151
|
Shi P, Tang B, Xie W, Li K, Guo D, Li Y, Yao Y, Cheng X, Xu C, Wang QK. LncRNA-induced lysosomal localization of NHE1 promotes increased lysosomal pH in macrophages leading to atherosclerosis. J Biol Chem 2025:110246. [PMID: 40383150 DOI: 10.1016/j.jbc.2025.110246] [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: 01/31/2025] [Revised: 04/30/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025] Open
Abstract
ANRIL, also referred to as CDKN2B-AS1, is a lncRNA gene implicated in the pathogenesis of multiple human diseases including atherosclerotic coronary artery disease, however, definitive in vivo evidence is lacking and the underlying molecular mechanism is largely unknown. In this study, we show that ANRIL overexpression causes atherosclerosis in vivo as transgenic mouse overexpression of full-length ANRIL (NR_003529) increases inflammation and aggravates atherosclerosis under ApoE-/- background (ApoE-/-ANRIL mice). Mechanistically, ANRIL reduces the expression of miR-181b-5p, which leads to increased TMEM106B expression. TMEM106B is significantly up-regulated in atherosclerotic lesions of both human CAD patients and ApoE-/-ANRIL mice. TMEM106B interacts and co-localizes with Na+-H+ exchanger NHE1, which results in mis-localization of NHE1 from cell membranes to lysosomal membranes, leading to increased lysosomal pH in macrophages. Large truncation and point mutation analyses define the critical amino acids for TMEM106B-NHE1 interaction and lysosomal pH regulation as F115 and F117 on TMEM106B and I537, C538, and G539 on NHE1. Topological analysis suggests that both N-terminus and C-terminus of NHE1 are located inside lysosomal lumen, and NHE1 is an important new proton efflux channel involved in raising lysosomal pH. A short TMEM106B peptide (YGRKKRRQRRR-L111A112V113F114F115L116F117) disrupting the TMEM106B-NHE1 interaction normalized lysosomal pH in macrophages with ANRIL overexpression. Our data demonstrate that ANRIL promotes atherosclerosis in vivo and identify the ANRIL/miR-181b-5p/TMEM106B-NHE1/lysosomal pH axis as the underlying molecular pathogenic mechanism for the chromosome 9p21.3 genetic locus for coronary artery disease.
Collapse
Affiliation(s)
- Pengcheng Shi
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Bo Tang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Wen Xie
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Ke Li
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Di Guo
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Yining Li
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Yufeng Yao
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College
| | - Chengqi Xu
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology
| | - Qing K Wang
- Center for Human Genome Research, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology; Maternal and Child Health Hospital of Hubei Province, Women and Children's Hospital of Hubei Province, Huazhong University of Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
152
|
Yang F, Qu G, Wu Y, Zhong P, Chu Z, He Z, Wang Y, Tang Y, Sun S, Luo F. A novel peptide from yak ameliorates hypoxia-induced cardiac dysfunction via targeting gut microbiota and HIF-1α pathway. J Dairy Sci 2025:S0022-0302(25)00358-3. [PMID: 40383391 DOI: 10.3168/jds.2024-26058] [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: 11/22/2024] [Accepted: 04/28/2025] [Indexed: 05/20/2025]
Abstract
Due to the high altitude and low oxygen levels, individuals residing or traveling in high-altitude regions often experience hypoxic cardiac dysfunction, which significantly affects their overall well-being and quality of life. Our previous investigations showed that peptide from yak milk residue exhibits notable antioxidant, anti-inflammatory, and anti-apoptotic properties that may have a good regulatory effect on hypoxic cardiac dysfunction. In this study, our results suggest that oral administration of yak milk peptide T3 improves the cardiac dysfunction of mice by the hypoxia-inducible factor 1α (HIF-1α) pathway, and these results may be related to the regulation of T3 on the gut microbiota of mice. Additionally, oral administration T3 enhances the permeability of the intestinal barrier and reduces intestinal inflammation. Further analysis revealed that the genera Oscillospira, Clostridium, and Staphylococcus are associated with aspartate aminotransferase, lactate dehydrogenase, and reactive oxygen species levels in heart tissues, which could ameliorate hypoxia-induced myocardial injury in mice. In vitro cell models have also confirmed that T3 intervention can activate the HIF-1α pathway and inhibit myocardial inflammation and cardiomyocyte apoptosis. These findings suggest that T3 may be a potential candidate for developing functional foods to reduce hypoxia-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Feiyan Yang
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Guangfan Qu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Yuchi Wu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Pingsheng Zhong
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Zhongxing Chu
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Zeyu He
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Yuyan Wang
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Yiping Tang
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China
| | - Shuguo Sun
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China.
| | - Feijun Luo
- National Engineering Laboratory for Deep Process of Rice and Byproducts, Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, China.
| |
Collapse
|
153
|
Singh A, Chaudhary R. Potentials of peroxisome proliferator-activated receptor (PPAR) α, β/δ, and γ: An in-depth and comprehensive review of their molecular mechanisms, cellular Signalling, immune responses and therapeutic implications in multiple diseases. Int Immunopharmacol 2025; 155:114616. [PMID: 40222274 DOI: 10.1016/j.intimp.2025.114616] [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: 01/22/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Peroxisome proliferator-activated receptors (PPARs), ligand-activated transcription factors, have emerged as a key regulator of various biological processes, underscoring their relevance in the pathophysiology and treatment of numerous diseases. PPARs are primarily recognized for their critical role in lipid and glucose metabolism, which underpins their therapeutic applications in managing type 2 diabetes mellitus. Beyond metabolic disorders, they have gained attention for their involvement in immune modulation, making them potential targets for autoimmune-related inflammatory diseases. Furthermore, PPAR's ability to regulate proliferation, differentiation, and apoptosis has positioned them as promising candidates in oncology. Their anti-inflammatory and anti-fibrotic properties further highlight their potential in dermatological and cardiovascular conditions, where dysregulated inflammatory responses contribute to disease progression. Recent advancements have elucidated the molecular mechanisms of different PPAR isoforms, including their regulation of key signalling pathways such as NF-κB and MAPK, which are crucial in inflammation and cellular stress responses. Additionally, their interactions with co-factors and post-translational modifications further diversify their functional roles. The therapeutic potential of various PPAR agonists has been extensively explored, although challenges related to side effects and target specificity remain. This growing body of evidence underscores the significance of PPARs in understanding the molecular basis of diseases and advancing therapeutic interventions, paving way for targeted treatment approach across a wide spectrum of medical conditions. Here, we provide a comprehensive and detailed perspective of PPARs and their potential across different health conditions to advance our understanding, elucidate underlying mechanisms, and facilitate the development of potential treatment strategies.
Collapse
Affiliation(s)
- Alpana Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
154
|
Liss A, Siddiqi MT, Marsland P, Varodayan FP. Neuroimmune regulation of the prefrontal cortex tetrapartite synapse. Neuropharmacology 2025; 269:110335. [PMID: 39904409 DOI: 10.1016/j.neuropharm.2025.110335] [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: 11/14/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
The prefrontal cortex (PFC) is an essential driver of cognitive, affective, and motivational behavior. There is clear evidence that the neuroimmune system directly influences PFC synapses, in addition to its role as the first line of defense against toxins and pathogens. In this review, we first describe the core structures that form the tetrapartite PFC synapse, focusing on the signaling microdomain created by astrocytic cradling of the synapse as well as the emerging role of the extracellular matrix in synaptic organization and plasticity. Neuroimmune signals (e.g. pro-inflammatory interleukin 1β) can impact the function of each core structure within the tetrapartite synapse, as well as promote intra-synaptic crosstalk, and we will provide an overview of recent advances in this field. Finally, evidence from post mortem human brain tissue and preclinical studies indicate that inflammation may be a key contributor to PFC dysfunction. Therefore, we conclude with a mechanistic discussion of neuroimmune-mediated maladaptive plasticity in neuropsychiatric disorders, with a focus on alcohol use disorder (AUD). Growing recognition of the neuroimmune system's role as a critical regulator of the PFC tetrapartite synapse provides strong support for targeting the neuroimmune system to develop new pharmacotherapeutics.
Collapse
Affiliation(s)
- Andrea Liss
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Mahum T Siddiqi
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Paige Marsland
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA
| | - Florence P Varodayan
- Developmental Exposure Alcohol Research Center and Behavioral Neuroscience Program, Department of Psychology, Binghamton University-SUNY, Binghamton, NY, USA.
| |
Collapse
|
155
|
Al-Kuraishy HM, Jabir MS, Al-Gareeb AI, Albuhadily AK, Klionsky DJ, Rafeeq MF. Epilepsy and autophagy modulators: a therapeutic split. Autophagy 2025. [PMID: 40375490 DOI: 10.1080/15548627.2025.2506292] [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: 06/27/2024] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
Epilepsy is a neurological disease characterized by repeated unprovoked seizure. Epilepsy is controlled by anti-epileptic drugs (AEDs); however, one third of epileptic patients have symptoms that are not controlled by AEDs in a condition called refractory epilepsy. Dysregulation of macroautophagy/autophagy is involved in the pathogenesis of epilepsy. Autophagy prevents the development and progression of epilepsy through regulating the balance between inhibitory and excitatory neurotransmitters. Induction of autophagy and autophagy-related proteins could be a novel therapeutic strategy in the management of epilepsy. Despite the protective role of autophagy against epileptogenesis and epilepsy, its role in status epilepticus is perplexing and might reflect its nature as a double-edged sword. Autophagy inducers play a critical role in reducing seizure frequency and severity, and could be an adjuvant treatment in the management of epilepsy. However, autophagy inhibitors also have an anticonvulsant effect. Therefore, the aim of the present mini-review is to discuss the potential role of autophagy in the pathogenesis of epileptogenesis and epilepsy, and how autophagy modulators affect epileptogenesis and epilepsy.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Majid S Jabir
- Department of Applied Science, University of Technology-Iraq, Baghdad, Iraq
| | | | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | | | - Mayyadah F Rafeeq
- Department of Applied Science, University of Technology-Iraq, Baghdad, Iraq
| |
Collapse
|
156
|
Yi X, Abas R, Raja Muhammad Rooshdi RAW, Yan J, Liu C, An J, Daut UN. Time-restricted feeding attenuated hypertension-induced cardiac remodeling by modulating autophagy levels in spontaneously hypertensive rats. Sci Rep 2025; 15:16973. [PMID: 40374761 PMCID: PMC12081920 DOI: 10.1038/s41598-025-01587-x] [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: 11/14/2024] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
To investigate whether time-restricted feeding (TRF) can alleviate cardiac remodeling in spontaneously hypertensive rats (SHRs) by regulating autophagy levels. A 16-week TRF intervention was conducted on Wistar Kyoto (WKY) rats and SHRs, with dietary intake confined to the interval from 9:00 am to 5:00 pm each day. The study examined the impact of TRF on blood pressure (BP), cardiac morphology and function, and the expression levels of key proteins involved in autophagy and its associated signaling cascades. Transmission Electron Microscopy (TEM) was utilized to further evaluate autophagic changes in left ventricular (LV) tissues. TRF significantly mitigated systolic blood pressure (SBP), diastolic blood pressure (DBP), and mean blood pressure (MBP) in SHRs. Additionally, TRF improved ejection fraction (EF) and diminished interventricular septal thickness at end-diastole (IVS-d). The study further revealed that TRF enhanced the expression of microtubule-associated protein-I light chain 3 (LC3-I), while reducing that of microtubule-associated protein-II light chain 3 (LC3-II). Moreover, TRF suppressed the expression levels of Beclin-1, phosphorylated phosphoinositide 3-kinase (p-PI3K), phosphorylated protein kinase B (p-AKT), and phosphorylated mechanistic target of rapamycin (p-mTOR) in the LV tissues. TEM analysis confirmed that TRF could inhibit autophagy levels in the LV tissues. TRF can attenuate cardiac remodeling in SHRs by regulating autophagy levels.
Collapse
Affiliation(s)
- Xin Yi
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Department 1 of Cardiovasology, North China University of Science and Technology Affiliated Hospital, Tangshan City, 063000, Hebei Province, China
| | - Razif Abas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | | | - Jie Yan
- Department 1 of Cardiovasology, North China University of Science and Technology Affiliated Hospital, Tangshan City, 063000, Hebei Province, China
| | - Canzhang Liu
- Department 1 of Cardiovasology, North China University of Science and Technology Affiliated Hospital, Tangshan City, 063000, Hebei Province, China
| | - Jiaxu An
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Ummi Nadira Daut
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
157
|
Xiong L, Huang YX, Mao L, Xu Y, Deng YQ. Targeting gut microbiota and its associated metabolites as a potential strategy for promoting would healing in diabetes. World J Diabetes 2025; 16:98788. [DOI: 10.4239/wjd.v16.i5.98788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
Impaired healing of diabetic wounds is one of the most important complications of diabetes, often leading to lower limb amputations and incurring significant economic and psychosocial costs. Unfortunately, there are currently no effective prevention or treatment strategies available. Recent research has reported that an imbalance in the gut microbiota, known as dysbiosis, was linked to the onset of type 2 diabetes, as well as the development and progression of diabetic complications. Indeed, the gut microbiota has emerged as a promising therapeutic approach for treating type 2 diabetes and related diseases. However, there is few of literatures specifically discussing the relationship between gut microbiota and diabetic wounds. This review aims to explore the potential role of the gut microbiota, especially probiotics, and its associated byproducts such as short chain fatty acids, bile acids, hydrogen sulfide, and tryptophan metabolites on wound healing to provide fresh insights and novel perspectives for the treatment of chronic wounds in diabetes.
Collapse
Affiliation(s)
- Ling Xiong
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ya-Xin Huang
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Lan Mao
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong-Qiong Deng
- Department of Dermatology & STD, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu 610000, Sichuan Province, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
158
|
Ye XW, Zhang HX, Li Q, Li CS, Zhao CJ, Xia LJ, Ren HM, Wang XX, Yang C, Wang YJ, Jiang SL, Xu XF, Li XR. Scientometric analysis and historical review of diabetic encephalopathy research: Trends and hotspots (2004-2023). World J Diabetes 2025; 16:91200. [DOI: 10.4239/wjd.v16.i5.91200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 12/18/2024] [Accepted: 02/20/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Diabetic encephalopathy (DE) is a common and serious complication of diabetes that can cause death in many patients and significantly affects the lives of individuals and society. Multiple studies investigating the pathogenesis of DE have been reported. However, few studies have focused on scientometric analysis of DE.
