1
|
Zhang Y, Ge Y, Wu S, Shao Y, Lu Y, Zhao X, Gu J, Wang Y. Superoxide anion-responsive persulfide and all-trans retinoic acid co-donating peptide assemblies attenuate myocardial ischemia-reperfusion injury. Biomaterials 2025; 320:123276. [PMID: 40120175 DOI: 10.1016/j.biomaterials.2025.123276] [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: 11/06/2024] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) has become a severe threat to human health due to its high mortality rate and poor prognosis. Mutually entangled issues including ROS over-production, excessive inflammatory responses, and myocardial apoptosis are involved during MIRI. Effective inhibition of ROS burst at the beginning of reperfusion has been proved as the key for MIRI treatment. In this work, we report a superoxide anion-responsive peptide co-assembly (S/A-P) capable of delivering the H2S donor (i.e., superoxide-responsive persulfide donor) and all-trans retinoic acid (ATRA) simultaneously for the treatment. Our results suggest that compared with its single peptidic counterparts, the as-prepared system can significantly lower ROS production and repair myocardial mitochondrial dysfunction due to the synergy effect from the persulfides/H2S and ATRA. Moreover, S/A-P can reduce excessive inflammatory response through regulating macrophage polarization, which is further mapped by RNA sequencing. In vivo assessment of the co-assembly also displays an excellent therapeutic effect of MIRI on rats. In terms of good biocompatibility and outstanding efficacy, we believe that S/A-P will have a bright future for the treatment of cardiovascular diseases or other related diseases.
Collapse
Affiliation(s)
- Yanwen Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuxuan Ge
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shiqi Wu
- Department of Plastic and Reconstructive Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200125, China
| | - Yiyang Shao
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yujia Lu
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xueshan Zhao
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, 610000, China
| | - Jun Gu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, 610000, China.
| | - Yin Wang
- Engineering Research Center of Cell & Therapeutic Antibody, Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
2
|
Boginskaya I, Safiullin R, Tikhomirova V, Kryukova O, Afanasev K, Efendieva A, Bulaeva N, Golukhova E, Ryzhikov I, Kost O, Kurochkin I. The surface-enhanced Raman scattering method for point-of-care atrial fibrillation diagnostics. Comput Biol Med 2025; 189:109923. [PMID: 40043416 DOI: 10.1016/j.compbiomed.2025.109923] [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: 09/20/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 04/01/2025]
Abstract
We suggest a new method for the detection of paroxysmal atrial fibrillation by analyzing surface-enhanced Raman scattering (SERS) spectra of blood serum of patients in question in comparison with SERS spectra of the serum of healthy donors. Spectral measurements were carried out on compact SERS substrates in dried blood serum droplets with immediate subsequent processing. To process the spectra, machine learning methods were used, in particular, the logistic regression method and the principal component method. Furthermore, thanks to the possibility of the physical-chemical interpretation of the coefficients of the method, the vibrational bands responsible for the signs of atrial fibrillation were identified and their correlation was carried out. Evaluation metrics were presented for the classification, among which the accuracy value was 0.82, that is a high indicator when analyzing samples directly from the blood serum of patients with the disease under study. It was shown that a small number of measured spectra for each sample (near 35 measurements) was sufficient to carry out the study. A comparative analysis of the logistic regression method and other commonly used machine learning methods was also carried out: support vector machines and random forest. Each method was evaluated and the advantages of logistic regression in solving the problem presented in this study were shown. The receiver operating characteristic curve (ROC) analysis was also used for graphical representation and comparison of methods. The presented study shows the prospects for using the described method for the analysis of diseases associated with cardiac risks.
Collapse
Affiliation(s)
- I Boginskaya
- Institute for Theoretical and Applied Electromagnetics RAS, 125412, Moscow, Russia.
| | - R Safiullin
- Institute for Theoretical and Applied Electromagnetics RAS, 125412, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudny, 141700, Russia
| | - V Tikhomirova
- Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119991, Moscow, Russia
| | - O Kryukova
- Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119991, Moscow, Russia
| | - K Afanasev
- Institute for Theoretical and Applied Electromagnetics RAS, 125412, Moscow, Russia
| | - A Efendieva
- Bakulev Scientific Center for Cardiovascular Surgery, Cardiology Department, 121552, Moscow, Russia
| | - N Bulaeva
- Bakulev Scientific Center for Cardiovascular Surgery, Cardiology Department, 121552, Moscow, Russia
| | - E Golukhova
- Bakulev Scientific Center for Cardiovascular Surgery, Cardiology Department, 121552, Moscow, Russia
| | - I Ryzhikov
- Institute for Theoretical and Applied Electromagnetics RAS, 125412, Moscow, Russia; FMN Laboratory, Bauman Moscow State Technical University, 105005, Moscow, Russia
| | - O Kost
- Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119991, Moscow, Russia
| | - I Kurochkin
- Faculty of Chemistry, M.V. Lomonosov Moscow State University, 119991, Moscow, Russia; Emanuel Institute of Biochemical Physics RAS, 119334, Moscow, Russia
| |
Collapse
|
3
|
Ameling S, Van der Auwera S, Holtfreter S, Wiechert A, Michalik S, Friedrich N, Hammer E, Völzke H, Nauck M, Grabe HJ, Bröker BM, Völker U. Cytokine atlas of the population-based cohort SHIP-TREND-0 - Associations with age, sex, and BMI. Cytokine 2025; 189:156896. [PMID: 40020520 DOI: 10.1016/j.cyto.2025.156896] [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: 11/05/2024] [Revised: 12/20/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND The characterization of physiological immune signatures in a population-based cohort is a prerequisite for identifying pathological immune signatures associated with inflammatory or autoimmune diseases. METHODS Here, 47 plasma cytokines, chemokines, and growth factors were quantified with a bead-based multiplex-assay (Merck HCYTA-60 K) using a FLEXMAP 3D™ instrument in 1175 individuals of the Study of Health in Pomerania (SHIP; TREND cohort, 532 men and 643 women, age: 20 to 81, BMI: 17.7 to 53.6). Associations of cytokine concentrations with age, sex, BMI, season, and blood cell parameters (BCP) were examined by multivariate regression models. RESULTS The physiological cytokine concentrations differed strongly between analytes, with median concentrations ranging from 0.6 to 7820 pg/mL. Many cytokine levels showed a large dynamic range within the study population. Higher levels of the pro-inflammatory cytokines and chemokines IL-6, IL-8, CXCL9, CXCL10, IL-12p40, CCL2, CCL4, CCL11, IL-27, FLT3LG, and TNFα were significantly associated with increasing age. The strongest age-associated effects were seen for CXCL9 (βst = 0.4, p < 0.001) and CXLC10 (βst = 0.3, p < 0.001). Significant sex differences were detected for CCL2, CCL3, CCL4, CCL11, CCL22, IL-12p40, IL-1RA, IL-18, IL-27, and TNFα levels among which CCL11 showed the strongest effect (βst = -0.24, p < 0.001) with a lower level in women compared to men. Moreover, seven cytokines and chemokines, i.e. CCL4, CCL22, CXCL10, IL-1RA, IL-18, IL-6, and TNFα, displayed higher levels with increasing BMI. Among those, the strongest effect was seen for IL-1RA (βst = 0.19, p < 0.001), CCL4 (βst = 0.16, p < 0.001) and CXCL10 (βst = 0.14, p < 0.001). Only CCL11 (βst = -0.17, p < 0.001) decreased with increasing BMI. Subjects categorized as obese exhibited significantly elevated levels of CCL4, CCL22, CXCL10, and IL-1RA, while only CCL11 showed significantly reduced levels compared to normal weight. Certain cytokines such as IL-6, IL-18, or TNFα showed decreased significance levels after adjustment for blood cell components indicating blood cell components (BCPs) as potential confounders. We observed no significant non-linear seasonal effects for the investigated cytokines. CONCLUSION The generated cytokine atlas provides detailed information on cytokine variations in the general population and will provide a reference base for disease-related studies in the future. Furthermore, BCPs should be considered as potential confounders in association studies based on plasma cytokine levels.
Collapse
Affiliation(s)
- Sabine Ameling
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Straße 8, D-17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany.
| | - Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1, D-17475 Greifswald, Germany; German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, D-17475 Greifswald, Germany
| | - Anja Wiechert
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Straße 8, D-17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Stephan Michalik
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Straße 8, D-17475 Greifswald, Germany
| | - Nele Friedrich
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, D-17475 Greifswald, Germany
| | - Elke Hammer
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Straße 8, D-17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany; Institute for Community Medicine, University Medicine Greifswald, Walther-Rathenau-Straße 48, D-17475 Greifswald, Germany
| | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, D-17475 Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstraße 1, D-17475 Greifswald, Germany; German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Barbara M Bröker
- Institute of Immunology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, D-17475 Greifswald, Germany
| | - Uwe Völker
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Straße 8, D-17475 Greifswald, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| |
Collapse
|
4
|
Li S, He P, Liu J, Zang S, Luo J, Luo Y, Zhu S, Zang L. Ferulic acid protects against stress-induced myocardial injury in mice. Toxicol Appl Pharmacol 2025; 498:117309. [PMID: 40120650 DOI: 10.1016/j.taap.2025.117309] [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: 10/25/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
Excessive stress is a known contributor to cardiovascular diseases (CVD), and ferulic acid (FA), a natural phenolic compound, has demonstrated significant antioxidant and anti-inflammatory properties. This study investigates the protective effects of FA against stress-induced myocardial injury (SIMI) and elucidates the underlying mechanisms. An acute SIMI model was established in mice using low-temperature water immersion restraint. Cardiac function was assessed via cardiac index and histopathological analysis. Serum levels of corticosterone (CORT), lactate dehydrogenase (LDH), and brain natriuretic peptide (BNP) were quantified using enzyme-linked immunosorbent assay (ELISA), along with inflammatory markers TNF-α and IL-1β. The oxidative stress parameters, including malondialdehyde (MDA), glutathione (GSH), superoxide dismutase (SOD), and reactive oxygen species (ROS), were analyzed using colorimetric methods and fluorescent probes. Immunohistochemistry (IHC) and Western Blot were used to analyze the expression of proteins related to TNF, MAPK, PPAR-α/PGC-1α, and Nrf2 signaling pathways. Results indicated that FA pretreatment improved cardiac index, myocardial structural integrity, and reduced inflammatory cell infiltration. Serum levels of LDH, BNP, CORT, TNF-α, and IL-1β were significantly decreased in FA-treated SIMI mice. Elevated MDA and ROS levels, along with decreased GSH and SOD levels in the SIMI group, were reversed by FA pretreatment, likely through activation of the PPARα/PGC-1α and Nrf2 signaling pathways. Additionally, FA inhibited the TNF-α/TNFR1 and ERK/JNK MAPK pathways, contributing to its protective effects. In conclusion, FA mitigates SIMI by alleviating oxidative stress and inflammatory responses.
Collapse
Affiliation(s)
- Siyong Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Peiyi He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiahe Liu
- The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | | | - Jiahao Luo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Luo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shuguang Zhu
- The First Affiliated Hospital cardiothoracic surgery department, Guangdong Pharmaceutical University, Guangzhou 510080, China.
| | - Linquan Zang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
5
|
Wang Y, Bi L, Li Q, Wang Q, Lv T, Zhang P. Remnant cholesterol inflammatory index and its association with all-cause and cause-specific mortality in middle-aged and elderly populations: evidence from US and Chinese national population surveys. Lipids Health Dis 2025; 24:155. [PMID: 40275392 DOI: 10.1186/s12944-025-02580-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND The remnant cholesterol inflammatory index (RCII) is a novel metric that combines remnant cholesterol and high-sensitivity C-reactive protein, reflecting the metabolic and inflammatory risk. This study investigates the association between RCII and long-term risks of all-cause and cause-specific mortality in middle-aged and elderly populations in the US and China. METHOD We analyzed data from the National Health and Nutrition Examination Survey (NHANES) and the China Health and Retirement Longitudinal Study (CHARLS), including 7,565 and 12,932 participants aged 45 years and older, respectively. The participants were categorized into quartiles based on natural log-transformed RCII (lnRCII) values. Kaplan-Meier survival analysis, Cox proportional hazards models, restricted cubic splines (RCS) and mediation analysis were used to examine the relationship between lnRCII and mortality outcomes, adjusting for potential covariates. RESULT The mean age of the participants was 59.90 ± 10.44 years (NHANES) and 58.64 ± 9.78 years (CHARLS), with 53.28% and 52.50% female, respectively. Kaplan-Meier survival analysis showed that higher lnRCII quartiles (≥ 0.79 in NHANES, ≥ -0.13 in CHARLS) were significantly associated with increased all-cause mortality risk (p < 0.001). Each standard deviation (SD) increase in lnRCII corresponded to a higher risk of all-cause mortality, and the hazard ratios (HRs) and 95% confidence interval (CI) were 1.29 (95% CI: 1.21-1.36) in NHANES and 1.26 (95% CI: 1.15-1.38) in CHARLS. In NHANES, lnRCII was also associated with elevated risks of cardiovascular mortality (HR = 1.21, 95% CI: 1.08-1.35) and cancer mortality (HR = 1.30, 95% CI: 1.09-1.55). RCS analysis indicated a J-shaped relationship between lnRCII and both all-cause and cardiovascular mortality, and a linear association with cancer mortality. Mediation analysis showed that systolic blood pressure and fasting plasma glucose partially mediated these associations. Subgroup analyses suggested a stronger association between lnRCII and all-cause mortality in middle-aged US participants (p for interaction = 0.010). CONCLUSIONS Elevated RCII levels are significantly associated with increased all-cause mortality risk middle-aged and elderly populations in both the US and China. In the US population, RCII is also associated with increased risks of cardiovascular and cancer mortality. By integrating metabolic and inflammatory risk factors, RCII may serve as a valuable tool for mortality risk stratification and clinical decision-making.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Lei Bi
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Qing Li
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Qiuyu Wang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China
| | - Tingting Lv
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China.
| | - Ping Zhang
- Department of Cardiology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Changping District, 102218, China.
| |
Collapse
|
6
|
Mrochen A, Meuth SG, Pfeuffer S. Should we stay or should we go? Recent insights on drug discontinuation in multiple sclerosis. Neurol Res Pract 2025; 7:25. [PMID: 40254626 PMCID: PMC12010584 DOI: 10.1186/s42466-025-00379-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/14/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND The decision to discontinue disease-modifying therapies (DMTs) in patients with multiple sclerosis (PwMS) is a critical clinical challenge. Historically, DMTs were discontinued due to side effects, treatment limitations, or progression to secondary progressive MS. However, advancements in MS therapies, particularly high-efficacy DMTs (HE-DMTs) and the increased knowledge on disease courses and phenotypes have resulted in more personalized treatment approaches and introduced discussion on scheduled DMT discontinuation. This review explores the current evidence on DMT discontinuation, focusing on its implications for aging populations and the interplay between cardiovascular diseases (CVD) and MS. CURRENT EVIDENCE AND INTERPLAY WITH CVD Randomized trials such as DISCOMS and DOT-MS have provided insights into discontinuing DMTs in stable patients. In summary, both randomized clinical trials highlight the risk of disease reactivation following treatment discontinuation. Due to the limited sample size, neither study was able to conduct subgroup analyses based on age groups. Additionally, DOT-MS was terminated prematurely, direct comparisons with other studies should be avoided. While older studies and observational data (e.g., OFSEP) have shown relapse risks associated with discontinuation, particularly for drugs like natalizumab and fingolimod, there is limited data on HE-DMT discontinuation outcomes. Comorbidities, particularly CVDs, further complicate decisions regarding the continuation of DMTs in older adults. MS patients bear a higher burden of CVD, which is also associated with unfavorable disease courses. While optimizing cardiovascular risk profiles appears advisable, it remains unclear whether DMTs themselves have a positive impact on CVDs. CONCLUSION Given the complexities associated with discontinuing DMTs in MS patients, it is essential to balance the avoidance of polypharmacy with the potential risks of disease reactivation and the impact of comorbidities, especially CVDs, on disease progression. The interplay between MS and CVD highlights the importance of a holistic risk assessment when considering DMT discontinuation.
