS-induced enterocyte apoptosis by downregulating TNFR pathway and executioner caspase 3 in endotoxemic mice: insights from in vivo and in silico study. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03794-9. [PMID: 39812770 DOI: 10.1007/s00210-025-03794-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
This study was designed to evaluate the dose-dependent efficacy of neurotensin receptor-1 (NTSR1) agonist PD149163 in the amelioration of the lipopolysaccharide (LPS)-induced apoptosis in the gastrointestinal tract (GIT) of mice. PD149163 is an analogue of NTS, a GIT tri-decapeptide with anti-inflammatory and anti-oxidative effects. Swiss-albino mice (female/8 weeks/25 ± 2.5 g) were divided into six groups: control; LPS, LPS + PD149163L, and LPS + PD149163H groups were treated with LPS (0.2 μmol/L/kgBW; 5 days), followed by exposure of PD149163 to LPS + PD149163L (10.6 μmol/L/kgBW), and LPS + PD149163H (21.2 μmol/L/kgBW) for 28 days. OnlyPD149163L (10.6 μmol/L/kgBW) and onlyPD149163H (21.2 μmol/L/kgBW) groups were maintained for 28 days. Both the LPS and PD149163 were given intraperitoneally. PD149163 treatment for 4 weeks alleviated the LPS-induced enterocyte apoptosis in a dose-dependent manner. LPS-induced excessive levels of caspase-3, tumour necrosis factor-α, and leptin (biomarkers of LPS-induced apoptosis) in plasma were decreased by PD149163H treatment. Moreover, LPS-induced gut oxidative stress was ameliorated by PD149163H supplementation, as evidenced by the decreased content of malondialdehyde, lipid-hydroperoxide and increased level of superoxide-dismutase, catalase. Furthermore, PD149163H mediated elevation of the plasma anti-apoptotic protein (B-cell leukaemia/lymphoma-2) along with the NTS level contributed to the modulation of LPS-induced enterocyte apoptosis, reflected in histopathology. In vivo results were substantiated with in silico molecular docking analysis that predicted the binding of PD149163-TLR4 complex, suggesting that PD149163 can act as a TLR4 modulator and inhibit the activation of TLR4. The role of PD149163 in ameliorating GIT apoptosis by its anti-apoptotic and antioxidative effects is suggested. Further research may provide significant insights into the therapeutic intervention of PD149163 in apoptosis-related diseases of GIT.
Collapse
Affiliation(s)
- Priya Singh
- Department of Zoology, University of Allahabad, Senate House, University Road, Old Katra, Prayagraj, Uttar Pradesh, 211002, India
| | - Banalata Mohanty
- Department of Zoology, University of Allahabad, Senate House, University Road, Old Katra, Prayagraj, Uttar Pradesh, 211002, India.
| |
Collapse
|
19
|
Greygoose E, Metharom P, Kula H, Seckin TK, Seckin TA, Ayhan A, Yu Y. The Estrogen-Immune Interface in Endometriosis. Cells 2025; 14:58. [PMID: 39791759 PMCID: PMC11720315 DOI: 10.3390/cells14010058] [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: 12/16/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Endometriosis is a gynecologic condition characterized by the growth of endometrium-like stroma and glandular elements outside of the uterine cavity. The involvement of hormonal dysregulation, specifically estrogen, is well established in the initiation, progression, and maintenance of the condition. Evidence also highlights the association between endometriosis and altered immune states. The human endometrium is a highly dynamic tissue that undergoes frequent remodeling in response to hormonal regulation during the menstrual cycle. Similarly, endometriosis shares this propensity, compounded by unclear pathogenic mechanisms, presenting unique challenges in defining its etiology and pathology. Here, we provide a lens to understand the interplay between estrogen and innate and adaptive immune systems throughout the menstrual cycle in the pathogenesis of endometriosis. Estrogen is closely linked to many altered inflammatory and immunomodulatory states, affecting both tissue-resident and circulatory immune cells. This review summarizes estrogenic interactions with specific myeloid and lymphoid cells, highlighting their implications in the progression of endometriosis.
Collapse
Affiliation(s)
- Emily Greygoose
- Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
| | - Pat Metharom
- Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
| | - Hakan Kula
- Department of Obstetrics and Gynecology, Faculty of Medicine, Dokuz Eylul University, Izmir 35340, Turkey
| | - Timur K. Seckin
- Burnett School of Medicine, Texas Christian University, Fort Worth, TX 76104, USA;
| | - Tamer A. Seckin
- Department of Gynecology, Lenox Hill Hospital, and Hofstra University, New York, NY 10075, USA
| | - Ayse Ayhan
- Department of Tumor Pathology, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yu Yu
- Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
- Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6102, Australia
- Discipline of Obstetrics and Gynaecology, Medical School, University of Western Australia, Crawley, WA 6009, Australia
| |
Collapse
|
20
|
Wen X, Mao Y, Li Z, Chen G, Zhou S. Association between weight-adjusted waist index and Hashimoto's thyroiditis: insights from NHANES 2007-2012. Front Nutr 2025; 11:1520440. [PMID: 39834468 PMCID: PMC11743686 DOI: 10.3389/fnut.2024.1520440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Objective While previous studies have explored the relationship between obesity and levels of thyroid autoantibodies, research using novel indicators such as weight-adjusted waist index (WWI) remains limited. This study aimed to evaluate the potential relationship between WWI and thyroid autoantibody levels, with the objective of improving our understanding of the links between central obesity and Hashimoto's thyroiditis (HT). Methods We conducted a cross-sectional study using data from the National Health and Nutrition Examination Survey (NHANES) cycles from 2007 to 2012. We analyzed the relationship between WWI and levels of thyroid peroxidase antibodies (TPOAb) and thyroglobulin antibodies (TgAb) through multivariate linear regression and subgroup analyses. Results The study included 7,056 participants with an average age of 49.71 ± 17.66 years, comprising 49.18% females. Mean WWI across the cohort was 11.04 ± 0.84. Analysis revealed a significant positive association between WWI and TPOAb levels (β: 4.78, 95% CI: 1.52, 8.05, p = 0.0041), which remained consistent across all multivariate linear regression models. In contrast, no significant correlation was found between WWI and TgAb levels after adjusting for covariates. Subgroup analysis stratified by gender demonstrated a notable gender-specific effect, where the positive correlation between WWI and TPOAb levels was evident only in females (β: 8.13, 95% CI: 4.14, 12.12, p < 0.0001). Conclusion This study used WWI as a novel indicator of central obesity and identified a strong association with HT, particularly notable in females. However, further high-quality studies are needed to confirm these findings and explore the underlying biological mechanisms.