AIM To analyze literature on DE using scientometrics to provide a comprehensive picture of research directions and progress in this field.
METHODS We reviewed studies on DE or cognitive impairment published between 2004 and 2023. The latter were used to identify the most frequent keywords in the keyword analysis and explore the hotspots and trends of DE.
RESULTS Scientometric analysis revealed 1308 research papers on DE, a number that increased annually over the past 20 years, and that the primary topics explored were domain distribution, knowledge structure, evolution, and emergence of research topics related to DE. The inducing factors, comorbidities, pathogenesis, treatment, and animal models of DE help clarify its occurrence, development, and treatment. An increasing number of studies on DE may be a result of the recent increase in patients with diabetes, unhealthy lifestyles, and unhealthy eating habits, which have aggravated the incidence of this disease.
CONCLUSION We identified the main inducing factors and comorbidities of DE, though other complex factors undoubtedly increase social and economic burdens. These findings provide vital references for future studies.
Collapse
Affiliation(s)
- Xian-Wen Ye
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Hai-Xia Zhang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qian Li
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Chun-Shuai Li
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, Jiangxi Province, China
| | - Chong-Jun Zhao
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Liang-Jing Xia
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Hong-Min Ren
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xu-Xing Wang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Chao Yang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu-Jie Wang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shui-Lan Jiang
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xin-Fang Xu
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiang-Ri Li
- Traditional Chinese Medicine Processing Technology Inheritance Base of the National Administration of Traditional Chinese Medicine/Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
159
|
Chen Y, Dai MT, Gong GH. L-arginine overdose is a potential risk factor for myocardial injury in patients with type 2 diabetes. World J Diabetes 2025; 16:104409. [DOI: 10.4239/wjd.v16.i5.104409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 04/25/2025] Open
Abstract
We comment on an article published by Mansouri et al in the World Journal of Diabetes. L-arginine (L-Arg), a dietary supplement, is a precursor of nitric oxide, can improve cardiovascular disease, and it is important for treating heart disease and hypertension. Previous studies have demonstrated a beneficial effect of L-Arg on diabetes. In the study by Mansouri et al, L-Arg moderately increased blood glucose levels in normal rats. However, in diabetic rats, L-Arg significantly increased lipid levels, which is different from the findings of previous studies. This study demonstrated that a safe dose of 0.5 g/kg in diabetic rats can improve the lipid profile and decrease body weight. However, high doses (1 g/kg or higher) may aggravate damage to myocardial tissue in diabetic rats by increasing blood glucose, inflammation, and oxidative stress. Therefore, this study further demonstrated that high doses of L-Arg can exacerbate myocardial injury in diabetic patients.
Collapse
Affiliation(s)
- Yan Chen
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Meng-Ting Dai
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Guo-Hua Gong
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| |
Collapse
|
160
|
Sousa AAPD, Chaves LDS, Tarso Facundo H. Mitochondrial electron transport chain disruption and oxidative stress in lipopolysaccharide-induced cardiac dysfunction in rats and mice. Free Radic Res 2025:1-15. [PMID: 40337855 DOI: 10.1080/10715762.2025.2503844] [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: 07/15/2024] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/09/2025]
Abstract
Sepsis, characterized by severe systemic inflammation and an excessive immune response to infection, is frequently triggered by bacterial endotoxins like lipopolysaccharide (LPS) from Gram-negative bacteria. Moreover, sepsis-induced cardiac dysfunction remains a leading cause of mortality. This study aims to elucidate the effects of LPS-induced cardiac injury on mitochondrial damage, oxidative stress, and subsequent cardiac dysfunction. LPS injections (in rats and mice) for three days (1.5 mg/kg) impacted the body weight and increased cardiac TNF-α. Additionally, it decreased mitochondrial complexes I and II activities while complexes III and IV remained unaffected. Disturbed in mitochondrial electron transport chain leads to an increase in reactive oxygen species (ROS). Indeed, LPS treatment significantly increased mitochondrial hydrogen peroxide production, reduced the activity of antioxidant enzymes catalase, superoxide dismutase, glutathione peroxidase, and glutathione reductase activity. This was accompanied by decreased mitochondrial and cytosolic sulfhydryl proteins and parallel increased cellular lipid peroxidation in the presence or absence of Fe2+. LPS-treated samples had increased glutathione s-transferase activity, which may be an attempt of the cell to remove toxic lipid peroxidation products. In a more acute Langendorff-perfused rat hearts, LPS infusion (0.5 μg/mL) induced a significant elevation in left ventricular end-diastolic pressure and a decrease in left ventricular developed pressure. These findings elucidate the harmful mitochondrial and oxidative effects of LPS in cardiac tissue and could help the development of targeted therapies to mitigate the adverse effects of sepsis-induced cardiac dysfunction.
Collapse
|
161
|
Karuna N, Kerrigan L, Edgar K, Ledwidge M, McDonald K, Grieve DJ, Watson CJ. Sacubitril/Valsartan attenuates progression of diabetic cardiomyopathy through immunomodulation properties: an opportunity to prevent progressive disease. Cardiovasc Diabetol 2025; 24:206. [PMID: 40369551 PMCID: PMC12079907 DOI: 10.1186/s12933-025-02741-5] [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: 12/18/2024] [Accepted: 04/14/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND AND AIMS Diabetic cardiomyopathy (DbCM) is recognised as a key mediator and determinant of heart failure (HF), particularly HF with preserved ejection fraction (HFpEF). Improved understanding of mechanisms underlying transition from early-stage DbCM to HFpEF will inform innovative evidence-based treatment approaches, which are urgently required to alleviate increasing disease burden. This study aimed to determine whether inhibition of neprilysin activity by Sacubitril/Valsartan in both experimental and clinical DbCM attenuates adverse remodelling through promotion of cardioprotective signalling. METHODS AND RESULTS Sacubitril/Valsartan effectively reduced plasma neprilysin activity in both diabetic patients with pre-clinical HFpEF from the PARABLE trial (baseline (Val n = 25; Sac/Val n = 35) and 3 months after treatment (Val n = 21/25; Sac/Val n = 33/35)) and DbCM (high-fat diet and streptozotocin) mice. Plasma neprilysin activity at baseline was correlated with worsening cardiac performance at 18 months indicated by left atrial stiffness index in patients (n = 44/60), whilst diastolic dysfunction and pathological remodelling in DbCM mice were improved by Sacubitril/Valsartan, but not Valsartan. snRNA-sequencing showed that progressive experimental DbCM is characterised by chronic low-grade inflammation, reflected by increased infiltration of pro-inflammatory monocytes (Ccr2+ Ly6chi) and reduction in MHC-II macrophages, which was prevented by Sacubitril/Valsartan. Informatics analysis implicated IRF7 as a central mediator of Sacubitril/Valsartan-induced immunomodulation in DbCM, whilst treatment of M2-like pro-repair macrophages with the neprilysin inhibitor, LBQ657 and Valsartan suppressed glucose-induced IRF7 expression and paracrine activation of cardiac fibroblast differentiation in vitro. CONCLUSION Immune cells are significantly involved in DbCM progression, impacting myocardial homeostasis and HF progression. Neprilysin inhibition by Sacubitril/Valsartan improved adverse cardiac remodelling in experimental DbCM through direct regulation of inflammation, highlighting immunomodulation as a novel mechanism underlying established its cardioprotective actions.
Collapse
Affiliation(s)
- Narainrit Karuna
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
- Department of Pharmaceutical Care, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Lauren Kerrigan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Kevin Edgar
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Mark Ledwidge
- STOP-HF Unit, St. Vincent's University Healthcare Group and University College Dublin, Dublin, Ireland
| | - Ken McDonald
- STOP-HF Unit, St. Vincent's University Healthcare Group and University College Dublin, Dublin, Ireland
| | - David J Grieve
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK.
- STOP-HF Unit, St. Vincent's University Healthcare Group and University College Dublin, Dublin, Ireland.
| |
Collapse
|
162
|
Delalat S, Sultana I, Osman H, Sieme M, Zhazykbayeva S, Herwig M, Budde H, Kovács Á, Kaçmaz M, Göztepe E, Borgmann N, Shahriari G, Sasko B, Wintrich J, Haldenwang P, Schmidt WE, Fenske W, Khan M, Jaquet K, Mügge A, Máthé D, Tóth VE, Varga ZV, Ferdinandy P, El-Battrawy I, van Heerebeek L, Hamdani N. Dysregulated inflammation, oxidative stress, and protein quality control in diabetic HFpEF: unraveling mechanisms and therapeutic targets. Cardiovasc Diabetol 2025; 24:211. [PMID: 40369521 PMCID: PMC12080046 DOI: 10.1186/s12933-025-02734-4] [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: 02/19/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) represents a significant risk factor for cardiovascular disease, particularly heart failure with preserved ejection fraction (HFpEF). HFpEF predominantly affects elderly individuals and women, and is characterized by dysfunctions associated with metabolic, inflammatory, and oxidative stress pathways. Despite HFpEF being the most prevalent heart failure phenotype in patients with T2DM, its underlying pathophysiological mechanisms remain inadequately elucidated. OBJECTIVE This study aims to investigate the effects of diabetes mellitus on myocardial inflammation, oxidative stress, and protein quality control (PQC) mechanisms in HFpEF, with particular emphasis on insulin signaling, autophagy, and chaperone-mediated stress responses. METHODS We conducted an analysis of left ventricular myocardial tissue from HFpEF patients, both with and without diabetes, employing a range of molecular, biochemical, and functional assays. The passive stiffness of cardiomyocytes (Fpassive) was assessed in demembranated cardiomyocytes before and after implementing treatments aimed at reducing inflammation (IL-6 inhibition), oxidative stress (Mito-TEMPO), and enhancing PQC (HSP27, HSP70). Inflammatory markers (NF-κB, IL-6, TNF-α, ICAM-1, VCAM-1, NLRP3), oxidative stress markers (ROS, GSH/GSSG ratio, lipid peroxidation), and components of signaling pathways (PI3K/AKT/mTOR, AMPK, MAPK, and PKG) were evaluated using western blotting, immunofluorescence, and ELISA techniques. RESULTS Hearts from diabetic HFpEF patients exhibited significantly heightened inflammation, characterized by the upregulation of NF-κB, IL-6, and the NLRP3 inflammasome. This increase in inflammation was accompanied by elevated oxidative stress, diminished nitric oxide (NO) bioavailability, and impaired activation of the NO-sGC-cGMP-PKG signaling pathway. Notably, dysregulation of insulin signaling was observed, as indicated by decreased AKT phosphorylation and impaired autophagy regulation mediated by AMPK and mTOR. Additionally, PQC dysfunction was evidenced by reduced expression levels of HSP27 and HSP70, which correlated with increased cardiomyocyte passive stiffness. Targeted therapeutic interventions effectively reduced Fpassive, with IL-6 inhibition, Mito-TEMPO, and HSP administration leading to improvements in cardiomyocyte mechanical properties. CONCLUSION The findings of this study elucidate a mechanistic relationship among diabetes, inflammation, oxidative stress, and PQC impairment in the context of HFpEF. Therapeutic strategies that target these dysregulated pathways, including IL-6 inhibition, mitochondrial antioxidants, and chaperone-mediated protection, may enhance myocardial function in HFpEF patients with T2DM. Addressing these molecular dysfunctions could facilitate the development of novel interventions specifically tailored to the diabetic HFpEF population.
Collapse
Affiliation(s)
- Simin Delalat
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Innas Sultana
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Hersh Osman
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Marcel Sieme
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Saltanat Zhazykbayeva
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Melissa Herwig
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Heidi Budde
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Árpád Kovács
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Mustafa Kaçmaz
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Eda Göztepe
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Natalie Borgmann
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Gelareh Shahriari
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Benjamin Sasko
- Medical Department II, Marien Hospital Herne, Ruhr University Bochum, Bochum, Germany
| | - Jan Wintrich
- Medical Department II, Marien Hospital Herne, Ruhr University Bochum, Bochum, Germany
| | - Peter Haldenwang
- Department of Cardiothoracic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Wolfgang E Schmidt
- Department of Medicine I, St. Josef Hospital, UK RUB, Ruhr University Bochum, 44801, Bochum, Germany
| | - Wiebke Fenske
- Department of Internal Medicine I- General Internal Medicine, Endocrinology and Diabetology, Gastroenterology and Hepatology, BG University Hospital Bergmannsheil, Bochum, Germany
| | - Muchtiar Khan
- Department of Cardiology, OLVG, 1091 AC, Amsterdam, The Netherlands
| | - Kornelia Jaquet
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Andreas Mügge
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Semmelweis University, Tűzoltó utca 37-47, 1094, Budapest, 1085, Hungary
- In Vivo Imaging Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Budapest, Tűzoltó utca 37-47, 1094, Hungary
| | - Viktória E Tóth
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and pharmacotherapy,, Semmelweis University, Budapest, 1089, Hungary
- Center for Pharmacology and Drug Research & Development,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - Zoltán V Varga
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and pharmacotherapy,, Semmelweis University, Budapest, 1089, Hungary
- Center for Pharmacology and Drug Research & Development,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - Péter Ferdinandy
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and pharmacotherapy,, Semmelweis University, Budapest, 1089, Hungary
- Center for Pharmacology and Drug Research & Development,Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, 1089, Hungary
| | - Ibrahim El-Battrawy
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany
- Department of Medicine I, St. Josef Hospital, UK RUB, Ruhr University Bochum, 44801, Bochum, Germany
| | | | - Nazha Hamdani
- Medical Faculty, Department of Cellular and Translational Physiology, Institute of Physiology, Molecular and Experimental Cardiology, Institut Für Forschung und Lehre (IFL), Ruhr University Bochum, 44801, Bochum, Germany.
- Department of Physiology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands.
- Medical Faculty, Department Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, MA 2/156, 44780, Bochum, Germany.
- Institut Für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Gudrunstraße 56, 44791, Bochum, Germany.