Collapse
Affiliation(s)
- Anne Mrochen
- Department of Neurology, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Klinikstr. 33, 35392, Giessen, Germany
| | - Sven G Meuth
- Department of Neurology, University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, Germany
| | - Steffen Pfeuffer
- Department of Neurology, University Hospital Giessen and Marburg, Justus-Liebig-University Giessen, Klinikstr. 33, 35392, Giessen, Germany.
| |
Collapse
|
7
|
Yoon J, Kim D, Jeong NH, Choi YA, Kwon TK, Lee S, Khang D, Kim SH. Protectin D1, an omega-3-derived lipid mediator, resolves mast cell-driven allergic inflammation via FcεRⅠ signaling. Biomed Pharmacother 2025; 187:118060. [PMID: 40253829 DOI: 10.1016/j.biopha.2025.118060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025] Open
Abstract
Protectin D1 (PD1) derived from docosahexaenoic acid (DHA) has shown promise in resolving inflammation. Mast cells are critical drivers of allergic inflammation, releasing inflammatory mediators such as histamine and pro-inflammatory cytokines. This study assesses the effectiveness of PD1 in counteracting mast cell-mediated allergic inflammation. In vivo, two well-established mouse models were employed: IgE-mediated passive cutaneous anaphylaxis (PCA) and ovalbumin-induced active systemic anaphylaxis (ASA). The oral administration of PD1 markedly suppressed PCA reactions, including ear swelling, plasma extravasation of Evans blue and mast cell degranulation. In the ASA model, oral PD1 administration dose-dependently alleviated hypothermia and reduced elevated serum levels of IgE, histamine, and IL-4. Mechanistic insights were gained through studies in the RBL-2H3 and primary mast cells derived from mouse bone marrow, where PD1 inhibited IgE-mediated degranulation and decreased intracellular calcium influx by blocking FcεRⅠ signaling pathways involving Lyn, Fyn, and Syk kinases. Additionally, PD1 suppressed pro-inflammatory cytokine production by inhibiting the activity of critical transcription factor; nuclear factor-κB. These findings suggest that PD1, a bioactive lipid derived from DHA, is a very promising therapeutic candidate for mast cell-derived allergic inflammation.
Collapse
Affiliation(s)
- Jieun Yoon
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Daheen Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Na-Hee Jeong
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Young-Ae Choi
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Soyoung Lee
- Department of Innovative Pharmaceutical Sciences, Advanced Institute of Science and Technology, Kyungpook National University, Daegu 41566, Republic of Korea.
| | - Dongwoo Khang
- Department of Gachon Advanced Institute for Health Science & Technology (GAIHST), Lee Gil Ya Cancer and Diabetes Institute, and Department of Physiology, College of Medicine, Gachon University, Incheon 21999, Republic of Korea.
| | - Sang-Hyun Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
8
|
Dabravolski SA, Popov MA, Utkina AS, Babayeva GA, Maksaeva AO, Sukhorukov VN, Orekhov AN. Preclinical and mechanistic perspectives on adipose-derived stem cells for atherosclerotic cardiovascular disease treatment. Mol Cell Biochem 2025:10.1007/s11010-025-05285-0. [PMID: 40234340 DOI: 10.1007/s11010-025-05285-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Adipose-derived mesenchymal stem cells (AD-MSCs) are a promising therapeutic modality for cardiovascular diseases due to their immunomodulatory, anti-inflammatory, and pro-angiogenic properties. This manuscript explores the current status, challenges, and future directions of AD-MSC therapies, focusing on their application in atherosclerosis (AS), myocardial infarction (MI), and heart failure (HF). Preclinical studies highlight AD-MSC's ability to stabilise atherosclerotic plaques, reduce inflammation, and enhance myocardial repair through mechanisms such as macrophage polarisation, endothelial protection, and angiogenesis. Genetically and pharmacologically modified AD-MSCs, including those overexpressing SIRT1, IGF-1, and PD-L1 or primed with bioactive compounds, exhibit superior efficacy compared to unmodified cells. These modifications enhance cell survival, immunopotency, and reparative capacity, showcasing the potential for tailored therapies. However, clinical translation faces significant hurdles. While recent clinical trials have confirmed the safety of AD-MSC therapy, their efficacy remains inconsistent, necessitating further optimisation of patient selection, dosing strategies, and delivery methods. Donor variability, particularly in patients with co-morbidities like type 2 diabetes (T2D) or obesity, impairs AD-MSC efficacy. Emerging research on extracellular vesicles (EVs) derived from AD-MSC offers a promising cell-free alternative, retaining the therapeutic benefits while mitigating risks. Future perspectives emphasise the need for multidisciplinary approaches to overcome these limitations. Strategies include refining genetic modifications, exploring EV-based therapies, and integrating personalised medicine and advanced diagnostic tools. By addressing these challenges, AD-MSC therapies hold the potential to revolutionise the treatment of cardiovascular diseases, providing innovative solutions to improve patient outcomes.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51P.O. Box 78, 2161002, Karmiel, Israel.
| | - Mikhail A Popov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, 125315, Russia
| | - Aleksandra S Utkina
- Department of Commodity Expertise and Customs Business, Plekhanov Russian University of Economics, 36, Stremyanny Lane, 115054, Moscow, Russia
| | - Gulalek A Babayeva
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Str., 121552, Moscow, Russia
| | - Anastasia O Maksaeva
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, 125315, Russia
- Sechenov First Moscow State Medical University, 8, Trubetskaya Street Building 2, 119991, Moscow, Russia
| | - Vasily N Sukhorukov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow, 125315, Russia
- Institute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, 119991, Moscow, Russia
| | - Alexander N Orekhov
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street Building 4, 117418, Moscow, Russia
| |
Collapse
|
9
|
Mansoori R, Ashrafpour M, Asghari MH, Golchoobian R, Hosseini SM, Reiter RJ, Karim B, Moghadamnia AA, Kazemi S. Protective effects of melatonin against 5-fluorouracil-induced cardiotoxicity in rats: A comprehensive evaluation of oxidative, inflammatory, and apoptotic pathways. Toxicol Appl Pharmacol 2025; 499:117343. [PMID: 40239743 DOI: 10.1016/j.taap.2025.117343] [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: 11/24/2024] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Cardiotoxicity is a serious adverse effect of 5-fluorouracil (5-FU) a common chemotherapeutic agent. This study aimed to evaluate the protective effects of melatonin (MLT) against 5-fluorouracil (5-FU)-induced cardiotoxicity in rats, focusing on oxidative stress, inflammatory pathways, gene expression, electrocardiographic and histopathological changes. MATERIALS AND METHODS Twenty-five male Wistar rats were divided into five groups. The animals received either MLT at doses of 2.5, 5, or 10 mg/kg/day, 5-FU at 50 mg/kg (i.p.), or a combination of both treatments. Cardiotoxicity was assessed through electrocardiography, cardiac enzymes, oxidative stress markers, and histopathology. RESULTS 5-FU treatment significantly increased oxidative stress markers and inflammatory mediators while causing histopathological damage in heart tissues. Co-administration of MLT with 5-FU significantly mitigated these effects by reducing oxidative damage, as evidenced by lower levels of malondialdehyde (MDA), nitric oxide (NO), and myeloperoxidase (MPO). Additionally, MLT enhanced antioxidant activity, as reflected by increased levels of superoxide dismutase (SOD) and glutathione peroxidase (GPx) in heart tissues. Gene expression analysis further confirmed that MLT treatment reduced the elevated levels of COX-2 and VEGF, which are critical players in the inflammatory process. Histopathological examination demonstrated that MLT preserved the structural integrity of myocardial tissues, reducing 5-FU-induced damage score in a dose-dependent manner. Furthermore, MLT co-administration significantly attenuated the rise in cardiac biomarkers, including LDH, AST, and CK-MB, associated with 5-FU-induced cardiotoxicity. CONCLUSION These findings highlight that MLT, through its antioxidant and anti-inflammatory properties, exerts a protective effect against 5-FU-induced toxicity, suggesting its therapeutic potential for improving cardiovascular health during chemotherapy.
Collapse
Affiliation(s)
- Razieh Mansoori
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Manoochehr Ashrafpour
- Mobility Impairment Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Hossien Asghari
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ravieh Golchoobian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Bardia Karim
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Ali Akbar Moghadamnia
- Pharmaceutical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
10
|
Altyar AE, Bhardwaj S, Ghaboura N, Kaushik P, Alenezi SK, Mantargi MJS, Afzal M. Role of IL-2, IL-6, and TNF-α as Potential Biomarkers in Ischemic Heart Disease: A Comparative Study of Patients with CAD and Non-CAD. Med Sci (Basel) 2025; 13:40. [PMID: 40265387 PMCID: PMC12015866 DOI: 10.3390/medsci13020040] [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: 01/31/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
Background: Ischemic heart disease (CAD), a leading global health burden, arises primarily from atherosclerosis, an inflammatory condition characterized by lipid accumulation and metabolic dysregulation. The precise contribution of inflammatory cytokines (IL-2, IL-6, and TNF-α) to CAD pathogenesis remains an area of significant research. Aim: The primary aim of this study is to examine the IL-2, IL-6, and TNF-α in patients with coronary artery disease (CAD) and compare them with Non-CAD individuals to evaluate their potential as diagnostic biomarkers for CAD. Methodology: A prospective observational study was conducted over 3 years, involving 100 participants divided into CAD and non-CAD groups. Blood samples were isolated and analyzed for IL-2, IL-6, and TNF-α levels utilizing ELISA kits. Biochemical parameters, including lipid profiles, were also assessed. Results: This study observed significantly elevated IL-6 in patients with CAD compared with controls, while IL-2 and TNF-α levels did not reach statistical significance. The CAD group exhibited dyslipidemia characterized by elevated triglycerides and reduced HDL. Furthermore, the CAD group demonstrated alterations in biochemical parameters, including lower albumin and calcium levels, higher urea and uric acid levels, and an elevated erythrocyte sedimentation rate. These findings suggest a systemic inflammatory state and metabolic disturbances in patients with CAD. Conclusions: This study highlights IL-6 as a potential biomarker and key player in CAD pathogenesis. These findings warrant further investigation into the therapeutic potential of targeting inflammatory pathways for cardiovascular risk reduction.
Collapse
Affiliation(s)
- Ahmed E. Altyar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia;
- Pharmacy Program, Department of Pharmacy Practice, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Shilpa Bhardwaj
- Department of Neurochemistry, Institute of Human Behaviour and Allied Sciences Hospital, Delhi 110095, India
| | - Nehmat Ghaboura
- Pharmacy Program, Department of Pharmacy Practice, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Priya Kaushik
- Pacific Institute of Medical Sciences, Udaipur 313015, India
| | - Sattam Khulaif Alenezi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Mohammed Jaffar Sadiq Mantargi
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Muhammad Afzal
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| |
Collapse
|
11
|
Zengwei C, Shiyi G, Pinfang K, Dasheng G, Jun W, Sigan H. Associations of Gla-rich protein and interleukin-1β with coronary artery calcification risk in patients with suspected coronary artery disease. Front Endocrinol (Lausanne) 2025; 16:1504346. [PMID: 40241989 PMCID: PMC11999850 DOI: 10.3389/fendo.2025.1504346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Background Gla-rich protein (GRP) and interleukin-1β (IL-1β) are recognized as reliable biomarkers for evaluating inflammation and are effective predictors of cardiovascular disease. However, the relationship between GRP, IL-1β, and coronary artery calcification (CAC) in patients with suspected coronary artery disease (CAD) remains unclear. Therefore, we investigated the association between these inflammatory biomarkers (GRP and IL-1β) and CAC in patients with suspected CAD. Methods This prospective study included patients with suspected CAD who underwent coronary computed tomography angiography (CTA). Fasting venous blood samples were collected at admission, and GRP and IL-1β levels were quantified using enzyme-linked immunosorbent assays (ELISA). The Agatston score was calculated to assess coronary artery calcification (CAC) based on coronary CTA findings. Results A total of 120 patients were included in this study. Multivariate logistic regression analysis revealed that GRP [odds ratio (OR), 1.202; 95% confidence interval (CI), 1.065-1.356; p = 0.003] and IL-1β (OR, 1.011; 95% CI, 1.002-1.020; p = 0.015) were independent risk factors for CAC severity. Receiver operating characteristic (ROC) curve analysis demonstrated that GRP had a predictive ability for CAC, with an area under the curve (AUC) of 0.830 [95% CI (0.755, 0.904)]. IL-1β exhibited an AUC of 0.753 [95% CI (0.660, 0.847)]. The combination of GRP and IL-1β in a predictive model improved the AUC to 0.835. Additionally, GRP and IL-1β levels showed a strong positive correlation (r = 0.6861, p < 0.05), and GRP was significantly associated with CAC severity (r = 0.5018, p < 0.05). Conclusions Elevated levels of GRP and IL-1β, as inflammatory biomarkers, were associated with CAC in patients with suspected CAD. These biomarkers may provide valuable insights into the pathophysiology of coronary artery calcification and contribute to improved risk stratification in this patient population.
Collapse
Affiliation(s)
- Cheng Zengwei
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Cardiology, Wuhe County People’s Hospital, Bengbu, China
| | - Gao Shiyi
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Kang Pinfang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Gao Dasheng
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Wang Jun
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Hu Sigan
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
12
|
Zhao C, Fu J, Wang Y, Zhou Y. Latest Evidence and Perspectives of Panax Notoginseng Extracts and Preparations for the Treatment of Cardiovascular Diseases. J Cardiovasc Pharmacol 2025; 85:248-260. [PMID: 39903802 DOI: 10.1097/fjc.0000000000001670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/11/2025] [Indexed: 02/06/2025]
Abstract
ABSTRACT Cardiovascular diseases are a major cause of death worldwide, and their high incidence poses a significant threat to human health and public health systems. Panax notoginseng , a traditional Chinese medicinal herb with a long history, has shown promise in treating cardiovascular diseases. This review examines the diverse mechanisms through which Panax notoginseng addresses cardiovascular diseases, including anti-inflammatory, antiplatelet aggregation, anticoagulation, anti-oxidative stress, regulation of angiogenesis, antiatherosclerosis, improvement of microcirculatory disorders, and protection against myocardial ischemia-reperfusion injury, highlighting saponins as the principal active components. It also summarizes studies involving Panax notoginseng preparations like Xueshuantong and Xuesaitong in treating coronary heart disease and myocardial infarction, and discusses the safety, limitations, and future research directions of these extracts. In conclusion, the cardiovascular protective mechanism of Panax notoginseng is multitargeted and multipathways, and its clinical application is relatively safe, with rare and mild adverse drug reactions, suggesting a promising therapeutic potential.
