1
|
Qi W, Cao X, Chen Y, Chen H, Zhang N, Liu R, Wang W, Liu Q, Zheng S, Li S, Li X, Zao X, Ye Y. JiGuCao capsule formula alleviates metabolic fatty liver disease by regulating the gut-liver axis and lipid metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156559. [PMID: 40064115 DOI: 10.1016/j.phymed.2025.156559] [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: 10/17/2024] [Revised: 02/12/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Metabolic-associated fatty liver disease (MAFLD) is a prevalent chronic liver condition globally, characterized by suboptimal treatment outcomes. Traditional therapies often fail to address the multifaceted pathogenesis of MAFLD, which involves lipid metabolism, inflammation, and gut-liver axis dysregulation. JiGuCao Capsule formula (JCF), a patented Chinese medicine, has demonstrated clinical efficacy in liver disease treatment, indicating its potential as a new therapeutic option for MAFLD. PURPOSE This study aimed to investigate the therapeutic effects and underlying mechanisms of JCF in treating MAFLD, particularly focusing on its impact on liver pathology, intestinal health, and gut microbiota composition. METHODS A MAFLD mouse model was developed by administering a high-fat diet and 5% fructose water for 16 weeks. At week 8, mice exhibited significant steatosis, inflammation, and insulin resistance. Fifty mice were allocated into two groups: the normal diet (ND) group with 19 mice and the high-fat feed diet (HFD) group with 31 mice. Seven mice from each group were sacrificed at week 8 for serological and histopathological assessments. The remaining mice were allocated into ND (n = 6), HFD (n = 6), HFD + JCFL (human equivalent dose,780 mg/kg, n = 6), HFD + JCFH (threefold the human equivalent dose, 2340 mg/kg, n = 6), HFD + Polyene Phosphatidylcholine (PPC) (human equivalent dose,177.84 mg/kg, n = 6) and ND+ JCF (human equivalent dose,780 mg/kg, n = 6) groups. Daily gavage started at week 9. At week 16, after fasting, body weight and liver condition were recorded, and mice were euthanized with pentobarbital sodium. Mouse tissues and feces were collected for histopathological, molecular biological, and multi-omics analyses. RESULTS JCF effectively slowed MAFLD progression in mice by decreasing hepatic lipid accumulation and inflammation. Treatment with JCF significantly reduced hepatic triglycerides and inflammatory markers, including TNF-α and IL-6. JCF enhanced lipid metabolism, repaired the intestinal barrier, and lowered inflammatory cytokines in the intestines, as indicated by reduced serum LPS and restored tight junction proteins expression, such as claudin-1 and occludin. Fecal microbiota analysis indicated that JCF treatment elevated Lactobacillus levels and reduced Colidextribacter levels, correlating with enhanced metabolic profiles. The primary bioactive compounds identified in JCF responsible for these therapeutic effects were betulinic acid, cholic acid, deoxycholic acid, oleanolic acid, and pectolinarigenin. Transcriptomic analysis showed that JCF regulated key pathways involved in lipid metabolism, including the pparγ-cd36 axis and modulation of ox-LDL levels. The results indicate that JCF effectively mitigates MAFLD by influencing the gut-liver axis and lipid metabolism. CONCLUSION JCF alleviates MAFLD by modulating the gut-liver axis and lipid metabolism. Its effects involve improving gut barrier function, regulating microbiota, and targeting the pparγ-cd36 axis. Active compounds like betulinic acid support its therapeutic potential. JCF shows promise as a novel treatment for MAFLD, with further clinical studies needed.
Collapse
Affiliation(s)
- Wenying Qi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Xu Cao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Yue Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Hening Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Ningyi Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Ruijia Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Wei Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Qiyao Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Shihao Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Size Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Xiaoke Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Xiaobin Zao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, PR China.
| | - Yong'an Ye
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China; Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, PR China.
| |
Collapse
|
2
|
Chen CH, Sawamura T, Akhmedov A, Tsai MH, Akyol O, Kakino A, Chiang HH, Kraler S, Lüscher TF. Evolving concepts of low-density lipoprotein: From structure to function. Eur J Clin Invest 2025; 55:e70019. [PMID: 40045739 DOI: 10.1111/eci.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/17/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Low-density lipoprotein (LDL) is a central player in atherogenesis and has long been referred to as 'bad cholesterol.' However, emerging evidence indicates that LDL functions in multifaceted ways beyond cholesterol transport that include roles in inflammation, immunity, and cellular signaling. Understanding LDL's structure, metabolism and function is essential for advancing cardiovascular disease research and therapeutic strategies. METHODS This narrative review examines the history, structural properties, metabolism and functions of LDL in cardiovascular health and disease. We analyze key milestones in LDL research, from its early identification to recent advancements in molecular biology and omics-based investigations. Structural and functional insights are explored through imaging, proteomic analyses and lipidomic profiling, providing a deeper understanding of LDL heterogeneity. RESULTS Low-density lipoprotein metabolism, from biosynthesis to receptor-mediated clearance, plays a crucial role in lipid homeostasis and atherogenesis. Beyond cholesterol transport, LDL contributes to plaque inflammation, modulates adaptive immunity and regulates cellular signaling pathways. Structural studies reveal its heterogeneous composition, which influences its pathogenic potential. Evolving perspectives on LDL redefine its clinical significance, affecting cardiovascular risk assessment and therapeutic interventions. CONCLUSIONS A holistic understanding of LDL biology challenges traditional perspectives and underscores its complexity in cardiovascular health. Future research should focus on further elucidating LDL's structural and functional diversity to refine risk prediction models and therapeutic strategies, ultimately improving cardiovascular outcomes.
Collapse
Affiliation(s)
- Chu-Huang Chen
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, USA
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | - Ming-Hsien Tsai
- Department of Child Care, College of Humanities and Social Sciences, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Omer Akyol
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, USA
| | - Akemi Kakino
- Department of Molecular Pathophysiology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Huan-Hsing Chiang
- Molecular Cardiology Research Laboratories, Vascular and Medicinal Research, The Texas Heart Institute, Houston, Texas, USA
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Department of Internal Medicine and Cardiology, Cantonal Hospital Baden, Baden, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals, GSTT and King's College, London, UK
| |
Collapse
|
3
|
Tu L, Zou Z, Yang Y, Wang S, Xing B, Feng J, Jin Y, Cheng M. Targeted drug delivery systems for atherosclerosis. J Nanobiotechnology 2025; 23:306. [PMID: 40269931 DOI: 10.1186/s12951-025-03384-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Atherosclerosis is a complex cardiovascular disease driven by multiple factors, including aging, inflammation, oxidative stress, and plaque rupture. The progression of this disease is often covert, emphasizing the need for early biomarkers and effective intervention measures. In recent years, advancements in therapeutic strategies have highlighted the potential of targeting specific processes in atherosclerosis, such as plaque localization, macrophage activity, and key enzymes. Based on this, this review discusses the potential role of targeted drugs in the treatment of atherosclerosis. It also focuses on their clinical efficacy in anti-atherosclerosis treatment and their ability to provide more precise therapeutic approaches. The findings underscore that future research can concentrate on exploring newer drug delivery systems and biomarkers to further refine clinical treatment strategies and enhance the long-term dynamic management of atherosclerosis.