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
163
|
Thakare AM, Nerurkar AS. Comparison of Anti-Biofilm Potential of Rhamnolipid Biosurfactant, Chemical Agents, and Coliphage Against E. coli Biofilm. Appl Biochem Biotechnol 2025:10.1007/s12010-025-05257-6. [PMID: 40366538 DOI: 10.1007/s12010-025-05257-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
Biosurfactants are amphipathic microbial products that are released extracellularly or remain attached to the cell surface. The strong biofilm anti-adhesive and anti-biofilm properties of biosurfactants make them suitable candidates for application aimed at destroying troublesome bacterial biofilm. To investigate the anti-adhesion and biofilm disruptive properties of natural rhamnolipid biosurfactant, targeted isolation of a hydrocarbonoclastic bacteria from hydrocarbon-contaminated soil of Dakor, Gujarat, India, led to the isolation of bacteria producing biosurfactant, identified as Pseudomonas aeruginosa. DKR. The orcinol test preliminarily indicated that the biosurfactant was indeed rhamnolipid. The Fourier transform infrared spectroscopy and nuclear magnetic resonance further confirmed the biosurfactant as rhamnolipid. Pseudomonas aeruginosa DKR produced 25 mg/ml rhamnolipid that reduced the surface tension to 22.4 mN/m and possessed CMC (critical micellar concentration) of 130 mg/L. Sub-inhibitory dilution (0.25 mg/ml) of purified rhamnolipid DKR demonstrated superior antiadhesive and antibiofilm properties against biofilm-forming E. coli strains isolated from drinking water coolers in comparison to subinhibitory concentrations of common chemical surfactants, chelating agents, and weak acids used. Coliphage AM isolated on selected E. coli strains as hosts also demonstrated an appreciable biofilm anti-adhesive and anti-biofilm effect at 106 Pfu/ml. This study emphasizes the utility of rhamnolipid biosurfactant DKR and Coliphage AM in lieu of chemicals as natural and eco-friendly agents in applications to eradicate biofilm from drinking water cooling containers, etc.
Collapse
Affiliation(s)
- Ajinkya M Thakare
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, India
| | - Anuradha S Nerurkar
- Department of Microbiology and Biotechnology Centre, Faculty of Science, The Maharaja Sayajirao University of Baroda, Gujarat, India.
| |
Collapse
|
164
|
Sahu K, Dash P. Chromium dynamics in soil and detoxification of chromite belts using rhizospheric soil-plant interface. ENVIRONMENTAL MONITORING AND ASSESSMENT 2025; 197:654. [PMID: 40360753 DOI: 10.1007/s10661-025-14085-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 04/29/2025] [Indexed: 05/15/2025]
Abstract
The chromium-contaminated soil expresses its severe eco-toxicity on living organisms of the locality and adjoining regions. This review has focused on the chemical interactions of chromium variants in soil and the sequestration of chromium using the soil-plant interface in the rhizosphere. The application of plant hyper-accumulators on chromium-contaminated soil for chromium sequestration is an attempt to minimize chromium toxicity of mining and industrial belts. This review utilized the PRISMA 2009 systematic review methodology. The literature screening was conducted by searching databases such as Scopus, Google Scholar, and Web of Science up to 2025 using specific keywords. In countries like Kazakhstan, South Africa, and India, more than 90% of world shipping-grade mine reserves of chromium are present. The mining and metallurgy of chromium can threaten the environmental quality and the region's public health. The Sukinda chromite mines in India are globally known for their rich chromite mining, metallurgy, and eco-toxicity. The present article analyzes the ecological challenges and searches for possible interactions of chromium variants in soil. The solution to mitigate chromium toxicity is possible using the rhizospheric soil-plant interface. This article's findings and discussion section help solve ecological challenges and strive for healthy soil at chromium-polluted sites. This review article can contribute to sustainable soil quality improvement at mining and industrial belts. Further research on the isotopic tracer technique is recommended to enhance the understanding of chromium dynamics in soil.
Collapse
Affiliation(s)
- Khusboo Sahu
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India
| | - Patitapaban Dash
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, 751003, Odisha, India.
| |
Collapse
|
165
|
de Souza MC, Agneis MLG, das Neves KA, de Almeida MR, Feltran GDS, Souza Cruz EM, Schoffen JPF, Chuffa LGDA, Seiva FRF. Melatonin Improves Lipid Homeostasis, Mitochondrial Biogenesis, and Antioxidant Defenses in the Liver of Prediabetic Rats. Int J Mol Sci 2025; 26:4652. [PMID: 40429795 PMCID: PMC12111231 DOI: 10.3390/ijms26104652] [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: 03/14/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Type 2 diabetes mellitus represents a major global health burden and is often preceded by a prediabetic state characterized by insulin resistance and metabolic dysfunction. Mitochondrial alterations, oxidative stress, and disturbances in lipid metabolism are central to the prediabetes pathophysiology. Melatonin, a pleiotropic indolamine, is known to regulate metabolic and mitochondrial processes; however, its therapeutic potential in prediabetes remains poorly understood. This study investigated the effects of melatonin on energy metabolism, oxidative stress, and mitochondrial function in a rat model of prediabetes induced by chronic sucrose intake and low-dose streptozotocin administration. Following prediabetes induction, animals were treated with melatonin (20 mg/kg) for four weeks. Biochemical analyses were conducted to evaluate glucose and lipid metabolism, and mitochondrial function was assessed via gene expression, enzymatic activity, and oxidative stress markers. Additionally, hepatic mitochondrial dynamics were examined by quantifying key regulators genes associated with biogenesis, fusion, and fission. Prediabetic animals exhibited dyslipidemia, hepatic lipid accumulation, increased fat depots, and impaired glucose metabolism. Melatonin significantly reduced serum glucose, triglycerides, and total cholesterol levels, while enhancing the hepatic high-density lipoprotein content. It also stimulated β-oxidation by upregulating hydroxyacyl-CoA dehydrogenase and citrate synthase activity. Mitochondrial dysfunction in prediabetic animals was evidenced by the reduced expression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha and mitochondrial transcription factor A, both of which were markedly upregulated by melatonin. The indolamine also modulated mithocondrial dynamics by regulating fusion and fission markers, including mitosuin 1 and 2, optic atrophy protein, and dynamin-related protein. Additionally, melatonin mitigated oxidative stress by enhancing the activity of superoxide dismutase and catalase while reducing lipid peroxidation. These findings highlight melatonin's protective role in prediabetes by improving lipid and energy metabolism, alleviating oxidative stress, and restoring mitochondrial homeostasis. This study provides novel insights into the therapeutic potential of melatonin in addressing metabolic disorders, particularly in mitigating mitochondrial dysfunction associated with prediabetes.
Collapse
Affiliation(s)
- Milena Cremer de Souza
- Department of Parasitology, Immunology and General Pathology, State University of Londrina (UEL), Londrina 86057-970, Paraná, Brazil
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Maria Luisa Gonçalves Agneis
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Karoliny Alves das Neves
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Matheus Ribas de Almeida
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Geórgia da Silva Feltran
- Department of Chemistry and Biochemistry, São Paulo State University (UNESP), Botucatu 18618-693, São Paulo, Brazil
| | - Ellen Mayara Souza Cruz
- Department of Parasitology, Immunology and General Pathology, State University of Londrina (UEL), Londrina 86057-970, Paraná, Brazil
| | - João Paulo Ferreira Schoffen
- Center of Biological Sciences, State University of Northern Paraná (UENP), Bandeirantes 86360-000, Paraná, Brazil
| | | | | |
Collapse
|
166
|
Zhou S, Zhang L, Duan X, Liu K, Yingnan Y, Ma M, Han B. MiR-425-5p intervenes in autoimmune myocarditis by regulating Treg cell differentiation through NRAS. Front Cell Dev Biol 2025; 13:1600103. [PMID: 40433545 PMCID: PMC12106460 DOI: 10.3389/fcell.2025.1600103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Aim Our Previous research revealed significant differences in exosome-mediated intercellular miR-425a-5p between normal children and those with fulminant myocarditis. We sought to elucidate the molecular underpinnings and functional implications of miR-425a-5p in the context of myocarditis progression. Methods Bioinformatics techniques were employed to predict NRAS as the target gene of miR-425a-5p. We constructed a cellular myocarditis paradigm through LPS-mediated provocation of AC16 cardiomyocyte cultures. MiR-425a-5p was overexpressed, and the expressions of NRAS, cell apoptosis, and proinflammatory cytokine profiles, encompassing IL-1β, IL-6, and TNF-α, were comprehensively quantified. An experimental autoimmune myocarditis (EAM) mouse model was created using adeno-associated virus (AAV) for miR-425a-5p overexpression. Comprehensive histopathological analyses were conducted utilizing multiple staining techniques, including hematoxylin-eosin (HE), immunohistochemical, and Masson trichrome methodologies to characterize tissue responses. Results The study demonstrated that miR-425a-5p alleviated the inflammatory response in both AC16 cells and EAM mice through NRAS mediation. Single-cell data analysis of cardiac immune cells revealed that miR-425a-5p promoted Treg cell differentiation and improved cardiac function. Conclusion MiR-425a-5p plays a crucial role in modulating inflammatory responses in myocarditis, potentially offering a novel therapeutic strategy for managing the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
167
|
Pekayvaz K, Kilani B, Joppich M, Eivers L, Brambs S, Knottenberg V, Akgöl S, Yue K, Li L, Martinez-Navarro A, Kaiser R, Meißner N, Schulz H, Belz L, Akhalkatsi A, Stockhausen S, Mueller TT, Millonig S, Hartelt L, Gold C, Janjic A, Polewka V, Wendler F, Droste Zu Senden A, Titova A, Leunig A, Voelkl M, Engelmann B, Hernandez Petzsche MR, Boeckh-Behrens T, Liebig T, Winning S, Fandrey J, Dichgans M, Enard W, Zimmer R, Tiedt S, Massberg S, Nicolai L, Stark K. Immunothrombolytic monocyte-neutrophil axes dominate the single-cell landscape of human thrombosis and correlate with thrombus resolution. Immunity 2025; 58:1343-1358.e13. [PMID: 40280129 DOI: 10.1016/j.immuni.2025.03.020] [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: 08/30/2024] [Revised: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Thrombotic diseases remain the major cause of death and disability worldwide, and the contribution of inflammation is increasingly recognized. Thromboinflammation has been identified as a key pathomechanism, but an unsupervised map of immune-cell states, trajectories, and intercommunication at a single-cell level has been lacking. Here, we reveal innate leukocyte substates with prominent thrombolytic properties by employing single-cell omics measures on human stroke thrombi. Using in vivo and in vitro thrombosis models, we propose a pro-resolving monocyte-neutrophil axis, combining two properties: (1) NR4A1hi non-classical monocytes acquire a thrombolytic and neutrophil-chemoattractive phenotype, and (2) blood neutrophils are thereby continuously recruited to established thrombi through CXCL8-CXCR1 and CXCR2 and adopt a hypoxia-induced thrombus-resolving urokinase receptor (PLAUR)+ phenotype. This immunothrombolytic axis results in thrombus resolution. Together, with this immune landscape of thrombosis, we provide a valuable resource and introduce the concept of "immunothrombolysis" with broad mechanistic and translational implications at the crossroad of inflammation and thrombosis.
Collapse
Affiliation(s)
- Kami Pekayvaz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Badr Kilani
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luke Eivers
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Sophia Brambs
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sezer Akgöl
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Keyang Yue
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Li
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Rainer Kaiser
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Nina Meißner
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Heiko Schulz
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Larissa Belz
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Sven Stockhausen
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Tonina T Mueller
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Simon Millonig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Lea Hartelt
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Christoph Gold
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Vivien Polewka
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Franziska Wendler
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | | | - Anna Titova
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Leunig
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Voelkl
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany; Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
| | - Bernd Engelmann
- Institute of Laboratory Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Moritz R Hernandez Petzsche
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Tobias Boeckh-Behrens
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas Liebig
- Institute for Diagnostic and Interventional Neuroradiology, University Hospital, LMU Munich, Munich, Germany
| | - Sandra Winning
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Joachim Fandrey
- University of Duisburg-Essen, Institute for Physiology, Essen, Germany
| | - Martin Dichgans
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany; Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Massberg
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| | - Konstantin Stark
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
168
|
Tsai LT, Wu CT, Liu CY, Chiang CK, Liu SH. Zinc protoporphyrin accumulation as a positive regulator of renal heme oxygenase-1 participates in the progression of chronic kidney disease. Biochem Biophys Res Commun 2025; 770:152014. [PMID: 40381240 DOI: 10.1016/j.bbrc.2025.152014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
Chronic kidney disease (CKD) has become a major global public health concern, with both its incidence and prevalence continuing to rise. Zinc protoporphyrin (ZnPP) is formed during heme biosynthesis when zinc is incorporated into the protoporphyrin IX ring in place of iron, a process that is markedly enhanced under conditions of iron deficiency or impaired iron metabolism. Elevated ZnPP levels observed in patients with renal failure result from a variety of pathogenic mechanisms. Heme oxygenase (HO)-1, a key enzyme in heme catabolism, degrades heme into biliverdin (subsequently converted to bilirubin), carbon monoxide, and ferrous iron. However, the relationship between ZnPP and HO-1 in the kidney, as well as their roles in CKD progression, still remains to be clarified. In the present study, an adenine-induced CKD mouse model was utilized to investigate the regulatory role of ZnPP in HO-1 expression and activity and its involvement in CKD progression in vivo. CKD mice exhibited substantial ZnPP accumulation, accompanied by significant upregulation of renal HO-1 protein expression and enzymatic activity, along with pronounced renal dysfunction. To further elucidate the role of ZnPP, N,N,N',N'-tetrakis (2-pyridinylmethyl)-1,2-ethanediamine (TPEN), a potent zinc chelator as a ZnPP formation inhibitor, was administered. TPEN treatment markedly attenuated ZnPP accumulation, decreased HO-1 protein expression and enzymatic activity, and ameliorated renal pathological changes in CKD mice. Collectively, these findings suggest that endogenous ZnPP may act as an activator of HO-1 in the kidney and contribute to the pathogenesis of CKD. Targeting ZnPP-mediated HO-1 pathway may offer a novel therapeutic strategy for CKD management.