Collapse
Affiliation(s)
- Chenyu Zhao
- The First Clinical Medical College of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | | | | | | |
Collapse
|
13
|
Yan L, Huang Y, Xie B, Liu Z, Luo L, He B, Ding C, Fang W, Lin Y, Kang D, Chen F. Association of periodontitis and periodontal parameters with migraine and mortality in people with migraine disease: A nationally representative observational study. Headache 2025; 65:578-588. [PMID: 39739848 DOI: 10.1111/head.14893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 01/02/2025]
Abstract
OBJECTIVE To investigate the association of periodontitis and clinical periodontal parameters with migraine as well as mortality among people with migraine disease. BACKGROUND Periodontitis has been shown to increase the systemic inflammatory burden thereby promoting various systemic health outcomes; however, the evidence regarding the relationship between periodontitis and migraine is scarce. METHODS A cross-sectional study was performed, and it included 13,108 participants from the National Health and Nutrition Examination Survey (1999-2004). Weighted logistic regression analysis was used to evaluate the association between periodontitis/clinical periodontal parameters and migraine. Mediation analysis was performed to explore the potential mediating role of inflammatory response. A cohort study including 1909 participants with migraine disease was further conducted to assess the associations between periodontitis/clinical periodontal parameters and mortality from all causes, cardiovascular disease (CVD), and cancer in participants with migraine disease using Cox proportional hazards models. Death outcomes were ascertained by linkage to National Death Index records through December 31, 2018. RESULTS Periodontitis was positively associated with migraine (odds ratio [OR] 1.29, 95% confidence interval [CI] 1.01-1.65). Each 1-unit rise in attachment loss and pocket depth was linked to a 17.5% (OR 1.18, 95% CI 1.08-1.29) and 28.1% (OR 1.28, 95% CI 1.08-1.51) increase in migraine risk, respectively. Mediation analyses revealed that leukocyte, monocyte, and lymphocyte counts mediated 17.9%, 7.3%, and 20.1%, respectively, of the association between periodontitis and migraine. During a median follow-up of 17.7 years among 1909 participants with migraine disease, periodontitis was associated with greater all-cause mortality (hazard ratio 1.82, 95% CI 1.25-2.66), but was not significantly associated with mortality from CVD or cancer among participants with migraine disease. Similar association patterns were also observed for attachment loss and pocket depth. CONCLUSIONS This study provides evidence that periodontitis and clinical periodontal parameters were significantly associated with migraine as well as all-cause mortality in people with migraine disease. These findings underscore the importance of considering periodontal health in the prevention and management strategies for migraine disease.
Collapse
Affiliation(s)
- Lingjun Yan
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yu Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Bingqin Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Zilin Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Lan Luo
- Fujian Key Laboratory of Oral Diseases, Department of Periodontology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Baochang He
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Chenyu Ding
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenhua Fang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yuanxiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Dezhi Kang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Department of Neurosurgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Fa Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, China
| |
Collapse
|
14
|
Tcholadze G, Pantsulaia I, Ratiani L, Kopaleishvili L, Bolotashvili T, Jorbenadze A, Chikovani T. The Prognostic Value of Circulating Cytokines and Complete Blood Count-Based Inflammatory Markers in COVID-19 Patients With Atrial Fibrillation. Cardiol Res 2025; 16:153-160. [PMID: 40051670 PMCID: PMC11882233 DOI: 10.14740/cr2027] [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: 12/06/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
Background Atrial fibrillation (AF) is associated with a high burden of cardiovascular disease, which has been worsened during the coronavirus disease 2019 (COVID-19) pandemic. The purpose of this study was to assess the association between clinical markers, especially interleukin-6 (IL-6) and other inflammatory biomarkers, and the severity of COVID-19 in patients with AF. Methods This retrospective cohort study categorized patients based on clinical presentations and laboratory results to investigate the prognostic significance of inflammatory markers in COVID-19 outcomes among those with AF. The study included 100 hospitalized COVID-19 patients aged between 40 to 80 years and was conducted at the Chapidze Hospital in Tbilisi, Georgia. Patients were then grouped by disease severity according to computed tomography (CT) scores, clinical symptoms, respiratory rate and oxygen saturation. Levels of IL-6 were obtained at three time points during hospitalization. A broad range of laboratory tests, including C-reactive protein (CRP), ferritin, and D-dimer, were also conducted. Results Patients with AF demonstrated significantly elevated levels of IL-6 (P = 0.024), CRP (P = 0.001), and ferritin (P < 0.001), suggesting a severe inflammatory response. D-dimer levels were also notably higher in the AF group (P < 0.005), indicating an increased risk of thrombotic complications. Oxygen saturation levels were significantly lower (P = 0.004) and CT scores higher in patients with AF. Furthermore, the length of hospitalization was longer among patients with AF (median duration significantly higher, P = 0.032), indicating a more severe disease course. Conclusions The proinflammatory markers such as IL-6 are independent predictive markers of COVID-19 severity in AF patients. Overall, it highlights urgent treatment approaches, such as available anti-inflammatory drugs, for COVID-19 patients with arrhythmias. Combining these biomarkers into clinical routines helps us better identify patients at risk and how to treat them.
Collapse
Affiliation(s)
- Giorgi Tcholadze
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| | - Ia Pantsulaia
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
- Vl. Bakhutashvili Institute of Medical Biotechnology, Tbilisi State Medical University, Tbilisi 0159, Georgia
| | | | | | | | | | - Tinatin Chikovani
- Department of Immunology, Tbilisi State Medical University, Tbilisi 0177, Georgia
| |
Collapse
|
15
|
Pizziolo PG, Clemente LM, Ribeiro AB, Oliveira VDC, Macedo AP, Salgado HC, Fazan-Junior R, Watanabe E, Silva-Lovato CH, Ribeiro AB. The interplay of edentulism, smoking, microbiota, oral rehabilitation on cytokineprofile and different conditions of hypertension. Arch Oral Biol 2025; 172:106176. [PMID: 39827717 DOI: 10.1016/j.archoralbio.2025.106176] [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: 08/05/2024] [Revised: 12/19/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE To evaluate the influence of edentulism, smoking, microbiota, and oral rehabilitation on the cytokine profile in healthy and hypertensive edentulous individuals using complete dentures. DESIGN This case-control study was divided into four groups: normotensives (control group - NH), controlled hypertensives (case group 1 - CH), unreported hypertensives (case group 2 - UnrH), and uncontrolled hypertensives (case group 3 - UncH). The participants were characterized by sociodemographic data, clinical and behavioral information, and systolic and diastolic blood pressure. The microbial load of Candida spp., Staphylococcus spp., enterobacteria, and mutans streptococci was evaluated by quantifying colony-forming unit. Salivary flow and salivary cytokines (IL-2, IL-4, IL-6, IL-10, TNF-α, IFN-γ, and IL-17) were quantified in unstimulated saliva by mL per minute and by flow cytometry, respectively. RESULTS Eighty patients (66 ± 7.2 years) were evaluated. The duration of edentulism was positively associated with systolic blood pressure (p = 0.012). Patients with non-functional denture rehabilitation, those with only upper complete dentures, showed significantly higher systolic blood pressure (p = 0.024) and levels of IL-2 (p = 0.024), predominantly in UncH. The colony-forming unit of mutans streptococci on the denture was higher in UncH and showed a negative association with smoking habit, and this had a positive association with salivary cytokines (IL-4, IL-2, IL-17, IFN-γ) and CVD. CONCLUSIONS The interplay between edentulism, smoking, and oral rehabilitation significantly impacts the cytokine profile, particularly in hypertensive conditions. Smoking habits modulated microbiota and interleukin profile, especially in cardiovascular patients. Moreover, non-functional dentures are associated with uncontrolled hypertension, marked by increased systolic blood pressure and IL-2.
Collapse
Affiliation(s)
- Pillar Gonçalves Pizziolo
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Lorena Mosconi Clemente
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | | | - Viviane de Cássia Oliveira
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Ana Paula Macedo
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Hélio César Salgado
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Rubens Fazan-Junior
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Evandro Watanabe
- Department of Restorative Dentistry, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Cláudia Helena Silva-Lovato
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Adriana Barbosa Ribeiro
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
16
|
Han H. Harnessing NLP to investigate biomarker interactions and CVD risks in elderly chronic kidney disease patients. SLAS Technol 2025; 31:100243. [PMID: 39798697 DOI: 10.1016/j.slast.2025.100243] [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: 10/30/2024] [Revised: 12/06/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Chronic kidney disease (CKD) significantly increases the risk of CVD diseases, particularly among elderly patients. Understanding the interaction between several biomarkers and cardiovascular (CVD) risks is crucial for improving patient outcomes and tailoring personalized treatment strategies. There is much more to learn about the intricate relationship between biomarkers and CVD risks in elderly CKD patients. Research aims to harness natural language processing (NLP) strategies to investigate the interaction between biomarkers and CVD risks in elderly patients with CKD. This research examined how changes in baseline values of four novel and classic cardiac biomarkers relate to the danger of CVD, and all-cause death in a large cohort of patients with CKD. Initially, medical data were collected from EHR of elderly CKD patients. NLP technique, such as Named Entity Recognition (NER), is used to extract the relevant biomarkers and CVD risk factors from the data. Statistical techniques were applied to examine the associations between biomarkers and CVD risks. The predictive models, using a combination of structured and NLP-extracted features demonstrated improved accuracy in forecasting CVD outcomes compared to traditional methods. This investigation highlights the critical role of specific biomarkers like PTH and FGF-23 in predicting CVD outcomes, providing insights into the possibility of using EHR data for better patient management and enhancing predictive models for this high-risk population.
Collapse
Affiliation(s)
- Hongli Han
- Department of General Medicine, The First Afiliated Hospital of Jinan University, Guangzhou, Guangdong, 510000, China.
| |
Collapse
|
17
|
Mrisho II, Musazade E, Chen H, Zhao H, Xing J, Li X, Han J, Cai E. Unlocking the Therapeutic Potential of Patchouli Leaves: A Comprehensive Review of Phytochemical and Pharmacological Insights. PLANTS (BASEL, SWITZERLAND) 2025; 14:1034. [PMID: 40219102 PMCID: PMC11990424 DOI: 10.3390/plants14071034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025]
Abstract
Plant-based products play an increasingly vital role in the pharmaceutical industry, including Pogostemon cablin (Blanco) Benth. (patchouli), which is notable for its rich history and extensive use in traditional medicine. Patchouli has a longstanding historical use as a remedy for a wide range of health conditions, including colds, fevers, headaches, inflammation, digestive disorders, and insect and snake bites. Comprehensive phytochemical studies have revealed that patchouli leaves contain diverse valuable bioactive compounds, notably patchouli alcohol, β-patchoulene, pogostone, α-bulnesene, and β-caryophyllene. Recent studies have demonstrated that patchouli leaves exhibit various pharmacological properties, including anti-oxidant, anti-inflammatory, antimicrobial, antidepressant, and anticancer effects. Despite robust traditional knowledge, specific therapeutic applications of patchouli leaves require scientific validation and standardization of their bioactive compounds. This review provides a comprehensive overview of the existing literature on the phytochemical composition, pharmacological properties, and underlying mechanisms of action of patchouli essential oil (PEO) and plant extracts obtained from patchouli leaves. It offers detailed insights into potential therapeutic applications, aiming to inform and guide future research across multiple medical disciplines. Ultimately, this review underscores the need for further research to validate and develop the medicinal applications of patchouli leaves, providing a foundation for future healthcare advancements.
Collapse
Affiliation(s)
- Isack Ibrahim Mrisho
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun 130118, China; (I.I.M.); (H.C.); (H.Z.); (J.X.); (X.L.)
| | - Elshan Musazade
- Key Laboratory of Soybean Molecular Design Breeding, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Changchun 130102, China;
- College of Life Science, Jilin Agricultural University, Changchun 130118, China
| | - Haobo Chen
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun 130118, China; (I.I.M.); (H.C.); (H.Z.); (J.X.); (X.L.)
| | - Huixuan Zhao
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun 130118, China; (I.I.M.); (H.C.); (H.Z.); (J.X.); (X.L.)
| | - Junjia Xing
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun 130118, China; (I.I.M.); (H.C.); (H.Z.); (J.X.); (X.L.)
| | - Xue Li
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun 130118, China; (I.I.M.); (H.C.); (H.Z.); (J.X.); (X.L.)
| | - Jiahong Han
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun 130118, China; (I.I.M.); (H.C.); (H.Z.); (J.X.); (X.L.)
| | - Enbo Cai
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun 130118, China; (I.I.M.); (H.C.); (H.Z.); (J.X.); (X.L.)
| |
Collapse
|
18
|
Karakasis P, Patoulias D, Theofilis P, Pamporis K, Sagris M, Vlachakis PK, Koufakis T, Antoniadis AP, Fragakis N. GLP-1 Receptor Agonists and Myocardial Perfusion: Bridging Mechanisms to Clinical Outcomes. Int J Mol Sci 2025; 26:3050. [PMID: 40243679 PMCID: PMC11988964 DOI: 10.3390/ijms26073050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Coronary microvascular dysfunction (CMD) is a key contributor to myocardial ischemia and adverse cardiovascular outcomes, particularly in individuals with metabolic disorders such as type 2 diabetes (T2D). While conventional therapies primarily target epicardial coronary disease, effective treatments for CMD remain limited. Glucagon-like peptide-1 receptor (GLP-1R) agonists have emerged as promising agents with cardiovascular benefits extending beyond glycemic control. Preclinical and clinical evidence suggests that GLP-1R activation enhances coronary microvascular function through mechanisms including improved endothelial function, increased nitric oxide bioavailability, attenuation of oxidative stress, and reduced vascular inflammation. Moreover, GLP-1R agonists have been shown to improve myocardial blood flow, myocardial perfusion reserve, and coronary endothelial function, particularly in high-risk populations. Despite these promising findings, inconsistencies remain across studies due to variability in patient populations, study designs, and imaging methodologies. This review summarizes current evidence on the role of GLP-1R agonists in myocardial perfusion, bridging mechanistic insights with clinical outcomes. Further large-scale, well-designed trials are needed to clarify their long-term impact on coronary microcirculation and explore their potential as targeted therapies for CMD.