Collapse
Affiliation(s)
- Xiaoyong Wen
- Department of Thyroid Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Mao
- Department of Thyroid Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zeyu Li
- Department of Thyroid Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
- Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangji Chen
- Department of Surgery, University Hospital, Central South University, Changsha, Hunan, China
| | - Shiwei Zhou
- Department of Thyroid Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan, China
| |
Collapse
|
21
|
Wang P, Huang J, Xu L, Hu R. The association between the neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and lymphocyte-to-monocyte ratio and delirium in ischemic stroke patients. Front Med (Lausanne) 2025; 11:1456742. [PMID: 39835091 PMCID: PMC11743177 DOI: 10.3389/fmed.2024.1456742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Background Delirium is a severe neuropsychiatric symptom following acute ischemic stroke (IS) and is associated with poor outcomes. Systemic inflammation and immune dysregulation are believed to contribute to the pathophysiology of delirium. The neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR) are widely recognized as convenient and reliable biomarkers of systemic inflammation. However, their association with delirium after IS remains unclear. Methods In this study, we identified IS patients requiring ICU admission from the Medical Information Mart for Intensive Care (MIMIC)-IV database. We employed multivariable logistic regression and restricted cubic splines (RCS) to assess the association between the NLR, PLR, and LMR and delirium. Two-sample Mendelian randomization (MR) analysis was performed to further explore their causal relationship at the genetic level. Results A total of 1,436 patients with IS were included in this study, of whom 214 (14.9%) had delirium. In the multivariate logistic regression analysis, after adjustment for confounders, the patients in the highest quartile of the NLR (odds ratio [OR] 2.080, 95% confidence interval [CI], 1.282-3.375) and LMR (OR 0.503, 95% CI 0.317-0.798) and the patients in the second quartile of the PLR (OR 1.574, 95% CI 1.019-2.431) were significantly associated with delirium. The RCS function showed a progressive increase in the risk of delirium with higher NLR and PLR and lower LMR. In the MR analysis, only the PLR was negatively associated with the risk of delirium. Conclusion The observational studies found significant associations between the NLR, PLR, and LMR and delirium. However, the MR analysis only demonstrated a potential protective causal relationship between the PLR and delirium. Further prospective studies are needed to validate their association and to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Pangbo Wang
- State Key Laboratory of Trauma, Burn, and Combined Injury, Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Trauma Neurosurgery, NO. 946 Hospital of PLA Land Force, Yining, China
| | - Jing Huang
- School of Nursing, Peking University, Beijing, China
| | - Liwei Xu
- Department of Burn Plastic Surgery, NO. 946 Hospital of PLA Land Force, Yining, China
| | - Rong Hu
- State Key Laboratory of Trauma, Burn, and Combined Injury, Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
22
|
Diao M, Wang Y, Wu S, He S, Liao Y. Estrogen, estrogen receptor and the tumor microenvironment of NSCLC. Int J Cancer 2025. [PMID: 39754298 DOI: 10.1002/ijc.35309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/06/2025]
Abstract
Lung cancer remains the foremost cause of cancer-related mortality worldwide. Clinical observations reveal a notable increase in both the proportion and mortality rate among female non-small cell lung cancer (NSCLC) patients compared to males, a trend that continues to escalate. Extensive preclinical research underscores the pivotal role of estrogen in the initiation, progression, prognosis, and treatment response of NSCLC. Estrogen receptors are widely expressed in stromal and immune cells, influencing cellular activities across innate and adaptive immune systems. Immune evasion mechanisms significantly impact tumor development and outcomes, with immunotherapy offering promise in NSCLC by targeting these mechanisms. The intriguing gender disparities in immunotherapy responses prompt an exploration into the data on NSCLC occurrence, progression, and gender-specific immunotherapy. Evidence highlights estrogen's contribution to a tumor-permissive microenvironment, influencing various cells including cancer-associated fibroblasts, macrophages, neutrophils, dendritic cells, natural killer cells, B cells, and T cells. Gender-specific variations in NSCLC occurrence, development, prognosis, and treatment efficacy likely stem from interactions between estrogen, lung cancer cells, and these estrogen-responsive cells, shaping a microenvironment conducive to tumor progression. Clarifying estrogen's role and its signaling pathways in NSCLC may unveil novel therapeutic strategies to modify the tumor microenvironment or enhance immunotherapy efficacy.
Collapse
Affiliation(s)
- Mingxin Diao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangwei Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shihao Wu
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiwen He
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Corti C, Binboğa Kurt B, Koca B, Rahman T, Conforti F, Pala L, Bianchini G, Criscitiello C, Curigliano G, Garrido-Castro AC, Kabraji SK, Waks AG, Mittendorf EA, Tolaney SM. Estrogen Signaling in Early-Stage Breast Cancer: Impact on Neoadjuvant Chemotherapy and Immunotherapy. Cancer Treat Rev 2025; 132:102852. [PMID: 39571402 DOI: 10.1016/j.ctrv.2024.102852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/14/2024] [Accepted: 11/10/2024] [Indexed: 01/01/2025]
Abstract
Neoadjuvant chemoimmunotherapy (NACIT) has been shown to improve pathologic complete response (pCR) rates and survival outcomes in stage II-III triple-negative breast cancer (TNBC). Promising pCR rate improvements have also been documented for selected patients with estrogen receptor-positive (ER+) human epidermal growth factor receptor 2-negative (HER2-) breast cancer (BC). However, one size does not fit all and predicting which patients will benefit from NACIT remains challenging. Accurate predictions would be useful to minimize immune-related toxicity, which can be severe, irreversible, and potentially impact fertility and quality of life, and to identify patients in need of alternative treatments. This review aims to capitalize on the existing translational and clinical evidence on predictors of treatment response in patients with early-stage BC treated with neoadjuvant chemotherapy (NACT) and NACIT. It summarizes evidence suggesting that NACT/NACIT effectiveness may correlate with pre-treatment tumor characteristics, including mutational profiles, ER expression and signaling, immune cell presence and spatial organization, specific gene signatures, and the levels of proliferating versus quiescent cancer cells. However, the predominantly qualitative and descriptive nature of many studies highlights the challenges in integrating various potential response determinants into a validated, comprehensive, and multimodal predictive model. The potential of novel multi-modal approaches, such as those based on artificial intelligence, to overcome current challenges remains unclear, as these tools are not free from bias and shortcut learning. Despite these limitations, the rapid evolution of these technologies, coupled with further efforts in basic and translational research, holds promise for improving treatment outcome predictions in early HER2- BC.
Collapse
Affiliation(s)
- Chiara Corti
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy.
| | - Busem Binboğa Kurt
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Beyza Koca
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Tasnim Rahman
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Fabio Conforti
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Laura Pala
- Department of Medical Oncology, Humanitas Gavazzeni, Bergamo, Italy
| | - Giampaolo Bianchini
- Department of Medical Oncology, San Raffaele Hospital, IRCCS, Milan, Italy; School of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology (DIPO), University of Milan, Milan, Italy
| | - Ana C Garrido-Castro
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sheheryar K Kabraji
- Department of Medicine, Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Adrienne G Waks
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Breast Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Sara M Tolaney
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
24
|
Viswanath M, Peter MCS. Thyroid hormones activate TH/E 2 receptor/regulator system and drive Na +/K +-ATPase in the ovarian wall of hypothyroid air-breathing fish (Anabas testudineus Bloch). Gen Comp Endocrinol 2025; 360:114640. [PMID: 39536982 DOI: 10.1016/j.ygcen.2024.114640] [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: 12/29/2023] [Revised: 11/09/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
In fish, as in other vertebrates, thyroid hormones (THs) act on many biological processes including growth and reproduction. Primary THs such as thyroxine (T4) and triiodothyronine (T3) are known for their direct action on osmoregulatory organs regulating ion osmotic homeostasis in many teleost fishes. However, it is unclear how these hormones interact with estradiol-17β (E2), an ovarian hormone that regulates the development of oocytes. We thus examined the short-term in vivo action of varied THs such as T4, T3 and T2, a potent TH metabolite diiodothyronine, on the expression pattern of receptors of THs and E2 in the ovarian wall of the hypothyroid climbing perch to identify the interactive pattern of TH/E2 receptor system and the molecular dynamics of Na+/K+-ATPase (NKA) subunits in the ovarian wall that provides structural and functional support to ovary. We found differential pattern of transcript abundance of NKA subunits isoforms such as nkaα1a, nkaα1b, nkaα1c atp1b1, atp1b2 and fxyd3, fxyd5, fxyd6, TH receptor isoforms (tr<, trβ, tr
Collapse
Affiliation(s)
- Meenu Viswanath
- Inter-University Centre for Evolutionary and Integrative Biology-iCEIB, School of Life Sciences, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Department of Zoology, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India
| | - M C Subhash Peter
- Inter-University Centre for Evolutionary and Integrative Biology-iCEIB, School of Life Sciences, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Department of Zoology, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India; Sastrajeevan Integrative Bioresearch and Education-SIBE, F17-Gandhipuram, Sreekariyam, Thiruvananthapuram 695017, Kerala, India.
| |
Collapse
|
25
|
Tchalla EYI, Betadpur A, Khalil AY, Bhalla M, Bou Ghanem EN. Sex-based difference in immune responses and efficacy of the pneumococcal conjugate vaccine. J Leukoc Biol 2024; 117:qiae177. [PMID: 39141715 PMCID: PMC11684992 DOI: 10.1093/jleuko/qiae177] [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: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 08/16/2024] Open
Abstract
Vaccine-mediated protection and susceptibility to Streptococcus pneumoniae (pneumococcus) infections are influenced by biological sex. The incidence of invasive pneumococcal disease remains higher in males compared to females even after the introduction of the pneumococcal conjugate vaccine. However, sex-based differences in the immune response to this conjugate vaccine remain unexplored. To investigate those differences, we vaccinated adult male and female mice with pneumococcal conjugate vaccine and assessed cellular and humoral immune responses. Compared to females, male mice displayed lower levels of T follicular helper cells, germinal center B cells, and plasmablasts, which are all required for antibody production following vaccination. This was linked to lower IgG and IgM levels against pneumococci and lower isotype switching to IgG3 in vaccinated males. Due to lower antibody levels, sera of vaccinated male mice had lower efficacy in several anti-pneumococcal functions, including neutralization of bacterial binding to pulmonary epithelial cells as well as direct cytotoxicity against S. pneumoniae. Importantly, while the vaccine was highly protective in females, vaccinated males succumbed to infection more readily and were more susceptible to both lung-localized infection and systemic spread following S. pneumoniae challenge. These findings identify sex-based differences in immune responses to pneumococcal conjugate vaccine that can inform future vaccine strategies.