Collapse
Affiliation(s)
- Li-Ting Tsai
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Cheng-Tien Wu
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chieh-Yun Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chih-Kang Chiang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan; Departments of Integrated Diagnostics & Therapeutics and Internal Medicine, College of Medicine and Hospital, National Taiwan University, Taipei, Taiwan.
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University & Hospital, Taipei, Taiwan.
| |
Collapse
|
169
|
Acharya D, Sayyad Z, Hoenigsperger H, Hirschenberger M, Zurenski M, Balakrishnan K, Zhu J, Gableske S, Kato J, Zhang SY, Casanova JL, Moss J, Sparrer KMJ, Gack MU. TRIM23 mediates cGAS-induced autophagy in anti-HSV defense. Nat Commun 2025; 16:4418. [PMID: 40360474 PMCID: PMC12075517 DOI: 10.1038/s41467-025-59338-5] [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: 07/16/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
The cGAS-STING pathway, well-known to elicit interferon (IFN) responses, is also a key inducer of autophagy upon virus infection or other stimuli. Whereas the mediators for cGAS-induced IFN responses are well characterized, much less is known about how cGAS elicits autophagy. Here, we report that TRIM23, a unique TRIM protein harboring both ubiquitin E3 ligase and GTPase activity, is crucial for cGAS-STING-dependent antiviral autophagy. Genetic ablation of TRIM23 impairs autophagic control of HSV-1 infection. HSV-1 infection or cGAS-STING stimulation induces TBK1-mediated TRIM23 phosphorylation at S39, which triggers TRIM23 autoubiquitination and GTPase activity and ultimately elicits autophagy. Fibroblasts from a patient with herpes simplex encephalitis heterozygous for a dominant-negative, kinase-inactivating TBK1 mutation fail to activate autophagy by TRIM23 and cGAS-STING. Our results thus identify the cGAS-STING-TBK1-TRIM23 axis as a key autophagy defense pathway and may stimulate new therapeutic interventions for viral or inflammatory diseases.
Collapse
Affiliation(s)
- Dhiraj Acharya
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
| | - Zuberwasim Sayyad
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | | | | | - Matthew Zurenski
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
| | - Kannan Balakrishnan
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Junji Zhu
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Sebastian Gableske
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
- Eisai GmbH, Frankfurt am Main, Germany
| | - Jiro Kato
- The Critical Care Medicine and Pulmonary Branch; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| | - Joel Moss
- The Critical Care Medicine and Pulmonary Branch; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Konstantin M J Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Michaela U Gack
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA.
- Department of Microbiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
170
|
Aliyu U, Toor SM, Abdalhakam I, Elrayess MA, Abou−Samra AB, Albagha OME. Evaluating indices of insulin resistance and estimating the prevalence of insulin resistance in a large biobank cohort. Front Endocrinol (Lausanne) 2025; 16:1591677. [PMID: 40421243 PMCID: PMC12104043 DOI: 10.3389/fendo.2025.1591677] [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: 03/11/2025] [Accepted: 04/11/2025] [Indexed: 05/28/2025] Open
Abstract
Introduction Insulin resistance (IR) is involved in the pathogenesis of various metabolic disorders. Several surrogate indices of IR have been proposed. We assessed the performance of seven clinically relevant indirect measures of IR and estimated the prevalence of IR in a large population-based cohort. Methods The study was conducted on fasting individuals from the Qatar biobank (QBB) participants (n = 7,875). Individuals were considered insulin sensitive (IS) if lean, not diagnosed with diabetes, no hypertriglyceridemia, and not on lipid-lowering drugs, while individuals with Type 2 diabetes (T2D) were considered insulin resistant (IR). Cut-offs were determined as the top or lowest quartile values in the IS participants. The performance of IR indices was based on area under the curve (AUC), sensitivity and specificity. Results The cut-off for HOMA-IR was determined at 1.878, HOMA2-IR (insulin); 1.128, HOMA2-IR (C-peptide); 1.307, QUICKI; 0.347, TyG; 8.281, McAi; 7.727 and 1.718 for TG/HDL. All IR indices analyzed yielded AUC values ranging from 0.83 to 0.92. TyG was the most robust measure for IR (AUC = 0.92, Sensitivity = 0.90, Specificity = 0.79). The overall prevalence of IR in Qatar was estimated at ~51 - 65%. Conclusions TyG index was the most robust index for determining IR in the Qatari population. The proposed cut-offs could serve as a reference in Middle Eastern populations for IR screening.
Collapse
Affiliation(s)
- Usama Aliyu
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M. Toor
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | | | - Mohamed A. Elrayess
- Biomedical Research Center, QU Health, Qatar University, Doha, Qatar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Omar M. E. Albagha
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
171
|
Zaidi SA, Fan Z, Chauhdari T, Ding Y. MicroRNA regulatory dynamic, emerging diagnostic and therapeutic frontier in atherosclerosis. Microvasc Res 2025; 160:104818. [PMID: 40368159 DOI: 10.1016/j.mvr.2025.104818] [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: 03/01/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
MicroRNAs (miRNAs), a class of non-coding RNAs, are pivotal post-transcriptional regulators of gene expression with profound implications in the pathogenesis of atherosclerosis (AS). As a progressive arterial disease driven by vascular cells dysfunction, lipid dysregulation and subsequent chronic inflammation, AS remains a leading cause of global morbidity. Recent studies have demonstrated how important miRNAs are in regulating central biological processes in the vascular wall, such as endothelial function, vascular smooth muscle cell (VSMC) phenotypic switching, and macrophage polarization. This review provides comprehensive insight into the role of miRNAs in the development and complexity of atherosclerotic plaques according to their effects on endothelial cells, macrophages, and VSMCs. We also go over the growing prospects of miRNAs as therapeutic targets and diagnostic biomarkers, providing information to be used in the study of vascular diseases. Lastly, we address recent complications and potential applications of miRNA-based approaches in clinical practice.
Collapse
Affiliation(s)
- Syeda Armana Zaidi
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Zhiyu Fan
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Talha Chauhdari
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| | - Yongsheng Ding
- College of Life Sciences, University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, PR China.
| |
Collapse
|
172
|
Hartmann S, Radochonski L, Ye C, Martinez-Sobrido L, Chen J. SARS-CoV-2 ORF3a drives dynamic dense body formation for optimal viral infectivity. Nat Commun 2025; 16:4393. [PMID: 40355429 PMCID: PMC12069715 DOI: 10.1038/s41467-025-59475-x] [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: 05/08/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
SARS-CoV-2 hijacks multiple organelles for virion assembly, of which the mechanisms have not been fully understood. Here, we identified a SARS-CoV-2-driven membrane structure named the 3a dense body (3DB). 3DBs are unusual electron-dense and dynamic structures driven by the accessory protein ORF3a via remodeling a specific subset of the trans-Golgi network (TGN) and early endosomal membrane. 3DB formation is conserved in related bat and pangolin coronaviruses but was lost during the evolution to SARS-CoV. During SARS-CoV-2 infection, 3DB recruits the viral structural proteins spike (S) and membrane (M) and undergoes dynamic fusion/fission to maintain the optimal unprocessed-to-processed ratio of S on assembled virions. Disruption of 3DB formation resulted in virions assembled with an abnormal S processing rate, leading to a dramatic reduction in viral entry efficiency. Our study uncovers the crucial role of 3DB in maintaining maximal SARS-CoV-2 infectivity and highlights its potential as a target for COVID-19 prophylactics and therapeutics.
Collapse
Affiliation(s)
- Stella Hartmann
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Lisa Radochonski
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Jueqi Chen
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA.
| |
Collapse
|
173
|
Zhu XX, Zhao CY, Lu QB, Zhang AY, Meng XY, Su JB, Chen G, Xu AJ, Sun HJ, Nie XW. Hydrogen sulfide as a new therapeutic target of pulmonary hypertension: an overview with update on immunomodulation. Front Pharmacol 2025; 16:1510275. [PMID: 40421210 PMCID: PMC12104183 DOI: 10.3389/fphar.2025.1510275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Pulmonary hypertension (PH) is a complex and progressive vascular disease characterized by elevated pulmonary arterial pressure (PAP) and vascular resistance, leading to right ventricular failure and, ultimately, death. Current therapies primarily focus on vasodilation and symptom management, but there remains a critical need for treatments that address the underlying pathophysiological mechanisms of PH. Numerous studies have identified hydrogen sulfide (H2S) as a potential therapeutic target in PH. Traditionally recognized for its toxic effects at high concentrations, H2S is now known to play crucial roles in various physiological processes, including vasodilation, anti-inflammation, and antioxidation, which are relevant to PH pathogenesis. Given its multifaceted roles in the pathophysiology of PH, H2S represents a promising therapeutic target. Strategies to enhance endogenous H2S production or administer exogenous H2S donors are being explored as potential treatments for PH. These approaches aim to harness the vasodilatory, anti-inflammatory, antioxidant, and anti-remodeling properties of H2S to mitigate disease progression and improve patient outcomes. Future research should focus on optimizing H2S-based therapies and exploring their clinical efficacy and safety in PH patients.
Collapse
Affiliation(s)
- Xue-Xue Zhu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, China
| | - Chen-Yang Zhao
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, China
| | - Qing-Bo Lu
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China
| | - Ao-Yuan Zhang
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, China
| | - Xin-Yu Meng
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, China
| | - Jia-Bao Su
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, China
| | - Guo Chen
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, China
| | - An-Jing Xu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, China
| | - Hai-Jian Sun
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, China
| | - Xiao-Wei Nie
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
174
|
Cao C, Hua W, Xian R, Liu Y. Fumonisin B 1 Exposure Causes Intestinal Tissue Damage by Triggering Oxidative Stress Pathways and Inducing Associated CYP Isoenzymes. Toxins (Basel) 2025; 17:239. [PMID: 40423322 DOI: 10.3390/toxins17050239] [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: 03/24/2025] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
Fumonisin B1 (FB1) is considered the most toxic fumonisin produced by fungi and is commonly found in contaminated feed and crops. Fumonisin and its metabolites extensively exist in feed and crops, where FB1-polluted crop ingestion can do harm to livestock and poultry, causing poultry intestinal toxicity in the latter. For investigating FB1-mediated intestinal toxicity, we assessed the function of FB1 exposure in quail intestines and explored its possible molecular mechanisms. In total, 120 quail pups were classified into two groups, where those in the control group were given a typical control diet, and those in the experimental group were given a typical diet that contained 30 mg/kg FB1. We evaluated the histopathological and ultrastructural changes in quails' intestines on days 14, 28, and 42, and studied the molecular mechanisms by assessing oxidative stress, inflammation, and nuclear xenobiotic receptors (NXRs). Our results suggest that FB1 exposure causes intestinal inflammation by triggering oxidative stress pathways and modulating NXRs to induce Cytochrome P450 proteins (CYP) isoforms, leading to intestinal histopathological damage. The results of this study shed novel light on the molecular mechanism underlying FB1-induced intestinal injury in juvenile quails.
Collapse
Affiliation(s)
- Changyu Cao
- College of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Weiping Hua
- College of Animal Science and Technology, Foshan University, Foshan 528225, China
- Foshan University Veterinary Teaching Hospital, Foshan 528231, China
| | - Runxi Xian
- College of Animal Science and Technology, Foshan University, Foshan 528225, China
- Foshan University Veterinary Teaching Hospital, Foshan 528231, China
| | - Yang Liu
- Quality Control Technical Center (Foshan) of National Famous and Special Agricultural Products (CAQS-GAP-KZZX043)/South China Food Safety Research Center, Foshan 528231, China
| |
Collapse
|
175
|
Marín de Jesús S, Vigueras-Villaseñor RM, Cortés-Barberena E, Hernández-Rodríguez J, Pérez-Aguirre SG, Montes S, Carrizales-Yáñez L, Arrieta-Cruz I, Arteaga-Silva M. Early Zinc Supplementation Enhances Epididymal Sperm Glycosylation, Endocrine Activity, and Antioxidant Activity in Rats Exposed to Cadmium. Int J Mol Sci 2025; 26:4589. [PMID: 40429736 PMCID: PMC12111536 DOI: 10.3390/ijms26104589] [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: 03/28/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Sperm maturation involves changes in plasma membrane glycosylation for fertilization. Cadmium (Cd) exerts a negative effect by disrupting testicular and epididymal function, altering antioxidant activity. Zinc (Zn) is an essential element known for its antioxidant properties, role in testosterone synthesis, and support of spermatogenesis. However, its effect on sperm membrane glycosylation, as well as endocrine and antioxidant activity, after exposure to Cd has remained unexplored. This study evaluated the impact of Zn on epididymal sperm glycosylation, endocrine activity, and antioxidant activity in Cd-exposed rats. Four groups of male Wistar rats were analyzed: control, Cd-exposed, Zn-supplemented, and Zn + Cd groups. On postnatal day 90, tissues and blood were collected for Zn and Cd quantification, testosterone levels, antioxidant activity, histological analysis, and sperm quality. The results showed that Cd concentration increased significantly, reduced testosterone levels, modified antioxidant activity, and caused structural damage in the epididymis. The Cd-exposed group showed disrupted glycosylation and distribution patterns and reduced sperm quality. The Zn + Cd group showed lower Cd accumulation, preserved testosterone levels, restored antioxidant activity, and preserved glycosylation patterns and sperm quality. This study highlights the protective role of Zn in mitigating Cd-induced reproductive toxicity, probably through the competitive inhibition of Cd uptake and antioxidant support, thereby preserving fertility.