Collapse
Affiliation(s)
- Paschalis Karakasis
- Second Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (A.P.A.); (N.F.)
| | - Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Faculty of Medicine, School of Health Sciences Aristotle, University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Panagiotis Theofilis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (K.P.); (M.S.); (P.K.V.)
| | - Konstantinos Pamporis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (K.P.); (M.S.); (P.K.V.)
| | - Marios Sagris
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (K.P.); (M.S.); (P.K.V.)
| | - Panayotis K. Vlachakis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (K.P.); (M.S.); (P.K.V.)
| | - Theocharis Koufakis
- Second Propedeutic Department of Internal Medicine, Faculty of Medicine, School of Health Sciences Aristotle, University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Antonios P. Antoniadis
- Second Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (A.P.A.); (N.F.)
| | - Nikolaos Fragakis
- Second Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (A.P.A.); (N.F.)
| |
Collapse
|
19
|
Czajkowski M, Wierzbicki PM, Dolny M, Matuszewski M, Hakenberg OW. Inflammation in Penile Squamous Cell Carcinoma: A Comprehensive Review. Int J Mol Sci 2025; 26:2785. [PMID: 40141426 PMCID: PMC11943298 DOI: 10.3390/ijms26062785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Inflammation appears to play a crucial role in the development and progression of penile cancer (PeCa). Two molecular pathways of PeCa are currently described: HPV-dependent and HPV-independent. The tumor immune microenvironment (TIME) of PeCa is characterized by the presence of tumor-associated macrophages, cancer-associated fibroblasts, and tumor-infiltrating lymphocytes. The components of the TIME produce pro-inflammatory cytokines and chemokines, which have been found to be overexpressed in PeCa tissues and are associated with tumor progression and unfavorable prognoses. Additionally, the nuclear factor kappa B (NF-κB) pathway and secreted phosphoprotein 1 (SPP1) have been implicated in PeCa pathogenesis. Elevated C-reactive protein (CRP) levels and the neutrophil-to-lymphocyte ratio (NLR) have been identified as potential prognostic biomarkers in PeCa. This overview presents the complex contribution of the inflammatory process and collates projects aimed at modulating TIME in PeCa.
Collapse
Affiliation(s)
- Mateusz Czajkowski
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Piotr M. Wierzbicki
- Department of Histology, Medical University of Gdańsk, Dębinki, 80-211 Gdansk, Poland;
| | - Maciej Dolny
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Marcin Matuszewski
- Department of Urology, Medical University of Gdańsk, Mariana Smoluchowskiego 17 Street, 80-214 Gdansk, Poland; (M.D.); (M.M.)
| | - Oliver W. Hakenberg
- Department of Urology, University Medical Center Rostock, 18055 Rostock, Germany;
- Department of Urology, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
20
|
Aaronson PI. The Role of Hydrogen Sulfide in the Regulation of the Pulmonary Vasculature in Health and Disease. Antioxidants (Basel) 2025; 14:341. [PMID: 40227402 PMCID: PMC11939758 DOI: 10.3390/antiox14030341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
The gasotransmitter hydrogen sulfide (H2S; also termed sulfide) generally acts as a vasodilator in the systemic vasculature but causes a paradoxical constriction of pulmonary arteries (PAs). In light of evidence that a fall in the partial pressure in oxygen (pO2) increases cellular sulfide levels, it was proposed that a rise in sulfide in pulmonary artery smooth muscle cells (PASMCs) is responsible for hypoxic pulmonary vasoconstriction, the contraction of PAs which develops rapidly in lung regions undergoing alveolar hypoxia. In contrast, pulmonary hypertension (PH), a sustained elevation of pulmonary artery pressure (PAP) which can develop in the presence of a diverse array of pathological stimuli, including chronic hypoxia, is associated with a decrease in the expression of sulfide -producing enzymes in PASMCs and a corresponding fall in sulfide production by the lung. Evidence that PAP in animal models of PH can be lowered by administration of exogenous sulfide has led to an interest in using sulfide-donating agents for treating this condition in humans. Notably, intracellular H2S exists in equilibrium with other sulfur-containing species such as polysulfides and persulfides, and it is these reactive sulfur species which are thought to mediate most of its effects on cells through persulfidation of cysteine thiols on proteins, leading to changes in function in a manner similar to thiol oxidation by reactive oxygen species. This review sets out what is currently known about the mechanisms by which H2S and related sulfur species exert their actions on pulmonary vascular tone, both acutely and chronically, and discusses the potential of sulfide-releasing drugs as treatments for the different types of PH which arise in humans.
Collapse
Affiliation(s)
- Philip I Aaronson
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9RT, UK
| |
Collapse
|
21
|
Dhalla NS, Ostadal P, Tappia PS. Involvement of Oxidative Stress and Antioxidants in Modification of Cardiac Dysfunction Due to Ischemia-Reperfusion Injury. Antioxidants (Basel) 2025; 14:340. [PMID: 40227421 PMCID: PMC11939711 DOI: 10.3390/antiox14030340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
Delayed reperfusion of the ischemic heart (I/R) is known to impair the recovery of cardiac function and produce a wide variety of myocardial defects, including ultrastructural damage, metabolic alterations, subcellular Ca2+-handling abnormalities, activation of proteases, and changes in cardiac gene expression. Although I/R injury has been reported to induce the formation of reactive oxygen species (ROS), inflammation, and intracellular Ca2+ overload, the generation of oxidative stress is considered to play a critical role in the development of cardiac dysfunction. Increases in the production of superoxide, hydroxyl radicals, and oxidants, such as hydrogen peroxide and hypochlorous acid, occur in hearts subjected to I/R injury. In fact, mitochondria are a major source of the excessive production of ROS in I/R hearts due to impairment in the electron transport system as well as activation of xanthine oxidase and NADPH oxidase. Nitric oxide synthase, mainly present in the endothelium, is also activated due to I/R injury, leading to the production of nitric oxide, which, upon combination with superoxide radicals, generates nitrosative stress. Alterations in cardiac function, sarcolemma, sarcoplasmic reticulum Ca2+-handling activities, mitochondrial oxidative phosphorylation, and protease activation due to I/R injury are simulated upon exposing the heart to the oxyradical-generating system (xanthine plus xanthine oxidase) or H2O2. On the other hand, the activation of endogenous antioxidants such as superoxide dismutase, catalase, glutathione peroxidase, and the concentration of a transcription factor (Nrf2), which modulates the expression of various endogenous antioxidants, is depressed due to I/R injury in hearts. Furthermore, pretreatment of hearts with antioxidants such as catalase plus superoxide dismutase, N-acetylcysteine, and mercaptopropionylglycerine has been observed to attenuate I/R-induced subcellular Ca2+ handling and changes in Ca2+-regulatory activities; additionally, it has been found to depress protease activation and improve the recovery of cardiac function. These observations indicate that oxidative stress is intimately involved in the pathological effects of I/R injury and different antioxidants attenuate I/R-induced subcellular alterations and improve the recovery of cardiac function. Thus, we are faced with the task of developing safe and effective antioxidants as well as agents for upregulating the expression of endogenous antioxidants for the therapy of I/R injury.
Collapse
Affiliation(s)
- Naranjan S. Dhalla
- St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Department of Physiology & Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R2H 2A6, Canada
| | - Petr Ostadal
- Department of Cardiology, 2nd Faculty of Medicine, Charles University, Motol University Hospital, V Uvalu 84, 15000 Prague, Czech Republic;
| | - Paramjit S. Tappia
- Asper Clinical Research Institute, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
| |
Collapse
|
22
|
Mbizvo GK, Buchan IE, Marson AG, Lip GYH. Epilepsy-heart syndrome: Concept, clinical context, and opportunity for integrated care. Epilepsia 2025. [PMID: 40079478 DOI: 10.1111/epi.18356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/15/2025]
Abstract
In this concept paper, we introduce epilepsy-heart syndrome as a shared burden of illness between epilepsy and cardiac disorders. This pragmatic definition is agnostic of which condition came first (the epilepsy or the cardiac disorder), recognising that these conditions can each serve as a risk factor for the other owing to a bidirectional relationship that exists between the brain and the heart. To provide clinical context, we include ictal asystole as an example phenotype of epilepsy-heart syndrome. We highlight evidence of patients with ictal asystole coming to harm owing to the failure of integrated care between neurology and cardiology. This underscores epilepsy-heart syndrome as an unmet need for collaborative care between neurology and cardiology. To address this, we propose a framework for integrated care, drawing upon our own centre's recently established and successful multidisciplinary team meeting (MDT) between neurologists and cardiologists, our joint cardiology-neurology PhD programme, and our work developing a joint national guideline on ictal asystole management between the Association of British Neurologists (ABN) and the British Heart Rhythm Society (BHRS).
Collapse
Affiliation(s)
- Gashirai K Mbizvo
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, UK
- Pharmacology and Therapeutics, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
- Walton Centre National Health Service (NHS) Foundation Trust, Liverpool, UK
| | - Iain E Buchan
- Department of Public Health, Policy, and Systems, Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Anthony G Marson
- Pharmacology and Therapeutics, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
- Walton Centre National Health Service (NHS) Foundation Trust, Liverpool, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
23
|
Huang Y, Zhao D, Yang Z, Wei C, Qiu X. The relationship between VAI, LAP, and depression and the mediation role of sleep duration-evidence from NHANES 2005-2020. BMC Psychiatry 2025; 25:228. [PMID: 40069662 PMCID: PMC11899296 DOI: 10.1186/s12888-025-06631-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The relationship between obesity and mental health has attracted attention. However, large sample studies on the relationship between visceral fat obesity and depression are lacking. This study aimed to explore the relationship between visceral fat obesity and depression by using visceral adiposity index (VAI) and lipid accumulation product (LAP). Additionally, it sought to explore the potential mediating role of sleep duration in these associations. METHODS The data used in the current cross-sectional investigation are from the National Health and Nutrition Examination Survey (NHANES) spanning from 2005 to 2020, including 19,659 participants. Depression was measured using the nine-item Patient Health Questionnaire. Weighted multivariable regression analysis was used to evaluate the correlation of VAI and LAP with depression. The potential non-linear relationship was determined using smooth curve fitting and threshold effect analysis. Additionally, mediation analysis was performed to investigate the potential mediating role of sleep duration. The stability of the relationship was assessed through sensitivity analysis. RESULTS VAI and LAP were closely related to depression. In the fully adjusted model, VAI and LAP in the highest quartile increased the association of depression by 52% (OR = 1.52, 95% CI 1.20-1.92, P < 0.001) and 51% (OR = 1.51, 95% CI 1.19-1.91, P < 0.001), respectively, compared with the lowest quartile. Specific saturation effects for VAI, LAP, and depression were identified by smoothed curve fitting, with inflection points of 3.81 and 98.55, respectively. Additionally, mediation analysis revealed that 5.1% and 2.8% of the associations between VAI and LAP with depression were mediated through sleep duration. The results of the sensitivity analysis showed interactions between hypertension and cardiovascular disease in the associations of VAI, and depression (P < 0.05). CONCLUSION VAI and LAP are associated with depression in US adults. The associations between VAI and LAP with depression are non-linear, which may be mediated through sleep duration. The study highlights the potential of VAI and LAP as valuable tools for the prevention and management of depression.
Collapse
Affiliation(s)
- Yiqing Huang
- Medical School, Shenzhen University, No.1066, Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong Province, 518060, People's Republic of China
| | - Dan Zhao
- Centre for Health Management and Policy Research, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhongfang Yang
- School of Nursing, Fudan University, 305 Fenglin Road, Shanghai, 200032, China
| | - Changning Wei
- School of Tech X Academy, Shenzhen Polytechnic University, Shenzhen, China
| | - Xichenhui Qiu
- Medical School, Shenzhen University, No.1066, Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong Province, 518060, People's Republic of China.
- Rory Meyers College of Nursing, New York University, New York, NY, 10010, USA.
| |
Collapse
|
24
|
Yu JF, Dong Q, Du YM. Interleukin-6: Molecular Mechanisms and Therapeutic Perspectives in Atrial Fibrillation. Curr Med Sci 2025:10.1007/s11596-025-00021-7. [PMID: 40035997 DOI: 10.1007/s11596-025-00021-7] [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: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/06/2025]
Abstract
Atrial fibrillation (AF) is a prevalent cardiac arrhythmia with a multifactorial pathophysiology involving electrical, structural, and autonomic remodeling of the atria. AF is closely associated with elevated interleukin-6 (IL-6) levels, which contribute to atrial remodeling and the progression of AF. This review summarizes the mechanisms by which IL-6 promotes AF through inflammatory pathways, atrial fibrosis, electrical remodeling, and calcium mishandling. Experimental models have demonstrated that IL-6 neutralization reduces the incidence of AF, highlighting its potential as a therapeutic target. Future studies should focus on IL-6 blockade strategies to manage AF, aiming to improve patient outcomes.
Collapse
Affiliation(s)
- Jin-Fang Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qian Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yi-Mei Du
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Research Center of Ion Channelopathy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Lab for Biological Targeted Therapy of Education Ministry and Hubei Province, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
25
|
Du X, Wang X, Zhang R, Chen Y, Chen Q, Yao J, Shi L, Ren Y. Standardized Rhodiola rosea injection for left ventricular remodeling and inflammation in patients with HFrEF: a systematic review and meta-analysis. Front Pharmacol 2025; 16:1536686. [PMID: 40098619 PMCID: PMC11911375 DOI: 10.3389/fphar.2025.1536686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Background Heart failure with reduced ejection fraction (HFrEF) affects a substantial portion of the global population, with left ventricular remodeling (LVR) and inflammation identified as key contributors to disease progression. Standardized Rhodiola rosea Injection (SRRI) is a pharmacopoeia-based botanical drug preparation derived from Rhodiola rosea, widely used in China for heart failure treatment. It is standardized in composition and quality control, with known antioxidant, anti-inflammatory, and anti-fibrotic properties. However, comprehensive evaluations of SRRI's effects on LVR and inflammatory mediators in HFrEF patients are limited. Purpose To evaluate the effects of SRRI on LVR and inflammatory mediators in patients with HFrEF. Methods A systematic review and meta-analysis were conducted following PRISMA and Cochrane guidelines. Eight databases were searched for randomized controlled trials (RCTs) on SRRI in HFrEF treatment with studies identified from inception to 31 October 2024. Quality assessment of the included studies was conducted using the Cochrane Collaboration's risk of bias tool and the modified Jadad scale. Statistical analysis was performed using Stata version 17.0, with sensitivity analyses conducted by sequentially excluding studies to assess the robustness of findings. Publication bias was evaluated using Egger's test. Results Twenty-five RCTs with 2,325 participants were included. SRRI significantly improved LVR, indicated by increased LVEF (MD = 6.81, 95% CI: 5.71 to 7.91, P < 0.00001), reduced LVEDD (MD = -4.37, 95% CI: -5.42 to -3.33, P < 0.00001), and decreased LVESD (MD = -4.48, 95% CI: -5.42 to -3.58, P < 0.00001). Additionally, SRRI effectively reduced inflammatory mediators, including TNF-α (MD = -10.37, 95% CI: -12.96 to -7.78, P < 0.00001), IL-6 (MD = -6.99, 95% CI: -8.88 to -5.11, P < 0.00001), and hs-CRP (MD = -2.58, 95% CI: -3.37 to -1.79, P < 0.00001). SRRI also significantly reduced BNP (MD = -105.10, 95% CI: -132.29 to -77.90, P < 0.00001) and NT-pro BNP (MD = -415.95, 95% CI: -553.00 to -278.89, P < 0.00001). Clinical effectiveness was improved, with no significant increase in adverse reactions (RR = 0.86, 95% CI: 0.59 to 1.25, P = 0.44). Sensitivity analyses confirmed the robustness of these findings, and no significant publication bias was detected. Conclusion SRRI appears to effectively enhance LVR, reduce inflammatory mediators, and improve clinical effectiveness in HFrEF patients while maintaining a favorable safety profile. However, the current evidence is limited by methodological shortcomings, and further well-designed, multicenter RCTs are needed to validate these findings, especially in diverse populations and over long-term treatment durations. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=603884, Identifier CRD42024603884.