Collapse
Affiliation(s)
- Liangxing Tu
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
| | - Zijian Zou
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
| | - Ye Yang
- Wenzhou Yining Geriatric Hospital, Wenzhou, 325041, P.R. China
| | - Siying Wang
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
| | - Banghuai Xing
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
| | - Jianfang Feng
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
- Guangxi University of Chinese Medicine, Nanning, 530200, P.R. China
| | - Yi Jin
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China.
| | - Meng Cheng
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China.
| |
Collapse
|
4
|
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
|
5
|
Ynalvez RA, Rangel RA, Gutierrez JA. Mercury toxicity resulting from enzyme alterations- minireview. Biometals 2025; 38:357-370. [PMID: 39820948 DOI: 10.1007/s10534-025-00663-z] [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: 08/20/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Mercury is widely known for its detrimental effects on living organisms, whether in its elemental or bonded states. Recent comparative studies have shed light on the biochemical implications of mercury ingestion, both in low, persistent concentrations and in elevated acute dosages. Studies have presented models that elucidate how mercury disrupts healthy cells. Mercury's unique ability to interfere with crucial enzymatic processes at deposition sites is a vital feature of these models. The strong affinity for the sulfhydryl moieties of enzyme catalytic sites leads to enzyme inactivation through permanent covalent modifications. This inactivation can have catastrophic effects on an organism's metabolic functions. Moreover, it has been found that mercury's binding to sulfhydryl moieties is highly nonspecific and can occur in various ways. This review aimed to explore the effects of mercury on a broad spectrum of enzymes with a specific focus on how these alterations can detrimentally affect several metabolic pathways.
Collapse
Affiliation(s)
- Ruby A Ynalvez
- Department of Biology and Chemistry, Texas A&M International University, Laredo, TX, USA.
| | - Rene A Rangel
- Paul L Foster School of Medicine, Texas Tech Health Sciences Center El Paso, El Paso, TX, USA
| | - Jose A Gutierrez
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
6
|
Bakillah A, Soliman AF, Al Subaiee M, Obeid KK, Al Hussaini A, Bashir SF, Al Arab M, Al Otaibi A, Mubarak SAS, Al Qarni AA. Adiponectin and TNF-Alpha Differentially Mediate the Association Between Cystatin C and Oxidized LDL in Type 2 Diabetes Mellitus Patients. Int J Mol Sci 2025; 26:3001. [PMID: 40243674 PMCID: PMC11988364 DOI: 10.3390/ijms26073001] [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: 02/20/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
In individuals with type 2 diabetes mellitus (T2DM), elevated levels of both plasma and urinary cystatin C (Cys-C) contribute to increased oxidation, which in turn accelerates the oxidation of low-density lipoprotein (LDL). This process may worsen the development of atherosclerosis and cardiovascular disease by promoting endothelial dysfunction and inflammation. Despite its potential significance, the relationship between Cys-C and oxidized LDL (ox-LDL) in T2DM remains poorly understood. This study investigated the relationship between plasma and urinary Cys-C and ox-LDL levels in T2DM patients. The cohort included 57 patients with T2DM (mean age 61.14 ± 9.99 years; HbA1c 8.66 ± 1.60% and BMI 35.15 ± 6.65 kg/m2). Notably, 95% of the patients had hypertension, 82% had dyslipidemia, 59% had an estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73 m2, 14% had coronary artery disease (CAD), and 5% had a history of stroke. Plasma and urinary Cys-C and ox-LDL levels were measured using ELISA. Adipokine and cytokine levels were measured using the multiplex® MAP Human Adipokine Magnetic Bead Panels. Spearman's correlation analysis revealed a significant positive correlation of plasma and urinary Cys-C with ox-LDL (r = 0.569, p = 0.0001 and r = 0.485, p = 0.0001, respectively). Multivariable regression analysis indicated that both plasma and urinary Cys-C were independently associated with ox-LDL, after adjusting for confounding factors (β = 0.057, p = 0.0001 and β = 0.486, p = 0.003, respectively). Stepwise linear regression identified TNFα and adiponectin as the strongest predictors of the relationship between urinary Cys-C and ox-LDL (β = 0.382, p = 0.0001; r2 = 0.64), while adiponectin alone was the best predictor of the plasma Cys-C and ox-LDL association (β = 0.051, p = 0.005; r2 = 0.46). Furthermore, adiponectin partly mediated the relationship between plasma Cys-C and ox-LDL, explaining 18% of the variance in this association. In contrast, TNFα partly mediated the relationship between urinary Cys-C and ox-LDL, accounting for 28% of the variance. This study emphasizes the complex interaction between Cys-C and ox-LDL in T2DM. It highlights the need for additional research involving larger patient cohorts to improve our understanding of the therapeutic potential of plasma and urinary Cys-C in conjunction with ox-LDL for managing complications associated with T2DM.