Collapse
Affiliation(s)
- Sergio Marín de Jesús
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco 186, Col. Leyes de Reforma 1ª. Sección, Alcaldía Iztapalapa, Ciudad de México C.P. 09340, Mexico; (S.M.d.J.); (S.G.P.-A.)
| | - Rosa María Vigueras-Villaseñor
- Laboratorio de Biología de la Reproducción, Instituto Nacional de Pediatría, Av. Insurgentes Sur 3700-Letra C, Insurgentes Cuicuilco, Coyoacán, Ciudad de México C.P. 04530, Mexico;
| | - Edith Cortés-Barberena
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud de la Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco 186, Col. Leyes de Reforma 1ª. Sección, Alcaldía Iztapalapa, Ciudad de México C.P. 09340, Mexico;
| | - Joel Hernández-Rodríguez
- Cuerpo Académico en Quiropráctica, Universidad Estatal del Valle de Ecatepec, Av. Central s/n, Esq. Leona Vicario, Col. Valle de Anáhuac, Secc. A, Ecatepec C.P. 55210, Mexico;
| | - Sonia Guadalupe Pérez-Aguirre
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco 186, Col. Leyes de Reforma 1ª. Sección, Alcaldía Iztapalapa, Ciudad de México C.P. 09340, Mexico; (S.M.d.J.); (S.G.P.-A.)
| | - Sergio Montes
- Unidad Académica Multidisciplinaria Reynosa-Aztlán, Universidad Autónoma de Tamaulipas, Calle 16 y Lago de Chapala, Col. Aztlán, Reynosa C.P. 88740, Mexico;
| | - Leticia Carrizales-Yáñez
- Coordinación de Innovación y Aplicación de la Ciencia y la Tecnología (CIACYT)-Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí C.P. 78210, Mexico;
| | - Isabel Arrieta-Cruz
- Departamento de Investigación Básica, Instituto Nacional de Geriatría, Secretaria de Salud, Ciudad de México C.P. 10200, Mexico;
| | - Marcela Arteaga-Silva
- Laboratorio de Neuroendocrinología Reproductiva, Departamento de Biología de la Reproducción, División de Ciencias Biológicas y de la Salud de la Universidad Autónoma Metropolitana-Iztapalapa, Av. Ferrocarril San Rafael Atlixco 186, Col. Leyes de Reforma 1ª. Sección, Alcaldía Iztapalapa, Ciudad de México C.P. 09340, Mexico
| |
Collapse
|
176
|
Wang J, Zhang Y, Cao C, Hua J, Xing L, Wu C. The anti-atherosclerosis effect and molecular mechanism of AMPKα1 by polarizing monocytes to an M2 phenotype via cell-intrinsic lysosomal lipolysis. Cardiovasc Pathol 2025; 78:107742. [PMID: 40354887 DOI: 10.1016/j.carpath.2025.107742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025] Open
Abstract
Regulating the differentiation of monocytes into M2 macrophages can promote the regression of Atherosclerosis (AS) plaque. However, the key molecules regulating the differentiation of monocytes to M2 are unknown. In this study, we reported that adenosine-activated protein kinase α1 (AMPKα1) plays an anti-AS role by polarizing monocytes to an M2 phenotype via promoting fatty acid oxidation (FAO). AMPKα1 enhances the decomposition of cholesterol esters by increasing lysosomal acid lipase expression to provide fatty acids for FAO. Furthermore, AMPKα1 can induce lysosomal biogenesis and enhance lipolysis by promoting the transcription factor EB (TFEB) expression and facilitating TFEB nuclear translocation. In conclusion, AMPKα1 enhances the decomposition of cholesterol esters by increasing lysosomal acid lipase expression to produce fatty acids, which may represent a mechanism to promote FAO and inflammatory monocytes differentiation towards M2 phenotype.
Collapse
Affiliation(s)
- Jing Wang
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China.
| | - Yahui Zhang
- School of Life Science, Shanxi University, Taiyuan, 030006, China
| | - Caixing Cao
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Jiale Hua
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Li Xing
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi University, 92 Wucheng Road, Taiyuan, 030006, Shanxi province, China.
| |
Collapse
|
177
|
Wu M, Yu C, Wen F, Li Y, Zhang X, Wang Y, Chen X, Chen X. NLRP3 inflammasome inhibits mitophagy during the progression of temporal lobe epilepsy. Sci Rep 2025; 15:16341. [PMID: 40348802 PMCID: PMC12065917 DOI: 10.1038/s41598-025-01087-y] [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: 07/04/2024] [Accepted: 05/02/2025] [Indexed: 05/14/2025] Open
Abstract
Epilepsy is a neurological disorder involving mitochondrial dysfunction and neuroinflammation. This study examines the relationship between NLRP3 inflammasome activation and mitophagy in the temporal lobe epilepsy, which has not been reported before. A pilocarpine-induced epileptic rat model was used to assess seizure activity and neuronal loss. Pyroptosis markers (NLRP3, cleaved Gasdermin D, IL-1β/IL-18), and autophagy/mitophagy activity (LC3B-II/I, BNIP3, TOMM20/LC3B colocalization) were analyzed via immunofluorescence, Western blot, and transmission electron microscopy. NLRP3 inhibitors and anti-IL-1β antibodies were administered to evaluate therapeutic effects. Epileptic rats exhibited progressive neuronal loss and seizure aggravation, correlating with NLRP3 inflammasome activation and pyroptosis. While general autophagy was upregulated, mitophagy was selectively impaired in the hippocampus. NLRP3 activation promoted IL-1β release, which suppressed mitophagy via PPTC7 upregulation. NLRP3 activation inhibitor (MCC950) and anti-IL-1β treatment restored mitophagy and reduced seizures. NLRP3 inflammasome-driven pyroptosis exacerbates epilepsy by impairing mitophagy activity via IL-1β/PPTC7. Targeted NLRP3 inhibition mitigates this cascade, offering a promising strategy for refractory epilepsy.
Collapse
Affiliation(s)
- Mengqian Wu
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Cong Yu
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Fuli Wen
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Yunfei Li
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Xu Zhang
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Yinzhou Wang
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China
| | - Xiaoqian Chen
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| | - Xingyong Chen
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou University Affiliated Provincial Hospital, Fuzhou, 350001, China.
| |
Collapse
|
178
|
Kang HL, Várkonyi Á, Csonka Á, Szász A, Várkonyi T, Pósa A, Kupai K. Endothelial-Mesenchymal Transition and Possible Role of Cytokines in Streptozotocin-Induced Diabetic Heart. Biomedicines 2025; 13:1148. [PMID: 40426976 PMCID: PMC12109261 DOI: 10.3390/biomedicines13051148] [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: 02/10/2025] [Revised: 05/04/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Although endothelial mesenchymal transition (EndMT) has been characterized as a basic process in embryogenesis, EndMT is the mechanism that accelerates the development of cardiovascular diseases, including heart failure, aging, and complications of diabetes or hypertension as well. Endothelial cells lose their distinct markers and take on a mesenchymal phenotype during EndMT, expressing distinct products. Methods: In this study, type 1 Diabetes mellitus (T1DM) was induced in rats with streptozotocin (STZ) by intraperitoneal injection at a 60 mg/kg dose. Diabetic rats were randomly divided into two groups, namely, control and diabetic rats, for 4 weeks. Heart, aorta, and plasma samples were collected at the end of 4 weeks. Sequentially, biochemical parameters, cytokines, reactive oxygen species (ROS), protein expression of EndMT markers (Chemokine C-X-C motif ligand-1 (CXCL-1), vimentin, citrullinated histone H3 (H3Cit), α-smooth muscle actin (α-SMA), and transforming growth factor beta (TGF-β) and versican), components of the extracellular matrix (matrix metalloproteinase 2 (MMP-2), tissue inhibitor of metalloproteinase-1(TIMP-1), and discoidin domain tyrosine kinase receptor 2 (DDR-2)) were detected by ELISA or Western blot, respectively. Results: Cytokines and ROS were increased in diabetic hearts, which induced partial EndMT. Among EndMT markers, histone citrullination, α-SMA, and CXCL-1 were increased; vimentin was decreased in DM. The endothelial marker endothelin-1 was significantly higher in the aortas of DM rats. Interestingly, TGF-β showed a significant decrease in the diabetic heart, plasma, and aorta. Additionally, MMP-2/TIMP-1 levels also decreased in DM. Conclusions: To sum up, the identification of molecules and regulatory pathways involved in EndMT provided novel therapeutic approaches for cardiac pathophysiological conditions.
Collapse
Affiliation(s)
- Hsu Lin Kang
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
| | - Ákos Várkonyi
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
| | - Ákos Csonka
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary; (Á.C.); (T.V.)
| | - András Szász
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
| | - Tamás Várkonyi
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary; (Á.C.); (T.V.)
| | - Anikó Pósa
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
| | - Krisztina Kupai
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary (Á.V.); (A.S.); (A.P.)
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary; (Á.C.); (T.V.)
| |
Collapse
|
179
|
DeLano K, Sprague AC, Jandarov R, Jackson BP, Shatz R, Langevin SM, Sawyer RP. Association of plasma concentration of trace metals with frontotemporal degeneration. Front Neurol 2025; 16:1593821. [PMID: 40417119 PMCID: PMC12098025 DOI: 10.3389/fneur.2025.1593821] [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: 03/14/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025] Open
Abstract
Objective Compare the burden of heavy metals in plasma from people with frontotemporal degeneration (FTD) and healthy controls. Methods A cross-sectional study of 14 FTD cases and 28 healthy controls recruited from the University of Cincinnati. Plasma samples were sent to the Trace Element Analysis Core at Dartmouth College for assessment of 24 metals or metalloids via inductively coupled plasma mass spectrometry (ICP-MS). Unconditional logistic regression models were performed with adjustments for age (centered at the median) and sex. Results After adjusting for age and sex, there was a significant positive association of FTD with the highest tertile of Manganese (ORadjusted = 11.1, 95% CI: 1.57-132) and Chromium (ORadjusted = 9.86, 95% CI: 1.24-218). There was significant inverse associations observed between FTD and the highest tertile of Barium (ORadjusted = 0.06, 95% CI: <0.01-0.47) and Mercury (ORadjusted = 0.13, 95% CI: 0.01-0.74), with a significant inverse trend (ptrend = 0.03). Conclusion Significant associations between plasma concentration of several trace metals and FTD. The significantly elevated levels of Manganese and Chromium may suggest a role of environmental exposure in the pathogenesis of FTD. However, larger, well-designed prospective studies, along with complementary experimental work, are needed to better elucidate this relationship.
Collapse
Affiliation(s)
- Kelly DeLano
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Alex C. Sprague
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Department of Environmental and Public Health Sciences, Division of Epidemiology, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, Division of Epidemiology, Cincinnati, OH, United States
| | - Roman Jandarov
- Department of Environmental and Public Health Sciences, University of Cincinnati College of Medicine, Department of Environmental and Public Health Sciences, Division of Epidemiology, Cincinnati, OH, United States
- Department of Environmental and Public Health Sciences, Division of Epidemiology, Cincinnati, OH, United States
| | - Brian P. Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH, United States
| | - Rhonna Shatz
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Scott M. Langevin
- Division of Hematology & Oncology, University of Vermont Larner College of Medicine, Burlington, VT, United States
- University of Vermont Cancer Center, Burlington, VT, United States
| | - Russell P. Sawyer
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
180
|
Zhou S, Han B. Biological disturbance of MiR-425 and its application prospects in cardiovascular diseases. Front Cell Dev Biol 2025; 13:1593241. [PMID: 40417179 PMCID: PMC12098596 DOI: 10.3389/fcell.2025.1593241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/28/2025] [Indexed: 05/27/2025] Open
Abstract
MiR-425 is a biological molecule that has potential applications in cardiovascular diseases. It can regulate biological functions by combining with LncRNAs, binding with proteins, and changing the differentiation of immune cells. MiR-425 also has a role as a biomarker of disease. In cardiovascular diseases, it has clinical significance in reducing inflammation and heart repair, inducing angiogenesis, improving the prediction of atherosclerosis, reducing cardiac fibrosis, and regulating atrial natriuretic peptide to affect cardiovascular function. Target gene prediction and KEGG enrichment analysis are also mentioned.
Collapse
Affiliation(s)
- Shan Zhou
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong first Medical University, Jinan, Shandong, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong first Medical University, Jinan, Shandong, China
- The Laboratory of Medical Science and Technology Innovation Center (Institute of Biomedical Engineering and Interdisciplinary Studies), Shandong First Medical University, jinan, China
| |
Collapse
|
181
|
Venturi S, Rendine M, Marino M, Klimis-Zacas D, Riso P, Del Bo' C. Differential Effects of Wild Blueberry (Poly)Phenol Metabolites in Modulating Lipid Metabolism and Oxidative Stress in 3T3-L1 Adipocytes. Mol Nutr Food Res 2025:e70101. [PMID: 40345987 DOI: 10.1002/mnfr.70101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/01/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Adipocyte hypertrophy, driven by lipid accumulation, is crucial in the development of obesity. Wild blueberry (WB; Vaccinium angustifolium) (poly)phenols (PPs) metabolites may modulate adipogenesis and the development of obesity. This study examines WB PP metabolites' effects on lipid accumulation, lipid metabolism, and oxidative stress in mature 3T3-L1 adipocytes. Differentiated 3T3-L1 adipocytes were treated for 48 h with free fatty acids (FFAs; oleic/palmitic acid 750 µM, 2:1 ratio) and WB-derived PPs, including ferulic acid (FA), isoferulic acid (IA), vanillic acid (VA), and syringic acid (SA) at physiological and supra-physiological concentrations. Assessments included lipid accumulation, glycerol release, and markers of lipid metabolism (sterol regulatory element-binding protein 1c [SREBP-1], fatty acid synthase [FASN], FAB4) and oxidative stress (DNA damage, 8-hydroxy 2-deoxyguanosine [8OHdG], nuclear erythroid factor 2-related factors 2 (NRF2), heme oxygenase 1 [HO-1]). FFAs significantly increased lipid accumulation, glycerol release, and FASN levels, while reducing HO-1 levels, without affecting other markers. WB PP metabolites did not reduce lipid accumulation, but IA and VA reduced FASN levels (-25% and -26%; p < 0.05), and SA improved HO-1 levels (+150%; p < 0.05). Despite the different effects observed, the findings obtained under our experimental conditions seem to suggest that IA, VA, and SA may modulate lipid metabolism and oxidative stress markers. However, further studies are fundamental to corroborate the findings obtained and support the contribution of these BB PPs metabolites and other compounds in the prevention and management of obesity.
