1
|
Duan Y, Yang F, Zhang Y, Zhang M, Shi Y, Lang Y, Sun H, Wang X, Jin H, Kang X. Role of mitophagy in spinal cord ischemia-reperfusion injury. Neural Regen Res 2026; 21:598-611. [PMID: 39665804 DOI: 10.4103/nrr.nrr-d-24-00668] [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: 06/18/2024] [Accepted: 10/29/2024] [Indexed: 12/13/2024] Open
Abstract
Spinal cord ischemia-reperfusion injury, a severe form of spinal cord damage, can lead to sensory and motor dysfunction. This injury often occurs after traumatic events, spinal cord surgeries, or thoracoabdominal aortic surgeries. The unpredictable nature of this condition, combined with limited treatment options, poses a significant burden on patients, their families, and society. Spinal cord ischemia-reperfusion injury leads to reduced neuronal regenerative capacity and complex pathological processes. In contrast, mitophagy is crucial for degrading damaged mitochondria, thereby supporting neuronal metabolism and energy supply. However, while moderate mitophagy can be beneficial in the context of spinal cord ischemia-reperfusion injury, excessive mitophagy may be detrimental. Therefore, this review aims to investigate the potential mechanisms and regulators of mitophagy involved in the pathological processes of spinal cord ischemia-reperfusion injury. The goal is to provide a comprehensive understanding of recent advancements in mitophagy related to spinal cord ischemia-reperfusion injury and clarify its potential clinical applications.
Collapse
Affiliation(s)
- Yanni Duan
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Fengguang Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yibao Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Mingtao Zhang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yujun Shi
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Yun Lang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hongli Sun
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xin Wang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Hongyun Jin
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| | - Xuewen Kang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, China
- Orthopaedics Key Laboratory of Gansu Province, The Second Hospital of Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
2
|
Liu Z, Guo Y, Zhang Y, Gao Y, Ning B. Metabolic reprogramming of astrocytes: Emerging roles of lactate. Neural Regen Res 2026; 21:421-432. [PMID: 39688570 DOI: 10.4103/nrr.nrr-d-24-00776] [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: 07/16/2024] [Accepted: 10/25/2024] [Indexed: 12/18/2024] Open
Abstract
Lactate serves as a key energy metabolite in the central nervous system, facilitating essential brain functions, including energy supply, signaling, and epigenetic modulation. Moreover, it links epigenetic modifications with metabolic reprogramming. Nonetheless, the specific mechanisms and roles of this connection in astrocytes remain unclear. Therefore, this review aims to explore the role and specific mechanisms of lactate in the metabolic reprogramming of astrocytes in the central nervous system. The close relationship between epigenetic modifications and metabolic reprogramming was discussed. Therapeutic strategies for targeting metabolic reprogramming in astrocytes in the central nervous system were also outlined to guide future research in central nervous system diseases. In the nervous system, lactate plays an essential role. However, its mechanism of action as a bridge between metabolic reprogramming and epigenetic modifications in the nervous system requires future investigation. The involvement of lactate in epigenetic modifications is currently a hot research topic, especially in lactylation modification, a key determinant in this process. Lactate also indirectly regulates various epigenetic modifications, such as N6-methyladenosine, acetylation, ubiquitination, and phosphorylation modifications, which are closely linked to several neurological disorders. In addition, exploring the clinical applications and potential therapeutic strategies of lactic acid provides new insights for future neurological disease treatments.
Collapse
Affiliation(s)
- Zeyu Liu
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yijian Guo
- Department of Spinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yulei Gao
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Bin Ning
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
- Department of Spinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
3
|
Guo HH, Ou HN, Yu JS, Rosa JM, Formolo DA, Cheng T, Yau SY, Tsang HWH. Adiponectin as a potential mediator of the pro-cognitive effects of physical exercise on Alzheimer's disease. Neural Regen Res 2026; 21:96-106. [PMID: 39885660 PMCID: PMC12094572 DOI: 10.4103/nrr.nrr-d-23-00943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 01/11/2024] [Accepted: 12/19/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease is the primary cause of dementia and imposes a significant socioeconomic burden globally. Physical exercise, as an effective strategy for improving general health, has been largely reported for its effectiveness in slowing neurodegeneration and increasing brain functional plasticity, particularly in aging brains. However, the underlying mechanisms of exercise in cognitive aging remain largely unclear. Adiponectin, a cell-secreted protein hormone, has recently been found to regulate synaptic plasticity and mediate the antidepressant effects of physical exercise. Studies on the neuroprotective effects of adiponectin have revealed potential innovative treatments for Alzheimer's disease. Here, we reviewed the functions of adiponectin and its receptor in the brains of human and animal models of cognitive impairment. We summarized the role of adiponectin in Alzheimer's disease, focusing on its impact on energy metabolism, insulin resistance, and inflammation. We also discuss how exercise increases adiponectin secretion and its potential benefits for learning and memory. Finally, we highlight the latest research on chemical compounds that mimic exercise-enhanced secretion of adiponectin and its receptor in Alzheimer's disease.
Collapse
Affiliation(s)
- Hui-Hui Guo
- Department of Rehabilitation Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, China
- Department of Rehabilitation Medicine, Shaoxing People’s Hospital, Shaoxing, Zhejiang Province, China
| | - Hai-Ning Ou
- Department of Rehabilitation Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, China
- Department of Rehabilitation, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
- The Second Institute of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jia-Sui Yu
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Julia Macedo Rosa
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Douglas Affonso Formolo
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Tong Cheng
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Hector Wing Hong Tsang
- Department of Rehabilitation Medicine, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Mental Health Research Center, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| |
Collapse
|
4
|
Shan Y, Hu H, Yang A, Zhao W, Chu Y. An integrative approach to identifying NPC1 as a susceptibility gene for gestational diabetes mellitus. J Matern Fetal Neonatal Med 2025; 38:2445665. [PMID: 39746811 DOI: 10.1080/14767058.2024.2445665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE The objective of this study was to identify a novel gene and its potential mechanisms associated with susceptibility to gestational diabetes mellitus (GDM) through an integrative approach. METHODS We analyzed data from genome-wide association studies (GWAS) of GDM in the FinnGen R11 dataset (16,802 GDM cases and 237,816 controls) and Genotype Tissue Expression v8 expression quantitative trait locus data. We used summary-data-based Mendelian randomization to determine associations between transcript levels and phenotypes, transcriptome-wide association studies to provide insights into gene-trait associations, multi-marker analysis of genomic annotation to perform gene-based analysis, genome-wide complex trait analysis-multivariate set-based association test-combo to determine gene prioritization, and polygenic priority scores to prioritize the causal genes to screen candidate genes. Subsequent Mendelian randomization analysis was performed to infer causality between the candidate genes and GDM and phenome-wide association study (PheWAS) analysis was used to explore the associations between selected genes and other characteristics. Furthermore, to gain a deeper understanding of the functional implications of these susceptibility genes, GeneMANIA analysis was used to determine the fundamental biological functions of the therapeutic targets and protein-protein interaction network analysis was performed to identify intracellular protein interactions. RESULTS We identified two novel susceptibility genes associated with GDM: NPC1 and KIAA1191. Magnetic resonance imaging revealed a strong correlation between NPC1 expression levels and a lower incidence of GDM (odds ratio: 0.922, 95% confidence interval: 0.866-0.981, p = 0.011). PheWAS at the gene level indicated that NPC1 was not associated with any other trait. The biological significance of this gene was evidenced by its strong association with sterol metabolism. CONCLUSION Our study identified NPC1 as a novel gene whose predicted expression level is linked to a reduced risk of GDM, providing new insights into the genetic framework of this disease.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Hu
- Clinical medicine, Nantong University, Nantong, China
| | - Anning Yang
- Department of Obstetrics and Gynecology, Qingdao Eighth People's Hospital, Qingdao, China
| | - Wendi Zhao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
5
|
Li S, Nan W, Peng Z, Huang Q, Chen Q, He B. Association between methylmalonic acid and prevalence of depression in US adults: evidence from NHANES 2011-2014. Eur J Psychotraumatol 2025; 16:2450109. [PMID: 39943880 PMCID: PMC11827031 DOI: 10.1080/20008066.2025.2450109] [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: 08/15/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 02/16/2025] Open
Abstract
Background: Depression is a prevalent mental disorder with high morbidity and mortality globally. Methylmalonic acid (MMA) is involved in the pathogenesis of numerous diseases. However, it is unclear whether there is an association between MMA and the prevalence of depression.Methods: This study enrolled 7866 US adults from the 2011-2014 survey of the National Health and Nutrition Examination Survey (NHANES). Individuals were categorized into depression group and non-depression group based on Patient's Health Questionnaire-9 (PHQ-9) score. The association between MMA concentrations and prevalence of depression was analysed by multivariate logistic and linear regression, restricted cubic spline regression, and subgroup analysis. Mediation analysis was used to explore the role of inflammation in the relationship between MMA and depression.Results: MMA concentrations were higher in participants with depression than those without depression. There was a positive and linear relationship of MMA concentrations with PHQ-9 score and depression risk, respectively. Moreover, the association was stable in most subgroups. Furthermore, inflammatory factors were positively correlated to MMA concentrations and prevalence of depression. In addition, white blood cell, neutrophil and alkaline phosphatase (ALP) mediated the relationship between MMA and depression.Conclusion: Our findings revealed that there was a linear and positive correlation between MMA and the prevalence of depression in US adults, which might be mediated by inflammation.
Collapse
Affiliation(s)
- Siqi Li
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Wenbin Nan
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People’s Republic of China
| | - Zhenyu Peng
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, People’s Republic of China
| | - Qiong Huang
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Qiong Chen
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Baimei He
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
6
|
Liu X, Pan X. ALKBH3-mediated m1A demethylation promotes the malignant progression of acute myeloid leukemia by regulating ferroptosis through the upregulation of ATF4 expression. Hematology 2025; 30:2451446. [PMID: 39803678 DOI: 10.1080/16078454.2025.2451446] [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] [Accepted: 01/05/2025] [Indexed: 05/02/2025] Open
Abstract
To investigate the role of ALKBH3 in acute myeloid leukemia (AML), we constructed an animal model of xenotransplantation of AML. Our study demonstrated that ALKBH3-mediated m1A demethylation inhibits ferroptosis in KG-1 cells by increasing ATF4 expression, thus promoting the development of AML. These findings suggest that reducing ALKBH3 expression may be a potential strategy to mitigate AML progression. Background: Acute myeloid leukemia (AML) is characterized by the unrestrained proliferation of myeloid cells. Studies have shown that ALKBH3 is upregulated in most tumors, but the role of ALKBH3 in AML remains unclear.Methods: In this study, we investigated the function of ALKBH3 in AML cells (KG-1) by immunofluorescence, ELISA, flow cytometry, HE staining, and Western blotting.Results: Our results revealed that ALKBH3 is upregulated in AML and that the downregulation of ALKBH3 inhibited KG-1 cell proliferation and promoted cell apoptosis; at the same time, ALKBH3 upregulated ATF4 expression through m1A demethylation, and the knockdown of ATF4 resulted in increased ferrous iron content; TFR1, ACSL4, and PTGS2 expression; and ROS and MDA levels, whereas SOD and GSH levels and the expression levels of ATF4, SLC7A11, GPX4, and FTH1 decreased in KG-1 cells, thereby promoting ferroptosis. Mechanistically, ALKBH3-mediated m1A demethylation suppressed ferroptosis in KG-1 cells by increasing ATF4 expression, thereby promoting the development of AML.Conclusions: Our study indicated that reducing the expression of ALKBH3 might be a potential target for improving AML symptoms.
Collapse
Affiliation(s)
- Xin Liu
- Clinical College of the 920th Hospital of Kunming Medical University, Kunming, Yunnan Province, People's Republic of China
| | - Xinghua Pan
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, Yunnan Province, People's Republic of China
| |
Collapse
|
7
|
Purificati-Fuñe RA, Lightning P, Lightning P, Bruno G, Littlechild J, Bell RC, Oster RT. Deadly Dads Support Society: understanding the development and impact of a culturally centred, group-led support strategy for nêhiyaw (Plains Cree) fathers and men. Int J Circumpolar Health 2025; 84:2506258. [PMID: 40410659 PMCID: PMC12107637 DOI: 10.1080/22423982.2025.2506258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 04/29/2025] [Accepted: 05/09/2025] [Indexed: 05/25/2025] Open
Abstract
Through a long-standing community-university partnership, we developed a culturally centred, group-led support strategy for nêhiyaw (Plains Cree) fathers and men to enhance their well-being. A community-based participatory research approach was adapted to honour nêhiyaw ways of knowing. Group-led and developed support activities for fathers and men took place from August 2021 to January 2023, with data gathered from Wisdom Circles, meeting minutes, reflexive journals, photos, implementation notes, and community reports. Data analysis was narrative, relational, and non-linear. Knowledge sharing efforts aimed to: 1) explore lessons from co-developing support activities; 2) understand the significance of gathering in safe and healthy ways; and 3) examine the impacts of support on members. The group's development was rooted in mutual generosity and overcoming institutional inequities to offer meaningful supports. This resulted in healthy ways of gathering and supporting one another through relational connections; learning from and identifying with one another; and breaking cycles of intergenerational trauma through cultural connections, sharing, and expressions of love. Experiential and pressure-free activities contributed to a sense of belonging, positivity, and collective ownership, and supported participants through life difficulties. The success and sustainability of the group relied on transcending Western academic approaches to embrace community ways of knowing and relationality.