Collapse
Affiliation(s)
- Essi Y I Tchalla
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| | - Anagha Betadpur
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| | - Andrew Y Khalil
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| | - Elsa N Bou Ghanem
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY 14203, United States
| |
Collapse
|
26
|
Zhao R, Lian W, Xu Q. Sex hormones and immune regulation in ovarian cancer. Discov Oncol 2024; 15:849. [PMID: 39738765 DOI: 10.1007/s12672-024-01675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
Ovarian cancer continues to be a major cause of morbidity and mortality in women, with immune regulation playing a critical role in its progression and treatment response. This review explores the interplay between sex hormones, particularly estrogen and progesterone, and immune regulation in ovarian cancer. We delve into the mechanisms by which these hormones influence immune cell function, modulate immune checkpoints, and alter the tumor microenvironment. Key pathways involving estrogen and progesterone receptors are examined, highlighting their impact on tumor growth and immune evasion. The review also discusses the therapeutic implications of these interactions, including the potential for combining hormone-based therapies with immune checkpoint inhibitors. Personalized medicine approaches, leveraging biomarkers for predicting treatment response, are considered essential for optimizing patient outcomes. Finally, we address current research gaps and future directions, emphasizing the need for advanced research technologies and novel therapeutic strategies to improve the treatment of ovarian cancer through a better understanding of hormone-immune interactions.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Tianhe District, Guangzhou, 510623, Guangdong, China
| | - Wenqin Lian
- Department of Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Tianhe District, Guangzhou, 510623, Guangdong, China.
| | - Qiong Xu
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Tianhe District, Guangzhou, 510623, Guangdong, China.
| |
Collapse
|
27
|
Skrzypczak M, Wolinska E, Adaszek Ł, Ortmann O, Treeck O. Epigenetic Modulation of Estrogen Receptor Signaling in Ovarian Cancer. Int J Mol Sci 2024; 26:166. [PMID: 39796024 PMCID: PMC11720219 DOI: 10.3390/ijms26010166] [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: 11/27/2024] [Revised: 12/21/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Ovarian cancer remains one of the leading causes of cancer-related deaths in women. There are several processes that are described to have a causal relationship in ovarian cancer development, progression, and metastasis formation, that occur both at the genetic and epigenetic level. One of the mechanisms involved in its pathogenesis and progression is estrogen signaling. Estrogen receptors (ER) α, ERβ, and G-protein coupled estrogen receptor 1 (GPER1), in concert with various coregulators and pioneer transcription factors, mediate the effects of estrogens primarily by the transcriptional regulation of estrogen responsive genes, thereby exerting pleiotropic effects including the regulation of cellular proliferation and apoptosis. The expression and activity of estrogen receptors and their coregulators have been demonstrated to be regulated by epigenetic mechanisms like histone modifications and DNA methylation. Here, we intend to summarize and to provide an update on the current understanding of epigenetic mechanisms regulating estrogen signaling and their role in ovarian cancer. For this purpose, we reviewed publications on this topic listed in the PubMed database. Finally, we assess to which extent drugs acting on the epigenetic level might be suitable for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Maciej Skrzypczak
- Chair and Department of Gynecology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Ewa Wolinska
- Department of Pathology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Łukasz Adaszek
- Clinic of Infectious Diseases, University of Life Sciences Lublin, 20-950 Lublin, Poland;
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany;
| | - Oliver Treeck
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93935 Regensburg, Germany;
| |
Collapse
|
28
|
Gao Z, Ding C, Huang X, Liu Y, Fan W, Song S. Estrogen receptor α aggravates intestinal inflammation via promoting the activation of NLRP3 inflammasome. Int Immunopharmacol 2024; 143:113425. [PMID: 39426237 DOI: 10.1016/j.intimp.2024.113425] [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: 08/26/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Activation of the NLRP3 inflammasome and estrogen receptor α (ERα) has been shown to increase the risk of inflammatory bowel diseases (IBD) or promote disease recurrence. In previous work, we demonstrated that ERα regulated the transcription of NLRP3. However, the precise mechanism by which ERα modulates NLRP3 in IBD models remains unclear. In this study, we induced IBD in wild-type mice using DSS or TNBS, followed by treatment with the ERα-specific agonist PPT. The results showed that IBD symptoms and intestinal inflammation responses were significantly exacerbated after PPT treatment. Furthermore, the activation of ERα by PPT led to a marked increase in the expression of NLRP3 and pro-inflammatory cytokines, including IL-1β and IL-18, suggesting that ERα activation exacerbated intestinal inflammation and impaired mucosal healing during the recovery phase of inflammation. In contrast, ERα-knockout mice exhibited only mild symptoms when exposed to DSS or TNBS, with a concurrent reduction in NLRP3 expression, indicating that ERα plays a role in inflammation susceptibility. Similar findings were observed in NCM-460 cells, where the inflammation response was attenuated in ERα-knockdown cells. Importantly, we demonstrated that ERα interacted with the NLRP3 inflammasome and promoted its assembly. Collectively, we propose an underlying pathogenesis of IBD, that is, ERα can interact with the NLRP3 inflammasome and promote its expression and assembly, thereby exacerbating intestinal inflammation in IBD models. Therefore, ERα could serve as a potential therapeutic target for NLRP3 inflammasome-associated intestinal inflammation.
Collapse
Affiliation(s)
- Zhangshan Gao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Chenchen Ding
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Xi Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Yapei Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Wentao Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China
| | - Suquan Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu Province, China.
| |
Collapse
|
29
|
Gong R, Wang S, Ding H, Yu L, Xu M, Xu S, Ling Y. Association between thyroid hormone sensitivity and carotid plaque risk: a health examination cohort-based study. Front Endocrinol (Lausanne) 2024; 15:1472752. [PMID: 39722809 PMCID: PMC11668592 DOI: 10.3389/fendo.2024.1472752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction The involvement of thyroid hormone in cardiovascular disease remains debated. The aim of our research was to ascertain whether thyroid hormone sensitivity indices are related to carotid plaque (CAP) risk in the general population. Methods We recruited 5,360 participants for health examinations to explore the correlation between thyroid hormone sensitivity indices and CAP risk. We then compared baseline characteristics of participants with CAP to those without CAP based on multivariate logistic regression analysis. Additionally, we conducted subgroup analyses stratified by gender and age to further elucidate this relationship. Results Among the 5,360 participants, 1,055 (19.7%) were diagnosed with CAP. After adjusting for various confounding factors, our results showed a positive association between CAP risk and the indices (TFQI, PTFQI, TSHI, and TT4RI). Conversely, the FT3/FT4 ratio showed a negative correlation with CAP risk. Sex-based subgroup analysis revealed a stronger correlation between thyroid hormone sensitivity and CAP in females compared to males. In the age subgroup, the significant association was observed in older individuals (age >60) compared to middle-aged participants (age ≤60). Conclusion Our study suggests a significant correlation between thyroid hormone sensitivity and CAP, particularly in females and participants over the age of 60.