Collapse
Affiliation(s)
- Ahmed Bakillah
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.H.); (S.F.B.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Ayman Farouk Soliman
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Maram Al Subaiee
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Khamis Khamees Obeid
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Arwa Al Hussaini
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.H.); (S.F.B.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Shahinaz Faisal Bashir
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.H.); (S.F.B.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Mohammad Al Arab
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.H.); (S.F.B.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Abeer Al Otaibi
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.H.); (S.F.B.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Sindiyan Al Shaikh Mubarak
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.H.); (S.F.B.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| | - Ali Ahmed Al Qarni
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Ahsa 31982, Saudi Arabia; (A.A.H.); (S.F.B.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Ahsa 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNG-HA), King Abdulaziz Hospital, Al Ahsa 36428, Saudi Arabia; (A.F.S.); (M.A.S.); (K.K.O.)
| |
Collapse
|
7
|
Radić M, Belančić A, Đogaš H, Vučković M, Sener YZ, Sener S, Fajkić A, Radić J. Cardiometabolic Risk in Psoriatic Arthritis: A Hidden Burden of Inflammation and Metabolic Dysregulation. Metabolites 2025; 15:206. [PMID: 40137170 PMCID: PMC11943837 DOI: 10.3390/metabo15030206] [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: 02/14/2025] [Revised: 03/07/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease that extends beyond musculoskeletal and dermatologic involvement to elevate cardiometabolic risk. Emerging evidence highlights the critical role of systemic inflammation in metabolic dysregulation, accelerating insulin resistance, dyslipidemia, and oxidative stress, all of which contribute to the increased burden of cardiovascular disease in PsA. This review explores the intricate interplay between inflammatory mediators-such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-17 (IL-17),-adipokine imbalances, and lipid metabolism abnormalities, all of which foster endothelial dysfunction and atherosclerosis. The dysregulation of adipokines, including leptin, adiponectin, and resistin, further perpetuates inflammatory cascades, exacerbating cardiovascular risk. Additionally, the metabolic alterations seen in PsA, particularly insulin resistance and lipid dysfunction, not only contribute to cardiovascular comorbidities but also impact disease severity and therapeutic response. Understanding these mechanistic links is imperative for refining risk stratification strategies and tailoring interventions. By integrating targeted immunomodulatory therapies with metabolic and cardiovascular risk management, a more comprehensive approach to PsA treatment can be achieved. Future research must focus on elucidating shared inflammatory and metabolic pathways, enabling the development of innovative therapeutic strategies to mitigate both systemic inflammation and cardiometabolic complications in PsA.
Collapse
Affiliation(s)
- Mislav Radić
- Department of Internal Medicine, Division of Rheumatology, Allergology and Clinical Immunology, Center of Excellence for Systemic Sclerosis in Croatia, University Hospital of Split, 21000 Split, Croatia;
- Internal Medicine Department, School of Medicine, University of Split, 21000 Split, Croatia
| | - Andrej Belančić
- Department of Basic and Clinical Pharmacology with Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Hana Đogaš
- Department of Neurology, University Hospital of Split, 21000 Split, Croatia;
| | - Marijana Vučković
- Department of Internal Medicine, Division of Nephrology and Dialysis, University Hospital of Split, 21000 Split, Croatia;
| | - Yusuf Ziya Sener
- Department of Pediatric Rheumatology, Sophia Children’s Hospital, Erasmus University Medical Center, 3000 CB Rotterdam, The Netherlands;
| | - Seher Sener
- Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, 3000 CB Rotterdam, The Netherlands;
| | - Almir Fajkić
- Department of Pathophysiology, Faculty of Medicine, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Josipa Radić
- Internal Medicine Department, School of Medicine, University of Split, 21000 Split, Croatia
- Department of Internal Medicine, Division of Nephrology and Dialysis, University Hospital of Split, 21000 Split, Croatia;
| |
Collapse
|
8
|
Baissary J, Koberssy Z, Durieux JC, Atieh O, Daher J, Ailstock K, Labbato D, Foster T, Rodgers MA, Merheb A, Funderburg NT, McComsey GA. The Effect of COVID-19 on Arterial Stiffness and Inflammation: A Longitudinal Prospective Study. Viruses 2025; 17:394. [PMID: 40143322 PMCID: PMC11945347 DOI: 10.3390/v17030394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Data are limited for assessing the effect of COVID infection on endothelial function, pre- and post-pandemic. The objective of this study was to assess changes in pre-pandemic cardiovascular parameters after COVID-19 infection. This prospective cohort study used EndoPAT2000 Itamar Medical Ltd., Caesarea, Israel, to measure the augmentation index (AI; arterial elasticity) and reactive hyperemic index (RHI; endothelial function). Markers of endothelial function, inflammation, and gut integrity were collected at pre- and post-pandemic visits. COVID-negative and COVID-positive participants were matched on pre-pandemic covariates, and AI ≥ 5.0 was defined as having worse AI. Among the 156 participants, 50% had documented COVID-19 infection. Groups were balanced (p > 0.05) on pre-pandemic characteristics. Increases in oxLDL (p = 0.03) were observed in the COVID-positive group, and COVID infection had a negative effect on inflammatory markers (sVCAM-1, sTNF-RI, sTNF-RII, sCD14) and gut integrity (I-FABP, BDG) compared to COVID-negative participants (p < 0.05). There was a 16.7% (p = 0.02) increase in the proportion of COVID-positive participants with AI ≥ 5.0, without a significant change (p = 0.09) among the COVID-negative group. COVID-positive status, female sex, and higher IL-6 and sCD163 were associated (p < 0.05) with an increase in having worse AI. COVID infection is independently associated with arterial stiffness. For COVID survivors, female sex and higher markers of inflammation were associated with arterial stiffness.
Collapse
Affiliation(s)
- Jhony Baissary
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Ziad Koberssy
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Jared C. Durieux
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Ornina Atieh
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Joviane Daher
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
| | - Kate Ailstock
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; (K.A.); (N.T.F.)
| | - Danielle Labbato
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Theresa Foster
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Michael A. Rodgers
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| | - Alexander Merheb
- Harvard Faculty of Arts and Sciences, Harvard University, Cambridge, MA 02138, USA;
| | - Nicholas T. Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, OH 43210, USA; (K.A.); (N.T.F.)
| | - Grace A. McComsey
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (J.B.); (Z.K.); (O.A.); (J.D.)
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.); (T.F.); (M.A.R.)
| |
Collapse
|
9
|
Mulumba M, Le C, Schelsohn E, Namkung Y, Laporte SA, Febbraio M, Servant MJ, Chemtob S, Lubell WD, Marleau S, Ong H. Selective Azapeptide CD36 Ligand MPE-298 Regulates oxLDL-LOX-1-Mediated Inflammation and Mitochondrial Oxidative Stress in Macrophages. Cells 2025; 14:385. [PMID: 40072113 PMCID: PMC11898605 DOI: 10.3390/cells14050385] [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: 10/30/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Macrophage mitochondrial dysfunction, caused by oxidative stress, has been proposed as an essential event in the progression of chronic inflammation diseases, such as atherosclerosis. The cluster of differentiation-36 (CD36) and lectin-like oxLDL receptor-1 (LOX-1) scavenger receptors mediate macrophage uptake of oxidized low-density lipoprotein (oxLDL), which contributes to mitochondrial dysfunction by sustained production of mitochondrial reactive oxygen species (mtROS), as well as membrane depolarization. In the present study, the antioxidant mechanisms of action of the selective synthetic azapeptide CD36 ligand MPE-298 have been revealed. After binding to CD36, MPE-298 was rapidly internalized by and simultaneously induced CD36 endocytosis through activation of the Lyn and Syk (spleen) tyrosine kinases. Within this internalized complex, MPE-298 inhibited oxLDL/LOX-1-induced chemokine ligand 2 (CCL2) secretion, abolished the production of mtROS, and prevented mitochondrial membrane potential depolarization in macrophages. This occurred through the inhibition of the multiple-component enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) by oxLDL-activated LOX-1, which was further supported by the reduced recruitment of the p47phox subunit and small GTPase (Rac) 1/2/3 into the plasma membrane. A new mechanism for alleviating oxLDL-induced oxidative stress and inflammation in macrophages is highlighted using the CD36 ligand MPE-298.