Collapse
Affiliation(s)
- Reuel A. Purificati-Fuñe
- Department of Agricultural, Food & Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
| | - Patrick Lightning
- Ermineskin Cree Nation, Maskwacîs, AB, Canada
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Patrick Lightning
- Department of Agricultural, Food & Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
- Ermineskin Cree Nation, Maskwacîs, AB, Canada
| | - Grant Bruno
- Samson Cree Nation, Maskwacîs, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Josh Littlechild
- Ermineskin Cree Nation, Maskwacîs, AB, Canada
- Office of the Provost and Vice-President (Academic), University of Alberta, Edmonton, AB, Canada
| | - Rhonda C. Bell
- Department of Agricultural, Food & Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
| | - Richard T. Oster
- Department of Agricultural, Food & Nutritional Sciences, University of Alberta, Edmonton, AB, Canada
- Indigenous Wellness Core, Alberta Health Services, Edmonton, AB, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
8
|
Hou Y, Fu Z, Wang C, Kucharzewska P, Guo Y, Zhang S. 27-Hydroxycholesterol in cancer development and drug resistance. J Enzyme Inhib Med Chem 2025; 40:2507670. [PMID: 40401382 PMCID: PMC12100970 DOI: 10.1080/14756366.2025.2507670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/25/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
27-Hydroxycholesterol (27HC), a cholesterol metabolite, functions both as a selective oestrogen receptor (ER) modulator and a ligand for liver X receptors (LXRs). The discovery of 27HC involvement in carcinogenesis has unveiled new research avenues, yet its precise role remains controversial and context-dependent. In this review, we provide an overview of the biosynthesis and metabolism of 27HC and explore its cancer-associated signalling, with a particular focus on ER- and LXR-mediated pathways. Given the tissue-specific dual role of 27HC, we discuss its differential impact across various cancer types. Furthermore, we sort out 27HC-contributed drug resistance mechanisms from the perspectives of drug efflux, cellular proliferation, apoptosis, epithelial-mesenchymal transition (EMT), antioxidant defence, epigenetic modification, and metabolic reprogramming. Finally, we highlight the chemical inhibitors to mitigate 27HC-driven cancer progression and drug resistance. This review offers an updated role of 27HC in cancer biology, setting the stage for future research and the development of targeted therapeutics.
Collapse
Affiliation(s)
- Yaxin Hou
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| | - Zhiguang Fu
- Department of Tumor Radiotherapy, Air Force Medical Center, People’s Liberation Army of China (PLA), Beijing, China
| | - Chenhui Wang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| | - Paulina Kucharzewska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, Warsaw, Poland
| | - Yuan Guo
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
9
|
Flores Ventura E, Lane JA, Turjeman S, Vidra N, Weiss GA, Gross G, Chang CY, Koren O. ILSI Europe perspective review: site-specific microbiota changes during pregnancy associated with biological consequences and clinical outcomes: opportunities for probiotic interventions. Gut Microbes 2025; 17:2501186. [PMID: 40397816 PMCID: PMC12101587 DOI: 10.1080/19490976.2025.2501186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/11/2025] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
Pregnancy induces notable alterations in the gut, vaginal, and oral microbiota driven by hormonal, immune, metabolic, dietary, and environmental factors. During pregnancy, the gut microbiota is characterized by increased proportions of the genus Bifidobacterium and the phyla Pseudomonadota (formerly Proteobacteria) and Actinomycetota (formerly Actinobacteria). These changes occur alongside reduced alpha diversity and greater beta diversity, changes that influence maternal metabolism and fetal development. Shifts in gut and oral microbiota have been associated with complications such as preterm birth (PTB), pre-eclampsia, and gestational diabetes (GDM), though patterns are sometimes inconsistent. The vaginal microbiota remains Lactobacillus-dominant during pregnancy, with reduced diversity leading to reduced risk of pathogenic infection and increased diversity has been linked with a higher risk of PTB. Hormonal changes also affect the oral microbiota, potentially increasing pathogenic species and contributing to adverse outcomes like PTB. Probiotic supplementation during pregnancy has significant potential to reduce adverse pregnancy outcomes; however, clinical studies are still limited. Probiotics may be effective in alleviating maternal constipation and lead to lower PTB risk, particularly by modulating the vaginal microbiota, but they have limited impact on GDM. In the context of maternal mental health, some studies suggest benefits of probiotics in reducing anxiety, but effects on depression are inconclusive. This perspective examines how pregnancy-related microbial shifts, both natural and probiotic-induced, affect maternal and fetal health and highlights potential opportunities for the innovative use of probiotics during the gestation period.
Collapse
Affiliation(s)
- Eduard Flores Ventura
- Department of Biotechnology, Institute of Agrochemistry and Food Technology – Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Jonathan A. Lane
- Health and Happiness (H & H) Group, H & H Research, National Food Innovation Hub, Teagasc Moorepark, Fermoy, Co., Cork, Ireland
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | | | | | - Gabriele Gross
- Mead Johnson Nutrition Institute/Reckitt, R & D, Nutrition Science Platforms, Nijmegen, The Netherlands
| | - Ching-Yu Chang
- International Life Science Institute, International Life Science Institute, European Branch, Brussels, Belgium
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Kyung Hee University, Seoul, The Republic of Korea
| |
Collapse
|
10
|
He Y, Zhao G, Ouyang X, Wang S, Chen Y, Li C, He Y, Gao J, Han S, Zhao J, Wang J, Wang C. Creatine-mediated ferroptosis inhibition is involved in the intestinal radioprotection of daytime-restricted feeding. Gut Microbes 2025; 17:2489072. [PMID: 40205678 PMCID: PMC11988229 DOI: 10.1080/19490976.2025.2489072] [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: 10/26/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Ionizing radiation-induced intestinal injury (IRIII) is a catastrophic disease lack of sufficient medical countermeasures currently. Regulation of the gut microbiota through dietary adjustments is a potential strategy to mitigate IRIII. Time-restricted feeding (TRF) is an emerging behavioral nutrition intervention with pleiotropic health benefits. Whether this dietary pattern influences the pathogenesis of IRIII remains vague. We evaluated the impact of TRF on intestinal radiosensitivity in this study and discovered that only daytime TRF (DTRF), not nighttime TRF, could ameliorate intestinal damage in mice that received a high dose of IR. Faecal metagenomic and metabolomic studies revealed that the intestinal creatine level was increased by approximate 9 times by DTRF, to which the Bifidobacterium pseudolongum enrichment contribute. Further investigations showed that creatine could activate the energy sensor AMP-activated protein kinase in irradiated enterocytes and induce phosphorylation of acetyl-CoA carboxylase, resulting in reduced production of polyunsaturated fatty acids and reduced ferroptosis after IR. The administration of creatine mitigated IRIII and reduced bacteremia and proinflammatory responses. Blockade of creatine import compromised the ferroptosis inhibition and mitigation of DTRF on IRIII. Our study demonstrates a radioprotective dietary mode that can reshape the gut microbiota and increase intestinal creatine, which can suppress IR-induced ferroptosis, thereby providing effective countermeasures for IRIII prevention.
Collapse
Affiliation(s)
- Yingjuan He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Ouyang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Shaobo Wang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yin Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Chenwenya Li
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Yongwu He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jining Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
11
|
Cheng Y, Guo L. Lactate metabolism and lactylation in kidney diseases: insights into mechanisms and therapeutic opportunities. Ren Fail 2025; 47:2469746. [PMID: 40012230 PMCID: PMC11869332 DOI: 10.1080/0886022x.2025.2469746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025] Open
Abstract
The kidney is essential for lactate metabolism. Under normal conditions, the renal cortex mainly absorbs and metabolizes lactate, with minimal amounts excreted in urine. This process is part of a glucose-lactate recycling system between the cortex and medulla. In conditions such as acute kidney injury (AKI) and diabetic kidney disease (DKD), the kidney's ability to metabolize lactate is impaired, leading to lactate accumulation and exacerbated renal dysfunction. Novel post-translational modifications, such as lactylation, are critical in kidney disease pathophysiology by modulating gene transcription, protein function, and cellular metabolism. Lactylation is involved in inflammatory responses and tumor promotion in AKI, mitochondrial dysfunction in DKD, and tumor progression in clear cell renal cell carcinoma (ccRCC). The lactate-lactylation axis is central to the Warburg effect in ccRCC, where tumor cells preferentially rely on glycolysis rather than oxidative phosphorylation. Understanding the mechanisms of lactate metabolism and lactylation in kidney diseases may offer new therapeutic strategies. This review examines the role of lactate esters, especially lactylation, in kidney diseases, with a focus on their regulatory mechanisms and potential as therapeutic targets.
Collapse
Affiliation(s)
- Yuhua Cheng
- Department of Nephrology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| | - Linjuan Guo
- Department of Cardiology, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| |
Collapse
|
12
|
Xie J, Zheng Z, Wang B, Zhang J, Jiang J, Wu F, Zhong X, Chen J. LncRNA HOTAIR promotes aerobic glycolysis by recruiting Lin28 to induce inflammation and apoptosis in acute lung injury. RNA Biol 2025; 22:1-12. [PMID: 40052944 PMCID: PMC11901367 DOI: 10.1080/15476286.2025.2475255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Acute lung injury (ALI) is a life-threatening condition with high rates of morbidity and mortality. Recently, there has been growing evidence suggesting a link between lncRNA HOTAIR and ALI. Nonetheless, the precise role and mechanism of lncRNA HOTAIR in ALI remain to be fully elucidated. siHOTAIR transfection, qPCR detection (HOTAIR), ELISA (TNF-α, IL-6, and IL-1β), Lactate detection, Glucose uptake experiment, Cell Apoptosis Analysis, Fluorescence in situ hybridization (FISH) assay. Through siHOTAIR transfection, we discovered that HOTAIR plays a role in the secretion of inflammatory factors in ALI and further regulates glucose uptake and metabolism in lung epithelial cells. Moreover, a comparison between HOTAIR knockdown cells and HOTAIR overexpression cells revealed that HOTAIR promotes cellular aerobic sugar metabolism, leading to increased secretion of inflammatory factors and cell apoptosis. Our in-depth research also identified an interaction between HOTAIR and the LIN28 protein. Knocking down HOTAIR resulted in the downregulation of LIN28 protein expression, which subsequently inhibited the expression of the glucose transporter GLUT1. This indicates that HOTAIR facilitates glucose uptake and boosts cellular aerobic glycolysis by modulating the LIN28 protein, thereby promoting inflammation and apoptosis in acute lung injury. The research findings presented in this article offer significant insights into the function of HOTAIR in ALI and suggest a potential therapeutic target for the treatment of this condition.
Collapse
Affiliation(s)
- Junjie Xie
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Zhicong Zheng
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Bin Wang
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianfang Zhang
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Junqi Jiang
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Fengde Wu
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Xiangming Zhong
- Department of Pediatrics, Maternal and Child Health Hospital of Sanshui District, Foshan, China
| | - Jianfeng Chen
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Fumagalli A, Castells-Nobau A, Trivedi D, Garre-Olmo J, Puig J, Ramos R, Ramió-Torrentà L, Pérez-Brocal V, Moya A, Swann J, Martin-Garcia E, Maldonado R, Fernández-Real JM, Mayneris-Perxachs J. Archaea methanogens are associated with cognitive performance through the shaping of gut microbiota, butyrate and histidine metabolism. Gut Microbes 2025; 17:2455506. [PMID: 39910065 PMCID: PMC11810085 DOI: 10.1080/19490976.2025.2455506] [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: 09/30/2024] [Revised: 12/28/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
The relationship between bacteria, cognitive function and obesity is well established, yet the role of archaeal species remains underexplored. We used shotgun metagenomics and neuropsychological tests to identify microbial species associated with cognition in a discovery cohort (IRONMET, n = 125). Interestingly, methanogen archaeas exhibited the strongest positive associations with cognition, particularly Methanobrevibacter smithii (M. smithii). Stratifying individuals by median-centered log ratios (CLR) of M. smithii (low and high M. smithii groups: LMs and HMs) revealed that HMs exhibited better cognition and distinct gut bacterial profiles (PERMANOVA p = 0.001), characterized by increased levels of Verrucomicrobia, Synergistetes and Lentisphaerae species and reduced levels of Bacteroidetes and Proteobacteria. Several of these species were linked to the cognitive test scores. These findings were replicated in a large-scale validation cohort (Aging Imageomics, n = 942). Functional analyses revealed an enrichment of energy, butyrate, and bile acid metabolism in HMs in both cohorts. Global plasma metabolomics by CIL LC-MS in IRONMET identified an enrichment of methylhistidine, phenylacetate, alpha-linolenic and linoleic acid, and secondary bile acid metabolism associated with increased levels of 3-methylhistidine, phenylacetylgluamine, adrenic acid, and isolithocholic acid in the HMs group. Phenylacetate and linoleic acid metabolism also emerged in the Aging Imageomics cohort performing untargeted HPLC-ESI-MS/MS metabolic profiling, while a targeted bile acid profiling identified again isolithocholic acid as one of the most significant bile acid increased in the HMs. 3-Methylhistidine levels were also associated with intense physical activity in a second validation cohort (IRONMET-CGM, n = 116). Finally, FMT from HMs donors improved cognitive flexibility, reduced weight, and altered SCFAs, histidine-, linoleic acid- and phenylalanine-related metabolites in the dorsal striatum of recipient mice. M. smithii seems to interact with the bacterial ecosystem affecting butyrate, histidine, phenylalanine, and linoleic acid metabolism with a positive impact on cognition, constituting a promising therapeutic target to enhance cognitive performance, especially in subjects with obesity.