Collapse
Affiliation(s)
- Rui Gong
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi Wang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongqiong Ding
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lixia Yu
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Xu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sanping Xu
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Ling
- Health Management Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Li J, Chen Y, Li S, Zhang X, Cheng Y, Fu X, Li J, Zhu L. Estrogen Receptor β Alleviates Colitis by Inhibiting Ferroptosis in Intestinal Epithelial Cells. J Inflamm Res 2024; 17:10785-10805. [PMID: 39677282 PMCID: PMC11645966 DOI: 10.2147/jir.s492290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Background Ulcerative colitis (UC), a major type of inflammatory bowel disease, is characterized by chronic inflammation of the colonic mucosa and submucosa. Estrogen receptor β (ERβ) predominates in the colon and exerts anti-inflammatory effects. Ferroptosis, a recently discovered form of iron-dependent programmed cell death, is implicated in the pathogenesis of several diseases, including UC. However, the link between ferroptosis and the anti-inflammatory actions of ERβ in UC remains to be elucidated. Methods We analyzed colonic mucosal samples from inflammatory and non-inflammatory regions of UC patients to assess ferroptosis levels. Experimental colitis was induced in wild-type C57BL/6 mice and intestinal epithelial cell-specific ERβ knockout (ERβ-/-) mice using dextran sulfate sodium (DSS). We measured body weight, colon length, disease activity index (DAI), and histopathological scores. RNA sequencing was performed to identify differentially expressed genes and related signaling pathways, with additional ferroptosis assessment in vivo and in vitro through biochemical markers and cellular assays. Results In UC patients, ferroptosis was significantly elevated in inflammatory mucosal regions compared to non-inflammatory areas. Compared to the wild-type counterparts, ERβ-/- mice exacerbated DSS-induced experimental colitis, including reduced body weight, shortened colon length, and higher DAI scores. RNA sequencing showed enrichment of inflammatory and immune response pathways, with significant activation of JAK/STAT, NF-κB, and TNF signaling in ERβ-/- mice. ERβ deficiency induced ferroptosis in both in vitro and in vivo models. Ferroptosis indicators such as PTGS2 were upregulated, GPX4 expression was downregulated, and there were increases in malondialdehyde, iron content, reactive oxygen species, and mitochondrial damage. Conclusion Our findings demonstrate that ERβ deficiency exacerbates colitis and enhances ferroptosis in IECs. ERβ positively regulates GPX4 transcription, thereby inhibiting ferroptosis and alleviating colitis. These insights suggest that modulation of ERβ and its regulation of ferroptosis may represent a novel therapeutic strategy for UC.
Collapse
Affiliation(s)
- Junrong Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Division of Gastroenterology, Chongqing Hospital Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Chongqing, People’s Republic of China
| | - Yidong Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shuang Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiaopeng Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yiyu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiaoyu Fu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jiamin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Liangru Zhu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
31
|
Caneparo C, Carignan L, Lonina E, Goulet SM, Pellerin FA, Chabaud S, Bordeleau F, Bolduc S, Pelletier M. Impact of Endocrine Disruptors on the Genitourinary Tract. J Xenobiot 2024; 14:1849-1888. [PMID: 39728407 DOI: 10.3390/jox14040099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/04/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Over the last decades, the human species has seen an increase in the incidence of pathologies linked to the genitourinary tract. Observations in animals have allowed us to link these increases, at least in part, to changes in the environment and, in particular, to an increasing presence of endocrine disruptors. These can be physical agents, such as light or heat; natural products, such as phytoestrogens; or chemicals produced by humans. Endocrine disruptors may interfere with the signaling pathways mediated by the endocrine system, particularly those linked to sex hormones. These factors and their general effects are presented before focusing on the male and female genitourinary tracts by describing their anatomy, development, and pathologies, including bladder and prostate cancer.
Collapse
Affiliation(s)
- Christophe Caneparo
- Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, Geneva University Hospitals, University of Geneva, CH-1205 Geneva, Switzerland
| | - Laurence Carignan
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - Elena Lonina
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Sarah-Maude Goulet
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Felix-Antoine Pellerin
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - Stéphane Chabaud
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
| | - François Bordeleau
- Oncology Division, CHU de Québec-Université Laval Research Center and Université Laval Cancer Research Center, Quebec, QC G1R 3S3, Canada
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Stéphane Bolduc
- Regenerative Medicine Division, Centre de Recherche en Organogénèse Expérimentale/LOEX, CHU de Québec-Université Laval Research Center, Université Laval, Quebec, QC G1J 5B3, Canada
- Department of Surgery, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Martin Pelletier
- Infectious and Immune Diseases Division, CHU de Québec-Université Laval Research Center, Quebec, QC G1V 4G2, Canada
- Intersectorial Centre for Endocrine Disruptors Analysis, Institut National de La Recherche Scientifique (INRS), Montreal, QC H4V 1B7, Canada
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University; ARThrite Research Center, Université Laval, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
32
|
D'Onofrio V, Sékaly RP. The immune-endocrine interplay in sex differential responses to viral infection and COVID-19. Trends Immunol 2024; 45:943-958. [PMID: 39562265 DOI: 10.1016/j.it.2024.10.004] [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: 08/21/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 11/21/2024]
Abstract
Men are at higher risk for developing severe COVID-19 than women, while women are at higher risk for developing post-acute sequelae of COVID-19 (PASC). This highlights the impact of sex differences on immune responses and clinical outcomes of acute COVID-19 or PASC. A dynamic immune-endocrine interface plays an important role in the development of effective immune responses impacting the control of viral infections. In this opinion article we discuss mechanisms underlying the transcriptional and epigenetic regulation of immune responses by sex hormones during viral infections. We propose that disruption of this delicate immune-endocrine interplay can result in worsened outcomes of viral disease. We also posit that insights into these immune mechanisms can propel the development of novel immunomodulatory interventions that leverage immune-endocrine pathways to treat viral infections.
Collapse
Affiliation(s)
- Valentino D'Onofrio
- Center for Vaccinology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Rafick Pierre Sékaly
- Pathology Advanced Translational Research Unit, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
33
|
Niu Q, Hao J, Li Z, Zhang H. Helper T cells: A potential target for sex hormones to ameliorate rheumatoid arthritis? (Review). Mol Med Rep 2024; 30:215. [PMID: 39370806 PMCID: PMC11450432 DOI: 10.3892/mmr.2024.13339] [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: 07/17/2024] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease whose etiology is not fully understood. Defective peripheral immune tolerance and subsequent mis‑differentiation and aberrant infiltration of synovium by various immune cells, especially helper T (Th) cells, play an important role in the development of RA. There are significant sex differences in RA, but the results of studies on the effects of sex hormones on RA have been difficult to standardize and hormone replacement therapy has been limited by the potential for serious side effects. Existing research has amply demonstrated that cellular immune responses are largely determined by sex and that sex hormones play a key role in Th cell responses. Based on the aforementioned background and the plasticity of Th cells, it is reasonable to hypothesize that the action of sex hormones on Th cells will hopefully become a therapeutic target for RA. The present review discussed the role of various Th cell subsets in the pathogenesis of RA and also explored the role of sex hormones on the phenotype and function of these aberrantly regulated immune cells in RA as well as other pathologic effects on RA.
Collapse
Affiliation(s)
- Quanjun Niu
- Department of Orthopedics IV, Handan Hospital of Traditional Chinese Medicine, Handan, Hebei 056001, P.R. China
| | - Junhang Hao
- Department of Orthopedics IV, Handan Hospital of Traditional Chinese Medicine, Handan, Hebei 056001, P.R. China
| | - Zhen Li
- Department of Orthopedics IV, Handan Hospital of Traditional Chinese Medicine, Handan, Hebei 056001, P.R. China
| | - Huiping Zhang
- Department of Orthopedics IV, Handan Hospital of Traditional Chinese Medicine, Handan, Hebei 056001, P.R. China
| |
Collapse
|
34
|
Li L, Li H, Chen B. Chronobiological and neuroendocrine insights into dry eye. Trends Mol Med 2024:S1471-4914(24)00279-X. [PMID: 39551666 DOI: 10.1016/j.molmed.2024.10.012] [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: 08/28/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024]
Abstract
Dry eye, a prevalent ocular surface disease, is significantly influenced by modern lifestyle factors such as night-shift work and extended screen time. Emerging evidence suggests a strong correlation between disturbances in circadian rhythm, sleep disorders, and dry eye. However, the precise underlying mechanisms remain unclear. Recent studies have underscored the crucial role of circadian rhythms and neuroendocrine regulation in maintaining ocular surface health. Advances in treatment strategies targeting neuroendocrine pathways have shown promising developments. This review explores the interplay between circadian rhythms, neuroendocrine regulation, and the ocular surface, examines the impact of circadian disruption on the pathophysiology of dry eye, and proposes intervention strategies to alleviate dry eye associated with disturbances in circadian rhythms.