Collapse
Affiliation(s)
- Mukandila Mulumba
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| | - Catherine Le
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| | - Emmanuelle Schelsohn
- Institut des Sciences Pharmaceutiques de Suisse Occidentale (ISPSO), Section Sciences Pharmaceutiques, Département des Sciences, Université de Genève, 1205 Genève, Switzerland;
| | - Yoon Namkung
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (Y.N.); (S.A.L.)
| | - Stéphane A. Laporte
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada; (Y.N.); (S.A.L.)
| | - Maria Febbraio
- Department of Dentistry, University of Alberta, Edmonton, AB T6G 2H5, Canada;
| | - Marc J. Servant
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| | - Sylvain Chemtob
- Faculté de Médecine, Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC H3T 1C5, Canada;
| | - William D. Lubell
- Département de Chimie, Université de Montréal, Montréal, QC H3C 3J7, Canada;
| | - Sylvie Marleau
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| | - Huy Ong
- Faculté de Pharmacie, Université de Montréal, Montréal, QC H3C 3J7, Canada; (M.M.); (C.L.); (M.J.S.); (S.M.)
| |
Collapse
|
10
|
Karasaki K. Effects of aged garlic extract on macrophage functions: a short review of experimental evidence (Review). Biomed Rep 2025; 22:47. [PMID: 39882336 PMCID: PMC11775638 DOI: 10.3892/br.2025.1925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025] Open
Abstract
Macrophages play crucial roles in both the innate and adaptive immune systems, contributing to the removal of pathogens and subsequent immune responses. Conversely, aberrant macrophage functions are associated with the onset and progression of various diseases, highlighting macrophages as potential therapeutic targets. Aged garlic extract (AGE) is derived from garlic that has undergone a maturation process of over 10 months in an ethanol solution and contains a variety of bioactive components which are produced in the aging process. Previous animal studies and clinical trials have demonstrated that AGE and its constituents exert a range of health benefits, including immune modulation and amelioration of disease conditions. Experimental studies indicate that AGE modulates macrophage functions associated with pathological conditions. To facilitate understanding of AGE's potential as a functional alleviation for macrophage-associated diseases, the present short review summarizes experimental evidence supporting the notion that AGE and its components modify macrophage functions, including phagocytosis, production of reactive oxygen species and polarization.
Collapse
Affiliation(s)
- Kohei Karasaki
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| |
Collapse
|
11
|
Yamamoto T, Okuno M, Kuwano K, Ogura Y. Mycoplasma pneumoniae drives macrophage lipid uptake via GlpD-mediated oxidation, facilitating foam cell formation. Int J Med Microbiol 2025; 318:151646. [PMID: 39862618 DOI: 10.1016/j.ijmm.2025.151646] [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: 10/03/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Cardiovascular diseases, primarily caused by atherosclerosis, are a major public health concern worldwide. Atherosclerosis is characterized by chronic inflammation and lipid accumulation in the arterial wall, leading to plaque formation. In this process, macrophages play a crucial role by ingesting lipids and transforming into foam cells, which contribute to plaque instability and cardiovascular events. Recent studies have suggested that various pathogens are involved in the development of atherosclerosis, with Mycoplasma pneumoniae considered one of the potential candidates. Therefore, this study investigated whether this bacterium induces lipid accumulation in macrophages, which play a crucial role in the development of atherosclerosis, using the Raw264.7 model. Our findings revealed that M. pneumoniae infection promotes lipid droplet formation in macrophages. Glycerol 3-phosphate oxidase, GlpD, in the bacterium is involved in this process by producing reactive oxygen species, which in turn causes the oxidation of low-density lipoprotein. Furthermore, the significant increase in the expression of oxidized lipid receptors involved in the uptake of this oxidized lipid indicates that the bacteria promote lipid uptake in infected macrophages. These results suggest that M. pneumoniae has a direct pro-atherogenic effect, promoting the formation of atherosclerotic lesions through foam cell formation. Understanding the mechanisms by which M. pneumoniae influences atherosclerosis provides valuable insights for devising new therapeutic strategies for the prevention and management of cardiovascular diseases.
Collapse
Affiliation(s)
- Takeshi Yamamoto
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan.
| | - Miki Okuno
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Koichi Kuwano
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Yoshitoshi Ogura
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| |
Collapse
|
12
|
Dabravolski SA, Churov AV, Ravani AL, Karimova AE, Luchinkin IG, Sukhorukov VN, Orekhov AN. The role of Epsins in atherosclerosis: From molecular mechanisms to therapeutic applications. Vascul Pharmacol 2025; 158:107457. [PMID: 39672315 DOI: 10.1016/j.vph.2024.107457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/29/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
Atherosclerosis is a multifaceted disease characterised by chronic inflammation and vascular remodelling, leading to plaque formation and cardiovascular complications. Recent evidence highlights the critical role of epsins, a family of endocytic proteins, in the pathogenesis of atherosclerosis. This manuscript explores the multifarious functions of epsins in atherosclerosis, focusing on their involvement in angiogenesis, lymphangiogenesis, and the modulation of key signalling pathways. We discuss how epsins facilitate EndoMT through their interaction with the TGFβ signalling pathway, which contributes to vascular smooth muscle cell-like phenotypes and plaque instability. Additionally, we examine the therapeutic potential of targeting epsins, elucidating their interactions with crucial partners such as LDLR, LRP-1, and TLR 2/4, among others, in mediating lipid metabolism and inflammation. Furthermore, we highlight the promising prospects of epsin-targeting peptides and small interfering RNAs as therapeutic agents for atherosclerosis treatment. Despite these advancements, the research faces limitations, including a reliance on specific mouse models and a need for comprehensive studies on the long-term effects of epsin modulation. Therefore, future investigations should focus on elucidating the detailed mechanisms of epsin function and their implications in cardiovascular health, fostering collaborations to translate basic research into innovative therapeutic strategies. This work underscores the necessity for further exploration of epsins to unlock their full therapeutic potential in combating atherosclerosis and related cardiovascular diseases.