Collapse
Affiliation(s)
- Xuqin Du
- School of Traditional Chinese Medicine, Chongqing University of Chinese Medicine, Chongqing, China
| | - Xiaorong Wang
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Ruodai Zhang
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Yong Chen
- Department of Classic Traditional Chinese Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Qian Chen
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Jing Yao
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Lipeng Shi
- Department of Classic Traditional Chinese Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Yi Ren
- Department of Classic Traditional Chinese Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
26
|
Astiawati T, Rohman MS, Wihastuti T, Sujuti H, Endharti A, Sargowo D, Oceandy D, Lestari B, Triastuti E, Nugraha RA. The Emerging Role of Colchicine to Inhibit NOD-like Receptor Family, Pyrin Domain Containing 3 Inflammasome and Interleukin-1β Expression in In Vitro Models. Biomolecules 2025; 15:367. [PMID: 40149903 PMCID: PMC11940210 DOI: 10.3390/biom15030367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/22/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
While the beneficial effects of colchicine on inflammation and infarcted myocardium have been documented, its impact on cardiac fibroblast activation in the context of myocardial infarction (MI) remains unknown. This study aimed to investigate the effect of colchicine on the regulation of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation and Interleukin-1β (IL-1β) expression in fibroblasts. 3T3 fibroblasts were exposed to 600 μM CoCl2 for 24 h to simulate hypoxia, with normoxic cells as controls. Colchicine (1 μM) was administered for 24 h. ASC-NLRP3 colocalization and IL-1β expression were evaluated using immunofluorescence and flow cytometry, respectively. Data were analyzed using t-tests and one-way ANOVA with post hoc tests. Hypoxia treatment significantly induced apoptosis-associated speck-like protein containing a CARD (ASC)-NLRP3 colocalization (p < 0.05). Colchicine treatment of hypoxic 3T3 cells reduced ASC-NLRP3 colocalization, although this reduction was not statistically significant. Additionally, IL-1β expression was significantly inhibited in colchicine-treated hypoxic 3T3 cells compared to those treated with placebo (p < 0.05). The findings of this study indicate that colchicine treatment inhibits the activation of the NLRP3 inflammasome by disrupting the colocalization of ASC and NLRP3, thereby reducing IL-1β expression in CoCl2-treated 3T3 cells.
Collapse
Affiliation(s)
- Tri Astiawati
- Doctoral Program of Medical Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
- Department of Cardiology, Doctor Iskak General Hospital, Tulungagung 62233, Indonesia
| | - Mohammad Saifur Rohman
- Department of Cardiology and Cardiovascular Medicine, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
- Cardiovascular Research Centre, Brawijaya University, Malang 65145, Indonesia
| | - Titin Wihastuti
- Department of Biomedical, Nursing Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Hidayat Sujuti
- Department of Biochemistry, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Agustina Endharti
- Doctoral Program of Medical Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Djanggan Sargowo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Delvac Oceandy
- Division of Cardiovascular Science, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Bayu Lestari
- Department of Pharmacology, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Efta Triastuti
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Ricardo Adrian Nugraha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Airlangga—Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia;
| |
Collapse
|
27
|
Wang F, Zhou J, Liu W, Wang W, Tian B, Liu J, Zhang H, He P, Yang X, Yang L, Wang Y. Mogrol Regulates the Expression of ATPase Na+/K+ Transport Subunit 3, Inhibits Cardiomyocyte Apoptosis, and Plays a Protective Role Against Myocardial Infarction. Drug Des Devel Ther 2025; 19:1489-1502. [PMID: 40051467 PMCID: PMC11884411 DOI: 10.2147/dddt.s490484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
Background With the advancements in medical technology, the death rate from myocardial infarction (MI), a prevalent heart illness, has gradually decreased; however, treatment hurdles and diagnostic issues remain. Mogrol is a naturally occurring plant extract with specific biological activities such as antioxidant, anti-inflammatory, antitumor, and hypoglycemic effects. These biological activities make it a potential therapeutic drug or research subject; however, its function in MI remains unclear. Methods Potential targets of mogrol were searched using the MI Disease Database through online databases. Among the three intersecting genes, we focused on ATPase Na+/K+ transporting subunit 3A3, which is expressed at low levels in patients with MI. The preventive effect of mogrol against MI was investigated using cardiac ultrasonography, Western blotting, qPCR assay, Cell counting kit-8, Ca2+ concentration measurement, Na+/K+-ATPase, and flow cytometry. Results The findings demonstrated that mogrol upregulated Ca2+ concentration and ATPase Na+/K+ transporting subunit 3 protein levels in cardiomyocytes and tissues, downregulated the apoptosis-related proteins B-cell lymphoma 2-like protein 4, cleaved-caspase-3, and upregulated B-cell lymphoma 2. These effects enhanced cardiac function, prevented cardiomyocyte apoptosis, encouraged cardiomyocyte proliferation, and protected mice from MI. Knocking down ATP1A3 can reverse the protective effect of Mogrol. Conclusion Mogrol may have a protective effect on myocardial infarction by regulating Ca2+ concentration and the level of the ATPase Na+/K+ transport subunit 3 protein, as well as by regulating apoptosis-related proteins. Further revealing the pharmacokinetics of mogrol in vivo is expected to make it a subsequent drug for the treatment of cardiac infarction.
Collapse
Affiliation(s)
- Feng Wang
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Jinling Zhou
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Weiwei Liu
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Wei Wang
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Boyan Tian
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Jinyu Liu
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Han Zhang
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Peina He
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Xiaoyun Yang
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Li Yang
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| | - Yueheng Wang
- Echocardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, People’s Republic of China
| |
Collapse
|
28
|
Blagov AV, Churov AV, Starodubtseva IA, Kovyanova TI, Pecherina TB, Sukhorukov VN. Cytokines are the Basis of the Development and Suppression of Inflammation in Atherosclerosis. Rev Cardiovasc Med 2025; 26:26421. [PMID: 40160593 PMCID: PMC11951496 DOI: 10.31083/rcm26421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 04/02/2025] Open
Abstract
Cardiovascular diseases continue to be the primary cause of mortality in industrialised countries, and atherosclerosis plays a role in their development. A persistent inflammatory condition affecting big and medium-sized arteries is known as atherosclerosis. It is brought on by dyslipidemia and is facilitated by the immune system's innate and adaptive components. At every stage of the progression of atherosclerosis, inflammation plays a crucial role. It has been demonstrated that soluble factors, or cytokines, activate cells involved in the pathophysiology of atherosclerosis and have a significant impact on disease progression. Anti-inflammatory cytokines (such as interleukin (IL)-5 and IL-13) mitigate atherosclerosis, whereas pro-inflammatory cytokines (such as IL-1, IL-6) quicken the disease's course. Of interest is the fact that a number of cytokines can exhibit both atherogenic and atheroprotective properties, which is the topic of study and discussion in this review. This review provides a comparative analysis of the functions of the main cytokines involved in the pathogenesis of atherosclerosis. Their functional relationships with each other are also shown. In addition, potential therapeutic strategies targeting these cytokines for the treatment of atherosclerosis are proposed, with an emphasis on recent clinical research in this area.
Collapse
Affiliation(s)
| | - Alexey V. Churov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute on Aging Research, Russian Gerontology Clinical Research Centre, Pirogov Russian National Research Medical University, 129226 Moscow, Russia
| | - Irina A. Starodubtseva
- Department of Polyclinic Therapy, NN Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Tatiana I. Kovyanova
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute for Atherosclerosis Research, 121609 Moscow, Russia
| | - Tamara B. Pecherina
- Laboratory for Cardiac Fibrosis, Research Institute for Complex Issues of Cardiovascular Diseases, 650002 Kemerovo, Russia
| | - Vasily N. Sukhorukov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute of Experimental Cardiology, Russian Medical Research Center of Cardiology, 121552 Moscow, Russia
| |
Collapse
|
29
|
Torres EM, Tellechea ML. Biomarkers of endothelial dysfunction and cytokine levels in hypothyroidism: a series of meta-analyses. Expert Rev Endocrinol Metab 2025; 20:119-128. [PMID: 39676305 DOI: 10.1080/17446651.2024.2438997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Hypothyroidism (HT) is associated with different comorbidities comprising increased arterial stiffness and decreased flow-mediated dilatation. The exact pathological mechanism of endothelial activation and dysfunction (ED) in HT remains unknown. We conducted a systematic review and meta-analyses to provide an overview of the pathogenesis of ED in HT. METHODS The literature search was done in February 2024 for studies analyzing traditional and novel circulating biomarkers of ED in patients with HT, including cytokines and chemokines. Random-effect models were used except when no heterogeneity was found. Protocol was registered under the number PROSPERO CRD42024540560. RESULTS 25 macromolecules and 66 studies were entered into analyses. HT was associated with increased levels of E-selectin, soluble intercellular adhesion molecule-1, osteoprotegerin, and oxidized-LDL (p < 0.02). Results were not conclusive for endothelin-1. Interleukin (IL)-6, IL-12 and CXCL10 were higher in HT (p < 0.05). Subjects with overt HT may display a proinflammatory tendency with increased levels of IL-6 and interferon-γ, and decreased levels of TGF-β (p < 0.05). CONCLUSIONS The data presented and discussed here highlights the association between HT and soluble biomarkers of ED. Inflammatory mediators released by activated T-cells and macrophages may aggravate local and systemic inflammation, which arouses more inflammation, forming a vicious circle leading to ED.
Collapse
Affiliation(s)
- Emiliana María Torres
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá", Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación de Endocrinología Infantil - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Mariana Lorena Tellechea
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá", Consejo Nacional de Investigaciones Científicas y Técnicas - Fundación de Endocrinología Infantil - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| |
Collapse
|
30
|
Wu J, Huang D, Li J, Yi J, Lei Y, Yin J. Predicting cardiovascular disease and all-cause mortality using the lymphocyte-to-monocyte ratio: Insights from explainable machine learning models. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2025; 24:200372. [PMID: 39995422 PMCID: PMC11849660 DOI: 10.1016/j.ijcrp.2025.200372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/24/2024] [Accepted: 01/26/2025] [Indexed: 02/26/2025]
Abstract
Background Cardiovascular disease (CVD) is a leading cause of death globally, with its incidence and mortality rates continuing to rise. While commonly used biomarkers such as low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and blood glucose are widely applied, they have certain limitations. This study investigates the lymphocyte-to-monocyte ratio (LMR), a simple immune biomarker associated with inflammation, to assess whether it can serve as a new marker for predicting chronic inflammation in cardiovascular disease, and compares it to traditional biomarkers. Methods We conducted a cross-sectional analysis of data from the National Health and Nutrition Examination Survey (NHANES) from 2007 to 2018, utilizing a cohort of 1518 participants with a median follow-up period of 150 months. During this time, 522 participants died, including 166 from cardiovascular disease. We employed various statistical methods, including weighted Cox proportional hazards models, restricted cubic spline models, and time-varying receiver operating characteristic curves, to examine the association between LMR and mortality risk. Results The analysis revealed an L-shaped relationship between LMR and the incidence of cardiovascular disease. Lower LMR levels were negatively correlated with all-cause and cardiovascular mortality. The XGBoost model yielded the best performance metrics (AUC and F1 scores), and SHAP value analysis indicated that LMR significantly contributes to CVD outcomes. Non-linear analyses confirmed a stable negative correlation between LMR and all-cause mortality. Conclusion The study concludes that LMR is a simple and practical indicator for predicting cardiovascular disease and its mortality. Low levels of LMR significantly increase the risk of both cardiovascular disease and all-cause mortality in patients.
Collapse
Affiliation(s)
- Jichao Wu
- Department of Laboratory, Second Affiliated Hospital of Shantou University Medical College, 69 Dongxia North Road, Shantou, 515041, China
| | - Die Huang
- Department of Laboratory, Second Affiliated Hospital of Shantou University Medical College, 69 Dongxia North Road, Shantou, 515041, China
| | - Jiefang Li
- Department of Laboratory, Second Affiliated Hospital of Shantou University Medical College, 69 Dongxia North Road, Shantou, 515041, China
| | - Jingxing Yi
- Department of Laboratory, Second Affiliated Hospital of Shantou University Medical College, 69 Dongxia North Road, Shantou, 515041, China
| | - Yu Lei
- Department of Hematology, Second Affiliated Hospital of Shantou University Medical College, 69 Dongxia North Road, Shantou, 515041, China
| | - Jun Yin
- Department of Laboratory, Second Affiliated Hospital of Shantou University Medical College, 69 Dongxia North Road, Shantou, 515041, China
| |
Collapse
|
31
|
Del Monte A, Sarkozy A, Verbrugge FH. Atrial Fibrillation Management with Guideline-Directed Medical Therapy and Comorbidity Treatment in Heart Failure Patients. Card Electrophysiol Clin 2025; 17:63-73. [PMID: 39893038 DOI: 10.1016/j.ccep.2024.09.001] [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] [Indexed: 02/04/2025]
Abstract
Atrial myopathy is the underlying pathophysiological substrate of atrial fibrillation and contributes to the risk of heart failure as well. Atrial myopathy is caused by classic risk factors such as obesity, inflammation, diabetes, hypertension, and frequent alcohol use, in addition to structural heart and lung diseases that cause atrial pressure or volume overload. An optimal management of atrial fibrillation includes careful assessment of contributors to atrial myopathy, which can be treated by guideline-recommended medical therapies for heart failure, adequate control of congestion, and treatment of comorbid conditions such as sleep apnea syndrome. This approach works synergistically with rhythm control.
Collapse
Affiliation(s)
- Alvise Del Monte
- Heart Rhythm Management Centre, University Hospital Brussels, Jette, Belgium
| | - Andrea Sarkozy
- Heart Rhythm Management Centre, University Hospital Brussels, Jette, Belgium; Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Frederik H Verbrugge
- Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium; Centre for Cardiovascular Diseases, University Hospital Brussels, Laarbeeklaan 101, Jette 1090, Belgium.
| |
Collapse
|
32
|
Fan D, Feng H, Song M, Tan P. Gene expression profiles, potential targets and treatments of cardiac remodeling. Mol Cell Biochem 2025; 480:1555-1567. [PMID: 39367915 DOI: 10.1007/s11010-024-05126-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/24/2024] [Indexed: 10/07/2024]
Abstract
Hypertensive and ischemic heart diseases have high morbidity all over the world, and they primarily contribute to heart failure associated with high mortality. Cardiac remodeling, as a basic pathological process in heart diseases, is mainly comprised of cardiac hypertrophy and fibrosis, as well as cell death which occurs especially in the ischemic cardiomyopathy. Myocardial remodeling has been widely investigated by a variety of animal models, including pressure overload, angiotensin II stimulation, and myocardial infarction. Pressure overload can cause compensatory cardiac hypertrophy at the early stage, followed by decompensatory hypertrophy and heart failure at the end. Recently, RNA sequencing and differentially expressed gene (DEG) analyses have been extensively employed to elucidate the molecular mechanisms of cardiac remodeling and related heart failure, which also provide potential targets for high-throughput drug screenings. In this review, we summarize recent advancements in gene expression profiling, related gene functions, and signaling pathways pertinent to myocardial remodeling induced by pressure overload at distinct stages, ischemia-reperfusion, myocardial infarction, and diabetes. We also discuss the effects of sex differences and inflammation on DEGs and their transcriptional regulatory mechanisms in cardiac remodeling. Additionally, we summarize emerging therapeutic agents and strategies aimed at modulating gene expression profiles during myocardial remodeling.