Collapse
Affiliation(s)
- Licheng Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Haoyu Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China
| | - Baihua Chen
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, China.
| |
Collapse
|
35
|
Venegas-Ramírez J, Mendoza-Cano O, Trujillo X, Huerta M, Ríos-Silva M, Lugo-Radillo A, Bricio-Barrios JA, Cuevas-Arellano HB, Uribe-Ramos JM, Solano-Barajas R, García-Solórzano LA, Camacho-delaCruz AA, Murillo-Zamora E. Sex differences in pneumonia risk during COVID-19 in Mexico. Sci Rep 2024; 14:27962. [PMID: 39543312 PMCID: PMC11564899 DOI: 10.1038/s41598-024-78200-0] [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: 05/22/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
This study aimed to evaluate the pneumonia risk based on the patient's sex during the COVID-19 pandemic and the early months of the endemic phase of the disease in Mexico. A retrospective cohort study was conducted using a dataset resulting from the epidemiological surveillance of COVID-19 (February 2020 to August 2023). Data from 1.6 million adults with laboratory-positive disease, were analyzed. Risk ratios (RR) and 95% confidence intervals (CI), computed through generalized linear regression models, were used. The overall risk of pneumonia was 9.3% (95% CI 9.2-9.4%), with sex-specific estimates of 7.0% (95% CI 6.9-7.1%) for women and 12.0% (95% CI 11.9-12.1%) for men. This disparity was consistently observed throughout all phases of the pandemic, including the endemic phase of the disease. After adjusting for age, predominant viral genotype at illness onset and preexisting medical conditions, men had a 3.3% higher risk of severe manifestations when compared to women (RR = 1.033, 95% CI 1.032-1.034). Our research highlights the potential role of patients' sex as a factor influencing pneumonia risk during and after the COVID-19 pandemic in Mexico. These findings may provide useful considerations for healthcare planning and policy development focused on addressing the impact of the disease on vulnerable populations.
Collapse
Affiliation(s)
- Jesús Venegas-Ramírez
- Coordinación de Investigación en Salud, Jefatura de Servicios de Prestaciones Médicas, Instituto Mexicano del Seguro Social, Doroteo López 442, Colima, 28030, Mexico
| | - Oliver Mendoza-Cano
- Facultad de Ingeniería Civil, Universidad de Colima, km. 9 Carretera Colima-Coquimatlán, Coquimatlán, 28400, Mexico
| | - Xóchitl Trujillo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de julio 965, Colima, 28045, Mexico
| | - Miguel Huerta
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Av. 25 de julio 965, Colima, 28045, Mexico
| | - Mónica Ríos-Silva
- Facultad de Medicina, Universidad de Colima, Av. Universidad 333, Colima, 28040, Mexico
| | - Agustin Lugo-Radillo
- CONAHCyT-Faculty of Medicine and Surgery, Universidad Autónoma Benito Juárez de Oaxaca, Ex Hacienda Aguilera S/N, Carr. a San Felipe del Agua, Oaxaca, 68020, Mexico
| | | | | | - Juan Manuel Uribe-Ramos
- Facultad de Ingeniería Civil, Universidad de Colima, km. 9 Carretera Colima-Coquimatlán, Coquimatlán, 28400, Mexico
| | - Ramón Solano-Barajas
- Facultad de Ingeniería Civil, Universidad de Colima, km. 9 Carretera Colima-Coquimatlán, Coquimatlán, 28400, Mexico
| | - Luis A García-Solórzano
- Tecnológico Nacional de México, Campus Colima, Av. Tecnológico No. 1, Villa de Álvarez, 28976, Mexico
| | - Arlette A Camacho-delaCruz
- Facultad de Ingeniería Civil, Universidad de Colima, km. 9 Carretera Colima-Coquimatlán, Coquimatlán, 28400, Mexico
| | - Efrén Murillo-Zamora
- Unidad de Investigación en Epidemiología Clínica, Instituto Mexicano del Seguro Social, Av. Lapislázuli 250, Villa de Álvarez, 28984, Mexico.
| |
Collapse
|
36
|
Kapic A, Zaman K, Nguyen V, Prokai-Tatrai K, Prokai L. Identification of Estrogen-Responsive Proteins in Mouse Seminal Vesicles Through Mass Spectrometry-Based Proteomics. Pharmaceuticals (Basel) 2024; 17:1508. [PMID: 39598420 PMCID: PMC11597337 DOI: 10.3390/ph17111508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Although estrogenic compounds promise therapeutic potential in treating various conditions, concerns regarding their endocrine-disrupting effects have been raised. Current methodologies for screening estrogenicity in rodent models are limited to the female-specific uterotrophic bioassay. Studies have reported enlargement of the seminal vesicles in orchiectomized males treated with estrogens. However, identifying estrogenicity strictly through changes in wet weights is uninformative regarding the molecular mechanisms of these agents. Therefore, protein-based biomarkers can complement and improve the sensitivity of weight-based assessments. To this end, we present a discovery-driven proteomic analysis of 17β-estradiol's effects on the seminal vesicles. Methods: We treated orchidectomized mice with the hormone for five days and used the vehicle-treated group as a control. Seminal vesicles were analyzed by shotgun approach using data-dependent nanoflow liquid chromatography-tandem mass spectrometry and label-free quantification. Proteins found to be differentially expressed between the two groups were processed through a bioinformatics pipeline focusing on pathway analyses and assembly of protein interaction networks. Results: Out of 668 identified proteins that passed rigorous validation criteria, 133 were regulated significantly by 17β-estradiol. Ingenuity Pathway Analysis® linked them to several hormone-affected pathways, including those associated with immune function such as neutrophil degranulation. The altered protein interaction networks were also related to functions including endocrine disruption, abnormal metabolism, and therapeutic effects. Conclusions: We identified several potential biomarkers for estrogenicity in mouse seminal vesicles, many of them not previously linked with exogenous 17β-estradiol exposure.
Collapse
Affiliation(s)
| | | | | | | | - Laszlo Prokai
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (A.K.); (K.Z.); (V.N.); (K.P.-T.)
| |
Collapse
|
37
|
Payne LA, Seidman LC, Granger SW, Edelman A, Ren B. A Prospective Study of the Relationship of COVID-19 Vaccination to Menstrual Cycle Characteristics in Adolescent Girls. J Adolesc Health 2024; 75:819-826. [PMID: 39152975 PMCID: PMC11490362 DOI: 10.1016/j.jadohealth.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 08/19/2024]
Abstract
PURPOSE The current study aimed to evaluate the impact of the COVID-19 booster vaccine on menstrual cycle characteristics in adolescent girls (aged 13-20) compared to those who did not receive a booster vaccine. METHODS This prospective study measured menstrual cycle length for three cycles prior to and four cycles after vaccination (booster group), seven cycles without vaccination (control group). Menstrual flow, menstrual pain, and menstrual symptoms were assessed at baseline and monthly for 3 months. Stress was assessed at baseline using the PROMIS Pediatric Psychological Stress Experiences scale. Generalized linear mixed effects models were used to examine the changes in menstrual characteristics. RESULTS 65 adolescent girls (47 booster; 18 control) were recruited via social media and from ongoing studies in the United States. Girls in the booster group experienced shorter postbooster cycles by an average 5.35 days (p = .03) compared to prebooster cycle lengths, specifically in the second postbooster cycle, while the control group did not show any changes in cycle length pre-to postbooster. Participants who received the booster in the follicular phase had shorter mean postbooster cycle length (p = .0157) compared to their prebooster cycle length. Higher stress was associated with shorter cycles (p = .03) and increased menstrual symptoms (p = <.001), regardless of group. There were no differences in menstrual flow, menstrual pain, or menstrual symptoms in either group. DISCUSSION The COVID-19 booster vaccine was associated with shorter cycles in adolescent girls. These data demonstrate the need for further investigation regarding potential mechanisms of these observed changes.