Collapse
Affiliation(s)
- Siarhei A Dabravolski
- Department of Biotechnology Engineering, Braude Academic College of Engineering, Snunit 51, P.O. Box 78, Karmiel 2161002, Israel.
| | - Alexey V Churov
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia; Pirogov Russian National Research Medical University, Russia Gerontology Clinical Research Centre, Moscow, Institute on Ageing Research, Russian Federation, 16 1st Leonova Street, 129226 Moscow, Russia
| | - Alessio L Ravani
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia
| | - Amina E Karimova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, 33, Profsoyuznaya Street, Building 4, 117418 Moscow, Russia
| | - Igor G Luchinkin
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, 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
- Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia
| |
Collapse
|
13
|
Bucci T, Menichelli D, Palumbo IM, Pastori D, Ames PRJ, Lip GYH, Pignatelli P. Statins as an Adjunctive Antithrombotic Agent in Thrombotic Antiphospholipid Syndrome: Mechanisms and Clinical Implications. Cells 2025; 14:353. [PMID: 40072082 PMCID: PMC11899080 DOI: 10.3390/cells14050353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/22/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
The thrombotic physiopathology of antiphospholipid syndrome (APS) is complex, heterogeneous, and dynamic. While venous thromboembolism (VTE) is the most common initial presentation, arterial thrombotic events (ATE) become more frequent in advanced stages and are associated with high morbidity and mortality. Despite the use of oral anticoagulants (OACs), thrombotic APS remains associated with a high risk of recurrent thrombosis. Given their potential antithrombotic effects capable of reducing the risk of both VTE and ATE, statins have been proposed as an adjunctive therapy to OACs for patients with APS and recurrent thrombosis. However, this recommendation is primarily based on studies not specifically conducted in APS populations, with only preclinical data or evidence from retrospective observational studies available from APS patients cohorts. For these reasons, this narrative review aims to synthesise the studies evaluating the potential antithrombotic effects of statins in patients with APS, highlighting the progress made and identifying areas for future research.
Collapse
Affiliation(s)
- Tommaso Bucci
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool, L7 8TX, UK; (T.B.); (D.P.); (G.Y.H.L.)
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (D.M.); (I.M.P.)
| | - Danilo Menichelli
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (D.M.); (I.M.P.)
- Department of General and Specialized Surgery “Paride Stefanini”, Sapienza University of Rome, 00185 Rome, Italy
| | - Ilaria Maria Palumbo
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (D.M.); (I.M.P.)
- Department of General and Specialized Surgery “Paride Stefanini”, Sapienza University of Rome, 00185 Rome, Italy
| | - Daniele Pastori
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool, L7 8TX, UK; (T.B.); (D.P.); (G.Y.H.L.)
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (D.M.); (I.M.P.)
- IRCCS Neuromed, Località Camerelle, 86077 Pozzilli, Italy
| | - Paul R. J. Ames
- Immune Response and Vascular Disease, iNOVA, 4Health, Nova Medical School, Nova University Lisbon, 1099-085 Lisbon, Portugal;
- Department of Haematology, Dumfries Royal Infirmary, Cargenbridge, Dumfries DG2 8RX, UK
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool John Moores University and Liverpool and Heart and Chest Hospital, Liverpool, L7 8TX, UK; (T.B.); (D.P.); (G.Y.H.L.)
- Danish Centre for Health Services Research, Department of Clinical Medicine, Aalborg University, 9220 Aalborg, Denmark
- Department of Cardiology, Lipidology and Internal Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (D.M.); (I.M.P.)
| |
Collapse
|
14
|
Berisha H, Hattab R, Comi L, Giglione C, Migliaccio S, Magni P. Nutrition and Lifestyle Interventions in Managing Dyslipidemia and Cardiometabolic Risk. Nutrients 2025; 17:776. [PMID: 40077646 PMCID: PMC11902110 DOI: 10.3390/nu17050776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Dyslipidemia, characterized by abnormal blood lipid levels, is a major public health concern due to its association with atherosclerotic cardiovascular disease (ASCVD) and other cardiometabolic disorders. In this context, appropriate nutrition patterns are pivotal as they represent the basic approach for providing a wide range of substantial advantages. The best evidence for dyslipidemia management is offered by the Mediterranean Diet, the Plant-Based Diet, the High-Fiber Diet and the Anti-inflammatory Diet, while the DASH Diet and the Ketogenic Diet have also been shown to target additional pathological features like hypertension and other comorbidities. The bioactive compounds that are enriched in these nutrition patterns and able to manage dyslipidemia include monounsaturated fatty acids such as ω-3, polyphenols such as oleuropein, resveratrol, flavonoids, and catechins, carotenoids, phytosterols and soluble and unsoluble fibers. Diets rich in these compounds can improve lipid profile by mitigating oxidative stress, reducing low-grade chronic inflammation, modulating macronutrient absorption and other mechanisms, thereby supporting cardiovascular health. Additionally, lifestyle interventions such as regular physical activity, weight loss, reduced alcohol consumption and smoking cessation further ameliorate lipid metabolism and manage circulated lipid profile. Furthermore, emerging insights from nutrigenomics underscore the potential for proper diet to address genetic factors and optimize treatment outcomes. The pivotal role of nutrition interventions in the context of dyslipidemia and its cardiometabolic implications is discussed in this review, emphasizing evidence-based and personalized approaches.
Collapse
Affiliation(s)
- Hygerta Berisha
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (H.B.); (R.H.); (L.C.); (C.G.)
| | - Reham Hattab
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (H.B.); (R.H.); (L.C.); (C.G.)
| | - Laura Comi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (H.B.); (R.H.); (L.C.); (C.G.)
| | - Claudia Giglione
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (H.B.); (R.H.); (L.C.); (C.G.)
| | - Silvia Migliaccio
- Dipartimento di Medicina Sperimentale, Sapienza Università di Roma, 00185 Roma, Italy;
| | - Paolo Magni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy; (H.B.); (R.H.); (L.C.); (C.G.)