Collapse
Affiliation(s)
- Dong Fan
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China.
| | - Han Feng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Mengyu Song
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Penglin Tan
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| |
Collapse
|
33
|
Yao H, Yu D, He Q, Ning X. Causal associations of inflammatory cytokines and urinary stones: a two-sample Mendelian randomization study. Transl Androl Urol 2025; 14:258-265. [PMID: 40114831 PMCID: PMC11921299 DOI: 10.21037/tau-24-445] [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: 08/24/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Background Prior research has shown potential changes in cytokine and growth factor levels in patients with urinary stones, but the causal relationship remains uncertain. The purpose of this study was to investigate the causal relationship between cytokine and growth factor levels and urinary tract stones. Methods This study used a two-sample Mendelian randomization (MR) with 41 circulating cytokines and growth factors datasets alongside urinary stone disease (USD) data. It employed various analytical methods including inverse variance weighted (IVW) analysis, maximum likelihood estimation, MR-Egger regression, weighted median approach, simple median method, and weighted mode to establish causal relationships. Sensitivity analysis included the MR-Egger regression intercept test and Cochrane's Q statistic. Results Using the IVW method, an increase in stem cell growth factor β levels was associated with decreased urinary stone risk [odds ratio (OR) =0.9990; 95% confidence interval (CI): 0.9980-0.9999; P=0.04]. Conversely, an increase in interleukin-18 levels elevated the risk of urinary stones (OR =1.0012; 95% CI: 1.0002-1.0022; P=0.01). Various analytical methods consistently supported these findings. Conclusions Our findings suggest a causal and unidirectional relationship between interleukin-18, stem cell growth factors, and USD. This indicates that these cytokines may actively contribute to the development or prevention of USD, offering a new avenue for clinical intervention based on cytokines modulation.
Collapse
Affiliation(s)
- Huan Yao
- Nephrology Department, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Nephrology Department, Guizhou Hospital of The First Affiliated Hospital of Sun Yat-sen University, Guiyang, China
| | - Dan Yu
- Nephrology Department, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qingyun He
- Nephrology Department, Guizhou Hospital of The First Affiliated Hospital of Sun Yat-sen University, Guiyang, China
| | - Xiaoya Ning
- Nephrology Department, Guizhou Hospital of The First Affiliated Hospital of Sun Yat-sen University, Guiyang, China
| |
Collapse
|
34
|
Hill AP, Iqbal SB, Case BC, Shankar AA, Merdler I. Acute Coronary Syndrome and Rheumatic Disease. J Clin Med 2025; 14:1490. [PMID: 40094966 PMCID: PMC11900616 DOI: 10.3390/jcm14051490] [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: 01/08/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Patients with rheumatic disease and immune disorders have been noted to show an earlier development of atherosclerosis and to present with acute coronary syndrome. These diseases disproportionately affect women, and patients frequently have a higher number of comorbidities and other risk factors. Inflammation has long been known to play a role in the development of coronary artery disease. Early studies have shown some benefit in treatment targeting inflammation. While this has not been realized for all populations, there remains potential in treatment with targeted and individualized therapies. Especially since these diseases are associated with a worse prognosis, management benefits from the multidisciplinary expertise of cardiologists, rheumatologists, and other providers. However, the prevention and treatment of underlying rheumatic disease remains essential. This review will seek to highlight prior studies and future directions in the treatment of acute coronary syndrome in patients with rheumatologic disease.
Collapse
Affiliation(s)
- Andrew P. Hill
- Section of Cardiology, MedStar Washington Hospital Center and Georgetown University, Washington, DC 20007, USA;
| | - Shaikh B. Iqbal
- Section of Internal Medicine, MedStar Union Memorial Hospital and Georgetown University, Washington, DC 20007, USA;
| | - Brian C. Case
- Section of Interventional Cardiology, MedStar Washington Hospital Center, Washington, DC 20010, USA;
| | | | | |
Collapse
|
35
|
Sivalingam AM, Sureshkumar DD, Pandurangan V. Cerebellar pathology in forensic and clinical neuroscience. Ageing Res Rev 2025; 106:102697. [PMID: 39988260 DOI: 10.1016/j.arr.2025.102697] [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: 11/19/2024] [Revised: 01/30/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Recent research underscores the cerebellum's growing importance in forensic science and neurology, showing its functions extend beyond motor control, especially in identifying causes of death. Critical neuropathological markers including alpha-synuclein and tau protein aggregates, cellular degeneration, inflammation, and vascular changes are vital for identifying neurodegenerative diseases, injuries, and toxic exposures. Advanced forensic methods, such as Magnetic resonance imaging (MRI), immunohistochemistry, and molecular analysis, have greatly improved the accuracy of diagnoses. Promising new therapies, including neuroprotective agents like resveratrol and transcranial magnetic stimulation (TMS), offer potential in treating cerebellar disorders. The cerebellum's vulnerability to toxins, drugs, and traumatic brain injuries (TBIs) highlights its forensic relevance. Moreover, advancements in genetic diagnostics, such as next-generation sequencing and CRISPR-Cas9, are enhancing the understanding and treatment of genetic conditions like Joubert syndrome and Dandy-Walker malformation. These findings emphasize the need for further research into cerebellar function and its broader significance in both forensic science and neurology.
Collapse
Affiliation(s)
- Azhagu Madhavan Sivalingam
- Natural Products & Nanobiotechnology Research Lab, Department of Community Medicine, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), (Saveetha University), Thandalam, Chennai, Tamil Nadu 602 105, India.
| | - Darshitha D Sureshkumar
- Department of Forensic Science, NIMS Institute of Allied Medical Science and Technology, (NIMS University), Jaipur, Rajasthan 303121, India
| | - Vijayalakshmi Pandurangan
- Department of Radiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), (Saveetha University), Thandalam, Chennai-602 105, Tamil Nadu, India
| |
Collapse
|
36
|
Ivanova MM, Dao J, Friedman A, Kasaci N, Goker-Alpan O. Sex Differences in Circulating Inflammatory, Immune, and Tissue Growth Markers Associated with Fabry Disease-Related Cardiomyopathy. Cells 2025; 14:322. [PMID: 40072051 PMCID: PMC11899294 DOI: 10.3390/cells14050322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/15/2025] Open
Abstract
Fabry disease (FD) is a lysosomal disorder due to alpha-galactosidase-A enzyme deficiency, accumulation of globotriaosylceramide (Gb3) and globotriaosylsphingosine (lyso-Gb3) which lead to proinflammatory effects. Males develop progressive hypertrophic cardiomyopathy (HCM) followed by fibrosis; females develop nonconcentric hypertrophy and/or early fibrosis. The inflammatory response to Gb3/lyso-Gb-3 accumulation is one of the suggested pathogenic mechanisms in FD cardiomyopathy when the secretion of inflammatory and transforming growth factors with infiltration of lymphocytes and macrophages into tissue promotes cardiofibrosis. This study aims to evaluate inflammation-driving cytokines and cardio-hypertrophic remodeling biomarkers contributing to sex-specific HCM progression. Biomarkers were studied in 20 healthy subjects and 45 FD patients. IL-2, IL-10, TNF-α, and IFN-γ were elevated in all patients, while IL-1α, MCP-1, and TNFR2 showed sex-specific differences. The increased cytokines were associated with the NF-kB pathway in FD males with HCM, revealing a correlation between MCP-1, IFN-γ, VEGF, GM-CSF, IL-10, and IL-2. In female patients, the impaired TNFα/TNFR2/TGFβ cluster with correlations to MCP-1, VEGF, GM-CSF, and IL-1α was observed. The activation of cytokines and the NF-kB pathway indicates significant inflammation during HCM remodeling in FD males. The TNFα/TNFR2/TGFβ signaling cluster may explain early fibrosis in females with FD cardiomyopathy. Sex-specific inflammatory responses in FD influence the severity and progression of HCM.
Collapse
Affiliation(s)
- Margarita M. Ivanova
- Lysosomal and Rare Disorders Research and Treatment Center, Fairfax, VA 22030, USA (O.G.-A.)
| | | | | | | | | |
Collapse
|
37
|
Xu Z, Wang Y, Li X, Hou X, Yue S, Wang J, Ye S, Wu J. Interacting and joint effects of frailty and inflammation on cardiovascular disease risk and the mediating role of inflammation in middle-aged and elderly populations. BMC Cardiovasc Disord 2025; 25:118. [PMID: 39979798 PMCID: PMC11841180 DOI: 10.1186/s12872-025-04567-1] [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: 09/02/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Frailty and inflammation may increase the risk of cardiovascular disease (CVD), but their interacting and joint effects on CVDs remain unclear. To explore the interaction effects of frailty and inflammation on CVDs and the role of inflammation in the relationship between frailty and CVDs to provide better understanding of the underlying pathogenesis of CVD. METHODS A total of 220,608 initially CVD-free participants were recruited from the UK Biobank database and were categorized into non-frailty, pre-frailty, and frailty groups based on Fried's criteria. The participants were also grouped according to the low-grade inflammation (INFLA) score and its components: the neutrophil-lymphocyte ratio, C-reactive protein, white blood cell count, and platelet count. Cox proportional hazards models with hazard ratios (HRs) and 95% confidence intervals (CIs) were used to assess the effects of frailty phenotypes and inflammation on CVD risk. Mediation analysis was used to quantify the role of inflammation in the association between frailty and CVDs. The potential interactions between frailty and inflammation in terms of CVD risk were also evaluated using additive and multiplicative scales. RESULTS During a median follow-up of 13.3 years, 48,978 participants developed CVDs. After adjusting for various confounders, participants with pre-frailty and frailty had a higher risk of CVDs than those with non-frailty (HRs: 1.20 (95% CI: 1.18-1.23) and 1.80 (95% CI: 1.69-1.91), respectively). A higher risk of CVDs was observed among participants with moderate and high INFLA scores than those with low INFLA scores (HRs: 1.09 (95% CI: 1.07-1.12) and 1.27 (95% CI: 1.24-1.30), respectively). The INFLA score and its components had limited mediating effects in the association between frailty and CVDs. Significant interactions were observed between frailty phenotypes and INFLA scores on CVDs on the multiplicative scale but not on the additive scale. CONCLUSION Inflammation may amplify the harmful effect of frailty on the incidence of CVDs. Improving frailty alone might not substantially reduce the risk of CVDs, but effectively controlling inflammation might help to reduce the negative effects of frailty on cardiovascular health.
Collapse
Affiliation(s)
- Zihan Xu
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Yingbai Wang
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Xiaolin Li
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Xuefei Hou
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Suru Yue
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Jia Wang
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China
| | - Shicai Ye
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| | - Jiayuan Wu
- Clinical Research Service Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, China.
| |
Collapse
|
38
|
Zhao HQ, Lv JL, Gao YZ, Hu B, Du ZD, Wang Y, Wang ML, Hou MD, Li F, Xing X, Sun MH. Association between inflammatory score, healthy lifestyle, and cardiovascular disease: a national cohort study. Front Nutr 2025; 12:1534458. [PMID: 40051968 PMCID: PMC11882400 DOI: 10.3389/fnut.2025.1534458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
Background The inflammation score is currently regarded as a reliable composite index for comprehensive assessment of inflammatory status. However, the relationship between inflammation score and cardiovascular disease (CVD) is unclear. Thus, we aimed to explore the association of inflammatory score with CVD, as well as to evaluate whether adhering to a healthy lifestyle could alleviate this association. Methods We analyzed 6,164 participants aged ≥45 years who entered a prospective cohort study of the China Longitudinal Study of Health and Retirement (CHARLS) between 2011 and 2012 and were followed up for CVD incidence untill 2018. The inflammatory score was measured by summing of the Z-scores for C-reactive protein and white blood cell count at baseline. The healthy lifestyle score was calculated by four factors, smoking status, alcohol consumption, body mass index, and sleep duration. Cox proportional hazard models were utilized to calculate the hazard ratios (HRs) and 95% confidence intervals (CIs) for the incidence of CVD. Results During the 7-year follow-up period, there were 761 incident cases of CVD. Compared with the lowest tertiles, the highest inflammatory score was associated with an elevated risk of CVD (HR = 1.25, 95% CI = 1.04-1.49). Compared to the unhealthy lifestyle, participants adhered to a healthy lifestyle was inversely associated with CVD risk (HR = 0.74, 95% CI = 0.60-0.93). Of note, when participants adhered to a healthy lifestyle, the higher inflammatory score was no longer significantly correlated with CVD risk (HR = 1.00, 95% CI = 0.76-1.34). Additionally, a multiplicative interaction was detected between inflammatory score and healthy lifestyle score for CVD risk (p interaction <0.05). Conclusion The inflammation score was associated with higher risk of CVD incidence, but adherence to a healthy lifestyle may mitigate the adverse association of inflammation score and CVD among the middle-aged and older participants.