Collapse
Affiliation(s)
- Andrea Fumagalli
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Salt, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III; Madrid, Spain
| | - Anna Castells-Nobau
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Salt, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III; Madrid, Spain
| | - Dakshat Trivedi
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Josep Garre-Olmo
- serra-hunter program Department of Nursing, University of Girona, Girona, Spain
| | - Josep Puig
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Institute of Diagnostic Imaging (IDI)-Research Unit (IDIR), Parc Sanitari Pere Virgili, Barcelona, Spain
- Medical Imaging, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- Department of Radiology (IDI), Dr. Josep Trueta University Hospital, Girona, Spain
| | - Rafel Ramos
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Vascular Health Research Group of Girona (ISV-Girona), Jordi Gol Institute for Primary Care Research (Institut Universitari per a la Recerca en Atenció Primària Jordi Gol I Gorina -IDIAPJGol), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud-RICAPPS- ISCIII Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, Girona, Catalonia, Spain
- Research in Vascular Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Dr. Josep Trueta University Hospital, Girona, Spain
| | - Lluís Ramió-Torrentà
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Dr. Josep Trueta University Hospital, Girona, Spain
- Neurodegeneration and Neuroinflammation Research Group, IDIBGI-CERCA, Girona, Spain
| | - Vicente Pérez-Brocal
- Area of Genomics and Health, Foundation for the Promotion of Sanitary and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Andrés Moya
- Area of Genomics and Health, Foundation for the Promotion of Sanitary and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and Spanish National Research Council (CSIC), Valencia, Spain
| | - Jonathan Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Elena Martin-Garcia
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III; Madrid, Spain
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Salt, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III; Madrid, Spain
| |
Collapse
|
14
|
Li J, Zhang Q, Wang X, Fu X, Chen L. Association between inflammatory biomarkers and gestational diabetes mellitus in women aged 20-44: a cross-sectional analysis of NHANES 2007-2018. Gynecol Endocrinol 2025; 41:2487492. [PMID: 40176742 DOI: 10.1080/09513590.2025.2487492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 01/15/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025] Open
Abstract
Inflammation exerts an essential role in gestational diabetes mellitus (GDM), but the relationship between peripheral blood inflammatory markers and GDM remains unclear. The purpose of this study was to explore the relationship between inflammatory markers and GDM in US adults. Data were extracted from the National Health and Nutrition Examination Survey. Five inflammatory markers were derived from complete blood count. Survey-weighted multivariable logistic regression models were used to assess the association between inflammatory markers and GDM. Restricted cubic splines and subgroup analyses were conducted to validate the stability of the results. Finally, a total of 2363 women aged 20-44 were included based on specific criteria, with 229 self-reported GDM cases (9.69%). The increased lymphocyte-monocyte ratio (LMR) was associated with the higher risk of GDM, aOR = 1.82 (CI:1.30-2.56). Compared with the lowest tertile, the highest tertile group of LMR showed a significantly increased risk of GDM, aOR = 2.24 (CI: 1.28-2.85). Conversely, the highest tertile group of systemic inflammation response index (SIRI) was negatively associated with GDM, aOR = 0.61 (95% CI: 0.40-0.94). And high platelet-lymphocyte ratio (PLR) levels are related to a lower risk of GDM. No non-linear relationships were observed. Furthermore, subgroup analysis revealed that the association between LMR, SIRI, and GDM remained consistent with the overall results. Our study indicated that LMR, PLR, and SIRI may be potential predictors of GDM. Further large-scale prospective study is needed to investigate the role of LMR, PLR and SIRI in GDM.
Collapse
Affiliation(s)
- Jinsheng Li
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- South China University of Technology School of Medicine, Guangzhou, China
| | - Qianwen Zhang
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- South China University of Technology School of Medicine, Guangzhou, China
| | - Xi Wang
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- South China University of Technology School of Medicine, Guangzhou, China
| | - Xin Fu
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
- The Second Affiliated Hospital, The Department of Gynecology and Obstetrics, Hengyang Medical School, University of South China, Hengyang, China
| | - Lei Chen
- Department of Obstetrics and Gynecology, The Sixth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Sun S, Arif Aslam M, Ma EB, Lee G, Muhammad Ahmad Javaid H, Yoon S, Huh JY. Activation of CXCR7 exerts an inhibitory effect on adipogenesis through regulation of β-arrestin2/Wnt and AKT signalling. Adipocyte 2025; 14:2490258. [PMID: 40302245 PMCID: PMC12045560 DOI: 10.1080/21623945.2025.2490258] [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: 08/27/2024] [Revised: 01/22/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025] Open
Abstract
CXCR7, an alternative receptor for the inflammatory chemokine SDF-1, is involved in cell proliferation and migration. Recent studies have reported that CXCR7 also plays a role in adipose tissue. However, evidence regarding the role of CXCR7 and its ligands in adipocyte differentiation is limited. In this study, we aimed to elucidate changes in CXCR7 expression during adipocyte differentiation and the role of the SDF-1/CXCR7/CXCR4 axis in adipogenesis using recombinant SDF-1, the CXCR7 ligand CCX771, and small interfering RNAs. The results indicated that the levels of SDF-1 and its receptors, CXCR7 and CXCR4, decreased during the early stages of adipogenesis. Treatment with recombinant SDF-1 and CCX771 inhibited adipogenesis and lipid accumulation by inducing β-arrestin2, Wnt expression, and AKT phosphorylation and downregulating C/EBPα, PPARγ, and FABP4 expression. In contrast, knockdown of SDF-1 and CXCR7 in preadipocytes downregulated the β-arrestin2/Wnt and AKT pathway, leading to the induction of adipogenesis. Meanwhile, knockdown of CXCR4 had no significant effect. In mice, basal gene expression levels of SDF-1 and CXCR7 were higher in the stromal vascular fraction compared to mature adipocytes and were significantly upregulated by a high-fat diet. Our results provide new insights into the local role of the SDF-1-CXCR7 axis in adipocytes and offer additional benefits for the prevention of obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Shiyue Sun
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Muhammad Arif Aslam
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Eun Bi Ma
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Gahui Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Hafiz Muhammad Ahmad Javaid
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Somy Yoon
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
16
|
Santos C, Bustamante A, Maia J, Vasconcelos O, Tani G, Hedeker D, Katzmarzyk PT, Pereira S. Secular trends in BMI, excess weight and body fat skinfolds in Peruvian children living at high altitude. Ann Hum Biol 2025; 52:2494565. [PMID: 40421916 DOI: 10.1080/03014460.2025.2494565] [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: 11/13/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND There is limited evidence on secular trends in adiposity among high-altitude children. AIMS To describe secular trends in body mass index (BMI), excess weight and body fat skinfolds (SKF) among Peruvian children living at high altitude and to investigate the risk for excess weight in 2019 relative to 2009. SUBJECTS AND METHODS We sampled 1585 Peruvians aged 6-11 years from the Junín region. Height and weight were measured, and BMI was calculated. WHO cut-offs were used to classify children by weight status. The sum of SKF was used for analysis. BMI and sum SKF were log-transformed. Two-factor ANOVA and post-hoc contrasts were used together with trend plots for BMIlog and SKFlog. Relative risks for excess weight were estimated with 95% confidence intervals. All analyses were done in STATA. RESULTS BMIlog and SKFlog means increased in both sexes from 2009 to 2019, with age-specific variation. Between 2009 and 2019, BMI values shifted from near P50 to P75 on WHO percentiles for both sexes in most age groups. The risk of excess weight was not significantly higher for most children in 2019 compared to 2009. CONCLUSIONS Positive secular trends in BMIlog and SKFlog were observed, with significant increases at specific ages.
Collapse
Affiliation(s)
- Carla Santos
- Centre of Research, Education, Innovation and Intervention in Sport (CIFI2D), Faculty of Sport, University of Porto, Porto, Portugal
- Research Center in Sport, Physical Education, and Exercise and Health (CIDEFES), Faculty of Physical Education and Sports, Lusófona University, Lisboa, Portugal
| | - Alcibíades Bustamante
- Research Group of Differential and Developmental Kinanthropometry, School of Physical Education and Sports, National University of Education, Lurigancho-Chosica, Peru
| | - José Maia
- Centre of Research, Education, Innovation and Intervention in Sport (CIFI2D), Faculty of Sport, University of Porto, Porto, Portugal
| | - Olga Vasconcelos
- Centre of Research, Education, Innovation and Intervention in Sport (CIFI2D), Faculty of Sport, University of Porto, Porto, Portugal
| | - Go Tani
- Motor Behavior Laboratory, School of Physical Education and Sports, University of São Paulo, São Paulo, Brazil
| | - Donald Hedeker
- Department of Public Health Sciences, University of Chicago, Chicago, IL, USA
| | | | - Sara Pereira
- Centre of Research, Education, Innovation and Intervention in Sport (CIFI2D), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
17
|
Baird F, Smith E, Beebeejaun Y, Subramanian V, Bhaduri M, Kametas N, Sarris I, Sunkara SK. Obstetric and perinatal outcomes in singleton pregnancies following medicated, stimulated and natural, frozen embryo transfer cycles: an updated systematic review and meta-analysis. HUM FERTIL 2025; 28:2448131. [PMID: 40356454 DOI: 10.1080/14647273.2024.2448131] [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: 07/04/2024] [Accepted: 12/24/2024] [Indexed: 05/15/2025]
Abstract
Frozen embryo transfer (FET) has been associated with higher risks of hypertensive disorders of pregnancy (HDP), large for gestational age and high birth weight. This systematic review and meta-analysis evaluates obstetric and perinatal outcomes in medicated, stimulated and natural FET cycles. A literature search was performed using MEDLINE, EMBASE, CINAHL and the Cochrane Library database; 152,590 FET cycles were analysed from 31 included studies. Risk ratios (RR) and 95% confidence intervals (CI) were calculated using fixed and random effects models determined by the heterogeneity (I2). There was significantly higher risk of obstetric and perinatal complications, including HDP (RR 1.84, CI:1.48-2.29, I2 87%) and low birth weight (LBW) (RR 1.25, CI:1.12-1.39, I2 57%) following medicated compared to natural FET cycles. A higher risk of HDP (RR 1.50, CI:1.33-1.64, I2 32%) and macrosomia (RR 1.28, CI:1.17-1.40, I2 37%) was noted following medicated compared to stimulated cycles. Natural demonstrated lower risk of HDP (RR 0.77, CI:0.60-0.99, I2 32%), gestational diabetes (RR 0.78, CI:0.68-0.89, I2 43%), LBW (RR 0.78, CI:0.64-0.95, I2 48%) and small for gestational age (RR 0.83, CI:0.70-0.98, I2 12%) than stimulated cycles. These findings indicate that medicated FET cycles convey greater risks of obstetric and perinatal complications than natural or stimulated cycles.
Collapse
Affiliation(s)
- Freya Baird
- King's Fertility, 1st Floor, Fetal Medicine Research Institute, London, UK
- Department of Women's Health, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Emilia Smith
- Department of Women's Health, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Yusuf Beebeejaun
- King's Fertility, 1st Floor, Fetal Medicine Research Institute, London, UK
- Department of Women's Health, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Venkatesh Subramanian
- King's Fertility, 1st Floor, Fetal Medicine Research Institute, London, UK
- Department of Women's Health, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Mahua Bhaduri
- Department of Women's Health, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Nick Kametas
- Department of Women's Health, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Harris Birthright Research Centre for Fetal Medicine, Fetal Medicine Research Institute, King's College Hospital, London, UK
| | - Ippokratis Sarris
- King's Fertility, 1st Floor, Fetal Medicine Research Institute, London, UK
- Department of Women's Health, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Sesh K Sunkara
- King's Fertility, 1st Floor, Fetal Medicine Research Institute, London, UK
- Department of Women's Health, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
18
|
Hu H, Wan S, Hu Y, Wang Q, Li H, Zhang N. Deciphering the role of APOE in cerebral amyloid angiopathy: from genetic insights to therapeutic horizons. Ann Med 2025; 57:2445194. [PMID: 39745195 DOI: 10.1080/07853890.2024.2445194] [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: 07/16/2024] [Revised: 10/26/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Cerebral amyloid angiopathy (CAA), characterized by the deposition of amyloid-β (Aβ) peptides in the walls of medium and small vessels of the brain and leptomeninges, is a major cause of lobar hemorrhage in elderly individuals. Among the genetic risk factors for CAA that continue to be recognized, the apolipoprotein E (APOE) gene is the most significant and prevalent, as its variants have been implicated in more than half of all patients with CAA. While the presence of the APOE ε4 allele markedly increases the risk of CAA, the ε2 allele confers a protective effect relative to the common ε3 allele. These allelic variants encode three APOE isoforms that differ at two amino acid positions. The primary physiological role of APOE is to mediate lipid transport in the brain and periphery; however, it has also been shown to be involved in a wide array of biological functions, particularly those involving Aβ, in which it plays a known role in processing, production, aggregation, and clearance. The challenges posed by the reliance on postmortem histological analyses and the current absence of an effective intervention underscore the urgency for innovative APOE-targeted strategies for diagnosing CAA. This review not only deepens our understanding of the impact of APOE on the pathogenesis of CAA but can also help guide the exploration of targeted therapies, inspiring further research into the therapeutic potential of APOE.
Collapse
Affiliation(s)
- Hantian Hu
- Tianjin Medical University, Tianjin, China
| | - Siqi Wan
- Tianjin Medical University, Tianjin, China
| | - Yuetao Hu
- Tianjin Medical University, Tianjin, China
| | - Qi Wang
- Tianjin Medical University, Tianjin, China
| | - Hanyu Li
- Tianjin Medical University, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
19
|
Cheng R, Zhu Y, Yang LQ, Xiao L, Fang XH, Zhang Q. Dysregulated serum levels of kisspeptin, NKB, GABA in women with polycystic ovary syndrome and their association with hormonal profiles. Gynecol Endocrinol 2025; 41:2463533. [PMID: 39932739 DOI: 10.1080/09513590.2025.2463533] [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: 11/28/2024] [Revised: 01/12/2025] [Accepted: 02/01/2025] [Indexed: 05/08/2025] Open
Abstract
INTRODUCTION The clinical study aimed to evaluate the levels of serum kisspeptin, NKB, and GABA in Chinese patients with polycystic ovary syndrome (PCOS) and explore their association with hormonal profiles, as well as the relationship between these levels in PCOS patients and controls. METHODS From December 2022 to December 2023, medical records of 60 individuals diagnosed with PCOS and 32 healthy subjects were obtained. Serum kisspeptin, NKB and GABA levels were quantified using enzyme-linked immunosorbent assay (ELISA) kits. To assess the correlation, either Pearson's or Spearman's analytical method was employed between serum kisspeptin, NKB, and GABA levels and hormonal profiles. RESULTS The results showed that serum kisspeptin and NKB levels were significantly lower in PCOS patients (p﹤.05), while GABA levels were elevated compared to those in the control group (p﹤.05). In PCOS patients, kisspeptin was notably positive-correlated with LH and LH/FSH (p﹤.05), but no significant correlation was found between NKB, GABA, and hormonal profiles. In PCOS, GABA levels had a strong positive correlation with NKB levels (r = 0.613, p = .000), but this was not observed in the control group. In control patients, kisspeptin levels were positively associated with NKB (r = 0.475, p = .011), strongly negatively correlated with GABA levels (r = -0.773, p = .000), but these were not observed in the PCOS group. CONCLUSION The study concluded that patients with PCOS have dysregulated levels of serum kisspeptin, NKB, and GABA and that they may have paradoxical effects under physiological and pathological situations.