Collapse
Affiliation(s)
- Laura A Payne
- McLean Hospital, Belmont, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| | | | | | | | - Boyu Ren
- McLean Hospital, Belmont, Massachusetts; Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
38
|
Bi Y, Xie Z, Cao X, Ni H, Xia S, Bao X, Huang Q, Xu Y, Zhang Q. Cedrol attenuates acute ischemic injury through inhibition of microglia-associated neuroinflammation via ERβ-NF-κB signaling pathways. Brain Res Bull 2024; 218:111102. [PMID: 39414157 DOI: 10.1016/j.brainresbull.2024.111102] [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: 01/29/2024] [Revised: 09/15/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Microglia-associated neuroinflammation plays essential roles in pathology of acute stroke. Cedrol, a natural compound extracted from ginger, has been shown to confer inhibitory effects on inflammation in various diseases. However, whether Cedrol suppresses neuroinflammation and protects brains from acute ischemic injury still remains unclear. In this study, we found that Cedrol inhibited microglia activation and the production of inflammatory factors in LPS-challenged microglia and the penumbra region of middle cerebral artery occlusion (MCAO) mice. We also found that Cedrol reduced the infarct size and mNSS scores and improved acute cerebral ischemia-induced behavioral outcomes, suggesting remarked neuroprotection of Cedrol. Molecular docking analysis showed that Cedrol bound to estrogen receptor β (ERβ) with moderate-strong affinity. Intriguingly, treatment with fulvestrant, an ER blocker, abolished the anti-inflammatory effects of Cedrol. Cedrol significantly reversed the LPS- and MCAO-induced increases in phosphorylation levels of IκB and NF-κB P65 in primary microglia and MCAO mice, respectively. Additionally, Cedrol was observed to rescue LPS-induced shuttling of NF-κB P65 from cytoplasm to nuclei in primary microglia, indicating inhibitory effects of Cedrol on NF-κB signaling. These results suggest microglia associated neuroinflammation may be mediated by ERβ-NF-κB signaling pathway. Together, our study reveals that Cedrol protected brain function from acute cerebral ischemia through inhibition of microglia-associated neuroinflammation via ERβ-NF-κB signaling pathways, and Cedrol may serve as an alternative option for treatment of acute stroke injury.
Collapse
Affiliation(s)
- Yu Bi
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Ziyi Xie
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Huanyu Ni
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Qinyue Huang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Qingxiu Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
39
|
Hua Y, Wang H, Chen T, Zhou Y, Chen Z, Zhao X, Mo S, Mao H, Li M, Wang L, Hong M. Antioxidant 1,2,3,4,6-Penta- O-galloyl-β-D-glucose Alleviating Apoptosis and Promoting Bone Formation Is Associated with Estrogen Receptors. Molecules 2024; 29:5110. [PMID: 39519751 PMCID: PMC11547736 DOI: 10.3390/molecules29215110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
1,2,3,4,6-penta-O-galloyl-β-D-glucose (PGG) is the main phenolic active ingredient in Paeoniae Radix Alba, which is commonly used for the treatment of osteoporosis (OP). PGG is a potent natural antioxidant, and its effects on OP remain unknown. This study aimed to investigate the effects of PGG on promoting bone formation and explore its estrogen receptor (ER)-related mechanisms. A hydrogen peroxide-induced osteoblast apoptosis model was established in MC3T3-E1 cells. The effects of PGG were assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, flow cytometry, alkaline phosphatase (ALP) staining, RT-qPCR, and Western blot methods. Furthermore, a prednisolone-induced zebrafish OP model was employed to study the effects in vivo. ER inhibitors and molecular docking methods were used further to investigate the interactions between PGG and ERs. The results showed that PGG significantly enhanced cell viability and decreased cell apoptosis by restoring mitochondrial function, attenuating reactive oxygen species levels, decreasing the mitochondrial membrane potential, and enhancing ATP production. PGG enhanced ALP expression and activity and elevated osteogenic differentiation. PGG also promoted bone formation in the zebrafish model, and these effects were reversed by ICI182780. These results provide evidence that the effects of PGG in alleviating apoptosis and promoting bone formation may depend on ERs. As such, PGG is considered a valuable candidate for treating OP.
Collapse
Affiliation(s)
- Yongqing Hua
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Haili Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Tingting Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yeru Zhou
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhiyuan Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinyue Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shaoqin Mo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hongyun Mao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Miao Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Linxia Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Min Hong
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
40
|
Carleton N, Lee S, Li R, Zou J, Brown DD, Hooda J, Chang A, Kumar R, Klei LR, Rigatti LH, Newsome J, John Mary DJS, Atkinson JM, West RE, Nolin TD, Oberly PJ, Huang Z, Poirier D, Diego EJ, Lucas PC, Tseng G, Lotze MT, McAuliffe PF, Zervantonakis IK, Oesterreich S, Lee AV. Systemic and local chronic inflammation and hormone disposition promote a tumor-permissive environment for breast cancer in older women. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.616978. [PMID: 39484485 PMCID: PMC11526964 DOI: 10.1101/2024.10.18.616978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Estrogen receptor positive (ER+) breast cancer is the most common subtype of breast cancer and is an age-related disease. The peak incidence of diagnosis occurs around age 70, even though these post-menopausal patients have low circulating levels of estradiol (E2). Despite the hormone sensitivity of age-related tumors, we have a limited understanding of the interplay between systemic and local hormones, chronic inflammation, and immune changes that contribute to the growth and development of these tumors. Here, we show that aged F344 rats treated with the dimethylbenz(a)anthracene / medroxyprogestrone acetate (DMBA/MPA) carcinogen develop more tumors at faster rates than their younger counterparts, suggesting that the aged environment promotes tumor initiation and impacts growth. Single-nuclei RNA-seq (snRNA-seq) of the tumors showed broad local immune dysfunction that was associated with circulating chronic inflammation. Across a broad cohort of specimens from patients with ER+ breast cancer and age-matched donors of normal breast tissue, we observe that even with an estrone (E1)-predominant estrogen disposition in the systemic circulation, tumors in older patients increase HSD17B7 expression to convert E1 to E2 in the tumor microenvironment (TME) and have local E2 levels similar to pre-menopausal patients. Concurrently, trackable increases in several chemokines, defined most notably by CCL2, promote a chronically inflamed but immune dysfunctional TME. This unique milieu in the aged TME, characterized by high local E2 and chemokine-enriched chronic inflammation, promotes both accumulation of tumor-associated macrophages (TAMs), which serve as signaling hubs, as well as polarization of TAMs towards a CD206+/PD-L1+, immunosuppressive phenotype. Pharmacologic targeting of estrogen signaling (either by HSD17B7 inhibition or with fulvestrant) and chemokine inflammation both decrease local E2 and prevent macrophage polarization. Overall, these findings suggest that chronic inflammation and hormonal disposition are critical contributors to the age-related nature of ER+ breast cancer development and growth and offer potential therapeutic insight to treat these patients. Translational Summary We uncover the unique underpinnings establishing how the systemic host environment contributes to the aged breast tumor microenvironment, characterized by high local estradiol and chronic inflammation with immune dysregulation, and show that targeting avenues of estrogen conversion and chronic inflammation work to restore anti-tumor immunity.
Collapse
|
41
|
Liu JC, Zeng Q, Duan YG, Yeung WSB, Li RHW, Ng EHY, Cheung KW, Zhang Q, Chiu PCN. B cells: roles in physiology and pathology of pregnancy. Front Immunol 2024; 15:1456171. [PMID: 39434884 PMCID: PMC11491347 DOI: 10.3389/fimmu.2024.1456171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
B cells constitute a diverse and adaptable immune cell population with functions that can vary according to the environment and circumstances. The involvement of B cells in pregnancy, as well as the associated molecular pathways, has yet to be investigated. This review consolidates current knowledge on B cell activities and regulation during pregnancy, with a particular focus on the roles of various B cell subsets and the effects of B cell-derived factors on pregnancy outcomes. Moreover, the review examines the significance of B cell-associated autoantibodies, cytokines, and signaling pathways in relation to pregnancy complications such as pregnancy loss, preeclampsia, and preterm birth.
Collapse
Affiliation(s)
- Jin-Chuan Liu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qunxiong Zeng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Raymond H. W. Li
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ernest H. Y. Ng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ka-Wang Cheung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
42
|
Yang L, Fu MF, Wang HY, Sun H. Research Advancements in the Interplay between T3 and Macrophages. Curr Med Sci 2024; 44:883-889. [PMID: 39446284 DOI: 10.1007/s11596-024-2935-6] [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: 12/14/2023] [Accepted: 09/03/2024] [Indexed: 10/25/2024]
Abstract
3,3',5-Triiodo-L-thyronine (T3) is a key endocrine hormone in the human body that plays crucial roles in growth, development, metabolism, and immune function. Macrophages, the key regulatory cells within the immune system, exhibit marked "heterogeneity" and "plasticity", with their phenotype and function subject to modulation by local environmental signals. The interplay between the endocrine and immune systems is well documented. Numerous studies have shown that T3 significantly target macrophages, highlighting them as key cellular components in this interaction. Through the regulation of macrophage function and phenotype, T3 influences immune function and tissue repair in the body. This review comprehensively summarizes the regulatory actions and mechanisms of T3 on macrophages, offering valuable insights into further research of the immunoregulatory effects of T3.