Collapse
Affiliation(s)
- Han-Qing Zhao
- Hunan Provincial Center for Disease Control and Prevention, Changsha, China
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jia-Le Lv
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan-Zhi Gao
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Bo Hu
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zong-Da Du
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Provincial Center for Disease Control and Prevention, Shenyang, China
| | - Yan Wang
- The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mei-Lin Wang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
| | - Meng-Di Hou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang Li
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xue Xing
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ming-Hui Sun
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
39
|
Sabit H, Arneth B, Altrawy A, Ghazy A, Abdelazeem RM, Adel A, Abdel-Ghany S, Alqosaibi AI, Deloukas P, Taghiyev ZT. Genetic and Epigenetic Intersections in COVID-19-Associated Cardiovascular Disease: Emerging Insights and Future Directions. Biomedicines 2025; 13:485. [PMID: 40002898 PMCID: PMC11852909 DOI: 10.3390/biomedicines13020485] [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: 12/19/2024] [Revised: 01/23/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
The intersection of COVID-19 and cardiovascular disease (CVD) has emerged as a significant area of research, particularly in understanding the impact of antiplatelet therapies like ticagrelor and clopidogrel. COVID-19 has been associated with acute cardiovascular complications, including myocardial infarction, thrombosis, and heart failure, exacerbated by the virus's ability to trigger widespread inflammation and endothelial dysfunction. MicroRNAs (miRNAs) play a critical role in regulating these processes by modulating the gene expressions involved in platelet function, inflammation, and vascular homeostasis. This study explores the potential of miRNAs such as miR-223 and miR-126 as biomarkers for predicting resistance or responsiveness to antiplatelet therapies in COVID-19 patients with cardiovascular disease. Identifying miRNA signatures linked to drug efficacy could optimize treatment strategies for patients at high risk of thrombotic events during COVID-19 infection. Moreover, understanding miRNA-mediated pathways offers new insights into how SARS-CoV-2 exacerbates CVD, particularly through mechanisms like cytokine storms and endothelial damage. The findings of this research could lead to personalized therapeutic approaches, improving patient outcomes and reducing mortality in COVID-19-associated cardiovascular events. With global implications, this study addresses the urgent need for effective management of CVD in the context of COVID-19, focusing on the integration of molecular biomarkers to enhance the precision of antiplatelet therapy.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Afaf Altrawy
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Aysha Ghazy
- Department of Agri-Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Rawan M. Abdelazeem
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Amro Adel
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza P.O. Box 77, Egypt
| | - Amany I. Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman bin Faisal University, Dammam 31441, Saudi Arabia
| | - Panos Deloukas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK;
| | - Zulfugar T. Taghiyev
- Department of Cardiovascular Surgery, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
40
|
Zhou K, Qin Q, Lu J. Pathophysiological mechanisms of ARDS: a narrative review from molecular to organ-level perspectives. Respir Res 2025; 26:54. [PMID: 39948645 PMCID: PMC11827456 DOI: 10.1186/s12931-025-03137-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) remains a life-threatening pulmonary condition with persistently high mortality rates despite significant advancements in supportive care. Its complex pathophysiology involves an intricate interplay of molecular and cellular processes, including cytokine storms, oxidative stress, programmed cell death, and disruption of the alveolar-capillary barrier. These mechanisms drive localized lung injury and contribute to systemic inflammatory response syndrome and multiple organ dysfunction syndrome. Unlike prior reviews that primarily focus on isolated mechanisms, this narrative review synthesizes the key pathophysiological processes of ARDS across molecular, cellular, tissue, and organ levels. MAIN BODY By integrating classical theories with recent research advancements, we provide a comprehensive analysis of how inflammatory mediators, metabolic reprogramming, oxidative stress, and immune dysregulation synergistically drive ARDS onset and progression. Furthermore, we critically evaluate current evidence-based therapeutic strategies, such as lung-protective ventilation and prone positioning, while exploring innovative therapies, including stem cell therapy, gene therapy, and immunotherapy. We emphasize the significance of ARDS subtypes and their inherent heterogeneity in guiding the development of personalized treatment strategies. CONCLUSIONS This narrative review provides fresh perspectives for future research, ultimately enhancing patient outcomes and optimizing management approaches in ARDS.
Collapse
Affiliation(s)
- Kaihuan Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Qianqian Qin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
| |
Collapse
|
41
|
Maidana D, Arroyo-Álvarez A, Barreres-Martín G, Arenas-Loriente A, Cepas-Guillen P, Brigolin Garofo RT, Caravaca-Pérez P, Bonanad C. Targeting Inflammation and Iron Deficiency in Heart Failure: A Focus on Older Adults. Biomedicines 2025; 13:462. [PMID: 40002874 PMCID: PMC11853203 DOI: 10.3390/biomedicines13020462] [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: 12/23/2024] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Heart failure (HF) is a leading cause of morbidity and mortality worldwide, with a higher prevalence among older adults. Iron deficiency (ID), affecting up to 50% of HF patients, is closely linked to chronic inflammation, exacerbating HF outcomes. This review aims to explore the interplay between inflammation, ID, and HF, focusing on older patients, and to identify therapeutic gaps and emerging treatment strategies. Methods: A comprehensive review of the literature was conducted, emphasizing the pathophysiological mechanisms of inflammation and ID in HF, the challenges of current diagnostic criteria, and the limitations of available treatments. Emerging pharmacological and diagnostic approaches were analyzed. Results: Chronic inflammation in HF, particularly in older adults, promotes functional ID through elevated hepcidin levels, impairing iron availability and worsening anemia. Current diagnostic criteria, relying heavily on ferritin, often misclassify ID due to inflammation. Intravenous (IV) iron therapy shows clinical benefits in patients with <50% left ventricular ejection fraction (LVEF), but the evidence is limited in heart failure with preserved ejection fraction (HFpEF). Emerging therapies, such as Sodium-Glucose Cotransporter-2 inhibitors (SGLT2is) and prolyl hydroxylase inhibitors like Roxadustat, offer promising avenues to improve iron metabolism and outcomes. Conclusions: ID and inflammation significantly impact HF progression, particularly inolder adults. Refining diagnostic criteria and exploring innovative therapies are critical to addressing these challenges. Future research should prioritize personalized approaches targeting inflammation and ID, especially in underrepresented populations, such as HFpEF and elderly patients.
Collapse
Affiliation(s)
- Daniela Maidana
- INCLIVA—Biomedical Research Institute, 46010 Valencia, Spain; (D.M.); (A.A.-Á.)
| | | | | | | | - Pedro Cepas-Guillen
- Quebec Heart & Lung Institute, Laval University, Quebec City, QC G1V 4G5, Canada;
| | | | - Pedro Caravaca-Pérez
- Cardiology Department, Hospital Clínic Barcelona, 08036 Barcelona, Spain (P.C.-P.)
| | - Clara Bonanad
- INCLIVA—Biomedical Research Institute, 46010 Valencia, Spain; (D.M.); (A.A.-Á.)
- Cardiology Department, University Clinical Hospital of Valencia, 46010 Valencia, Spain
| |
Collapse
|
42
|
Gao X, Ni C, Song Y, Xie X, Zhang S, Chen Y, Wu H, Shi H, Zhang B, Huang F, Wang C, Wu X. Dan-shen Yin attenuates myocardial fibrosis after myocardial infarction in rats: Molecular mechanism insights by integrated transcriptomics and network pharmacology analysis and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119070. [PMID: 39522849 DOI: 10.1016/j.jep.2024.119070] [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: 09/14/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dan-shen Yin (DSY) originated from the "Shi Fang Ge Kuo" is a Chinese formula composed of three medicines: Salvia miltiorrhiza (Dan-shen), Santalum album L. (Tan-xiang) and Amomum villosum Lour. (Sha-ren). It has many years of clinical experience in the prevention of myocardial fibrosis (MF). However, the specific mechanism of DSY in prevent MF is not clear. AIM OF THE STUDY This study aimed to assess the efficacy of DSY in the prevention of MF and reveal its underlying mechanism in a rat model of MF after myocardial infarction (MI) induced by ligation of the left anterior descending branch (LAD) of the coronary artery. MATERIALS AND METHODS The blood-entry components of DSY were analyzed by UHPLC-Q-TOF-MS/MS. LAD-ligated rats were used to assess the efficacy of DSY in the prevention of MF. Network pharmacology and transcriptomics analysis were used to predict possible target signaling pathways of DSY in MF. Echocardiography, immunohistochemistry and ELISA methods were used to evaluate the cardiac functions and biochemical changes of the rats. The mRNA expressions of target genes were measured by RT-qPCR. The proteins expressions, including Collagen I, Collagen III, α-smooth muscle actin (α-SMA), matrix metallopeptidase 2 (MMP 2), matrix metallopeptidase 9 (MMP 9), transforming growth factor-β (TGF-β), protein kinase B (AKT), phospho-AKT, extracellular regulated protein kinases (ERK), phospho-ERK, c-Jun N-terminal kinase (JNK), phospho-JNK, mothers against decapentaplegic protein (Smad3), and phospho-Smad3 were detected and quantified by Western Blot. RESULTS UHPLC-Q-TOF-MS/MS analysis disclosed that 20 components within DSY could be absorbed into blood of rats. DSY improved myocardial injury in the myocardial tissue of LAD-ligated rats, as evidenced by the elevation of left ventricular ejection fraction and left ventricular fractional shortening, and the decrease of the serum CK-MB and LDH levels. Network pharmacology and transcriptomics predicted that DSY could interfere biological processes, such as extracellular matrix organization, focal adhesion and ECM-receptor interaction, and modulate TGF-β mediated signaling pathways, including PI3K/AKT, MAPK, and Smad3. Further study confirmed that DSY reduced MF, accompanied by reduced TGF-β, Collagen I, Collagen III, α-SMA, MMP 2 and MMP 9. Moreover, DSY repressed the phosphorylation of AKT, MAPKs and Smad3. In addition, DSY reduced inflammation and suppressed the mRNA expressions of IL-1β, IL-6, TNF-α, COX2 and iNOS in MF rats. CONCLUSIONS Our study demonstrated that DSY prevented MF in vivo, the action of which was probably via reducing extracellular matrix organization, focal adhesion ECM-receptor interaction and inflammation by regulating TGF-β mediated PI3K/AKT, MAPK and Smad signaling pathways.
Collapse
Affiliation(s)
- Xuan Gao
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Chenyang Ni
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yingying Song
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Xueqing Xie
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Sitong Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yufeng Chen
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Hailian Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Beibei Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Changhong Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China.
| |
Collapse
|
43
|
Zhao H, Hua J, Lu W, Yan L, Zhang M, Chen C, Lv X. Effects of increasing levels of rubber seed cake on growth performance, nutrient digestion metabolism, serum biochemical parameters, and rumen microbiota of Hu sheep. BMC Vet Res 2025; 21:52. [PMID: 39910524 DOI: 10.1186/s12917-025-04503-7] [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: 11/11/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025] Open
Abstract
This study aimed to reveal the effects of increasing levels of rubber seed cake (RSC) on growth performance, nutrient digestion metabolism, serum biochemical parameters, and rumen microbiota in Hu sheep. In this study, 48 Hu sheep, weighing 17.01 ± 0.57 kg at 3 months of age, were randomly divided into four treatments: 0% rubber seed cake (RSC0%), 6% rubber seed cake (RSC6%), 12% rubber seed cake (RSC12%) and 18% rubber seed cake (RSC18%), with 12 sheep per group. Compared to the RSC0%, the ADG and DMI of the RSC6% and RSC12% were increased (P > 0.05). The apparent digestibility of OM and EE quadratically (P < 0.05) changed with the increase of RSC supplementation, with the greatest apparent digestibility of OM and EE observed in the RSC6% diet. With increased RSC supplementation, the N intake and fecal N increased linearly (P < 0.05), and the apparent digestibility of N reduced linearly (P < 0.05). As the increase of RSC supplementation, the serum levels of IgA, IgM, IgG, IL-4, T-AOC, and GSH-Px increased linearly (P < 0.05), and the serum level of IL-6 reduced linearly (P < 0.05). The serum level of IL-1β reduced quadratically (P < 0.05) with the increased RSC dose, and the serum level of SOD increased quadratically (P < 0.05) with the increased RSC dose. The ruminal NH3-N and the relative abundance of norank_Muribaculaceae quadratically (P < 0.05) changed with increased RSC supplementation, and the greatest relative abundance of norank_Muribaculaceae was observed in the RSC6% diet. In general, incorporating RSC into the diet of Hu sheep did not adversely affect growth performance and rumen fermentation characteristics. Supplementing with 6% RSC enhanced the relative abundance of norank_Muribaculacea in the rumen fluid and the immune and antioxidant capabilities. However, supplementing with 12 and 18% RSC might have negatively impacted nutrient digestion and metabolism. Therefore, this study recommended replacing corn and soybean meal with 6% RSC in the diet of Hu sheep.
Collapse
Affiliation(s)
- Huwei Zhao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Jinling Hua
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China.
| | - Wenwen Lu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Longfei Yan
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Min Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Chao Chen
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| | - Xiaokang Lv
- College of Animal Science, Anhui Science and Technology University, Chuzhou, 239000, China
| |
Collapse
|
44
|
Sun T, Song B, Li B. Gut microbiota and atrial cardiomyopathy. Front Cardiovasc Med 2025; 12:1541278. [PMID: 39968343 PMCID: PMC11832500 DOI: 10.3389/fcvm.2025.1541278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Atrial cardiomyopathy is a multifaceted heart disease characterized by structural and functional abnormalities of the atria and is closely associated with atrial fibrillation and its complications. Its etiology involves a number of factors, including genetic, infectious, immunologic, and metabolic factors. Recent research has highlighted the critical role of the gut microbiota in the pathogenesis of atrial cardiomyopathy, and this is consistent with the gut-heart axis having major implications for cardiac health. The aim of this work is to bridge the knowledge gap regarding the interactions between the gut microbiota and atrial cardiomyopathy, with a particular focus on elucidating the mechanisms by which gut dysbiosis may induce atrial remodeling and dysfunction. This article provides an overview of the role of the gut microbiota in the pathogenesis of atrial cardiomyopathy, including changes in the composition of the gut microbiota and the effects of its metabolites. We also discuss how diet and exercise affect atrial cardiomyopathy by influencing the gut microbiota, as well as possible future therapeutic approaches targeting the gut-heart axis. A healthy gut microbiota can prevent disease, but ecological dysbiosis can lead to a variety of symptoms, including the induction of heart disease. We focus on the pathophysiological aspects of atrial cardiomyopathy, the impact of gut microbiota dysbiosis on atrial structure and function, and therapeutic strategies exploring modulation of the microbiota for the treatment of atrial cardiomyopathy. Finally, we discuss the role of gut microbiota in the treatment of atrial cardiomyopathy, including fecal microbiota transplantation and oral probiotics or prebiotics. Our study highlights the importance of gut microbiota homeostasis for cardiovascular health and suggests that targeted interventions on the gut microbiota may pave the way for innovative preventive and therapeutic strategies targeting atrial cardiomyopathy.