Collapse
Affiliation(s)
- Ran Cheng
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Ying Zhu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liu-Qing Yang
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Lu Xiao
- Healthcare Office, Guang'anmen Hospital, China Academy of Chinese Medical, Beijing, China
| | - Xiao-Hong Fang
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| | - Qin Zhang
- Department of Traditional Chinese Medical Gynecology, Hangzhou TCM Hospital, Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Research Institute of Women's Reproductive Health, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Precise Protection and Promotion of Fertility, Hangzhou, China
| |
Collapse
|
20
|
Wang Z, Sofer T. Recent Progress in Omics Studies of Sleep and Circadian Phenotypes. CURRENT SLEEP MEDICINE REPORTS 2025; 11:17. [PMID: 40321983 PMCID: PMC12048028 DOI: 10.1007/s40675-025-00335-x] [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] [Accepted: 04/03/2025] [Indexed: 05/08/2025]
Abstract
Purpose of review Sleep and circadian biology is fundamental to human health. Following the advancement in sleep medicine and availability of multi-omics technology, this review outlines the current knowledge regarding genetic basis and multi-omics research on circadian rhythm and the two most prevalent sleep disorders, obstructive sleep apnea (OSA) and insomnia. Recent findings Genome wide association analyses identified variants across circadian genes and genes pertinent to inflammation, obesity and neuronal function associated with OSA and insomnia. Multi-omics integration has led to novel breakthroughs in identifying systemic biomarkers and elucidating cascades, and causal associations underpinning these complex traits. Summary Multi-omics studies in sleep and circadian rhythm possess great potential in unveiling molecular mechanisms behind circadian rhythm and sleep, thereby advancing personalized medicine in the long term. Nevertheless, researchers should remain mindful of existing challenges in genetic and multi-omics sleep research, including data harmonization and existing racial and ethnic disparities in data collection and availability, limiting research generalizability.
Collapse
Affiliation(s)
- Ziqing Wang
- Department of Medicine, Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, USA
| | - Tamar Sofer
- Department of Medicine, Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
21
|
Lee JS, Kao DJ, Worledge CS, Villamaria ZF, Wang RX, Welch NM, Kostelecky RE, Colgan SP. E. coli genetically modified for purine nucleobase release promotes butyrate generation and colonic wound healing during DSS insult. Gut Microbes 2025; 17:2490211. [PMID: 40247632 PMCID: PMC12013446 DOI: 10.1080/19490976.2025.2490211] [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: 02/03/2024] [Revised: 12/27/2024] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
The gut microbiota transforms energy stored as undigestible carbohydrates into a remarkable number of metabolites that fuel intestinal bacterial communities and the host tissue. Colonic epithelial cells at the microbiota-host interface depend upon such microbiota-derived metabolites (MDMs) to satisfy their energy requisite. Microbial dysbiosis eliciting MDM loss contributes to barrier dysfunction and mucosal disease. Recent work has identified a role for microbiota-sourced purines (MSPs), notably hypoxanthine, as an MDM salvaged by the colonic epithelium for nucleotide biogenesis and energy balance. Here, we investigated the role of MSPs in mice during disease-modeled colonic energetic stress using a strain of E. coli genetically modified for enhanced purine nucleobase release (E. coli Mutant). E. coli Mutant colonization protected against DSS-induced tissue damage and permeability while promoting proliferation for wound healing. Metabolite and metagenomic analyses suggested a colonic butyrate-purine nucleobase metabolic axis, wherein the E. coli Mutant provided purine substrate for Clostridia butyrate production and host purine salvage, altogether supplying the host substrate for efficient nucleotide biogenesis and energy balance.
Collapse
Affiliation(s)
- J. Scott Lee
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Daniel J. Kao
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Corey S. Worledge
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Zachary F. Villamaria
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Ruth X. Wang
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Nichole M. Welch
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| | - Rachael E. Kostelecky
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
| | - Sean P. Colgan
- Department of Medicine, Mucosal Inflammation Program, University of Colorado Anschutz Medical Campus
- Department of Medicine, Rocky Mountain Veterans Association, Aurora, CO, USA
| |
Collapse
|
22
|
Guan C, Chen R, Wang Y. Inflammatory markers mediate association of AIP with kidney failure risk: data from National Health and Nutrition Examination Survey (NHANES) 2005-2018. Ren Fail 2025; 47:2486565. [PMID: 40230193 PMCID: PMC12001854 DOI: 10.1080/0886022x.2025.2486565] [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: 07/25/2024] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/16/2025] Open
Abstract
Dyslipidemia and inflammation often coexist in the progression of kidney failure, with the atherosclerosis index of plasma (AIP) serving as a valuable marker for monitoring dyslipidemia. This cross-sectional study analyzed data from the National Health and Nutrition Examination Survey (NHANES) spanning 2005 to 2018, involving a total of 10,358 participants. AIP was calculated as the logarithmic ratio (base 10) of triglycerides to high-density lipoprotein cholesterol (log10[TG/HDL-C]), while kidney failure was assessed through self-reported physician diagnosis. Logistic regression models and restricted cubic splines (RCS) were utilized to examine the association between AIP and the risk of kidney failure, with additional subgroup analyses performed to explore potential interactions. Mediation analyses were conducted to investigate whether inflammatory markers mediated the relationship between AIP and kidney failure. In logistic regression, after adjusting for all covariates, AIP was found to be positively associated with the risk of kidney failure [OR = 1.74 (95% CI: 1.04-2.92)], and a linear relationship between AIP and kidney failure risk was observed (P-non-linear = 0.4050). Mediation analysis revealed that segmented neutrophils, eosinophils, and monocytes partially mediated the association between AIP and kidney failure, with mediation proportions of 19.65%, 2.44%, and 7.25%, respectively. These findings suggest that Higher AIP was associated with an increased risk of kidney failure, with segmented neutrophils, eosinophils, and monocytes serving as partial mediators. The results provide valuable insights into the role of inflammation in kidney failure and highlight potential avenues for its prevention.
Collapse
Affiliation(s)
- Chengjing Guan
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ruixue Chen
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yu Wang
- Department of Nephrology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
23
|
Akinborewa O, Quattrocelli M. Glucocorticoid receptor epigenetic activity in the heart. Epigenetics 2025; 20:2468113. [PMID: 40007064 PMCID: PMC11866966 DOI: 10.1080/15592294.2025.2468113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/23/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
The glucocorticoid receptor (GR) is a critical nuclear receptor that regulates gene expression in diverse tissues, including the heart, where it plays a key role in maintaining cardiovascular health. GR signaling influences essential processes within cardiomyocytes, including hypertrophy, calcium handling, and metabolic balance, all of which are vital for proper cardiac function. Dysregulation of GR activity has been implicated in various cardiovascular diseases (CVDs), highlighting the potential of GR as a therapeutic target. Remarkably, recent insights into GR's epigenetic regulation and its interaction with circadian rhythms reveal opportunities to optimize therapeutic strategies by aligning glucocorticoid administration with circadian timing. In this review, we provide an overview of the glucocorticoid receptor's role in cardiac physiology, detailing its genomic and non-genomic pathways, interactions with epigenetic and circadian regulatory mechanisms, and implications for cardiovascular disease. By dissecting these molecular interactions, this review outlines the potential of epigenetically informed and circadian-timed interventions that could change the current paradigms of CVD treatments in favor of precise and effective therapies.
Collapse
Affiliation(s)
- Olukunle Akinborewa
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
24
|
Zhang L, Fang L, Zou J, Zhou D, Xie H, Chen A, Wu Q. Causal associations of metabolic dysfunction-associated steatotic liver disease with gestational hypertension and preeclampsia: a two-sample Mendelian randomization study. Hypertens Pregnancy 2025; 44:2441862. [PMID: 39704480 DOI: 10.1080/10641955.2024.2441862] [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: 06/26/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Hypertensive disorders of pregnancy (HDPs), which include gestational hypertension (GH) and preeclampsia (PE), are the primary causes of maternal morbidity and mortality worldwide. Recent studies have found a correlation between metabolic dysfunction-associated steatotic liver disease (MASLD) and HDPs, but the causality of this association remains to be identified. Therefore, this study aims to evaluate the causal relationship between MASLD and HDPs through Mendelian randomization (MR) analysis. METHODS The summary statistics from genome-wide association studies were employed to conduct a two-sample MR analysis. Five complementary MR methods, including inverse variance weighting (IVW), MR-Egger, weighted median, simple mode and weighted mode were performed to assess the causality of MASLD on GH and PE. Furthermore, we conducted various sensitivity analyses to ensure the stability and reliability of the results. RESULTS Genetically predicted MASLD significantly increased the risk of GH (IVW: OR = 1.138, 95% CI: 1.062-1.220, p < 0.001), while there was little evidence of a causal relationship between MASLD and PE (IVW: OR = 0.980, 95% CI: 0.910-1.056, p = 0.594). The sensitivity analyses indicated no presence of heterogeneity and horizontal pleiotropy. CONCLUSION This MR study provided evidence supporting the causal effect of MASLD on GH. Our findings underscore the significance of providing more intensive prenatal care and early intervention for pregnant women with MASLD to prevent potential adverse obstetric outcomes.
Collapse
Affiliation(s)
- Lu Zhang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Liang Fang
- Department of Gastroenterology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Jiahua Zou
- Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Dong Zhou
- Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Haonan Xie
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Aihua Chen
- Department of Gastroenterology, China Resources & WISCO General Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Qingming Wu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Moore M, Whittington HD, Knickmeyer R, Azcarate-Peril MA, Bruno-Bárcena JM. Non-stochastic reassembly of a metabolically cohesive gut consortium shaped by N-acetyl-lactosamine-enriched fibers. Gut Microbes 2025; 17:2440120. [PMID: 39695352 DOI: 10.1080/19490976.2024.2440120] [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: 04/30/2024] [Revised: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Diet is one of the main factors shaping the human microbiome, yet our understanding of how specific dietary components influence microbial consortia assembly and subsequent stability in response to press disturbances - such as increasing resource availability (feeding rate) - is still incomplete. This study explores the reproducible re-assembly, metabolic interplay, and compositional stability within microbial consortia derived from pooled stool samples of three healthy infants. Using a single-step packed-bed reactor (PBR) system, we assessed the reassembly and metabolic output of consortia exposed to lactose, glucose, galacto-oligosaccharides (GOS), and humanized GOS (hGOS). Our findings reveal that complex carbohydrates, especially those containing low inclusion (~1.25 gL-1) components present in human milk, such as N-acetyl-lactosamine (LacNAc), promote taxonomic, and metabolic stability under varying feeding rates, as shown by diversity metrics and network analysis. Targeted metabolomics highlighted distinct metabolic responses to different carbohydrates: GOS was linked to increased lactate, lactose to propionate, sucrose to butyrate, and CO2, and the introduction of bile salts with GOS or hGOS resulted in butyrate reduction and increased hydrogen production. This study validates the use of single-step PBRs for reliably studying microbial consortium stability and functionality in response to nutritional press disturbances, offering insights into the dietary modulation of microbial consortia and their ecological dynamics.
Collapse
Affiliation(s)
- Madison Moore
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Hunter D Whittington
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Rebecca Knickmeyer
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, and UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jose M Bruno-Bárcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
26
|
Mi L, Yuan J, Jiang Y, Hu Y, Lv C, Xu Y, Liu M, Liu T, Liu X, Huang J, Jiang R, Quan W. Constructed transferrin receptor-targeted liposome for the delivery of fluvoxamine to improve prognosis in a traumatic brain injury mouse model. Drug Deliv 2025; 32:2486840. [PMID: 40230297 PMCID: PMC12001850 DOI: 10.1080/10717544.2025.2486840] [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: 01/01/2025] [Accepted: 01/22/2025] [Indexed: 04/16/2025] Open
Abstract
The dysregulation of blood-brain barrier (BBB) activates pathological mechanisms such as neuroinflammation after traumatic brain injury (TBI), and glymphatic system dysfunction accelerates toxic waste accumulation after TBI. It is essential to find an effective way to inhibit inflammation and repair BBB and glymphatic system after TBI; however, effective and lasting drug therapy remains challenging because BBB severely prevents drugs from being delivered to central nervous system. Transferrin receptors (TfRs) are mainly expressed on brain capillary endothelial cells. Here, we report a TfR-targeted nanomedicine for TBI treatment by penetrating BBB and delivering fluvoxamine (Flv). The TfR-targeted polypeptide liposome loaded with Flv (TPL-Flv) implements cell targeting ability on human umbilical vein endothelial cells (HUVECs) in vitro detected by flow cytometry, and drug safety was proved through cell viability analysis and blood routine and biochemistry analysis. Afterwards, we established a controlled cortical impact model to explore TPL-Flv administration effects on TBI mice. We confirmed that TPL-Flv could stimulate CXCR4/SDF-1 signaling pathway, activate Treg cells, and inhibit inflammation after TBI. TPL-Flv treatment also alleviated BBB disruption and restored aquaporin-4 (AQP4) polarization, as well as reversed glymphatic dysfunction. Furthermore, TPL-Flv accomplished remarkable improvement of motor and cognitive functions. These findings demonstrate that TPL-Flv can effectively cross BBB and achieve drug delivery to cerebral tissue, validating its potential to improve therapeutic outcomes for TBI.
Collapse
Affiliation(s)
- Liang Mi
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiangyuan Yuan
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuxing Jiang
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuqian Hu
- Department of Endoscopy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Chuanxiang Lv
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yongqiang Xu
- College of Biomedical Engineering, Hubei Key Laboratory of Medical Information Analysis and Tumor Diagnosis & Treatment, and Key Laboratory of Brain Cognitive Science (State Ethnic Affairs Commission), South-Central Minzu University, Wuhan, China
| | - Mingqi Liu
- Department of Rehabilitation Medicine, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Tao Liu
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
- Faculty of Medicine, The George Institute for Global Health, University of New South Wales, Sydney, Australia
| | - Xuanhui Liu
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhao Huang
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| | - Rongcai Jiang
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Quan
- Department of Neurosurgery, State Key Laboratory of Experimental Hematology, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin & Ministry of Education, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
27
|
Zhang Y, Wang A, Zhao W, Qin J, Zhang Y, Liu B, Yao C, Long J, Yuan M, Yan D. Microbial succinate promotes the response to metformin by upregulating secretory immunoglobulin a in intestinal immunity. Gut Microbes 2025; 17:2450871. [PMID: 39812329 PMCID: PMC11740685 DOI: 10.1080/19490976.2025.2450871] [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: 01/07/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Metformin is the first-line pharmacotherapy for type 2 diabetes mellitus; however, many patients respond poorly to this drug in clinical practice. The potential involvement of microbiota-mediated intestinal immunity and related signals in metformin responsiveness has not been previously investigated. In this study, we successfully constructed a humanized mouse model by fecal transplantation of the gut microbiota from clinical metformin-treated - responders and non-responders, and reproduced the difference in clinical phenotypes of responsiveness to metformin. The abundance of Bacteroides thetaiotaomicron, considered a representative differential bacterium of metformin responsiveness, and the level of secretory immunoglobulin A (SIgA) in intestinal immunity increased significantly in responder recipient mice following metformin treatment. In contrast, no significant alterations in B. thetaiotaomicron and SIgA were observed in non-responder recipient mice. The study of IgA-/- mice confirmed that downregulated expression or deficiency of SIgA resulted in non-response to metformin, meaning that metformin was unable to improve dysfunctional glucose metabolism and reduce intestinal and adipose tissue inflammation, ultimately leading to systemic insulin resistance. Furthermore, supplementation with succinate, a microbial product of B. thetaiotaomicron, potentially reversed the non-response to metformin by inducing the production of SIgA. In conclusion, we demonstrated that upregulated SIgA, which could be regulated by succinate, was functionally involved in metformin response through its influence on immune cell-mediated inflammation and insulin resistance. Conversely, an inability to regulate SIgA may result in a lack of response to metformin.