Collapse
Affiliation(s)
- Liu Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Meng-Fei Fu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Han-Yu Wang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China
| | - Hui Sun
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, 430022, China.
| |
Collapse
|
43
|
Guerra G, Nakase T, Kachuri L, McCoy L, Hansen HM, Rice T, Wiemels JL, Wiencke JK, Molinaro AM, Wrensch M, Francis SS. Association of immunoglobulin E levels with glioma risk and survival. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.09.24307132. [PMID: 38766059 PMCID: PMC11100947 DOI: 10.1101/2024.05.09.24307132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background Previous epidemiologic studies have reported an association of serum immunoglobulin E (IgE) levels with reduced glioma risk, but the association between IgE and glioma prognosis has not been characterized. This study aimed to examine how sex, tumor subtype, and IgE class modulate the association of serum IgE levels with glioma risk and survival. Methods We conducted a case-control study using participants from the University of California, San Francisco Adult Glioma Study (1997-2010). Serum IgE levels for total, respiratory and food allergy were measured in adults diagnosed with glioma (n=1319) and cancer-free controls (n=1139) matched based on age, sex, and race and ethnicity. Logistic regression was adjusted for patient demographics to assess the association between IgE levels and glioma risk. Multivariable Cox regression adjusted for patient-specific and tumor-specific factors compared survival between the elevated and normal IgE groups. All statistical tests were 2-sided. Results Elevated total IgE was associated with reduced risk of IDH-wildtype (RR=0.78, 95% CI: 0.71-0.86) and IDH-mutant glioma (RR=0.73, 95% CI: 0.63-0.85). In multivariable Cox regression, positive respiratory IgE was associated with improved survival for IDH-wildtype glioma (RR=0.79, 95% CI: 0.67-0.93). The reduction in mortality risk was significant in females only (RR=0.75, 95% CI: 0.57-0.98) with an improvement in median survival of 6.9 months (P<.001). Conclusion Elevated serum IgE was associated with improved prognosis for IDH-wildtype glioma, with a more pronounced protective effect in females than males, which has implications for the future study of IgE-based immunotherapies for glioma.
Collapse
Affiliation(s)
- Geno Guerra
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Taishi Nakase
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Linda Kachuri
- Department of Epidemiology & Population Health, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Lucie McCoy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Helen M. Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Terri Rice
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Joseph. L. Wiemels
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - John K. Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Annette M. Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Stephen S. Francis
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology & Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
44
|
Lu Y, Liang Z, Liu L, Zhou Y, Liu C, Zhao Z, Zheng T, Du Q, Liu W. Discovery of novel quinoline scaffold selective estrogen receptor degraders (SERDs) for treatment of ER positive breast cancer with enhanced antiproliferative bioactivity through immunogenic cell death (ICD) effects. Eur J Med Chem 2024; 275:116534. [PMID: 38870830 DOI: 10.1016/j.ejmech.2024.116534] [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: 02/15/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/15/2024]
Abstract
Combination therapy proven to be an effective therapeutic approach for estrogen receptor (ER)-positive breast cancer. Currently, cyclin-dependent kinase 4/6 (CDK4/6) inhibitors are combined with aromatase inhibitors (AIs) or selective estrogen receptor degraders (SERDs) as first-line therapy for advanced ER-positive breast cancer. Herein, a new family of quinoline scaffold SERDs was synthesized and evaluated in MCF-7 cells. Among them, compounds 18j and 24d exhibited remarkable MCF-7 inhibition, both alone and in combination with ribociclib (CDK4/6 inhibitor), in vitro and in vivo. Meanwhile, compounds 18j and 24d effectively degraded ER and inhibited ER downstream signaling pathways. Interestingly, compounds 18j and 24d induced endoplasmic reticulum stress (ERS) and triggered immunogenic cell death (ICD) via damage-associated molecular patterns (DAMPs) in MCF-7 cells. These findings highlight the immune-related and enhanced antiproliferative effects of oral SERDs in ER positive breast cancer treatment.
Collapse
Affiliation(s)
- Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, 210023, PR China
| | - Zhenlin Liang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Lijuan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Yanyu Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Chao Liu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029, PR China; School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Zhihao Zhao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Tianpeng Zheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, PR China; School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
45
|
Liang Z, Ning R, Wang Z, Kong X, Yan Y, Cai Y, He Z, Liu X, Zou Y, Zhou J. The emerging roles of UFMylation in the modulation of immune responses. Clin Transl Med 2024; 14:e70019. [PMID: 39259506 PMCID: PMC11389534 DOI: 10.1002/ctm2.70019] [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: 06/03/2024] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 09/13/2024] Open
Abstract
Post-translational modification is a rite of passage for cellular functional proteins and ultimately regulate almost all aspects of life. Ubiquitin-fold modifier 1 (UFM1) system represents a newly identified ubiquitin-like modification system with indispensable biological functions, and the underlying biological mechanisms remain largely undiscovered. The field has recently experienced a rapid growth of research revealing that UFMylation directly or indirectly regulates multiple immune processes. Here, we summarised important advances that how UFMylation system responds to intrinsic and extrinsic stresses under certain physiological or pathological conditions and safeguards immune homeostasis, providing novel perspectives into the regulatory framework and functions of UFMylation system, and its therapeutic applications in human diseases.
Collapse
Affiliation(s)
- Zhengyan Liang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular DiagnosticsSchool of Basic MedicineGuangdong Medical UniversityDongguanChina
| | - Rongxuan Ning
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular DiagnosticsSchool of Basic MedicineGuangdong Medical UniversityDongguanChina
| | - Zhaoxiang Wang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular DiagnosticsSchool of Basic MedicineGuangdong Medical UniversityDongguanChina
| | - Xia Kong
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular DiagnosticsSchool of Basic MedicineGuangdong Medical UniversityDongguanChina
| | - Yubin Yan
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular DiagnosticsSchool of Basic MedicineGuangdong Medical UniversityDongguanChina
| | - Yafei Cai
- Key Laboratory for Epigenetics of Dongguan City, China‐America Cancer Research InstituteGuangdong Medical UniversityDongguanChina
| | - Zhiwei He
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular DiagnosticsSchool of Basic MedicineGuangdong Medical UniversityDongguanChina
- Institute of Cancer ResearchShenzhen Bay LaboratoryShenzhenChina
| | - Xin‐guang Liu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular DiagnosticsSchool of Basic MedicineGuangdong Medical UniversityDongguanChina
| | - Yongkang Zou
- College of Animal Science and TechnologyNanjing Agricultural UniversityNanjingChina
| | - Junzhi Zhou
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular DiagnosticsSchool of Basic MedicineGuangdong Medical UniversityDongguanChina
- Institute of Cancer ResearchShenzhen Bay LaboratoryShenzhenChina
| |
Collapse
|
46
|
Monteiro FL, Stepanauskaite L, Archer A, Williams C. Estrogen receptor beta expression and role in cancers. J Steroid Biochem Mol Biol 2024; 242:106526. [PMID: 38657699 DOI: 10.1016/j.jsbmb.2024.106526] [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: 11/17/2023] [Revised: 04/06/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024]
Abstract
Estrogen drives the growth of some cancers, such as breast cancer, via estrogen receptor alpha (ERα). Estrogen also activates ERβ, but whether ERβ is expressed and has a role in different cancers is debated. The use of nonspecific antibodies has contributed to the confusion, and this review delves into ERβ's controversial role in cancer and focuses on tumor expression that can be supported by non-antibody-dependent assays. We discuss its expression at the transcript level and focus on its potential role in lymphoma, granulosa cell tumors, testicular, and adrenal cancers, emphasizing recent findings and the complexities that necessitate further research.