Collapse
Affiliation(s)
- Samuele Venturi
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Marco Rendine
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Mirko Marino
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | | | - Patrizia Riso
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| | - Cristian Del Bo'
- Division of Human Nutrition, Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
182
|
Pasut A, Lama E, Van Craenenbroeck AH, Kroon J, Carmeliet P. Endothelial cell metabolism in cardiovascular physiology and disease. Nat Rev Cardiol 2025:10.1038/s41569-025-01162-x. [PMID: 40346347 DOI: 10.1038/s41569-025-01162-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2025] [Indexed: 05/11/2025]
Abstract
Endothelial cells are multifunctional cells that form the inner layer of blood vessels and have a crucial role in vasoreactivity, angiogenesis, immunomodulation, nutrient uptake and coagulation. Endothelial cells have unique metabolism and are metabolically heterogeneous. The microenvironment and metabolism of endothelial cells contribute to endothelial cell heterogeneity and metabolic specialization. Endothelial cell dysfunction is an early event in the development of several cardiovascular diseases and has been shown, at least to some extent, to be driven by metabolic changes preceding the manifestation of clinical symptoms. Diabetes mellitus, hypertension, obesity and chronic kidney disease are all risk factors for cardiovascular disease. Changes in endothelial cell metabolism induced by these cardiometabolic stressors accelerate the accumulation of dysfunctional endothelial cells in tissues and the development of cardiovascular disease. In this Review, we discuss the diversity of metabolic programmes that control endothelial cell function in the cardiovascular system and how these metabolic programmes are perturbed in different cardiovascular diseases in a disease-specific manner. Finally, we discuss the potential and challenges of targeting endothelial cell metabolism for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Alessandra Pasut
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Eleonora Lama
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Amaryllis H Van Craenenbroeck
- Division of Nephrology, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Nephrology and Renal Transplantation Research Group, KU Leuven, Leuven, Belgium
| | - Jeffrey Kroon
- Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, The Netherlands.
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven, Belgium.
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
183
|
Li X, Sun YX, Tjahjono AW, Wei Y, Li X, Zheng QH, Qi WC, Liang FR. Acupuncture attenuates myocardial ischemia/reperfusion injury-induced ferroptosis via the Nrf2/HO-1 pathway. Chin Med 2025; 20:61. [PMID: 40346679 PMCID: PMC12065278 DOI: 10.1186/s13020-025-01114-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/27/2025] [Indexed: 05/11/2025] Open
Abstract
AIMS To observe the effect of electro-acupuncture (EA) on cardiomyocytes ferroptosis induced by myocardial ischemia/reperfusion injury (MIRI) in mice and to investigate whether this effect occurs via the nuclear factor-E2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) signalling pathway. MATERIALS AND METHODS Firstly, Fe2+ in the hearts and serum of mice from both the sham-operated (SO) group and MIRI group was measured to ascertain whether ferroptosis had occurred in the cardiomyocytes of mice in MIRI group. In the second phase, EA was administered, with sham acupuncture (SA) group as the comparator, to investigate the protective effects of EA on ferroptosis in MIRI cardiomyocytes and cardiac function. Additionally, we studied the levels of Nrf2 and HO-1 within the myocardium. In the third phase, Nrf2 inhibitor ML385 and agonist DMF were applied to observe the impact of inhibiting Nrf2 on the therapeutic efficacy of EA. RESULTS Compared with SO group, MIRI group showed increased iron deposition, along with a significant decrease in Nrf2 and HO-1 levels. Compared with MIRI group, MIRI + EA group exhibited significantly improved cardiac function and reduced cardiac iron deposition, accompanied by increased Nrf2 and HO-1 levels. Furthermore, the therapeutic effect of MIRI + EA group was superior to that of MIRI + SA group. Administration of ML385 partially blocked the anti-ferroptotic and cardioprotective effects of EA, while EA treatment exhibited similar effects to dimethyl fumarate (DMF) intervention. CONCLUSION EA alleviates ferroptosis-induced damage in MIRI in mice via the Nrf2/HO-1 pathway, providing modern scientific evidence for the application of acupuncture in the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao Li
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Yu-Xin Sun
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Adi Wirawan Tjahjono
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Ying Wei
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
| | - Xiang Li
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
- Sichuan Provincial Key Laboratory of Acupuncture and Chronobiology, Chengdu, 610075, People's Republic of China
| | - Qian-Hua Zheng
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China
- Sichuan Provincial Key Laboratory of Acupuncture and Chronobiology, Chengdu, 610075, People's Republic of China
| | - Wen-Chuan Qi
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China.
- Sichuan Provincial Key Laboratory of Acupuncture and Chronobiology, Chengdu, 610075, People's Republic of China.
| | - Fan-Rong Liang
- School of Acu-Mox and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, People's Republic of China.
- Sichuan Provincial Key Laboratory of Acupuncture and Chronobiology, Chengdu, 610075, People's Republic of China.
| |
Collapse
|
184
|
Bin H, Wen W. Metformin attenuates endoplasmic reticulum stress in diabetic kidney disease: mechanistic insights and future perspectives. Int Urol Nephrol 2025:10.1007/s11255-025-04562-7. [PMID: 40343634 DOI: 10.1007/s11255-025-04562-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Diabetic kidney disease (DKD) is a common microvascular complication of diabetes that can lead to end-stage renal failure. Emerging evidence suggests that endoplasmic reticulum (ER) stress plays a crucial role in the pathogenesis of DKD by affecting various renal parenchymal cells, including endothelial cells, podocytes, and mesangial cells. This review comprehensively examines the relationship between ER stress and DKD, focusing on how metformin, a first-line antidiabetic medication, ameliorates ER stress-induced kidney injury. Multiple factors, including reactive oxygen species (ROS), proteinuria, and advanced glycation end products (AGEs), contribute to ER stress in DKD. Metformin's renoprotective effects are primarily mediated through activation of the AMPK signaling pathway, which modulates ER stress response, reduction of oxidative stress and its impact on ER function, and improvement of mitochondrial function. These mechanisms collectively lead to decreased proteinuria, reduced cell apoptosis, and attenuated epithelial-mesenchymal transition in diabetic kidneys. Understanding these molecular mechanisms provides new insights into the therapeutic potential of metformin in DKD treatment. However, further research is needed to elucidate the precise molecular pathways through which metformin regulates ER stress in different renal cell types under diabetic conditions.
Collapse
Affiliation(s)
- Huang Bin
- Department of Endocrinology, Division of Life Science and Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, 230000, China
| | - Wenjie Wen
- Anhui Province Engineering Research Center for Dental Materials and Application, School of Stomatology, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
185
|
Albrecht E, Pelz K, Gress A, Trung HN, Kalinina OV, Kacprowski T, Baumbach J, List M, Tsoy O. DIGGER 2.0: digging into the functional impact of differential splicing on human and mouse disorders. Nucleic Acids Res 2025:gkaf384. [PMID: 40337913 DOI: 10.1093/nar/gkaf384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/11/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025] Open
Abstract
Changes in alternative splicing between groups or conditions contribute to protein-protein interaction rewiring, a consequence often neglected in data analysis. The web server and database DIGGER overcomes this limitation by augmenting a protein-protein interaction network with domain-domain interactions and splicing information. Here, we present DIGGER 2.0, which now features both experimental and newly added predicted domain-domain interactions. In addition to the human interactome, DIGGER 2.0 adds support for mouse as an important model organism. Additionally, we integrated the splicing analysis tool NEASE, which allows users to perform online splicing- and interactome-informed enrichment analysis on RNA-seq data. In two application cases (multiple sclerosis and mice models of cardiac diseases), we show the utility of DIGGER 2.0 for deeper exploration and functional interpretation of changes in alternative splicing in human and mouse disorders. DIGGER 2.0 is available at https://exbio.wzw.tum.de/digger/.
Collapse
Affiliation(s)
- Elias Albrecht
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany
| | - Konstantin Pelz
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
| | - Alexander Gress
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Graduate School of Computer Science, Saarland University, Campus E1.3, 66123 Saarbrücken, Germany
| | - Hieu Nguyen Trung
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
| | - Olga V Kalinina
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany
- Drug Bioinformatics, Medical Faculty, Saarland University, Gebäude 15, 66421 Homburg, Germany
- Center for Bioinformatics, Saarland University, Campus E2.1, 66123 Saarbrücken, Germany
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of Technische Universität Braunschweig and Hannover Medical School, Rebenring 56 Lower Saxony, 38106 Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56 Lower Saxony, 38106 Braunschweig, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany
- Institute of Mathematics and Computer Science, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Markus List
- Data Science in Systems Biology, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 3, 85354 Freising, Germany
- Munich Data Science Institute (MDSI), Technical University of Munich, Walther-von-Dyck-Straße 10, 85748 Garching, Germany
| | - Olga Tsoy
- Institute for Computational Systems Biology, University of Hamburg, Albert-Einstein-Ring 8-10, 22761 Hamburg, Germany
| |
Collapse
|
186
|
Li Y, Guo T, He J, Liu D, Peng S, Xu A. SLC35A2-mediated bisected GlcNAc-modified extracellular vesicles enhance immune regulation in breast cancer lung metastasis. Int Immunopharmacol 2025; 154:114505. [PMID: 40157085 DOI: 10.1016/j.intimp.2025.114505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
This study investigates the role of SLC35A2-mediated bisected GlcNAc-modified small extracellular vesicles (sEVs) in breast cancer (BC) lung metastasis. By modulating B3GALT1 expression, these sEVs regulate the pre-metastatic immune microenvironment, enhancing CD8+ T cell infiltration and reducing immune evasion. The use of β-peptide-loaded sEVs further amplifies anti-metastatic effects, as demonstrated in vivo mouse models and molecular analyses. These findings underscore the therapeutic potential of glycosylation-modified sEVs in enhancing immune responses and controlling BC metastasis.
Collapse
Affiliation(s)
- Yangyang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Tao Guo
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Juntong He
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Defeng Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Shihao Peng
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Aman Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
187
|
Wu Z, Jiang S, Li J, Wang P, Chen Y. Association between urinary cadmium levels and increased gallstone disease in US adults. Sci Rep 2025; 15:15974. [PMID: 40341372 PMCID: PMC12062283 DOI: 10.1038/s41598-025-00648-5] [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: 09/20/2024] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Heavy metal exposure is acknowledged as a risk factor for poor health. However, the effect of heavy metal exposure on the prevalence of gallstones is still unknown. Therefore, we investigated the relationship between heavy metal concentrations and the prevalence of gallstones among US adults. Multivariate logistic regression indicated that only urinary cadmium was an independent risk factor for gallstones. Compared to the low urine cadmium group, the high cadmium group had a elevated increased risk of gallstone formation. Furthermore, the weighted quantile sum model showed that heavy metal mixtures were not associated with gallstone prevalence. Additionally, urinary cadmium levels were associated with an increased risk of gallstone formation in young individuals, males, Mexican Americans, Non-Hispanic Whites, as well as smokers and drinkers. Moreover, nine machine learning methods were utilized to construct an interpretable predictive model for gallstone prevalence. Among these models, the XGBoost model exhibited the highest performance and was selected for further investigation. Subsequently, shapely additive explanations was used for model interpretation. The results also indicated that urinary cadmium concentrations were the most important variable for gallstones. Thus, our results indicated that long-term chronic cadmium exposure is a risk factor for gallstone prevalence.
Collapse
Affiliation(s)
- Zhaowei Wu
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China
| | - Shiming Jiang
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China
| | - Jinzhi Li
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China
| | - Panguo Wang
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China
| | - Yong Chen
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, Chongqing, 404100, China.
| |
Collapse
|
188
|
Anzic N, Stoiber W, Obermeyer A, Mertz KD, Stalder A, Haslbauer JD, Tzankov A. Two-sided effects of neutrophil extracellular traps and changes in the myeloid compartment in acute COVID-19: A histopathological study on autopsy cases from the first and second COVID-19 waves in Switzerland. J Leukoc Biol 2025; 117:qiaf056. [PMID: 40356380 DOI: 10.1093/jleuko/qiaf056] [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: 01/13/2025] [Revised: 03/05/2025] [Accepted: 05/12/2025] [Indexed: 05/15/2025] Open
Abstract
Severe COVID-19 is characterized by complex immunopathology that involves inflammation, endothelial dysfunction, and immunothrombosis. Neutrophil extracellular traps (NETs) have been recognized as key factors in the severity of the disease, with their emergence correlating to viral load, immmunothrombosis, and organ damage. In this study, we investigated the role of NETosis and macrophage activation in the course of severe COVID-19. We analyzed 23 autopsy samples from patients who died from COVID-19 and performed immunohistochemical staining and stereological point counting to quantify leukocyte infiltration and NET formation among other histopathological parameters. Our results showcase 2 evident immunophenotypes: lowNET and highNET. The lowNET group displayed lower NET formation, higher viral loads, and an increased incidence of secondary infections, as well as shorter survival times. In contrast, the highNET group exhibited increased neutrophil activation, pronounced endothelial damage and thrombotic complications, as well as prolonged survival times. Our data suggest a dual role of NETosis in COVID-19: initially protective, limiting viral replication, but later likely detrimental through immunothrombosis and tissue damage. These findings underline the need for tailored therapeutic actions, with early antiviral and immune-modulating interventions for lowNET patients and strategies aiming to limit excessive NETosis and coagulopathy in highNET patients. Further research is needed to define the timing of interventions based on the dynamics of NETosis.