Collapse
Affiliation(s)
- Tingting Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Beibei Song
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Bo Li
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| |
Collapse
|
45
|
Cheng D, Sheng S, Hu J, Cai S, Liu Y, Gan R, Zhu Z, Ge L, Chen W, Cheng X. Ershen Zhenwu Decoction suppresses myocardial fibrosis of chronic heart failure with heart-kidney Yang deficiency by down-regulating the Ras Homolog Gene Family Member A/Rho-Associated Coiled-Coil Kinases signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119146. [PMID: 39580131 DOI: 10.1016/j.jep.2024.119146] [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: 06/20/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
ETHNOPHARMACOLOGICAL SIGNIFICANCE The therapeutic efficacy of Ershen Zhenwu Decoction (ESZWD)-a famous formulation from Xin'an for patients with chronic heart failure heart-kidney Yang deficiency (CHF-HKYD)-is well established. Still, the underlying molecular mechanism is not clear. AIM OF THE STUDY This study investigated mechanisms by which ESZWD suppresses cardiac pathology, including myocardial fibrosis, in CHF-HKYD model rats and Ang II-stimulated cardiac fibroblasts (CFs). MATERIALS AND METHODS The components in ESZWD were analyzed by ultra-high-performance liquid chromatography coupled with Quadrupole Time-Of-Flight mass spectrometry (UHPLC-Q-TOF-MS). CHF-HKYD model was established in the male Sprague-Dawley rats through bilateral thyroidectomy and intraperitoneal administration of 0.02% doxorubicin (DOX), twice weekly for 3 weeks. Subsequently, the CHF-HKYD model rats were randomly categorized into the Model, ESZWD-L (3.96 g/kg/d ESZWD), ESZWD-M (7.92 g/kg/d ESZWD), ESZWD-H (15.84 g/kg/d ESZWD), and Sac/Val (68 mg/kg/d sacubitril/valsartan) groups and treated daily for 4 weeks. As a control, the sham surgery group (Sham) was used. Primary cardiac fibroblasts (CFs) were categorized into Control, Model, ESZWD, and Sac/Val groups. Then, the CFs were stimulated with Ang-II. The ESZWD and Sac/Val groups were incubated with different concentrations of drug-containing sera and their effects on CF viability were assessed via the CCK-8 assay. The ESZWD and Sac/Val groups received drug-containing serum concentrations determined by CCK-8 assay results. The cardioprotective effects of ESZWD were determined using echocardiography, Hematoxylin & Eosin (H&E) staining, Masson staining, and Sirius red staining, and the Enzyme Linked Immunosorbent Assay (ELISA). ESZWD's effects on the Ras Homolog Gene Family Member A (RhoA)/Rho-Associated Coiled-Coil Kinases (ROCKs) signaling pathway and myocardial fibrosis were assessed by Western blotting and Quantitative Real-Time PCR (qRT-PCR) analyses. Immunofluorescence was used to observe fibrotic markers in CFs. RESULTS ESZWD treatment improved cardiac function in the CHF-HKYD rats by significantly reducing myocardial fibrosis and ventricular remodeling. ESZWD treatment increased the rats' body temperature (Tb) and 24-h urine volume, left ventricular ejection fraction (LVEF) and LV fractional shortening (LVFS), and decreased LV internal systolic diameter (LVIDs), LV internal diastolic diameter (LVIDd), and heart weight/body weight (HW/BW) compared to the Model group. In comparison to the model rats, ESZWD treatment decreased serum levels of B-type natriuretic peptide precursor (NT-proBNP), tumor necrosis factor-alpha (TNF-α), interleukin-11 (IL-11), and IL-17A. ESZWD treatment significantly down-regulated the protein and mRNA expression levels of collagen I A1, α-SMA, RhoA, ROCK1, and ROCK2 in the heart tissues of the CHF-HKYD rats and the Ang II-stimulated CFs. CONCLUSION ESZWD significantly improved cardiac function and attenuated myocardial fibrosis and inflammation in the CHF-HKYD rats by inhibiting the RhoA/ROCKs signaling pathway.
Collapse
Affiliation(s)
- Dan Cheng
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Shuguang Hospital Affiliated with Shanghai University of Chinese Medicine, Anhui Hospital, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Sheng Sheng
- Shuguang Hospital Affiliated with Shanghai University of Chinese Medicine, Anhui Hospital, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Hu
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Shanshan Cai
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Yan Liu
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Ruixi Gan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Zhenpeng Zhu
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China
| | - Lan Ge
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China.
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China.
| | - Xiaoyu Cheng
- First Affiliated Hospital of Anhui University of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China; Xin'an Key Laboratory of Medical Science, Ministry of Education, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
46
|
De Grau-Bassal G, Calpena-Campmany AC, Silva-Abreu M, Suñer-Carbó J, Mallandrich-Miret M, Martínez-Ruiz S, Cordero C, Del Pozo A, Febrer NBD. Emulgel Containing Metronidazole and Clindamycin for the Treatment of Rosacea. Pharmaceutics 2025; 17:168. [PMID: 40006535 PMCID: PMC11859854 DOI: 10.3390/pharmaceutics17020168] [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: 12/13/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Rosacea is a common skin condition with quite a relevance. It currently affects at least 10% of the European population at some point after the age of 30. It is a chronic disorder that mainly affects the skin on the face and is characterized by outbreaks and remissions. Under normal circumstances, the skin face presents a wide range of commensal organisms, such as Staphylococcus epidermidis or Demodex folliculorum, but dysbiosis of the skin flora plays a relevant role in inflammatory processes and the development of the disease. Metronidazole (MD) is one of the main treatments indicated to reduce redness on the cheeks, nose, chin, or forehead and also to treat flushing, erythema, pimples, and other symptoms due in part to its anti-inflammatory action. On the other hand, clindamycin (CM) is another antibiotic used for rosacea, especially for its action against anaerobic and Gram-positive bacteria. Background/Objectives: This study aimed to develop an emulgel formulation that includes MD and CM, using excipients with non-comedogenic and non-irritating properties. Methods: The formulation was characterised physiochemically, rheological measurements were made, and short-term stability studies were carried out. In vitro release, permeation studies, toxicity an in vitro inflammation model were evaluated in a HaCaT cell model. To determine the interaction between the antibiotics, the minimum inhibitory concentration was determined separately and together using the broth microdilution method. To determine the formulation's antimicrobial activity, an agar diffusion method was used. Results: The MD-CM-gel droplet size was measured by laser diffraction and the diameter obtained was less than 2.68 ± 0.18 µm in 50% of the particles. Suitable results was observed for the short-term stability. Release and permeation data revealed sustained drug release and adequate permeation through human skin. Non-toxicity was detected and the MD showed an anti-inflammatory effect with non-interference of CM. Also, there is no antagonism between the two antibiotics and the MD-CM-gel shows better results when compared to the formulations with the antibiotics separately and to commercial formulations. Conclusions: It is suggested that, following detailed preclinical and clinical studies, MD-CM-gel could be considered as an alternative for treating rosacea.
Collapse
Affiliation(s)
- Guillermo De Grau-Bassal
- Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; (G.D.G.-B.); (N.B.-d.F.)
| | - Ana Cristina Calpena-Campmany
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (A.C.C.-C.); (J.S.-C.); (M.M.-M.); (A.D.P.)
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Marcelle Silva-Abreu
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (A.C.C.-C.); (J.S.-C.); (M.M.-M.); (A.D.P.)
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Joaquim Suñer-Carbó
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (A.C.C.-C.); (J.S.-C.); (M.M.-M.); (A.D.P.)
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Mireia Mallandrich-Miret
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (A.C.C.-C.); (J.S.-C.); (M.M.-M.); (A.D.P.)
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Sergio Martínez-Ruiz
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (S.M.-R.); (C.C.)
| | - Cecilia Cordero
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain; (S.M.-R.); (C.C.)
| | - Alfonso Del Pozo
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; (A.C.C.-C.); (J.S.-C.); (M.M.-M.); (A.D.P.)
| | - Núria Bozal-de Febrer
- Departament de Biologia, Sanitat i Medi Ambient, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), 08028 Barcelona, Spain; (G.D.G.-B.); (N.B.-d.F.)
| |
Collapse
|
47
|
Gong Z, Yang H, Gao L, Liu Y, Chu Q, Luo C, Kang L, Zhai H, Xu Q, Wu W, Li N, Li R. Mechanisms of wogonoside in the treatment of atherosclerosis based on network pharmacology, molecular docking, and experimental validation. BMC Complement Med Ther 2025; 25:28. [PMID: 39871254 PMCID: PMC11770944 DOI: 10.1186/s12906-025-04760-x] [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: 05/22/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Atherosclerosis serves as the fundamental pathology for a variety of cardiovascular disorders, with its pathogenesis being closely tied to the complex interplay among lipid metabolism, oxidative stress, and inflammation. Wogonoside is a natural flavonoid extracted from Scutellaria baicalensis with a variety of biological activities, including anti-inflammatory, hypolipidemic, and cardiac function improvement properties. Despite these known effects, the specific role of wogonoside in the context of atherosclerosis remains to be elucidated. PURPOSE To validate the efficacy of wogonoside in the treatment of atherosclerosis and to investigate its possible therapeutic mechanisms. METHODS Network pharmacology was used to obtain the core targets and signaling pathways that may be efficacious in the treatment of atherosclerosis with wogonoside, which were validated using molecular docking and molecular dynamics simulations. To further validate the core targets in the signaling pathway, we performed in vivo experiments using apolipoprotein E (ApoE)-/- mice. This included pathological morphology and lipid deposition analysis of mouse aorta, serum lipid level analysis, Elisa analysis, oxidative stress analysis, reactive oxygen species (ROS) fluorescence assay, immunohistochemical analysis and protein blot analysis. RESULTS Predictions were obtained that wogonoside treatment of atherosclerosis has 31 core targets, which are mainly focused on pathways such as Toll-like receptor (TLR) signaling pathway and NF-kappa B (NF-κB ) signaling pathway. Molecular docking and molecular dynamics simulations showed that wogonoside has good binding properties to the core targets. In vivo experimental results showed that wogonoside significantly inhibited aortic inflammatory response and lipid deposition, significantly reduced the release levels of total cholesterol (TC), triglycerides (TG), low-density-lipoprotein cholesterol (LDL-C), oxidized low density (ox-LDL) and free fatty acid (FFA), and significantly inhibited the release of inflammatory factors TNF-α, IL-1β, IL-6 and oxidative stress in ApoE-/- mice. Further molecular mechanism studies showed that wogonoside significantly inhibited the activation of TLR4/NF-κB signaling pathway in ApoE-/- mice. CONCLUSION Wogonoside may be an effective drug monomer for the treatment of atherosclerosis, and its mechanism of action is closely related to the inhibition of the activation of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhaohui Gong
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Haixin Yang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Li Gao
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Yi Liu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Qingmin Chu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Chuanjin Luo
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Liang Kang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Huiqi Zhai
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qiang Xu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Department of Rheumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Wei Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Nan Li
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China.
| | - Rong Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
48
|
Zhang Y, Guo C, Wang L, Wu L, Lv J, Huang X, Yang W. Mendelian Randomization Study Reveals Causal Pathways for Hypertrophic Cardiomyopathy, Cardiovascular Proteins, and Atrial Fibrillation. Br J Hosp Med (Lond) 2025; 86:1-19. [PMID: 39862032 DOI: 10.12968/hmed.2024.0504] [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] [Indexed: 01/27/2025]
Abstract
Aims/Background Research evidence has demonstrated a significant association between hypertrophic cardiomyopathy (HCM) and atrial fibrillation (AF), but the causality and pattern of this link remain unexplored. Therefore, this study investigated the causal relationship between HCM and AF using a two-sample and bidirectional Mendelian randomization (MR) approach. Additionally, this assessed the role of cardiovascular proteins (CPs) associated with cardiovascular diseases between HCM and AF by applying a two-step MR analysis. Methods Data for HCM, AF, and 90 CPs were obtained from the Finn Gen and IEU Open GWAS Project databases. MR-Egger, inverse variance weighting (IVW), weighted median estimator (WME), weighted mode, and simple mode were used to estimate causal inferences. Furthermore, Cochran's Q test, MR-Egger's intercept terms, and Leave-one-out methods determined the heterogeneity, horizontal pleiotropy, and sensitivity. Additionally, mediation effects were used to assess the role of CPs in the relationship between HCM and AF. Results Two-sample and bidirectional MR analysis revealed HCM as a risk factor for AF (odds ratio (OR) = 1.008, 95% confidence interval (CI): 1.001-1.016, p = 0.029) and AF was found to increase the risk of developing HCM (OR = 1.145, 95% CI: 0.963-1.361, p = 0.126). Moreover, Two-step MR analyses indicated that 5 CPs were causally associated with HCM; 12 CPs with AF and 1 CP (Melusin) with both HCM and AF. Additionally, Melusin was observed as a protective factor for both HCM and AF and may serve as a mediator variable for these two conditions (mediation effect 0.0004, mediation ratio 5.5178%, 95% CI: 5.4624-5.5731). Conclusion HCM may increase the risk of developing AF, with Melusin serving as a mediator for this risk.
Collapse
Affiliation(s)
- Yifei Zhang
- The Cardiology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chenyuan Guo
- The Cardiology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lanxin Wang
- The Cardiology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lei Wu
- The Oncology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jia Lv
- The Neurology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xia Huang
- The Laboratory Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wuxiao Yang
- The Cardiology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
49
|
Liu M, Chen R, Zheng Z, Xu S, Hou C, Ding Y, Zhang M, Bao M, He B, Li S. Mechanisms of inflammatory microenvironment formation in cardiometabolic diseases: molecular and cellular perspectives. Front Cardiovasc Med 2025; 11:1529903. [PMID: 39877020 PMCID: PMC11772298 DOI: 10.3389/fcvm.2024.1529903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Cardiometabolic diseases (CMD) are leading causes of death and disability worldwide, with complex pathophysiological mechanisms in which inflammation plays a crucial role. This review aims to elucidate the molecular and cellular mechanisms within the inflammatory microenvironment of atherosclerosis, hypertension and diabetic cardiomyopathy. In atherosclerosis, oxidized low-density lipoprotein (ox-LDL) and pro-inflammatory cytokines such as Interleukin-6 (IL-6) and Tumor Necrosis Factor-alpha (TNF-α) activate immune cells contributing to foam cell formation and arterial wall thickening. Hypertension involves the activation of the renin-angiotensin system (RAS) alongside oxidative stress-induced endothelial dysfunction and local inflammation mediated by T cells. In diabetic cardiomyopathy, a high-glucose environment leads to the accumulation of advanced glycation end products (AGEs), activating the Receptor for Advanced Glycation Endproducts (RAGE) and triggering inflammatory responses that further damage cardiac and microvascular function. In summary, the inflammatory mechanisms in different types of metabolic cardiovascular diseases are complex and diverse; understanding these mechanisms deeply will aid in developing more effective individualized treatment strategies.
Collapse
Affiliation(s)
- Menghua Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rumeng Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiwei Zheng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Shuling Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Chunyan Hou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yining Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mengling Zhang
- School of Stomatology, Changsha Medical University, Changsha, China
| | - Meihua Bao
- Hunan key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Binsheng He
- Hunan key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, School of Pharmaceutical Science, Changsha Medical University, Changsha, China
| | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
50
|
Lauinger AR, Sepe JJ. Vascularization, Innervation, and Inflammation: Pathways Connecting the Heart-Brain Axis and Implications in a Clinical Setting. Biomedicines 2025; 13:171. [PMID: 39857755 PMCID: PMC11762153 DOI: 10.3390/biomedicines13010171] [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: 12/20/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
With an aging population, the incidence of both ischemic heart disease and strokes have become the most prevalent diseases globally. These diseases have similar risk factors, such as hypertension, diabetes, and smoking. However, there is also evidence of a relationship between the heart and the brain, referred to as the heart-brain axis. In this relationship, dysfunction of either organs can lead to injury to the other. There are several proposed physiologies to explain this relationship. These theories usually involve vascular, neuromodulatory, and inflammatory processes; however, few articles have explored and compared these different mechanisms of interaction between the heart and brain. A better understanding of the heart-brain axis can inform physicians of current and future treatment and preventive care options in heart and brain pathologies. The relationship between the brain and heart depends on inflammation, vascular anatomy and function, and neuromodulation. The pathways connecting these organs often become injured or dysfunctional when a major pathology, such as a myocardial infarction or stroke, occurs. This leads to long-term impacts on the patient's overall health and risk for future disease. This study summarizes the current research involved in the heart-brain axis, relates these interactions to different diseases, and proposes future research in the field of neurocardiology. Conditions of the brain and heart are some of the most prevalent diseases. Through understanding the connection between these two organs, we can help inform patients and physicians of novel therapeutics for these pathologies.
Collapse
Affiliation(s)
- Alexa R. Lauinger
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA;
| | - Joseph J. Sepe
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|