- IRCCS MultiMedica, Sesto San Giovanni, 20099 Milan, Italy
| |
Collapse
|
15
|
Bakillah A, Al Subaiee M, Soliman AF, Obeid KK, Bashir SF, Al Hussaini A, Al Arab M, Al Otaibi A, Mubarak SAS, Al Qarni AA. Plasma Atrial Natriuretic Peptide Predicts Oxidized Low-Density Lipoprotein Levels in Type 2 Diabetes Mellitus Patients Independent of Circulating Adipokine and Cytokine. Int J Mol Sci 2025; 26:1859. [PMID: 40076485 PMCID: PMC11899485 DOI: 10.3390/ijms26051859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/09/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Atrial natriuretic peptide (ANP) and oxidized low-density lipoprotein (ox-LDL) play essential roles in the development and progression of vascular complications associated with type 2 diabetes mellitus (T2DM), and both are independently linked to cardiovascular diseases (CVD). However, the relationship between ANP and ox-LDL in patients with T2DM remains unclear as previous studies have primarily focused on circulating levels in various diseases. This study investigated the relationship between ANP and ox-LDL levels in obese individuals with T2DM. The cohort included 57 patients with T2DM (mean age 61.14 ± 9.99 years; HbA1c 8.66 ± 1.60%; BMI 35.15 ± 6.65 kg/m2). Notably, 95% of the patients had hypertension, 82% had dyslipidemia, 59% had an estimated glomerular filtration rate (eGFR) < 60 mL/min/1.73 m2, 14% had coronary artery disease (CAD), and 5% had a history of stroke. Plasma concentrations of ANP and ox-LDL were measured using ELISA. Adipokines and cytokines levels were measured using the multiplex® MAP Human Adipokine Magnetic Beads Spearman's correlation analysis which revealed a negative correlation between ANP and ox-LDL (r = -0.446, p = 0.001) as well as with the ox-LDL/apoB ratio (r = -0.423, p = 0.001) and ox-LDL/LDLc ratio (r = -0.307, p = 0.038). Multivariable regression analysis indicated that ANP was independently associated with ox-LDL (β = -115.736, p = 0.005). Stepwise linear regression further identified TNFα, leptin, and adiponectin as the strongest predictors influencing the relationship between ANP and ox-LDL levels (β = -64.664, p = 0.0311, and r2 = 0.546 for the model). However, these factors did not significantly mediate this association. This study emphasizes the need for further exploration of the complex interaction between ANP and ox-LDL in larger patient populations. This could provide valuable insights into potential therapeutic approaches for managing vascular complications in obese individuals with T2DM.
Collapse
Affiliation(s)
- Ahmed Bakillah
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Mubarraz 36428, Saudi Arabia; (S.F.B.); (A.A.H.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Mubarraz 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Maram Al Subaiee
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Ayman Farouk Soliman
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Khamis Khamees Obeid
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Shahinaz Faisal Bashir
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Mubarraz 36428, Saudi Arabia; (S.F.B.); (A.A.H.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Mubarraz 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Arwa Al Hussaini
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Mubarraz 36428, Saudi Arabia; (S.F.B.); (A.A.H.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Mubarraz 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Mohammad Al Arab
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Mubarraz 36428, Saudi Arabia; (S.F.B.); (A.A.H.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Mubarraz 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Abeer Al Otaibi
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Mubarraz 36428, Saudi Arabia; (S.F.B.); (A.A.H.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Mubarraz 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Sindiyan Al Shaikh Mubarak
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Mubarraz 36428, Saudi Arabia; (S.F.B.); (A.A.H.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Mubarraz 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| | - Ali Ahmed Al Qarni
- King Abdullah International Medical Research Center (KAIMRC), Eastern Region, Al Mubarraz 36428, Saudi Arabia; (S.F.B.); (A.A.H.); (M.A.A.); (A.A.O.); (S.A.S.M.); (A.A.A.Q.)
- Division of Biomedical Research Core Facility, King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), Al Mubarraz 36428, Saudi Arabia
- Ministry of National Guard-Health Affairs (MNGHA), King Abdulaziz Hospital, Al Mubarraz 36428, Saudi Arabia; (M.A.S.); (A.F.S.); (K.K.O.)
| |
Collapse
|
16
|
Alzahrani A, Alharbi AA, Alharbi AK, Alkhaldi A, Filimban AZ, Alfatni A, Kaifi R, Albngali A, Alkharaiji M, Alserihy O, Sultan SR. Evaluating Carotid Plaque Stiffness with Ultrasound 2D Shear-Wave Elastography in Patients Undergoing Coronary Artery Bypass Grafting. Diagnostics (Basel) 2025; 15:338. [PMID: 39941268 PMCID: PMC11817818 DOI: 10.3390/diagnostics15030338] [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: 01/07/2025] [Revised: 01/20/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Coronary and carotid artery diseases are manifestations of a systemic atherosclerotic process, often coexisting in patients affected by both conditions. This association emphasizes the importance of evaluating both coronary and carotid atherosclerosis in high-risk individuals. Ultrasound 2D shear-wave elastography (2D-SWE) has shown promise as a noninvasive technique for assessing carotid plaque stiffness. This prospective pilot study aimed to assess carotid plaque stiffness in patients undergoing coronary artery bypass grafting (CABG) and those not scheduled for the procedure as a control group. Methods: 32 patients (17 CABG and 15 controls) were recruited, collectively presenting 43 carotid plaques. Bilateral carotid ultrasound was performed using a high-resolution linear transducer. Plaque stiffness was quantified via 2D-SWE, expressed in shear-wave velocity (SWV, m/s) and Young's modulus (YM, kPa). Plaque characteristics, including GSM, were quantified. Intra-observer reproducibility was evaluated with intraclass correlation coefficients (ICCs) and Bland-Altman plots. Statistical differences and correlations were assessed using Mann-Whitney U and Spearman's correlation tests. Results: Carotid plaques in the CABG group exhibited significantly lower stiffness compared to controls (median stiffness SWV: 3.64 m/s vs. 4.91 m/s, p < 0.0001; YM: 20.96 kPa vs. 72.54 kPa, p < 0.0001). ICCs demonstrated excellent reproducibility for stiffness measurements (SWV: ICC = 0.992; YM: ICC = 0.992), with minimal bias in measurements. A positive correlation was observed between 2D-SWE and GSM values (SWV: r = 0.343, p = 0.024; YM: r = 0.340, p = 0.026). Conclusions: Ultrasound 2D-SWE has shown promise as a reliable tool for quantifying carotid plaque stiffness, demonstrating high reproducibility and a significant correlation with GSM. The observed reduction in plaque stiffness among CABG patients highlights its potential as a valuable parameter for identifying high-risk plaques and assessing cerebrovascular risk in patients undergoing CABG.