Collapse
Affiliation(s)
- Nina Anzic
- Institute of Pathology, Cantonal Hospital Luzern, Spitalstrasse 16, 6000 Luzern, Switzerland
| | - Walter Stoiber
- Department of Environment and Biodiversity, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Astrid Obermeyer
- Department of Environment and Biodiversity, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | - Kirsten D Mertz
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Anna Stalder
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Jasmin D Haslbauer
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital Basel, Schönbeinstrasse 40, 4031 Basel, Switzerland
| |
Collapse
|
189
|
Batista MP, Pimenta M, Fernández N, Duarte ARC, Bronze MDR, Marto J, Gaspar FB. Collagen-Chitosan Composites Enhanced with Hydroxytyrosol for Prospective Wound Healing Uses. Pharmaceutics 2025; 17:618. [PMID: 40430909 PMCID: PMC12114697 DOI: 10.3390/pharmaceutics17050618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Recent studies highlight the excellent wound-healing properties of collagen and chitosan materials. Combining these polymers with a bioactive compound could enhance their effectiveness as next-generation wound dressings. Hydroxytyrosol (HT), an antioxidant derived from olive oil, may aid wound healing due to its anti-inflammatory, antimicrobial, and angiogenesis-stimulating properties, making it a beneficial addition to collagen-chitosan dressings. It could be a beneficial addition to collagen-chitosan dressings, thus improving their therapeutic effects. This study screens the potential of collagen-chitosan composites with HT for wound-healing applications and assesses the influence of the compound's incorporation on the materials' properties. Methods: The material production involved incorporating chitosan and HT into a marine collagen extract. The resulting collagen-chitosan-HT material was obtained through freeze-drying. Prototype dressing characterization included morphology by scanning electron microscopy, solid and hydrated state by textural and rheological studies, and in vitro HT release studies. The materials' cytocompatibility screening was assessed using a mouse fibroblast cell line, and the antibacterial activity was evaluated against microorganisms commonly implicated in wound infections. Results: Burst strength, viscosity, frequency sweep test, tackiness, and adhesion results indicate that chitosan contributes to the material's mechanical robustness by maintaining a high viscosity and preserving the material's gel structure. The in vitro release studies suggest an HT-controlled release profile with a maximum release (70%) achieved after 10 h. Biological experiments confirmed the materials' cytocompatibility with skin cells and very promising antibacterial efficacy against Staphylococcus aureus and Pseudomonas aeruginosa. Conclusions: In conclusion, HT was successfully incorporated into a collagen-chitosan matrix, enhancing the therapeutic prospect of the resultant material. The collagen-chitosan-HT composite presents a promising potential as an advanced wound-healing material.
Collapse
Affiliation(s)
- Miguel P. Batista
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.P.B.); (M.P.); (N.F.); (M.d.R.B.)
- LAQV-REQUIMTE, Associated Laboratory for Green Chemistry—Network of Chemical and Technology, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Quinta da Torre, 2829-516 Caparica, Portugal;
| | - Margarida Pimenta
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.P.B.); (M.P.); (N.F.); (M.d.R.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Naiara Fernández
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.P.B.); (M.P.); (N.F.); (M.d.R.B.)
| | - Ana Rita C. Duarte
- LAQV-REQUIMTE, Associated Laboratory for Green Chemistry—Network of Chemical and Technology, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Quinta da Torre, 2829-516 Caparica, Portugal;
| | - Maria do Rosário Bronze
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.P.B.); (M.P.); (N.F.); (M.d.R.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- FFULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. das Forças Armadas, 1649-019 Lisboa, Portugal
| | - Joana Marto
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. das Forças Armadas, 1649-003 Lisboa, Portugal
| | - Frédéric Bustos Gaspar
- iBET—Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.P.B.); (M.P.); (N.F.); (M.d.R.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
190
|
Xiong Z, Liao Y, Zhang Z, Wan Z, Liang S, Guo J. Molecular Insights into Oxidative-Stress-Mediated Cardiomyopathy and Potential Therapeutic Strategies. Biomolecules 2025; 15:670. [PMID: 40427563 PMCID: PMC12108637 DOI: 10.3390/biom15050670] [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: 03/27/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiomyopathies comprise a heterogeneous group of cardiac disorders characterized by structural and functional abnormalities in the absence of significant coronary artery disease, hypertension, valvular disease, or congenital defects. Major subtypes include hypertrophic, dilated, arrhythmogenic, and stress-induced cardiomyopathies. Oxidative stress (OS), resulting from an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, has emerged as a key contributor to the pathogenesis of these conditions. ROS-mediated injury drives inflammation, protease activation, mitochondrial dysfunction, and cardiomyocyte damage, thereby promoting cardiac remodeling and functional decline. Although numerous studies implicate OS in cardiomyopathy progression, the precise molecular mechanisms remain incompletely defined. This review provides an updated synthesis of current findings on OS-related signaling pathways across cardiomyopathy subtypes, emphasizing emerging therapeutic targets within redox-regulatory networks. A deeper understanding of these mechanisms may guide the development of targeted antioxidant strategies to improve clinical outcomes in affected patients.
Collapse
Affiliation(s)
- Zhenyu Xiong
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Yuanpeng Liao
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhaoshan Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhengdong Wan
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
| | - Sijia Liang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiawei Guo
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
191
|
Huang X, Lu X, Wu Y, Wu Z, Li M, Miao Y, Xie Z, Gong Z, Cao Y. L-theanine prevents myocardial injury in sleep‑deprived mice by suppressing ferroptosis through SIRT1. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04206-8. [PMID: 40328913 DOI: 10.1007/s00210-025-04206-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025]
Abstract
Green tea is obtained from Camellia sinensis, and its role in promoting heart health has been widely recognized in traditional Chinese medicine. L-theanine, one of the main bioactive components of green tea, has antioxidant and cardiovascular protective effects. However, the effects of L-theanine on myocardial cells in sleep-deprived mice and its potential mechanisms have not been clearly elucidated. This study utilized a modified multiple-platform water environment method to establish an SD model and induce a ferroptosis model in H9c2 cells pretreated with erastin. The cardiac function parameters of mice were assessed using a small animal super-resolution ultrasound imaging system. H&E staining was used to evaluate pathological changes in tissue structure and cell morphology, while transmission electron microscopy (TEM) was employed to observe the extent of mitochondrial damage. Biochemical assays were employed to quantify myocardial damage markers, oxidative stress indicators, and Fe2⁺ concentrations, while immunofluorescence imaging assessed reactive oxygen species (ROS) levels. Western blot was used to analyze the expression of SIRT1 and proteins related to ferroptosis. The results demonstrate that L-theanine alleviates SD-induced tachycardia in mice, restores myocardial and mitochondrial integrity, and reduces oxidative damage markers, including ROS and Fe2⁺ in H9c2 cells. Furthermore, L-theanine reversed the abnormal expression of SIRT1 and ferroptosis-related proteins in cardiac tissue and H9c2 cells induced by SD and erastin. Notably, the SIRT1 inhibitor EX-527 can counteract the protective effect of L-theanine against ferroptosis in cardiomyocytes. These findings highlight that L-theanine mitigates SD-induced cardiac injury primarily by suppressing ferroptosis through SIRT1 in cardiomyocytes.
Collapse
Affiliation(s)
- Xuanxuan Huang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China
| | - Xinglong Lu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China
| | - Yi Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China
| | - Zhenghua Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China
| | - Mingrui Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China
| | - Yuping Miao
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, China.
| | - Zipeng Gong
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guian New Area 561113, 0851-86908468, 4 Beijing Road, Guiyang City, 550004, Guizhou Province, China.
| | - Yin Cao
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, 350 Longzihu Road, Xinzhan Area, Hefei, 230012, China.
- State Key Laboratory of Tea Plant Biology and Utilization, Anhui Agricultural University, 130 Changjiang West Road, Hefei, 230036, China.
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guian New Area 561113, 0851-86908468, 4 Beijing Road, Guiyang City, 550004, Guizhou Province, China.
| |
Collapse
|
192
|
Ferrara EF, Roik A, Wöhrmann-Zipf F, Ziegler M. Ex Situ Thermal Preconditioning Modulates Coral Physiology and Enhances Heat Tolerance: A Multispecies Perspective for Active Restoration. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:8527-8540. [PMID: 40279456 PMCID: PMC12060272 DOI: 10.1021/acs.est.4c08640] [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] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/27/2025]
Abstract
Global warming threatens reef-building corals by challenging their adaptive capacity. Therefore, interventions such as stress-hardening by thermal preconditioning could become crucial for their survival. This study aimed to systematically assess the effects of distinct thermal preconditioning regimes (stable-high at 29 °C, variable-high at 29 ± 1.5 °C, and stable-ambient control at 26 °C) on the baseline physiology and thermal tolerance of six stony coral species (Galaxea fascicularis, Porites rus, Acropora muricata, Montipora digitata, Pocillopora verrucosa, and Stylophora pistillata) to determine commonalities in the stress-hardening responses that transcend species-specific signatures. For this, we quantified changes in photosynthetic efficiency and bleaching intensity before and after a short-term heat stress assay and up to 30 days later. Stress-hardening was successful in all preconditioned corals, with the variable-high regime slightly outperforming the stable-high regime. Preconditioning reduced the heat stress response by up to 90%, yet species differed in receptiveness. It also improved resilience (survival and recovery), and corals with high inherent thermal tolerance recovered better than susceptible species. Notably, both preconditioning regimes affected baseline physiology, exclusively of the branching species, causing tissue paling and decreased photosynthetic efficiency. We conclude that implementing thermal stress-hardening protocols requires consideration of the species-specific receptiveness and potential physiological trade-offs.
Collapse
Affiliation(s)
- Erik F. Ferrara
- Marine
Holobiomics Lab, Department of Animal Ecology and Systematics, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32 IFZ, 35392 Giessen, Germany
| | - Anna Roik
- Helmholtz
Institute for Functional Marine Biodiversity at the University of
Oldenburg (HIFMB), 26129 Oldenburg, Germany
- Alfred
Wegener Institute, Helmholtz Center for Polar and Marine Research, 27570 Bremerhaven, Germany
| | - Franziska Wöhrmann-Zipf
- Marine
Holobiomics Lab, Department of Animal Ecology and Systematics, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32 IFZ, 35392 Giessen, Germany
| | - Maren Ziegler
- Marine
Holobiomics Lab, Department of Animal Ecology and Systematics, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32 IFZ, 35392 Giessen, Germany
| |
Collapse
|
193
|
McMullan A, Zwierzynski JB, Jain N, Haneline LS, Shou W, Kua KL, Hota SK, Durbin MD. Role of Maternal Obesity in Offspring Cardiovascular Development and Congenital Heart Defects. J Am Heart Assoc 2025; 14:e039684. [PMID: 40314345 DOI: 10.1161/jaha.124.039684] [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: 10/24/2024] [Accepted: 03/21/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Congenital heart disease is a leading cause of death in newborns, yet many of its molecular mechanisms remain unknown. Both maternal obesity and diabetes increase the risk of congenital heart disease in offspring, with recent studies suggesting these conditions may have distinct teratogenic mechanisms. The global prevalence of obesity is rising, and while maternal obesity is a known risk factor for fetal congenital heart disease, the specific mechanisms are largely unexplored. METHODS AND RESULTS We used a murine model of diet-induced maternal obesity, without diabetes, to produce dams that were overweight but had normal blood glucose levels. Embryos were generated and their developing hearts analyzed. Transcriptome analysis was performed using single-nucleus and bulk RNA sequencing. Global and phospho-enriched proteome analysis was performed using tandem mass tag-mass spectroscopy. Immunobloting and histologic evaluation were also performed. Analysis revealed disrupted oxidative phosphorylation and reactive oxygen species formation, with reduced antioxidant capacity, evidenced by downregulation of genes Sod1 and Gp4x, and disrupted Hif1a signaling. Evidence of oxidative stress, cell death signaling, and alteration in Rho GTPase and actin cytoskeleton signaling was also observed. Genes involved in cardiac morphogenesis, including Hand2, were downregulated, and fewer mature cardiomyocytes were present. Histologic analysis confirmed increased cardiac defects in embryos exposed to maternal obesity. CONCLUSIONS These findings demonstrate that maternal obesity alone can result in cardiac defects through mechanisms similar to those associated with maternal hyperglycemia. This study provides valuable insight into the role of maternal obesity, a growing and modifiable risk factor, in the development of the most common birth defect, congenital heart disease.
Collapse
Affiliation(s)
- Ashleigh McMullan
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | | | - Nina Jain
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | - Laura S Haneline
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | - Weinian Shou
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | - Kok Lim Kua
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
- Center for Diabetes and Metabolic Disease Research Indiana University School of Medicine Indianapolis IN USA
| | - Swetansu K Hota
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| | - Matthew D Durbin
- Department of Pediatrics Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis IN USA
| |
Collapse
|
194
|
Zhang S, Zhao R, Wang R, Lu Y, Xu M, Lin X, Lan R, Zhang S, Tang H, Fan Q, Yang J, Liu L, Xu J. Weissella viridescens Attenuates Hepatic Injury, Oxidative Stress, and Inflammation in a Rat Model of High-Fat Diet-Induced MASLD. Nutrients 2025; 17:1585. [PMID: 40362894 PMCID: PMC12073722 DOI: 10.3390/nu17091585] [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: 04/17/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Metabolic-dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disorder globally. Probiotic supplementation has shown promise in its prevention and treatment. Although Weissella viridescens, a lactic acid bacterium with immunomodulatory effects, has antibacterial and anti-inflammatory activities, there is a lack of direct evidence for its role in alleviating MASLD. This study aimed to investigate the protective effects of W. viridescens strain Wv2365, isolated from healthy human feces, in a high-fat diet (HFD)-induced rat model of MASLD. Methods: Rats were randomly assigned to a normal chow diet (NC), high-fat diet (HFD), and HFD supplemented with W. viridescens Wv2365 (Wv2365) groups. All groups were fed their respective diets for 8 weeks. During this period, the NC and HFD groups received a daily oral gavage of PBS, while the Wv2365 group received a daily oral gavage of Wv2365. Results: Wv2365 supplementation significantly reduced HFD-induced body weight gain, improved NAFLD activity scores, alleviated hepatic injury, and restored lipid metabolism. A liver transcriptomic analysis revealed the downregulation of inflammation-related pathways, along with decreased serum levels of TNF-α, IL-1β, IL-6, MCP-1, and LPS. Wv2365 also activated the Nrf2/HO-1 antioxidant pathway, enhanced hepatic antioxidant enzyme activities and reduced malondialdehyde levels. A gut microbiota analysis showed the enrichment of beneficial genera, including Butyricicoccus, Akkermansia, and Blautia. Serum metabolomic profiling revealed increased levels of metabolites including indole-3-propionic acid, indoleacrylic acid, and glycolithocholic acid. Conclusions: Wv2365 attenuates hepatic injury, oxidative stress, and inflammation in a rat model of high-fat-diet-induced MASLD, supporting its potential as a probiotic candidate for the modulation of MASLD.