Collapse
Affiliation(s)
- Ying Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiting Wang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Zhao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jia’an Qin
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu Zhang
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bing Liu
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chengcheng Yao
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jianglan Long
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingxia Yuan
- Department of Endocrinology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dan Yan
- Beijing Institute of Clinical Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Champroux A, Sadat-Shirazi M, Chen X, Hacker J, Yang Y, Feig LA. Astrocyte-derived exosomes regulate sperm miR-34c levels to mediate the transgenerational effects of paternal chronic social instability stress. Epigenetics 2025; 20:2457176. [PMID: 39868429 PMCID: PMC11776480 DOI: 10.1080/15592294.2025.2457176] [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: 09/27/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/28/2025] Open
Abstract
The effects of chronically stressing male mice can be transmitted across generations by stress-specific changes in their sperm miRNA content, which induce stress-specific phenotypes in their offspring. However, how each stress paradigm alters the levels of distinct sets of sperm miRNAs is not known. We showed previously that exposure of male mice to chronic social instability (CSI) stress results in elevated anxiety and reduced sociability specifically in their female offspring across multiple generations because it reduces miR-34c levels in sperm of stressed males and their unstressed male offspring. Here, we describe evidence that astrocyte-derived exosomes (A-Exos) carrying miR-34c mediate how CSI stress has this transgenerational effect on sperm. We found that CSI stress decreases miR-34c carried by A-Exos in the prefrontal cortex and amygdala, as well as in the blood of males. Importantly, miR-34c A-Exos levels are also reduced in these tissues in their F1 male offspring, who despite not being exposed to stress, exhibit reduced sperm miR-34c levels and transmit the same stress-associated traits to their male and female offspring. Furthermore, restoring A-Exos miR-34c content in the blood of CSI-stressed males by intravenous injection of miR-34c-containing A-Exos restores miR-34c levels in their sperm. These findings reveal an unexpected role for A-Exos in maintaining sperm miR-34c levels by a process that when suppressed by CSI stress mediates this example of transgenerational epigenetic inheritance.
Collapse
Affiliation(s)
- Alexandre Champroux
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Mitra Sadat-Shirazi
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Xuan Chen
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Jonathan Hacker
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Yongjie Yang
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Larry A. Feig
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
29
|
Luo Z, Huang Y, Yong K, Wu D, Zheng L, Yao X, Shen L, Yu S, Wang B, Cao S. Gut microbiota regulates hepatic ketogenesis and lipid accumulation in ketogenic diet-induced hyperketonemia by disrupting bile acid metabolism. Gut Microbes 2025; 17:2496437. [PMID: 40268803 PMCID: PMC12026136 DOI: 10.1080/19490976.2025.2496437] [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: 10/25/2024] [Revised: 01/13/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025] Open
Abstract
The ketogenic diet (KD) induces prolonged hyperketonemia, characterized by elevated circulating level of β-hydroxybutyrate. However, the KD can negatively affect host metabolic health by altering the gut microbial community. Despite this, the regulatory effect of the gut microbiota on hepatic ketogenesis and triacylglycerol (TAG) accumulation during a KD remains poorly understood. Here, we hypothesized that the commensal bacterium regulates hepatic lipid metabolism in association with KD-induced hyperketonemia. The KD disrupts the remodeling of the gut microbiota following antibiotic-induced depletion. The capacity for ketogenesis and the severity of TAG accumulation in the liver closely correlated with changes in the gut microbial composition and the up-regulation of hepatic farnesoid X receptor (FXR), peroxisome proliferator-activated receptor alpha (PPARα), and diacylglycerol O-acyltransferase 2 (DGAT2), which were modulated by bile acid metabolism through the gut-liver axis. The commensal bacterium Clostridium perfringens type A is particularly implicated in prolonged hyperketonemia, exacerbating hepatic ketogenesis and steatosis by disrupting secondary bile acid metabolism. The increased conversion of deoxycholic acid to 12-ketolithocholic acid represents a critical microbial pathway during C. perfringens colonization. These findings illuminate the adverse effects of the gut microbiota on hepatic adaptation to a KD and highlight the regulatory role of C. perfringens in ketonic states.
Collapse
Affiliation(s)
- Zhengzhong Luo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Yixin Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Kang Yong
- College of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, China
| | - Dan Wu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Linfeng Zheng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Xueping Yao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Liuhong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Shumin Yu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Baoning Wang
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Suizhong Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| |
Collapse
|
30
|
Xiang Z, Ma B, Pei X, Wang W, Gong W. Mechanism of action of genistein on breast cancer and differential effects of different age stages. PHARMACEUTICAL BIOLOGY 2025; 63:141-155. [PMID: 39996512 PMCID: PMC11864014 DOI: 10.1080/13880209.2025.2469607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
CONTEXT Genistein, a soy-derived isoflavone, exhibits structural similarities with 17β-estradiol and demonstrates antioxidant, anti-inflammatory, and estrogenic properties. Despite its low bioavailability limiting its clinical application, it shows potential for breast cancer prevention and treatment. OBJECTIVE This review aims to summarize the pharmacological effects and molecular mechanisms of genistein in breast cancer, focusing on its therapeutic potential, strategies to overcome bioavailability limitations, and its role in personalized medicine. Differential impacts among population subgroups are also discussed. METHODS A systematic review was conducted using PubMed, ScienceDirect, and Google Scholar databases. Studies were selected based on their focus on genistein's mechanisms of action, strategies to enhance its bioavailability, and interactions with other therapies. RESULTS Genistein exerted anticancer effects by modulating estrogen receptor β (ERβ), inhibiting angiogenesis, arresting the cell cycle, and inducing apoptosis. Its antioxidant properties help mitigate tumor-associated oxidative stress. Bioavailability enhancement strategies, such as nanoparticle and lipid-based formulations, show promise. Age-dependent effects were evident, with distinct responses observed in prepubertal, menopausal, and postmenopausal populations, underscoring its potential for personalized therapies. Furthermore, genistein influences epigenetic modifications, including DNA methylation and miRNA expression, bolstering its anticancer efficacy. CONCLUSION Genistein is a promising candidate for breast cancer therapy, particularly for personalized treatment. Strategies to enhance bioavailability and further clinical research are essential to optimize its therapeutic potential and evaluate its efficacy in combination therapies.
Collapse
Affiliation(s)
- Zhebin Xiang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Ma
- Zhejiang Hospital, Hangzhou, China
| | - Xiujun Pei
- Shandong Provincial Hospital, Shandong, China
| | - Wenjie Wang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Weilun Gong
- Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
31
|
Ke Z, Lu Z, Li F, Zhao Q, Jiang X, Hu Z, Sun F, He Z, Tang Y, Li Q, van Oostendorp S, Chen X, He Q, Wang Y, Zhu Z, Tong W. Gut microbiota alterations induced by Roux-en-Y gastric bypass result in glucose-lowering by enhancing intestinal glucose excretion. Gut Microbes 2025; 17:2473519. [PMID: 40028693 PMCID: PMC11881838 DOI: 10.1080/19490976.2025.2473519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/26/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Roux-en-Y gastric bypass (RYGB) results in glucose-lowering in patients with type 2 diabetes mellitus (T2DM) and may be associated with increased intestinal glucose excretion. However, the contribution of intestinal glucose excretion to glycemic control after RYGB and its underlying mechanisms are not fully elucidated. Here, we confirmed that intestinal glucose excretion significantly increased in obese rats after RYGB, which was negatively correlated with postoperative blood glucose levels. Moreover, we also found that the contribution of Biliopancreatic limb length, an important factor affecting glycemic control after RYGB, to the improvement of glucose metabolism after RYGB attributed to the enhancement of intestinal glucose excretion. Subsequently, we further determined through multiple animal models that intestinal glucose excretion is physiological rather than pathological and plays a crucial role in maintaining glucose homeostasis in the body. Finally, we employed germ-free mice colonized with fecal samples from patients and rats to demonstrate that enhanced intestinal glucose excretion after RYGB is directly modulated by the surgery-induced changes in the gut microbiota. These results indicated that the gut microbiota plays a direct causal role in the hypoglycemic effect of RYGB by promoting intestinal glucose excretion, which may provide new insights for developing gut microbiota-based therapies for T2DM.
Collapse
Affiliation(s)
- Zhigang Ke
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Daping Hospital, Center for Hypertension and Metabolic Diseases, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Fan Li
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qingyuan Zhao
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Xianhong Jiang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Zhihao Hu
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Daping Hospital, Center for Hypertension and Metabolic Diseases, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Zongcheng He
- Department of Digestive Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Tang
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Qing Li
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Stefan van Oostendorp
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiuyue He
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Yong Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Center for Hypertension and Metabolic Diseases, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Weidong Tong
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
32
|
Zabuliene L, Milionis C, Koukkou E, Ilias I. Exposure to artificial lighting at night: from an ecological challenge to a risk factor for glucose dysmetabolism and gestational diabetes? Narrative review. Ann Med 2025; 57:2477304. [PMID: 40065696 PMCID: PMC11899256 DOI: 10.1080/07853890.2025.2477304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
INTRODUCTION Artificial lighting at night (ALAN) leads to pervasive light pollution, affecting ecosystems and human health globally. Satellite assessments reveal widespread nocturnal illumination worldwide and research indicates adverse health effects. Environmental light pollution disrupts natural cycles, affecting the behavior and reproduction of various organisms. AIM/METHOD In this narrative review we aimed to present research on the effects of ALAN on glucose metabolism and diabetes and hone on its recently reported association with gestational diabetes (GDM). RESULTS Conflicting data exist on the effects of melatonin's administration vis-à-vis glycemia, with some studies suggesting beneficial outcomes for patients with type 2 diabetes mellitus and insomnia. Ambient light influences plasma glucose, with bright light increasing both fasting and postprandial glucose levels. Perinatal light exposure is linked to later-life health risks and prenatal exposure to ALAN is linked to fetal macrosomia. Analyzing European ALAN data in conjunction with epidemiological records for GDM reveals a notable probable association. Additionally, recent research from China (one case-control and two cohort studies) has shown that exposure to ALAN during pregnancy significantly increases the risk of GDM. DISCUSSION/CONCLUSION Despite progress, interdisciplinary research is needed to understand the impact of light pollution on health, especially regarding disrupted light-dark cycles and physiological functions relevant to conditions like GDM. At present, the simplest advice for all people and particularly for women who anticipate pregnancy, or for pregnant women, is to ensure a totally dark environment during sleep time.
Collapse
Affiliation(s)
- Lina Zabuliene
- Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Charalampos Milionis
- Department of Endocrinology, Diabetes and Metabolism, Elena Venizelou General and Maternity Hospital, Athens, Greece
| | - Eftychia Koukkou
- Department of Endocrinology, Diabetes and Metabolism, Elena Venizelou General and Maternity Hospital, Athens, Greece
| | - Ioannis Ilias
- Department of Endocrinology, Diabetes and Metabolism, Elena Venizelou General and Maternity Hospital, Athens, Greece
- Department of Endocrinology, Hippokration General Hospital, Athens, Greece
| |
Collapse
|
33
|
Farahani A, Farahani A, Kashfi K, Ghasemi A. Inhibition of hepatic gluconeogenesis in type 2 diabetes by metformin: complementary role of nitric oxide. Med Gas Res 2025; 15:507-519. [PMID: 40300886 PMCID: PMC12124709 DOI: 10.4103/mgr.medgasres-d-24-00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 10/15/2024] [Accepted: 01/21/2025] [Indexed: 05/01/2025] Open
Abstract
Metformin is the first-line treatment for type 2 diabetes mellitus. Type 2 diabetes mellitus is associated with decreased nitric oxide bioavailability, which has significant metabolic implications, including enhanced insulin secretion and peripheral glucose utilization. Similar to metformin, nitric oxide also inhibits hepatic glucose production, mainly by suppressing gluconeogenesis. This review explores the combined effects of metformin and nitric oxide on hepatic gluconeogenesis and proposes the potential of a hybrid metformin-nitric oxide drug for managing type 2 diabetes mellitus. Both metformin and nitric oxide inhibit gluconeogenesis through overlapping and distinct mechanisms. In hepatic gluconeogenesis, mitochondrial oxaloacetate is exported to the cytoplasm via various pathways, including the malate, direct, aspartate, and fumarate pathways. The effects of nitric oxide and metformin on the exportation of oxaloacetate are complementary; nitric oxide primarily inhibits the malate pathway, while metformin strongly inhibits the fumarate and aspartate pathways. Furthermore, metformin effectively blocks gluconeogenesis from lactate, glycerol, and glutamine, whereas nitric oxide mainly inhibits alanine-induced gluconeogenesis. Additionally, nitric oxide contributes to the adenosine monophosphate-activated protein kinase-dependent inhibition of gluconeogenesis induced by metformin. The combined use of metformin and nitric oxide offers the potential to mitigate common side effects. For example, lactic acidosis, a known side effect of metformin, is linked to nitric oxide deficiency, while the oxidative and nitrosative stress caused by nitric oxide could be counterbalanced by metformin's enhancement of glutathione. Metformin also amplifies nitric oxide -induced activation of adenosine monophosphate-activated protein kinase. In conclusion, a metformin-nitric oxide hybrid drug can benefit patients with type 2 diabetes mellitus by enhancing the inhibition of hepatic gluconeogenesis, decreasing the required dose of metformin for maintaining optimal glycemia, and lowering the incidence of metformin-associated lactic acidosis.