Collapse
Affiliation(s)
- Fátima L Monteiro
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Lina Stepanauskaite
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Amena Archer
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna 171 21, Sweden; Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden.
| |
Collapse
|
47
|
Wang L, Xu S, Chen R, Ding Y, Liu M, Hou C, Wu Z, Men X, Bao M, He B, Li S. Exploring the causal association between epigenetic clocks and menopause age: insights from a bidirectional Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1429514. [PMID: 39247918 PMCID: PMC11377254 DOI: 10.3389/fendo.2024.1429514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/02/2024] [Indexed: 09/10/2024] Open
Abstract
Background Evidence suggests a connection between DNA methylation (DNAm) aging and reproductive aging. However, the causal relationship between DNAm and age at menopause remains uncertain. Methods Employing established DNAm epigenetic clocks, such as DNAm Hannum age acceleration (Hannum), Intrinsic epigenetic age acceleration (IEAA), DNAm-estimated granulocyte proportions (Gran), DNAm GrimAge acceleration (GrimAgeAccel), DNAm PhenoAge acceleration (PhenoAgeAccel), and DNAm-estimated plasminogen activator inhibitor-1 levels (DNAmPAIadjAge), a bidirectional Mendelian randomization (MR) study was carried out to explore the potential causality between DNAm and menopausal age. The primary analytical method used was the inverse variance weighted (IVW) estimation model, supplemented by various other estimation techniques. Results DNAm aging acceleration or deceleration, as indicated by Hannum, IEAA, Gran, GrimAgeAccel, PhenoAgeAccel, and DNAmPAIadjAge, did not exhibit a statistically significant causal effect on menopausal age according to forward MR analysis. However, there was a suggestive positive causal association between age at menopause and Gran (Beta = 0.0010; 95% confidence interval (CI): 0.0004, 0.0020) in reverse MR analysis. Conclusion The observed increase in granulocyte DNAm levels in relation to menopausal age could potentially serve as a valuable indicator for evaluating the physiological status at the onset of menopause.
Collapse
Affiliation(s)
- Ling Wang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Shuling Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rumeng Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yining Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Menghua Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chunyan Hou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhu Wu
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Xiaoju Men
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Meihua Bao
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Binsheng He
- The Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
48
|
Harahap IA, Schmidt M, Pruszyńska-Oszmałek E, Sassek M, Suliburska J. Impact of Lactobacillus acidophilus and Its Combination with Isoflavone Products on Calcium Status, Calcium Transporters, and Bone Metabolism Biomarkers in a Post-Menopausal Osteoporotic Rat Model. Nutrients 2024; 16:2524. [PMID: 39125403 PMCID: PMC11314490 DOI: 10.3390/nu16152524] [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: 07/04/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Osteoporosis in menopausal women requires alternatives to current medications, considering their adverse effects. In this context, probiotics and isoflavone products are promising dietary interventions. The objective of our study was to examine the impacts of Lactobacillus acidophilus and its combination with daidzein and tempeh on calcium status, calcium transporters, and bone metabolism biomarkers in a post-menopausal osteoporotic rat model. A total of 48 female Wistar rats were exposed to a two-stage experiment involving calcium deficit induction and subsequent dietary interventions across six groups. Calcium levels, the gene expression of TRPV5 and TRPV6 calcium transporters, bone histopathology, serum bone metabolism markers, and blood biochemistry were evaluated. The results revealed that, while decreasing serum calcium levels, the groups that received the probiotic L. acidophilus and isoflavone combination exhibited increased bone metabolism biomarkers and decreased calcium transporter expressions, akin to the effects of bisphosphonate. Additionally, significant improvements in bone histopathology were observed in these groups. However, the group receiving probiotic L. acidophilus alone did not exhibit significant changes in bone resorption biomarkers, calcium transporter expression, or various blood parameters. Meanwhile, the combination of probiotic L. acidophilus with tempeh positively influenced hematological parameters and reduced cholesterol and triglyceride levels, but it led to elevated blood glucose levels. Correlation analyses highlighted associations between serum calcium levels, calcium transporter expression, and bone metabolism biomarkers. In conclusion, our findings suggest that the daily consumption of probiotic L. acidophilus in combination with isoflavone products may improve bone health in ovariectomized rats, warranting further research to elucidate potential interactions with other nutrients.
Collapse
Affiliation(s)
- Iskandar Azmy Harahap
- Department of Human Nutrition and Dietetics, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, 60-624 Poznan, Poland
| | - Marcin Schmidt
- Department of Biotechnology and Food Microbiology, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, 60-637 Poznan, Poland
| | - Ewa Pruszyńska-Oszmałek
- Department of Animal Physiology, Biochemistry and Biostructure, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, 60-637 Poznan, Poland
| | - Maciej Sassek
- Department of Animal Physiology, Biochemistry and Biostructure, Faculty of Veterinary Medicine and Animal Science, Poznan University of Life Sciences, 60-637 Poznan, Poland
| | - Joanna Suliburska
- Department of Human Nutrition and Dietetics, Faculty of Food Science and Nutrition, Poznan University of Life Sciences, 60-624 Poznan, Poland
| |
Collapse
|
49
|
Chupp DP, Rivera CE, Zhou Y, Xu Y, Ramsey PS, Xu Z, Zan H, Casali P. A humanized mouse that mounts mature class-switched, hypermutated and neutralizing antibody responses. Nat Immunol 2024; 25:1489-1506. [PMID: 38918608 PMCID: PMC11291283 DOI: 10.1038/s41590-024-01880-3] [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: 11/20/2023] [Accepted: 05/18/2024] [Indexed: 06/27/2024]
Abstract
Humanized mice are limited in terms of modeling human immunity, particularly with regards to antibody responses. Here we constructed a humanized (THX) mouse by grafting non-γ-irradiated, genetically myeloablated KitW-41J mutant immunodeficient pups with human cord blood CD34+ cells, followed by 17β-estradiol conditioning to promote immune cell differentiation. THX mice reconstitute a human lymphoid and myeloid immune system, including marginal zone B cells, germinal center B cells, follicular helper T cells and neutrophils, and develop well-formed lymph nodes and intestinal lymphoid tissue, including Peyer's patches, and human thymic epithelial cells. These mice have diverse human B cell and T cell antigen receptor repertoires and can mount mature T cell-dependent and T cell-independent antibody responses, entailing somatic hypermutation, class-switch recombination, and plasma cell and memory B cell differentiation. Upon flagellin or a Pfizer-BioNTech coronavirus disease 2019 (COVID-19) mRNA vaccination, THX mice mount neutralizing antibody responses to Salmonella or severe acute respiratory syndrome coronavirus 2 Spike S1 receptor-binding domain, with blood incretion of human cytokines, including APRIL, BAFF, TGF-β, IL-4 and IFN-γ, all at physiological levels. These mice can also develop lupus autoimmunity after pristane injection. By leveraging estrogen activity to support human immune cell differentiation and maturation of antibody responses, THX mice provide a platform to study the human immune system and to develop human vaccines and therapeutics.
Collapse
Affiliation(s)
- Daniel P Chupp
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
- Invivyd, Waltham, MA, USA
| | - Carlos E Rivera
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Yulai Zhou
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Yijiang Xu
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Patrick S Ramsey
- Department of Obstetrics & Gynecology, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Zhenming Xu
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Hong Zan
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
- Prellis Biologics, Berkeley, CA, USA
| | - Paolo Casali
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA.
- Department of Medicine, The University of Texas Long School of Medicine, San Antonio, TX, USA.
| |
Collapse
|
50
|
Dong H, Zeng X, Xu J, He C, Sun Z, Liu L, Huang Y, Sun Z, Cao Y, Peng Z, Qiu YA, Yu T. Advances in immune regulation of the G protein-coupled estrogen receptor. Int Immunopharmacol 2024; 136:112369. [PMID: 38824903 DOI: 10.1016/j.intimp.2024.112369] [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: 02/16/2024] [Revised: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
Estrogen and related receptors have been shown to have a significant impact on human development, reproduction, metabolism and immune regulation and to play a critical role in tumor development and treatment. Traditionally, the nuclear estrogen receptors (nERs) ERα and ERβ have been thought to be involved in mediating the estrogenic effects. However, our group and others have previously demonstrated that the G protein-coupled estrogen receptor (GPER) is the third independent ER, and estrogen signaling mediated by GPER is known to play an important role in normal physiology and a variety of abnormal diseases. Interestingly, recent studies have progressively revealed GPER involvement in the maintenance of the normal immune system, abnormal immune diseases, and inflammatory lesions, which may be of significant clinical value primarily in the immunotherapy of tumors. In this article, we review current advances in GPER-related immunomodulators and provide a theoretical basis and potential clinical targets to ameliorate immune-related diseases and immunotherapy for tumors.
Collapse
Affiliation(s)
- Hanzhi Dong
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Xiaoqiang Zeng
- Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Jiawei Xu
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Zhengkui Sun
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Liyan Liu
- Department of Pharmacy, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China
| | - Yanxiao Huang
- Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhe Sun
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Yuan Cao
- Department of Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhiqiang Peng
- Department of Lymphohematology, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| | - Yu-An Qiu
- Department of Critical Care Medicine, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Clinical Research Center for Cancer, Nanchang 330029, China.
| |
Collapse
|