Collapse
Affiliation(s)
- Adel Alzahrani
- Department of Diagnostic Radiology and Imaging, King Abdullah Medical City, Makkah 24246, Saudi Arabia; (A.A.)
| | - Amjad Ali Alharbi
- Department of Diagnostic Radiology and Imaging, King Abdullah Medical City, Makkah 24246, Saudi Arabia; (A.A.)
| | - Amjad Khalid Alharbi
- Department of Diagnostic Radiology and Imaging, King Abdullah Medical City, Makkah 24246, Saudi Arabia; (A.A.)
| | - Asma Alkhaldi
- Department of Diagnostic Radiology and Imaging, King Abdullah Medical City, Makkah 24246, Saudi Arabia; (A.A.)
| | - Asseel Z. Filimban
- Department of Radiologic Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Abrar Alfatni
- Department of Radiologic Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Reham Kaifi
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Jeddah 22384, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah 22384, Saudi Arabia
- Medical Imaging Department, Ministry of the National Guard—Health Affairs, Jeddah 11426, Saudi Arabia
| | - Ahmad Albngali
- Department of Radiologic Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammed Alkharaiji
- Department of Public Health, College of Health Sciences, Saudi Electronic University, Riyadh 93499, Saudi Arabia
| | - Omar Alserihy
- Department of Diagnostic Radiology and Imaging, King Abdullah Medical City, Makkah 24246, Saudi Arabia; (A.A.)
| | - Salahaden R. Sultan
- Department of Radiologic Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Radiology, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
17
|
Fedotova EI, Berezhnov AV, Popov DY, Shitikova EY, Vinokurov AY. The Role of mtDNA Mutations in Atherosclerosis: The Influence of Mitochondrial Dysfunction on Macrophage Polarization. Int J Mol Sci 2025; 26:1019. [PMID: 39940788 PMCID: PMC11817597 DOI: 10.3390/ijms26031019] [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: 11/27/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Atherosclerosis is a complex inflammatory process associated with high-mortality cardiovascular diseases. Today, there is a growing body of evidence linking atherosclerosis to mutations of mitochondrial DNA (mtDNA). But the mechanism of this link is insufficiently studied. Atherosclerosis progression involves different cell types and macrophages are one of the most important. Due to their high plasticity, macrophages can demonstrate pro-inflammatory and pro-atherogenic (macrophage type M1) or anti-inflammatory and anti-atherogenic (macrophage type M2) effects. These two cell types, formed as a result of external stimuli, differ significantly in their metabolic profile, which suggests the central role of mitochondria in the implementation of the macrophage polarization route. According to this, we assume that mtDNA mutations causing mitochondrial disturbances can play the role of an internal trigger, leading to the formation of macrophage M1 or M2. This review provides a comparative analysis of the characteristics of mitochondrial function in different types of macrophages and their possible associations with mtDNA mutations linked with inflammation-based pathologies including atherosclerosis.
Collapse
Affiliation(s)
- Evgeniya I. Fedotova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (E.I.F.); (A.V.B.)
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Alexey V. Berezhnov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (E.I.F.); (A.V.B.)
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Daniil Y. Popov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Elena Y. Shitikova
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Andrey Y. Vinokurov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| |
Collapse
|
18
|
Chandimali N, Bak SG, Park EH, Lim HJ, Won YS, Kim EK, Park SI, Lee SJ. Free radicals and their impact on health and antioxidant defenses: a review. Cell Death Discov 2025; 11:19. [PMID: 39856066 PMCID: PMC11760946 DOI: 10.1038/s41420-024-02278-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Free radicals, characterized by the presence of unpaired electrons, are highly reactive species that play a significant role in human health. These molecules can be generated through various endogenous processes, such as mitochondrial respiration and immune cell activation, as well as exogenous sources, including radiation, pollution, and smoking. While free radicals are essential for certain physiological processes, such as cell signaling and immune defense, their overproduction can disrupt the delicate balance between oxidants and antioxidants, leading to oxidative stress. Oxidative stress results in the damage of critical biomolecules like DNA, proteins, and lipids, contributing to the pathogenesis of various diseases. Chronic conditions such as cancer, cardiovascular diseases, neurodegenerative disorders, and inflammatory diseases have been strongly associated with the harmful effects of free radicals. This review provides a comprehensive overview of the characteristics and types of free radicals, their mechanisms of formation, and biological impacts. Additionally, we explore natural compounds and extracts studied for their antioxidant properties, offering potential therapeutic avenues for managing free radical-induced damage. Future research directions are also discussed to advance our understanding and treatment of free radical-associated diseases.
Collapse
Affiliation(s)
- Nisansala Chandimali
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
- Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Korea
| | - Seon Gyeong Bak
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
| | - Eun Hyun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Korea
| | - Hyung-Jin Lim
- Scripps Korea Antibody Institute, Chuncheon, 24341, Korea
| | - Yeong-Seon Won
- Division of Research Management, Department of Bioresource Industrialization, Honam National Institute of Biological Resource, Mokpo, 58762, Korea
| | - Eun-Kyung Kim
- Nutritional Education Major, Graduate School of Education, Dong-A University, Busan, 49315, Korea
| | - Sang-Ik Park
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Korea.
| | - Seung Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea.
- Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Korea.