Collapse
Affiliation(s)
- Arman Farahani
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aryan Farahani
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Wu H, Chen J, Guo S, Deng J, Zhou Z, Zhang X, Qi T, Yu F, Yang Q. Advances in the acting mechanism and treatment of gut microbiota in metabolic dysfunction-associated steatotic liver disease. Gut Microbes 2025; 17:2500099. [PMID: 40394806 PMCID: PMC12101596 DOI: 10.1080/19490976.2025.2500099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/17/2025] [Accepted: 04/25/2025] [Indexed: 05/22/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease(MASLD) is increasing in prevalence worldwide and has become the greatest potential risk for cirrhosis and hepatocellular liver cancer. Currently, the role of gut microbiota in the development of MASLD has become a research hotspot. The development of MASLD can affect the homeostasis of gut microbiota, and significant changes in the composition or abundance of gut microbiota and its metabolite abnormalities can influence disease progression. The regulation of gut microbiota is an important strategy and novel target for the treatment of MASLD with good prospects. In this paper, we summarize the role of gut microbiota and its metabolites in the pathogenesis of MASLD, and describe the potential preventive and therapeutic efficacy of gut microbiota as a noninvasive marker to regulate the pathogenesis of MASLD based on the "gut-hepatic axis", which will provide new therapeutic ideas for the clinic.
Collapse
Affiliation(s)
- Huaying Wu
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Jingjing Chen
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Shuyuan Guo
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jinhao Deng
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Zimeng Zhou
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - Xuan Zhang
- Department of Clinical Medicine, Shantou University Medical College, Shantou, China
| | - TianTian Qi
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Yu
- Department of Spine Surgery, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qi Yang
- Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
35
|
Chen W, Ge H, Cong J, Zhou W, Chang X, Quan X, Xia J, Tao X, Pu D, Wu J. Risk factors and prediction model for postpartum psychiatric disorders: a retrospective cohort study of 1418 Chinese women from 2020 to 2022. J Matern Fetal Neonatal Med 2025; 38:2438756. [PMID: 39667804 DOI: 10.1080/14767058.2024.2438756] [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: 05/28/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Postpartum psychiatric disorders (PPDs) have been deemed as a significant public health concern, affecting both maternal health and family dynamics. This study aimed to examine the current status of PPDs, identify the potential risk factors of PPDs, and further develop a clinical nomogram model for predicting PPDs in Chinese women. METHOD In this retrospective cohort study, 1418 postpartum women attending the routine postpartum examination at the 42nd day after delivery in Jiangsu Women and Children Health Hospital were recruited as participants from December 2020 to December 2022. The Symptom Checklist-90 (SCL-90) was utilized to assess the status of postpartum psychiatric disorders. A prediction model was constructed by multivariate logistic regression and presented as a nomogram. The performance of nomogram was measured by the receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA). The relationships between predictive factors of PPDs and SCL-90 were also evaluated using Pearson correlation analysis. The relationships between predictive factors of PPDs and SCL-90 were evaluated using Pearson correlation analysis. RESULTS With the SCL-90 cutoff value of 160, the incidence of postpartum psychiatric disorders was 9.17% among Chinese urban women. The univariate and multivariate logistic regression analyses indicated that age ≤ 25 years old (OR = 10.07, 95%CI = 1.83-55.33), prenatal mood disorder (OR = 4.12, 95%CI = 1.99-8.53), invasive prenatal diagnostic procedures (OR = 4.39, 95%CI = 1.16-16.56), poor relationship with husband (OR = 2.86, 95%CI = 1.58-5.16) and poor relationship with mother-in-law (OR = 5.10, 95%CI = 2.70-9.64) were significantly associated with PPDs. A nomogram prediction model for PPDs was further constructed based on these five independent risk factors, and the area under the receiver operating characteristic curve (AUC) of the nomogram model was 0.823 (95% CI = 0.781-0.865). The calibration curves showed remarkable accuracy of the nomogram and the DCA exhibited high clinical net benefit of the nomogram. Besides, we also explored the relationships between the five risk factors and different symptom dimensions of PPDs and found that the five risk factors were almost associated with increased levels of all symptom dimensions. CONCLUSIONS Five psychosocial risk factors for PPDs were identified in Chinese women and the nomogram prediction model constructed based on these five risk factors could predict the risk of PPDs intuitively and individually. Systematic screening these risk factors and further conducting psychosocial interventions earlier during the pregnancy period are crucial to prevent PPDs. For future research, we intend to incorporate additional risk factors, including blood biomarkers and facial expression indicators, to refine our risk model.
Collapse
Affiliation(s)
- Wenxi Chen
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Huan Ge
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Jing Cong
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Wenjie Zhou
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Xiaoxia Chang
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Xiaojie Quan
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Jing Xia
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Xincheng Tao
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Danhua Pu
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| | - Jie Wu
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanjing Medical University/Jiangsu Province Hospital/Jiangsu Women and Children Health Hospital, Nanjing, China
| |
Collapse
|
36
|
Schlicht K, Pape L, Rohmann N, Knappe C, Epe J, Geisler C, Pohlschneider D, Brodesser S, Kruse L, Rohlfing ME, Hartmann K, Türk K, Marquardt J, Beckmann J, von Schönfels W, Beckmann A, Wietzke-Braun P, Schulte DM, Hollstein T, Demetrowitsch T, Jensen-Kroll J, Brix F, Schreiber S, Franke A, Schwarz K, Waschina S, Laudes M. Prediabetes and type 2 diabetes but not obesity are associated with alterations in bile acid related gut microbe-microbe and gut microbe-host community metabolism. Gut Microbes 2025; 17:2474143. [PMID: 40045464 PMCID: PMC11901388 DOI: 10.1080/19490976.2025.2474143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
The interplay between bile acids (BAs) and metabolic diseases has gained importance in recent years, with a variety of studies investigating their relationship with diverging results. Therefore, in the present study we performed a detailed analysis of BA metabolism in 492 subjects with different metabolic phenotypes. Besides microbiomics and metabolomics this investigation included in silico analysis of community metabolism to examine metabolic interchange between different microbes as well as microbes and the human host. Our findings revealed distinct changes in the BA profiles of patients with diabetes and prediabetes, whereas obesity alone had no influence on circulating BAs. Impaired glycemic control led to increased circulating BAs, a shift toward more secondary BAs, and an increase in the ratio of glycine to taurine-conjugated BAs. Additional analyses revealed that the ratio of glycine to taurine conjugation demonstrated variations between the single BAs, cholic acid (CA), chenodeoxycholic acid (CDCA) and deoxycholic acid (DCA), regardless of the metabolic status, with CA having a higher fraction of taurine conjugation. Furthermore, we found that microbiome alterations are associated with BAs, independent of diabetes or obesity. Analysis of microbial community metabolism revealed differential relative pathway abundance in relation to diabetes, particularly those related to membrane and polyamine synthesis. Increased bacterial cross-feeding of polyamines, galactose, and D-arabinose also coincided with an increase in BA. Notably, our serum metabolome analysis mirrored several of the previously in silico predicted exchanged metabolites, especially amino acid metabolism. Therefore, targeting BA metabolism may be a future approach for the treatment of metabolic diseases, especially prediabetes and type 2 diabetes.
Collapse
Affiliation(s)
- Kristina Schlicht
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Lea Pape
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Nathalie Rohmann
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Carina Knappe
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Johannes Epe
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Corinna Geisler
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Daniela Pohlschneider
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Susanne Brodesser
- Faculty of Medicine and University Hospital of Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Lucy Kruse
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Maria-Elisabeth Rohlfing
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Katharina Hartmann
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Kathrin Türk
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Jens Marquardt
- Department of Internal Medicine 1, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Jan Beckmann
- Department of General and Abdominal Surgery, University Medical Center Schleswig-Holstein (UKSH), Kiel, Germany
| | - Witigo von Schönfels
- Department of General and Abdominal Surgery, University Medical Center Schleswig-Holstein (UKSH), Kiel, Germany
| | - Alexia Beckmann
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Perdita Wietzke-Braun
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Dominik M. Schulte
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Tim Hollstein
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Tobias Demetrowitsch
- Division of Food Technology, Institute of Human Nutrition and Food Science, Kiel University, Kiel, Germany
| | - Julia Jensen-Kroll
- Division of Food Technology, Institute of Human Nutrition and Food Science, Kiel University, Kiel, Germany
| | - Fynn Brix
- Division of Food Technology, Institute of Human Nutrition and Food Science, Kiel University, Kiel, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Karin Schwarz
- Division of Food Technology, Institute of Human Nutrition and Food Science, Kiel University, Kiel, Germany
| | - Silvio Waschina
- Division of Food Technology, Institute of Human Nutrition and Food Science, Kiel University, Kiel, Germany
| | - Matthias Laudes
- Institute of Diabetes and Clinical Metabolic Research, University Medical Center Schleswig-Holstein, Kiel, Germany
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine I, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
37
|
Zhao S, Lin H, Li W, Xu X, Wu Q, Wang Z, Shi J, Chen Y, Ye L, Xi L, Chen L, Yuan M, Su J, Gao A, Jin J, Ying X, Wang X, Ye Y, Sun Y, Zhang Y, Deng X, Shen B, Gu W, Ning G, Wang W, Hong J, Wang J, Liu R. Post sleeve gastrectomy-enriched gut commensal Clostridia promotes secondary bile acid increase and weight loss. Gut Microbes 2025; 17:2462261. [PMID: 39915243 PMCID: PMC11810084 DOI: 10.1080/19490976.2025.2462261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 01/07/2025] [Accepted: 01/29/2025] [Indexed: 02/12/2025] Open
Abstract
The gut microbiome is altered after bariatric surgery and is associated with weight loss. However, the commensal bacteria involved and the underlying mechanism remain to be determined. We performed shotgun metagenomic sequencing in obese subjects before and longitudinally after sleeve gastrectomy (SG), and found a significant enrichment in microbial species in Clostridia and bile acid metabolizing genes after SG treatment. Bile acid profiling further revealed decreased primary bile acids (PBAs) and increased conjugated secondary bile acids (C-SBAs) after SG. Specifically, glycodeoxycholic acid (GDCA) and taurodeoxycholic acid (TDCA) were increased at different follow-ups after SG, and were associated with the increased abundance of Clostridia and body weight reduction. Fecal microbiome transplantation with post-SG feces increased SBA levels, and alleviated body weight gain in the recipient mice. Furthermore, both Clostridia-enriched spore-forming bacteria and GDCA supplementation increased the expression of genes responsible for lipolysis and fatty acid oxidation in adipose tissue and reduced adiposity via Takeda G-protein-coupled receptor 5 (TGR5) signaling. Our findings reveal post-SG gut microbiome and C-SBAs as contributory to SG-induced weight loss, in part via TGR5 signaling, and suggest SBA-producing gut microbes as a potential therapeutic target for obesity intervention.
Collapse
Affiliation(s)
- Shaoqian Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huibin Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Qihan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Juan Shi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingxia Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liuqing Xi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijia Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingyang Yuan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junlei Su
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiabin Jin
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiayang Ying
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolin Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yaorui Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Yingkai Sun
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaxing Deng
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baiyong Shen
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
38
|
Le HM, Nguyen TA, Nguyen DK, Søndergaard Linde D, Bygbjerg IC, Søndergaard J, Duc Nguyen T, Quang Nguyen B, Thi Dang NA, Nguyen XB, Meyrowitsch DW, Vinter CA, Gammeltoft TM, Rasch V. Prevalence and risk factors of gestational diabetes mellitus among pregnant women in northern Vietnam: a cross-sectional study. Glob Health Action 2025; 18:2460339. [PMID: 39925195 PMCID: PMC11812109 DOI: 10.1080/16549716.2025.2460339] [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: 10/02/2024] [Accepted: 01/25/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Gestational diabetes mellitus (GDM) increases adverse neonatal and maternal outcomes. Understanding the prevalence and risk factors of GDM is necessary to plan health care interventions and policy. OBJECTIVE To determine the prevalence and risk factors of GDM in Thai Binh, Vietnam. METHODS A cross-sectional study was conducted in two health facilities in Thai Binh, Vietnam, with the participation of 1,106 pregnant women. Women were recruited at their first antenatal care visit where face-to-face interviews about socioeconomic and reproductive factors were performed. A 2-hour 75 g oral glucose tolerance test was conducted at 24-28 weeks of gestation. GDM was diagnosed according to the World Health Organization 2013 criteria. Logistic regression analyses were used to assess the factors associated with GDM. RESULTS The prevalence rate of GDM was 27.1%. Multivariate logistic regression analysis showed maternal age from 25 to 34 (adjusted OR 2.0; 95%CI 1.3-2.9), maternal age ≥ 35 (adjusted OR 3.0; 95%CI 1.7-5.4), pregestational body mass index ≥ 23 (adjusted OR 1.6; 95%CI 1.1-2.3), family history of diabetes (adjusted OR 1.9; 95%CI 1.3-2.9), fertility treatment (adjusted OR 2.3; 95%CI 1.3-3.8), and previous GDM (adjusted OR 3.1; 95%CI 1.4-6.9) were associated with increased odds of GDM. CONCLUSIONS More than one-fourth of pregnant women in Thai Binh, Vietnam, may have GDM. Advanced maternal age, high pregestational body mass index, family history of diabetes, and previous GDM were associated with increased risk of GDM. Additionally, fertility treatment appears to be strongly associated with an increased risk of GDM.