Collapse
Affiliation(s)
- Shuwei Zhang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruiqing Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruoshi Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Yao Lu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Mingchao Xu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, Shijiazhuang 050010, China
| | - Xiaoying Lin
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ruiting Lan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Suping Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Huijing Tang
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qianhua Fan
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Jing Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Liyun Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| | - Jianguo Xu
- School of Public Health, Nanjing Medical University, Nanjing 211166, China; (S.Z.)
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Research Units of Discovery of Unknown Bacteria and Function, Chinese Academy of Medical Sciences, Beijing 102206, China
- Hebei Key Laboratory of Intractable Pathogens, Shijiazhuang Center for Disease Control and Prevention, Shijiazhuang 050011, China
| |
Collapse
|
195
|
Olas B. The Cardioprotective Effect of Magnolia officinalis and Its Major Bioactive Chemical Constituents. Int J Mol Sci 2025; 26:4380. [PMID: 40362616 PMCID: PMC12072210 DOI: 10.3390/ijms26094380] [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: 04/01/2025] [Revised: 04/30/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
The genus Magnolia has been found to exhibit different biological properties, including antioxidant, anticancer, and others. For example, Magnolia officinalis is a classical traditional herb used in various Asian countries, especially China, South Korea, and Japan. Magnolia bark is the main medicinal part of this plant. This paper reviews the current state of knowledge regarding the M. officinalis bark and its active constituents, especially magnolol and honokiol, with a special emphasis on their cardioprotective activity in various models. This review paper also sheds new light on the cardioprotective mechanisms of magnolol and honokiol. However, their cardioprotective potential is limited to animal in vivo models and in vitro models. Only a single study has examined the cardiovascular properties of M. officinalis extract in obese mice. In addition, there is no clinical evidence for the absorption and bioavailability of M. officinalis extracts and their main bioactive compounds in humans. Moreover, there are no studies simultaneously comparing the activity of magnolol and honokiol. Therefore, there is a need for such studies. There are also no recommendations regarding their effective or safe doses for prophylaxis and the treatment of CVDs.
Collapse
Affiliation(s)
- Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
196
|
Wang X, Song SM, Lu WQ, Zhao Y, Lv RJ, He Y, Dong N, Yu Q, Yue HM. Alpha-lipoic acid alleviated intermittent hypoxia-induced myocardial injury in mice by promoting autophagy through Nrf2 signaling pathway. Eur J Pharmacol 2025; 994:177380. [PMID: 39954840 DOI: 10.1016/j.ejphar.2025.177380] [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: 10/11/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Obstructive sleep apnea syndrome (OSAS) is a prevalent sleep-related breathing disorder characterized by intermittent hypoxia (IH). Myocardial injury is a common complication associated with OSAS. Alpha-lipoic acid (LA), a potent antioxidant, has been utilized in various disease contexts and has demonstrated significant protective effects in myocardial infarction models. Given the limited treatment options available for OSAS-related myocardial injury, this study aimed to demonstrate the potential therapeutic effects of LA and to investigate the underlying mechanisms. IH is a widely employed method to simulate the pathophysiological conditions associated with OSAS. In vivo experiments were conducted using mice placed in a specialized hypoxic chamber to replicate IH conditions. Echocardiography indicated that exposure to IH severely impaired cardiac function. Treatment with LA activated the Nrf2 pathway and autophagy, which contributed to the improvement of cardiac function in mice with OSAS. Additionally, in vitro studies demonstrated that IH induced apoptosis and decreased cell viability in H9C2 cardiomyocytes. LA enhanced Nrf2 nuclear translocation and its downstream signaling pathways, thereby promoting autophagy, inhibiting apoptosis, and alleviating injury in H9C2 cardiomyocytes. Furthermore, in vitro inhibition of Nrf2 using ML385 reduced autophagy levels and attenuated the protective effects of LA against apoptosis in H9C2 cardiomyocytes. These findings suggest that LA may provide a promising therapeutic strategy for myocardial injury associated with OSAS. By elucidating these findings, new insights into the protective mechanisms of LA against IH-induced myocardial injury are provided, highlighting its potential as a therapeutic agent for diseases associated with OSAS.
Collapse
Affiliation(s)
- Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Shao-Ming Song
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Wen-Qiang Lu
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yan Zhao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Ren-Jun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China; Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Qin Yu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China; Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Hong-Mei Yue
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China; Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
197
|
Liu D, Zhao Y, Dai J, Li R, Yuan J, Shen K, He Z, Gu S. Changes of N6-methyladenosine and ferroptosis in cadmium-induced reproductive toxicity of male mice fed a high fat and high sugar diet. Toxicology 2025; 516:154172. [PMID: 40334773 DOI: 10.1016/j.tox.2025.154172] [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: 01/10/2025] [Revised: 05/02/2025] [Accepted: 05/03/2025] [Indexed: 05/09/2025]
Abstract
Cadmium (Cd) and high-fat and high-sugar diet (HFHS) are risk factors contributing to the decline in male sperm quality. N6-methyladenosine (m6A) is essential in the processes of testicular development and spermatogenesis. Ferroptosis, a form of cell death that depends on iron, is prone to causing testicular dysfunction. However, the changes in m6A modification regulatory proteins and ferroptosis signaling molecules in male mice with sperm abnormalities resulting from combined exposure to Cd and HFHS remain incompletely elucidate. Our present data indicate that the combined treatment of Cd and HFHS significantly reduced sperm quality in comparison to those in single Cd or HFHS treatment. In addition, indicators related to ferroptosis in the combined treatment of Cd and HFHS group have also undergone significant changes. In detail, the contents of malondialdehyde (MDA) and Fe2+ as well as Slc7a11 expression were increased while Gclc expressions were reduced in the testicular tissue of Cd and HFHS combined treatment mice. Further detect results showed that the combined exposure to Cd and HFHS synergistically elevated the m6A modification levels alongside a downregulation of the Mettl3, Fto, Alkbh5 and Ythdc2 at the protein level when compared with those in single Cd or HFHS treatment. Altogether, it can be inferred that Cd and HFHS combined treatment may alter the levels of m6A modification regulatory proteins in testicular tissue, leading to increased Fe2+ and MDA production, thus activating the Slc7a11/Gpx4 signaling pathway, ultimately decreasing the sperm quality in mice, providing preliminary evidence for the occurrence of ferroptosis in testicular cells. Our findings may provide direction for the study of reproductive toxicity of cadmium and offer reference for the selection of molecular targets.
Collapse
Affiliation(s)
- De Liu
- School of Public Health, Dali University, Dali, Yunnan, China
| | - Yuan Zhao
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Jiao Dai
- Qujing Medical College, Qujing, Yunnan, China
| | - Rongxian Li
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Jiamin Yuan
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Kaiyan Shen
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Zuoshun He
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China
| | - Shiyan Gu
- School of Public Health, Dali University, Dali, Yunnan, China; Institute of Preventive Medicine, Dali University, Dali, Yunnan, China.
| |
Collapse
|
198
|
Njale E, Shilla DJ, Dharsee N, Mahugija JAM, Mbare NS. Association between single and mixed exposure to potentially toxic trace metals and the risk of prostate cancer: a case-control study in Tanzania. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2025; 47:194. [PMID: 40316833 DOI: 10.1007/s10653-025-02497-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/03/2025] [Indexed: 05/04/2025]
Abstract
Metal contamination is a major environmental concern in Tanzania, where it has been linked to an increased risk of prostate cancer. However, there have been no epidemiological studies addressing this association. The aim of this study was to investigate the association between urinary toxic trace metals and prostate cancer. Inductively coupled plasma atomic emission spectrophotometer (ICP-AES) was employed to measure the concentrations of Pb, As, Ni, Al, and Cd in urine samples from histologically confirmed100 prostate cancer patients (n = 100) and 80 healthy controls (n = 80). The associations between individual metals and prostate cancer were assessed using unconditional logistic regression, while Bayesian kernel machine regression (BKMR) was employed to investigate the combined effects of multiple metals with adjustments of potential covariates. Cancer patients had significantly higher mean levels of Ni, Pb, and As in their urine compared to controls. In multivariable logistic models, the findings suggested that quartiles increase of As and Cd were positively associated with prostate cancer with ORs of 5.25 (1.33, 20.72) in Q3 for As and ORs of 2.87 (1.72, 11.52) in Q4 for Cd. The BKMR results revealed that the combined effect of five urinary metals exhibited a negative association with prostate cancer risk. In conclusion, this study offers preliminary evidence suggesting that exposure to trace metals particularly Cd and As may potentially be associated with prostate cancer. Pb and Al were found to have an inverse relationship with prostate cancer and overall metal mixture had no impact on prostate cancer. Since the study was preliminary, these results remain to be confirmed by further large-scale studies.
Collapse
Affiliation(s)
- Emmanuel Njale
- Department of Chemistry, University of Dar es Salaam, P.O. Box 35061, Dar es Salaam, Tanzania.
- Department of Chemistry, Mwalimu Julius K. Nyerere University of Agriculture and Technology, P.O. Box 976, Musoma, Tanzania.
| | - Dativa J Shilla
- Department of Chemistry, Dar es Salaam University College of Education, P.O. Box 2329, Dar es Salaam, Tanzania
| | - Nazima Dharsee
- Ocean Road Cancer Institute, P.O. Box 3592, Dar es Salaam, Tanzania
| | - John A M Mahugija
- Department of Chemistry, University of Dar es Salaam, P.O. Box 35061, Dar es Salaam, Tanzania
| | - Nyimvua S Mbare
- Department of Mathematics, University of Dar es Salaam, P.O. Box 35062, Dar es Salaam, Tanzania
| |
Collapse
|
199
|
Yeom S, Lee DH, Song J. Therapeutic Potential of Anti-Diabetes Drugs and Anti-Dyslipidemia Drugs to Mitigate Head and Neck Cancer Risk in Metabolic Syndrome. CNS Neurosci Ther 2025; 31:e70446. [PMID: 40387523 PMCID: PMC12087305 DOI: 10.1111/cns.70446] [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: 03/07/2025] [Revised: 04/24/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Head and neck cancer (HNC) encompasses a heterogeneous group of malignancies originating in the oral cavity, pharynx, nasopharynx, larynx, paranasal sinuses, and salivary glands. Accumulating evidence indicates that metabolic syndrome (MetS) characterized by a constellation of conditions including central adiposity, hyperglycemia, dyslipidemia, hypertension, and insulin resistance, may significantly influence cancer pathogenesis and progression. RESULTS MetS has been epidemiologically linked to elevated risk for multiple malignancies through various metabolic mechanisms involving chronic systemic inflammation, insulin resistance, and dysregulated lipid metabolism. Especially in HNC, recent studies demonstrated that MetS and metabolic imbalance conditions may contribute to carcinogenesis, disease progression, and clinical outcomes, but the exact mechanisms behind the association between excess fat accumulation and HNC risk remain unclear. Considering previous studies, pharmacological agents targeting metabolic pathways, including biguanides (metformin), thiazolidinediones, sodium-glucose cotransporter-2 (SGLT-2) inhibitors, and HMG-CoA reductase inhibitors (statins) are being investigated for potential repurposing in cancer prevention and adjuvant therapy. CONCLUSIONS Here, we summarize the latest evidence on the relationship between MetS and HNC, highlighting the therapeutic potential of anti-diabetes drugs and anti-dyslipidemia drugs in ameliorating various pathological problems in HNC patients with MetS.
Collapse
Affiliation(s)
- Sujung Yeom
- Department of Otolaryngology‐Head and Neck SurgeryChonnam National University Medical School & Hwasun HospitalHwasunRepublic of Korea
| | - Dong Hoon Lee
- Department of Otolaryngology‐Head and Neck SurgeryChonnam National University Medical School & Hwasun HospitalHwasunRepublic of Korea
| | - Juhyun Song
- Department of AnatomyChonnam National University Medical SchoolHwasunRepublic of Korea
| |
Collapse
|
200
|
Zhang J, Zhang M, Tatar M, Gong R. Keap1-independent Nrf2 regulation: A novel therapeutic target for treating kidney disease. Redox Biol 2025; 82:103593. [PMID: 40107017 PMCID: PMC11968292 DOI: 10.1016/j.redox.2025.103593] [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: 01/24/2025] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
The transcription factor NF-E2-related factor 2 (Nrf2) is a master regulator of antioxidant responses in mammals, where it plays a critical role in detoxification, maintaining cellular homeostasis, combating inflammation and fibrosis, and slowing disease progression. Kelch-like ECH-associated protein 1 (Keap1), an adaptor subunit of Cullin 3-based E3 ubiquitin ligase, serves as a critical sensor of oxidative and electrophilic stress, regulating Nrf2 activity by sequestering it in the cytoplasm, leading to its proteasomal degradation and transcriptional repression. However, the clinical potential of targeting the Keap1-dependent Nrf2 regulatory pathway has been limited. This is evidenced by early postnatal lethality in Keap1 knockout mice, as well as significant adverse events after pharmacological blockade of Keap1 in human patients with Alport syndrome as well as in those with type 2 diabetes mellitus and chronic kidney disease. The exact underlying mechanisms remain elusive, but may involve non-specific and systemic activation of the Nrf2 antioxidant response in both injured and normal tissues. Beyond Keap1-dependent regulation, Nrf2 activity is modulated by Keap1-independent mechanisms, including transcriptional, epigenetic, and post-translational modifications. In particular, GSK3β has emerged as a critical convergence point for these diverse signaling pathways. Unlike Keap1-dependent regulation, GSK3β-mediated Keap1-independent Nrf2 regulation does not affect basal Nrf2 activity but modulates its response at a delayed/late phase of cellular stress. This allows fine-tuning of the inducibility, magnitude, and duration of the Nrf2 response specifically in stressed or injured tissues. As one of the most metabolically active organs, the kidney is a major source of production of reactive oxygen and nitrogen species and also a vulnerable organ to oxidative damage. Targeting the GSK3β-mediated Nrf2 regulatory pathway represents a promising new approach for the treatment of kidney disease.
Collapse
Affiliation(s)
- Jiahui Zhang
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Mingzhuo Zhang
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Marc Tatar
- Division of Biology and Medicine, Brown University, Providence, RI, USA
| | - Rujun Gong
- Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA.
| |
Collapse
|