Collapse
Affiliation(s)
- Hieu Minh Le
- Department of Internal Medicine, Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
- Department of Internal Medicine, Hanoi Medical University, Hanoi, Vietnam
| | - Thi Ai Nguyen
- Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
| | - Dang Kien Nguyen
- Department of Gynaecology and Obstetrics, Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
| | - Ditte Søndergaard Linde
- Department of Gynaecology and Obstetrics, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ib Christian Bygbjerg
- Department of Public Health, Global Health Section, University of Copenhagen, Copenhagen, Denmark
| | - Jens Søndergaard
- Research Unit of General Practice, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Thanh Duc Nguyen
- Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
| | - Bay Quang Nguyen
- Department of Internal Medicine, Hanoi Medical University, Hanoi, Vietnam
- Department of Endocrinology, Bach Mai Hospital, Hanoi, Vietnam
| | - Ngoc-Anh Thi Dang
- Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
| | - Xuan Bai Nguyen
- Department of Histology, Thai Binh University of Medicine and Pharmacy, Thai Binh, Vietnam
| | - Dan W. Meyrowitsch
- Department of Public Health, Global Health Section, University of Copenhagen, Copenhagen, Denmark
| | - Christina A. Vinter
- Department of Gynaecology and Obstetrics, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Tine M. Gammeltoft
- Department of Anthropology, University of Copenhagen, Copenhagen, Denmark
| | - Vibeke Rasch
- Department of Gynaecology and Obstetrics, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
39
|
Xu T, Zhang H, Peng Z, Huang Y, Zhan Q, Ma Z, Zeng X, Liu C, Zeng Q, Dong Y, Xu D. The addition of alpha-ketoglutarate to NT-proBNP improves the prediction of long-term all-cause mortality in acute heart failure patients. Ann Med 2025; 57:2477827. [PMID: 40091615 PMCID: PMC11915736 DOI: 10.1080/07853890.2025.2477827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/21/2025] [Accepted: 02/09/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Alpha-ketoglutarate (AKG), is a major intermediate metabolite of the tricarboxylic acid cycle, and is closely associated with cardiometabolic disease prognosis. Previous studies indicated that AKG is related to myocardial energy expenditure levels and reflects adverse short-term outcomes in heart failure (HF) patients. In this prospective cohort study, we examined the long-term prognostic value of AKG levels in acute HF (AHF) patients. METHODS Plasma AKG levels were assessed in patients hospitalized with AHF. Hazard ratios (HRs) and 95% confidence intervals (CIs) for all-cause mortality were calculated via multiple Cox regression. All-cause mortality was compared between patients with NT-proBNP < 1000 pg/ml and those with NT-proBNP ≥ 1000 pg/ml via subgroup analysis. RESULTS Patients with AKG ≥ 9.83 μg/ml had higher heart rates and NT-proBNP and lower left ventricular ejection fraction (LVEF) and systolic blood pressure (SBP). After multiple adjustment, higher AKG was associated with an increased all-cause mortality risk (HR = 1.078, p < 0.001). Compared with AKG < 9.83 μg/ml, AKG ≥ 9.83 μg/ml nearly doubled (HR = 1.929, p < 0.001) and quadrupled (HR = 4.160, p < 0.001) the all-cause mortality risk in patients with NT-proBNP ≥ 1000 pg/ml and those with NT-proBNP < 1000 pg/ml, respectively. CONCLUSIONS AND RELEVANCE Plasma AKG was independently associated with greater all-cause mortality risk in patients with AHF. Higher AKG levels retained prognostic value for patients with relatively low NT-proBNP.
Collapse
Affiliation(s)
- Tianyu Xu
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hao Zhang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Cardiology, Shunde Hospital, Southern Medical University (the First People’s Hospital of Shunde), Foshan, China
| | - Zhengliang Peng
- Department of Emergency, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (the First People’s Hospital of Shunde), Foshan, China
| | - Qiong Zhan
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhuang Ma
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianghui Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Liu
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qingchun Zeng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yugang Dong
- NHC Key Laboratory of Assisted Circulation and Vascular Diseases, Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Dingli Xu
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
40
|
Jiang L, Yi R, Chen H, Wu S. Quercetin alleviates metabolic-associated fatty liver disease by tuning hepatic lipid metabolism, oxidative stress and inflammation. Anim Biotechnol 2025; 36:2442351. [PMID: 39718035 DOI: 10.1080/10495398.2024.2442351] [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: 08/22/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
The natural flavonoid quercetin, which exhibits a range of biological activities, has been implicated in liver disease resistance in recent research. In vivo study attesting to quercetin's protective effect against metabolic-associated fatty liver disease (MAFLD) is inadequate, however. Here, our investigation explored the potential benefits of quercetin in preventing MAFLD in C57BL/6 mice fed a high-fat diet (HFD). The results revealed that quercetin ameliorated the aberrant enhancement of body and liver weight. The hepatic histological anomalie induced by MAFLD were also mitigated by quercetin. HFD-induced imbalance in serum LDL, HDL, AST, ALT, TG, and LDH was mitigated by quercetin. Mechanically, we found that quercetin improved lipid metabolism by reducing lipogenesis proteins including ACC, FASN, and SREBP-1c and enhancing β-oxidation proteins including PPARα and CPT1A. In vitro study demonstrated that quercetin regulated hepatic lipid metabolism by targeting SREBP-1c and PPARα. Additionally, quercetin enhanced the antioxidant capacity in HFD-treated mice by downregulating Nrf2 and HO-1 expressions and upregulating SOD and GPX1 expressions. The hyper-activation of inflammation was also restored by quercetin via eliminating the phosphorylation of IκBα and NF-κB p65. Collectively, our observations highlight that quercetin exerts hepatoprotective properties in MAFLD mice by regulating hepatic lipid metabolism, oxidative stress and inflammatory response.
Collapse
Affiliation(s)
- Ling Jiang
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Rong Yi
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Huan Chen
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| | - Shuwu Wu
- Department of Endocrinology and Metabolism, People's Hospital of Yichun City, Yichun, Jiangxi, People's Republic of China
| |
Collapse
|
41
|
Wang H, Qin Y, Niu J, Chen H, Lu X, Wang R, Han J. Evolving perspectives on evaluating obesity: from traditional methods to cutting-edge techniques. Ann Med 2025; 57:2472856. [PMID: 40077889 PMCID: PMC11912248 DOI: 10.1080/07853890.2025.2472856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Objective: This review examines the evolution of obesity evaluation methods, from traditional anthropometric indices to advanced imaging techniques, focusing on their clinical utility, limitations, and potential for personalized assessment of visceral adiposity and associated metabolic risks. Methods: A comprehensive analysis of existing literature was conducted, encompassing anthropometric indices (BMI, WC, WHR, WHtR, NC), lipid-related metrics (LAP, VAI, CVAI, mBMI), and imaging technologies (3D scanning, BIA, ultrasound, DXA, CT, MRI). The study highlights the biological roles of white, brown, and beige adipocytes, emphasizing visceral adipose tissue (VAT) as a critical mediator of metabolic diseases. Conclusion: Although BMI and other anthropometric measurements are still included in the guidelines, indicators that incorporate lipid metabolism information can more accurately reflect the relationship between metabolic diseases and visceral obesity. At the same time, the use of more modern medical equipment, such as ultrasound, X-rays, and CT scans, allows for a more intuitive assessment of the extent of visceral obesity.
Collapse
Affiliation(s)
- Heyue Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaxin Qin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinzhu Niu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haowen Chen
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinda Lu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianli Han
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
42
|
Hornero-Ramirez H, Morisette A, Marcotte B, Penhoat A, Lecomte B, Panthu B, Lessard Lord J, Thirion F, Van-Den-Berghe L, Blond E, Simon C, Caussy C, Feugier N, Doré J, Sanoner P, Meynier A, Desjardins Y, Pilon G, Marette A, Cani PD, Laville M, Vinoy S, Michalski MC, Nazare JA. Multifunctional dietary approach reduces intestinal inflammation in relation with changes in gut microbiota composition in subjects at cardiometabolic risk: the SINFONI project. Gut Microbes 2025; 17:2438823. [PMID: 39710576 DOI: 10.1080/19490976.2024.2438823] [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: 07/03/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
The development of cardiometabolic (CM) diseases is associated with chronic low-grade inflammation, partly linked to alterations of the gut microbiota (GM) and reduced intestinal integrity. The SINFONI project investigates a multifunctional (MF) nutritional strategy's impact combining different bioactive compounds on inflammation, GM modulation and CM profile. In this randomized crossover-controlled study, 30 subjects at CM-risk consumed MF cereal-products, enriched with polyphenols, fibers, slowly-digestible starch, omega-3 fatty acids or Control cereal-products (without bioactive compounds) for 2 months. Metabolic endotoxemia (lipopolysaccharide (LPS), lipopolysaccharide-binding protein over soluble cluster of differentiation-14 (LBP/sCD14), systemic inflammation and cardiovascular risk markers, intestinal inflammation, CM profile and response to a one-week fructose supplementation, were assessed at fasting and post mixed-meal. GM composition and metabolomic analysis were conducted. Mixed linear models were employed, integrating time (pre/post), treatment (MF/control), and sequence/period. Compared to control, MF intervention reduced intestinal inflammation (fecal calprotectin, p = 0.007) and endotoxemia (fasting LPS, p < 0.05), without alteration of systemic inflammation. MF decreased serum branched-chain amino acids compared to control (p < 0.05) and increased B.ovatus, B.uniformis, A.butyriciproducens and unclassified Christensenellaceae.CAG-74 (p < 0.05). CM markers were unchanged. A 2-month dietary intervention combining multiple bioactive compounds improved intestinal inflammation and induced GM modulation. Such strategy appears as an effective strategy to target low-grade inflammation through multi-target approach.
Collapse
Affiliation(s)
- Hugo Hornero-Ramirez
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Arianne Morisette
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
- Nutrition, Health and Society Centre (NUTRISS), INAF, Laval University, Québec, QC, Canada
- Department of Plant Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Bruno Marcotte
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
- Nutrition, Health and Society Centre (NUTRISS), INAF, Laval University, Québec, QC, Canada
- Department of Plant Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Armelle Penhoat
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Béryle Lecomte
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Baptiste Panthu
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | | | | | - Laurie Van-Den-Berghe
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
| | - Emilie Blond
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
- Biochemistry Department, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Chantal Simon
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Cyrielle Caussy
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
- Endocrinology, Diabetes and Nutrition Department, Lyon South Hospital, Civil Hospices of Lyon, Pierre-Bénite, France
| | - Nathalie Feugier
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
| | - Joël Doré
- INRAE, MGP, Université Paris-Saclay, Jouy-en-Josas, France
| | - Philippe Sanoner
- iSymrise-Diana Food SAS, R&D, Naturals Food & Beverage, Rennes, France
| | - Alexandra Meynier
- Nutrition Research, Paris-Saclay Tech Center, Mondelez International R&D, Saclay, France
| | - Yves Desjardins
- Institute of Nutrition and Functional Foods (INAF), Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
- Nutrition, Health and Society Centre (NUTRISS), INAF, Laval University, Québec, QC, Canada
- Department of Plant Science, Faculty of Agriculture and Food Sciences, Laval University, Québec, QC, Canada
| | - Geneviève Pilon
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute, Université Laval, Québec, Canada
- Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Québec Heart and Lung Institute, Université Laval, Québec, Canada
- Centre Nutrition, santé et société (NUTRISS), Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, Canada
| | - Patrice D Cani
- UCLouvain, Université catholique de Louvain, Louvain Drug Research Institute, (LDRI) Metabolism and Nutrition Research Group (MNUT), Brussels, Belgium
- Louvain Drug Research Institute; Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
- UCLouvain, Université catholique de Louvain, Institute of Experimental and Clinical Research (IREC), Brussels, Belgium
| | - Martine Laville
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Sophie Vinoy
- Nutrition Research, Paris-Saclay Tech Center, Mondelez International R&D, Saclay, France
| | - Marie-Caroline Michalski
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine - Rhône-Alpes, INSERM, INRAe, Université Claude Bernard Lyon1, Hospices Civils de Lyon, Pierre Bénite, France
- Laboratoire CarMeN, INSERM U.1060, INRAe U. 1397, Université Claude Bernard Lyon1, Pierre Bénite, France
| |
Collapse
|
43
|
Chen X, Xiao Z, Cai Y, Pan Y. Reproductive hormone characteristics of obese Chinese patients with polycystic ovarian syndrome: a meta-analysis. Gynecol Endocrinol 2025; 41:2497854. [PMID: 40287875 DOI: 10.1080/09513590.2025.2497854] [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: 11/05/2024] [Revised: 03/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
The aim of this analysis is to assess the effect of obesity on reproductive hormones in Chineses patients with polycystic ovarian syndrome (PCOS). Seven databases were searched. The Newcastle-Ottawa Scale (NOS) assessed the quality of included studies. A meta-analysis was performed using random-effects model. The means and standard deviations of the outcomes were synthesized as standardized mean differences (SMDs) with corresponding 95% confidence intervals (CIs). A total of 23 studies involving 4554 patients with PCOS were included. No significant differences in follicle-stimulating hormone (FSH) (p = 0.51), estradiol (E2) (p = 0.48), and prolactin (PRL) (p = 0.46) levels were found between obese and nonobese PCOS patients. However, obese PCOS patients had significantly lower levels of luteinizing hormone (LH) (p < 0.00001), LH/FSH (p = 0.001), progesterone (P) (p = 0.009), and anti-mullerian hormone (AMH) (p = 0.001). Conversely, they exhibited significantly higher testosterone (T) (p = 0.001) levels. Obese PCOS patients exhibited lower levels of LH, LH/FSH, P, and AMH, but higher T levels compared to nonobese PCOS patients, and no significant difference were observed in FSH, E2, and PRL levels in PCOS patients with and without obesity.
Collapse
Affiliation(s)
- Xiaolin Chen
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhuoni Xiao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuli Cai
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yizhuo Pan
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
44
|
Luo Z, Yan X, Liu Y, Nan F, Lei Y, Ren Y, Li L. Prognostic significance of Ki-67 in assessing the risk of progression, relapse or metastasis in pheochromocytomas and paragangliomas. Ann Med 2025; 57:2478312. [PMID: 40079941 PMCID: PMC11984564 DOI: 10.1080/07853890.2025.2478312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 03/15/2025] Open
Abstract
INTRODUCTION Since the Fourth edition of the WHO classification, PPGLs have been recognized for their metastatic potential, though no clear features can accurately predict this behavior. The prognostic value of Ki-67 in assessing the risk of progression, relapse, or metastasis in PPGLs remains debated. METHODS This cohort study included 501 patients diagnosed with PPGLs at the First Hospital of Jilin University between 2000 and 2022, with clinical data, treatment details, pathological indicators, and germline gene test results collected. Bulk sequencing was performed on formalin-fixed paraffin-embedded (FFPE) primary tumor samples from 87 patients. Progression-free survival (PFS) was analyzed using multivariable Cox regression. RESULTS Among the 119 enrolled patients with PPGLs, the average age was 45.7 ± 14.0 years, and the median follow-up time was 46 months. A significant finding was the high expression of CDK1, a gene known to be significantly associated with the metastatic risk of PPGLs, in samples with Ki-67 ≥ 3% (p < 0.0001). More importantly, patients with PPGLs and a Ki-67 level ≥ 3% had a 3.59-fold higher risk of progression, relapse or metastasis compared to those with Ki-67 < 3% (HR = 4.59, 95% CI: 1.06-11.95), after adjusting for all confounding factors. In the composite model, the addition of Ki-67 enhanced the predictive ability of the combined model of SDHB, primary site, tumor size, and invade neighboring tissue (AUC = 0.888, 95% CI: 0.808-0.967 vs. AUC = 0.874, 95% CI: 0.783-0.965). CONCLUSION A Ki-67 level ≥ 3% is associated with an increased risk of progression, relapse or metastasis in patients with PPGLs.
Collapse
Affiliation(s)
- Zilan Luo
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Yan
- Pathology Department, The First Hospital of Jilin University, Changchun, China
| | - Yang Liu
- Tumor Immunotherapy Research Center of Jilin University, Changchun, China
| | - Fengrui Nan
- Tumor Immunotherapy Research Center of Jilin University, Changchun, China
| | - Yuhong Lei
- Tumor Immunotherapy Research Center of Jilin University, Changchun, China
| | - Yuan Ren
- Tumor Immunotherapy Research Center of Jilin University, Changchun, China
| | - Lingyu Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
45
|
Huang M, Ji Q, Huang H, Wang X, Wang L. Gut microbiota in hepatocellular carcinoma immunotherapy: immune microenvironment remodeling and gut microbiota modification. Gut Microbes 2025; 17:2486519. [PMID: 40166981 PMCID: PMC11970798 DOI: 10.1080/19490976.2025.2486519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality, with limited treatment options at advanced stages. The gut microbiota, a diverse community of microorganisms residing in the gastrointestinal tract, plays a pivotal role in regulating immune responses through the gut-liver axis. Emerging evidence underscores its impact on HCC progression and the efficacy of immunotherapy. This review explores the intricate interactions between gut microbiota and the immune system in HCC, with a focus on key immune cells and pathways involved in tumor immunity. Additionally, it highlights strategies for modulating the gut microbiota - such as fecal microbiota transplantation, dietary interventions, and probiotics - as potential approaches to enhancing immunotherapy outcomes. A deeper understanding of these mechanisms could pave the way for novel therapeutic strategies aimed at improving patient prognosis.
Collapse
Affiliation(s)
- Mingyao Huang
- School of Basic Medicine, Putian University, Putian, Fujian, China
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, Fujian, China
| | - Quansong Ji
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huiyan Huang
- Ward 3, De’an Hospital, Xianyou County, Putian, Fujian, China
| | - Xiaoqian Wang
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Lin Wang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
46
|
Liu H, Wang H, Gao W, Yuan Y, Tang T, Sang M, Liu F, Geng C. CircATP5C1 promotes triple-negative breast cancer progression by binding IGF2BP2 to modulate CSF-1 secretion. Cancer Biol Ther 2025; 26:2479926. [PMID: 40176374 PMCID: PMC11980513 DOI: 10.1080/15384047.2025.2479926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 04/04/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a common malignant disease among females and severely threatens the health of women worldwide. Nowadays, circular RNAs (circRNAs) aroused our interest for their functions in human cancers, including TNBC. However, the mechanism of most circRNAs in the progression of TNBC remains unclear. We found a novel circRNA named circATP5C1, whose function in TNBC remains uncovered. Tissue microarray was used to analyze the association between the expression of circATP5C1 and the prognoses of TNBC patients. Gain-and loss-of-function experiments were performed to validate the biological functions of circATP5C1 in different TNBC cell lines. RNA-seq analyses were conducted to find out the target genes regulated by circATP5C1. RNA pull-down assay and mass spectrometry were used to select the proteins associated with circATP5C1. RNA FISH-immunofluorescence and RNA immunoprecipitation (RIP) were complemented to validate the interaction between circATP5C1 and its binding protein. CircATP5C1 was identified to have predictive function in prognosis of TNBC patients. CircATP5C1 advanced the progression of TNBC cells. Mechanistically, Colony stimulating factor 1 (CSF-1) is a vital downstream gene regulated by circATP5C1. The alteration of CSF-1 expression level was validated due to the interaction between circATP5C1 and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2). Rescue experiments demonstrated that circATP5C1 accelerates the progression of TNBC partly via binding with IGF2BP2 to increase the secretion of CSF-1. This study uncovers a novel mechanism of circATP5C1/IGF2BP2/CSF-1 pathway in regulating progression of TNBC.
Collapse
Affiliation(s)
- Hongbo Liu
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Haoqi Wang
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Wei Gao
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yang Yuan
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Tiantian Tang
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Meixiang Sang
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Fei Liu
- Research Center and Tumor Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Cuizhi Geng
- Department of Breast Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
47
|
Li S, Ma M, Hu L, Lao J, Luo X, Pan J, Lu D, Wang M, Lin W, Fan Y, Wang F, Chen YH, Wang P, Wu F, Wei X, Xu J, Liu Y, Zheng L. Association between fish consumption and sleep disorders among Chinese adults: a cross-sectional study. Ann Med 2025; 57:2491663. [PMID: 40254933 PMCID: PMC12013145 DOI: 10.1080/07853890.2025.2491663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/22/2025] Open
Abstract
PURPOSE This study aimed to investigate the potential influence of fish consumption on sleep disorders and their specific dimensions among adults in China. METHODS A cross-sectional study was conducted involving 904 participants aged 28-95 from Wenling, China. Fish intake was assessed using a Food Frequency Questionnaire containing 10 items. Sleep quality was evaluated using the Insomnia Severity Index (ISI) and Pittsburgh Sleep Quality Index (PSQI). Participants were categorized into three groups based on weekly fish intake. Logistic regression analyses were employed to determine the association between fish intake and the prevalence of sleep disorders and their specific dimensions. RESULTS Higher marine fish intake was negatively associated with PSQI subdimensions daytime dysfunction, sleep latency and sleep quality scores compared to lower fish intake (adjusted odds ratio (OR): 0.316, 95% confidence interval (CI): 0.205-0.486; adjusted OR: 0.462, 95% CI: 0.302-0.706; and adjusted OR: 0.568, 95% CI: 0.369-0.861, respectively). Marine fish consumption appears to have a positive association with sleep quality, as well as short sleep latency and daytime functioning, among adults in China. CONCLUSIONS This study provides novel insights into the association between fish intake and sleep disorders and their specific dimensions.
Collapse
Affiliation(s)
- Siyuan Li
- Department of Clinical Pharmacy, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Mingwei Ma
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Liming Hu
- Department of Clinical Pharmacy, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Jiaying Lao
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xingguang Luo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Jie Pan
- Quzhou Center for Disease Control and Prevention, Quzhou Center for Public Health Service, Quzhou, China
| | - Dafeng Lu
- Quzhou Center for Disease Control and Prevention, Quzhou Center for Public Health Service, Quzhou, China
| | - Min Wang
- Quzhou Center for Disease Control and Prevention, Quzhou Center for Public Health Service, Quzhou, China
| | - Wenhui Lin
- Department of Cardiovascular Medicine, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Yuncao Fan
- Department of Cardiovascular Medicine, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, China
| | - Yu-Hsin Chen
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Penghui Wang
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, China
| | - Fenzan Wu
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, China
| | - Xiaojie Wei
- Affiliated Cixi Hospital, Wenzhou Medical University, Ningbo, China
| | - Jinzhong Xu
- Department of Clinical Pharmacy, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Lin Zheng
- Department of Cardiovascular Medicine, Affiliated Wenling Hospital, Wenzhou Medical University, Wenling, China
| |
Collapse
|
48
|
Ntiri ES, Chun Nin Wong A. Microbial metabolites as engines of behavioral variation across animals. Gut Microbes 2025; 17:2501191. [PMID: 40357979 PMCID: PMC12077453 DOI: 10.1080/19490976.2025.2501191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/07/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The microbiome, especially that present in the gut, has emerged as a key modulator of animal behavior. However, the extent of its influence across species and behavioral repertoires, as well as the underlying mechanisms, remains poorly understood. Increasing evidence suggests that microbial metabolites play an important role in driving behavioral variation. In this review, we synthesize findings from vertebrates to invertebrates, spanning both model and non-model organisms, to define key groups of microbial-derived metabolites involved in modulating seven distinct behaviors: nutrition, olfaction, circadian rhythms, reproduction, locomotion, aggression, and social interactions. We discuss how these microbial metabolites interact with host chemosensory systems, neurotransmitter signaling, and epigenetic modifications to shape behavior. Additionally, we highlight critical gaps in mechanistic understanding, including the need to map additional host receptors and signaling pathways, as well as the untapped potential of microbial biosynthetic gene clusters as sources for novel bioactive compounds. Advancing these areas will enhance understanding of the microbiome's role in behavioral modulation and open new avenues for microbiome-based interventions for behavioral disorders.
Collapse
Affiliation(s)
- Eric Siaw Ntiri
- Entomology and Nematology Department, University of Florida, Gainesville, FL, USA
| | - Adam Chun Nin Wong
- Entomology and Nematology Department, University of Florida, Gainesville, FL, USA
- Genetics Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
49
|
Chi J, Fan B, Li Y, Jiao Q, Li GY. Mitochondrial transplantation: a promising strategy for the treatment of retinal degenerative diseases. Neural Regen Res 2025; 20:3370-3387. [PMID: 39851134 PMCID: PMC11974652 DOI: 10.4103/nrr.nrr-d-24-00851] [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: 07/30/2024] [Revised: 09/11/2024] [Accepted: 10/24/2024] [Indexed: 01/26/2025] Open
Abstract
The retina, a crucial neural tissue, is responsible for transforming light signals into visual information, a process that necessitates a significant amount of energy. Mitochondria, the primary powerhouses of the cell, play an integral role in retinal physiology by fulfilling the high-energy requirements of photoreceptors and secondary neurons through oxidative phosphorylation. In a healthy state, mitochondria ensure proper visual function by facilitating efficient conversion and transduction of visual signals. However, in retinal degenerative diseases, mitochondrial dysfunction significantly contributes to disease progression, involving a decline in membrane potential, the occurrence of DNA mutations, increased oxidative stress, and imbalances in quality-control mechanisms. These abnormalities lead to an inadequate energy supply, the exacerbation of oxidative damage, and the activation of cell death pathways, ultimately resulting in neuronal injury and dysfunction in the retina. Mitochondrial transplantation has emerged as a promising strategy for addressing these challenges. This procedure aims to restore metabolic activity and function in compromised cells through the introduction of healthy mitochondria, thereby enhancing the cellular energy production capacity and offering new strategies for the treatment of retinal degenerative diseases. Although mitochondrial transplantation presents operational and safety challenges that require further investigation, it has demonstrated potential for reviving the vitality of retinal neurons. This review offers a comprehensive examination of the principles and techniques underlying mitochondrial transplantation and its prospects for application in retinal degenerative diseases, while also delving into the associated technical and safety challenges, thereby providing references and insights for future research and treatment.
Collapse
Affiliation(s)
- Jing Chi
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bin Fan
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yulin Li
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qing Jiao
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Guang-Yu Li
- Department of Ophthalmology, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
50
|
Lin S, Chen S, Zhang Q. Factors influencing premature ovarian insufficiency: a systematic review and meta-analysis. J OBSTET GYNAECOL 2025; 45:2469331. [PMID: 40013478 DOI: 10.1080/01443615.2025.2469331] [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: 01/08/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Premature ovarian insufficiency (POI) has multiple contributing factors. This study aims to systematically evaluate these factors and their mechanisms, identify high-risk groups, support early intervention, and provide a basis for future research. METHODS Following the PRISMA guidelines, a search was conducted across 11 Chinese and English databases, including PubMed, Embase, and the Cochrane Library, up to January 2024. Observational studies on patients with ovarian insufficiency were included. A meta-analysis using Stata 14 was performed to calculate odds ratios (OR) and 95% confidence intervals (CIs) for factors influencing POI. RESULTS A total of 38 studies with 4,968 cases and 5,158 controls were included. The following factors were identified as risk factors for POI: abnormal menstruation (OR = 2.707, 95% CI: 1.705-4.299), hair dyeing (OR = 4.725, 95% CI: 2.914-7.660), chemical exposure (OR = 3.314, 95% CI: 2.283-4.811), Type A personality (OR = 6.106, 95% CI: 4.696-7.939), survival stress (OR = 3.292, 95% CI: 2.380-4.555), sleep deprivation (OR = 3.340, 95% CI: 2.363-4.721), bad mood (OR = 4.654, 95% CI: 2.783-7.781), smoking history (OR = 2.748, 95% CI: 1.928-3.917), family history of POI (OR = 4.338, 95% CI: 2.422-7.768), history of mumps (OR = 5.802, 95% CI: 3.460-9.730), number of abortions (OR = 2.292, 95% CI: 1.887-2.785), and history of pelvic surgery (OR = 4.836, 95% CI: 3.066-7.628). Physical exercise (OR = 0.270, 95% CI: 0.106-0.692) and vegetable intake (OR = 0.434, 95% CI: 0.337-0.560) were protective factors. Sensitivity analysis and publication bias tests confirmed robustness. CONCLUSIONS POI is related with abnormal menstruation, hair dyeing, chemical exposure, Type A personality, survival stress, sleep deprivation, bad mood, smoking history, family history of POI, history of mumps, number of abortions, and history of pelvic surgery. Physical exercise and vegetable intake may provide protection. These findings inform clinical strategies for early identification and management.
Collapse
Affiliation(s)
- Shuling Lin
- Department of Obstetrics and Gynecology, Puning City Maternal and Child Health Hospital, Guangdong Province, China
| | - Shanyan Chen
- Department of Obstetrics and Gynecology, Puning City Maternal and Child Health Hospital, Guangdong Province, China
| | - Qin Zhang
- Reproductive Medicine Center, Shenzhen Luohu District People's Hospital, Guangdong Province, China
| |
Collapse
|