| |
Collapse
|
19
|
Fawzy El‐Sayed K, Mahlandt E, Schlicht K, Enthammer K, Tölle J, Wagner J, Hartmann K, Ebeling PR, Graetz C, Laudes M, Dörfer CE, Schulte DM. Effects of oxidized LDL versus IL-1ß/TNF-ɑ/INFɣ on human gingival mesenchymal stem cells properties. J Periodontal Res 2025; 60:77-89. [PMID: 38952262 PMCID: PMC11840472 DOI: 10.1111/jre.13319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 07/03/2024]
Abstract
AIMS Oxidized low-density lipoprotein (oxLDL) is an important player in the course of metabolic inflammatory diseases. oxLDL was identified in the gingival crevicular fluid, denoting possible associations between oxLDL-induced inflammation and periodontal disease. The current investigation compared for the first-time direct effects of oxLDL to a cytokine cocktail of IL-1ß/TNF-ɑ/INF-γ on gingival mesenchymal stem cells' (G-MSCs) attributes. METHODS Human third passage G-MSCs, isolated from connective tissue biopsies (n = 5) and characterized, were stimulated in three groups over 7 days: control group, cytokine group (IL-1β[1 ng/mL], TNF-α[10 ng/mL], IFN-γ[100 ng/mL]), or oxLDL group (oxLDL [50 μg/mL]). Next Generation Sequencing and KEGG pathway enrichment analysis, stemness gene expression (NANOG/SOX2/OCT4A), cellular proliferation, colony-formation, multilinear potential, and altered intracellular pathways were investigated via histochemistry, next-generation sequencing, and RT-qPCR. RESULTS G-MSCs exhibited all mesenchymal stem cells' characteristics. oxLDL group and cytokine group displayed no disparities in their stemness markers (p > .05). Next-generation-sequencing revealed altered expression of the TXNIP gene in response to oxLDL treatment compared with controls (p = .04). Following an initial boosting for up to 5 days by inflammatory stimuli, over 14 day, cellular counts [median count ×10-5 (Q25/Q75)] were utmost in control - [2.6607 (2.0804/4.5357)], followed by cytokine - [0.0433 (0.0026/1.4215)] and significantly lowered in the oxLDL group [0.0274 (0.0023/0.7290); p = .0047]. Osteogenic differentiation [median relative Ca2+ content(Q25/Q75)] was significantly lower in cytokine - [0.0066 (0.0052/0.0105)] compared to oxLDL - [0.0144 (0.0108/0.0216)] (p = .0133), with no differences notable for chondrogenic and adipogenic differentiation (p > .05). CONCLUSIONS Within the current investigation's limitations, in contrast to cytokine-mediated inflammation, G-MSCs appear to be minimally responsive to oxLDL-mediated metabolic inflammation, with little negative effect on their differentiation attributes and significantly reduced cellular proliferation.
Collapse
Affiliation(s)
- Karim Fawzy El‐Sayed
- Clinic for Conservative Dentistry and PeriodontologyUniversity Hospital of Schleswig‐HolsteinKielGermany
- Oral Medicine and Periodontology Department, Faculty of DentistryCairo UniversityCairoEgypt
- Stem Cells and Tissue Engineering Unit, Faculty of DentistryCairo UniversityCairoEgypt
| | - Elena Mahlandt
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Kristina Schlicht
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Kim Enthammer
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Johannes Tölle
- Clinic for Conservative Dentistry and PeriodontologyUniversity Hospital of Schleswig‐HolsteinKielGermany
- Department of DermatologyUniversity Hospital Schleswig‐HolsteinKielGermany
- Institute of ImmunologyUniversity Hospital Schleswig‐HolsteinKielGermany
| | - Juliane Wagner
- Department of Oral and Maxillofacial SurgeryUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Katharina Hartmann
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Peter R. Ebeling
- Department of Medicine, School of Clinical Sciences at Monash HealthMonash UniversityMelbourneVictoriaAustralia
| | - Christian Graetz
- Clinic for Conservative Dentistry and PeriodontologyUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Mathias Laudes
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine IUniversity Hospital Schleswig‐HolsteinKielGermany
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and PeriodontologyUniversity Hospital of Schleswig‐HolsteinKielGermany
| | - Dominik M. Schulte
- Institute of Diabetes and Clinical Metabolic ResearchUniversity Hospital of Schleswig‐HolsteinKielGermany
- Department of Medicine, School of Clinical Sciences at Monash HealthMonash UniversityMelbourneVictoriaAustralia
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Internal Medicine IUniversity Hospital Schleswig‐HolsteinKielGermany
| |
Collapse
|
20
|
Cohen Z, Williams RM. Single-Walled Carbon Nanotubes as Optical Transducers for Nanobiosensors In Vivo. ACS NANO 2024; 18:35164-35181. [PMID: 39696968 PMCID: PMC11697343 DOI: 10.1021/acsnano.4c13076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Semiconducting single-walled carbon nanotubes (SWCNTs) may serve as signal transducers for nanobiosensors. Recent studies have developed innovative methods of engineering molecularly specific sensors, while others have devised methods of deploying such sensors within live animals and plants. These advances may potentiate the use of implantable, noninvasive biosensors for continuous drug, disease, and contaminant monitoring based on the optical properties of single-walled carbon nanotubes (SWCNTs). Such tools have substantial potential to improve disease diagnostics, prognosis, drug safety, therapeutic response, and patient compliance. Outside of clinical applications, such sensors also have substantial potential in environmental monitoring or as research tools in the lab. However, substantial work remains to be done to realize these goals through further advances in materials science and engineering. Here, we review the current landscape of quantitative SWCNT-based optical biosensors that have been deployed in living plants and animals. Specifically, we focused this review on methods that have been developed to deploy SWCNT-based sensors in vivo as well as analytes that have been detected by SWCNTs in vivo. Finally, we evaluated potential future directions to take advantage of the promise outlined here toward field-deployable or implantable use in patients.
Collapse
Affiliation(s)
- Zachary Cohen
- Department
of Biomedical Engineering, The City College
of New York, New York, New York 10031, United States
| | - Ryan M. Williams
- Department
of Biomedical Engineering, The City College
of New York, New York, New York 10031, United States
- PhD
Program in Chemistry, The Graduate Center
of The City University of New York, New York, New York 10016, United States
| |
Collapse
|
21
|
Skau E, Wagner P, Leppert J, Ärnlöv J, Hedberg P. Determinants of growth differentiation factor 15 plasma levels in outpatients with peripheral arterial disease. Ups J Med Sci 2024; 129:11001. [PMID: 39780955 PMCID: PMC11708457 DOI: 10.48101/ujms.v129.11001] [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: 08/27/2024] [Revised: 11/27/2024] [Accepted: 08/27/2024] [Indexed: 01/11/2025] Open
Abstract
Background Growth differentiation factor 15 (GDF-15) is a robust prognostic biomarker in patients with cardiovascular (CV) disease, and a better understanding of its clinical determinants is desirable. We aimed to study the associations between GDF-15 levels and traditional CV risk factors, indicators of atherosclerotic burden, and cardiac geometry and dysfunction in outpatients with peripheral arterial disease (PAD). Methods An explorative cross-sectional study (Study of Atherosclerosis in Vastmanland, Västerås, Sweden) included 439 outpatients with carotid or lower extremity PAD. The mean age was 70 years (standard deviation [SD] 7), and 59% of the patients were men. Plasma levels of GDF-15 were obtained along with potential determinants, including medical history, biochemical data, echocardiographic measures of cardiac geometry and function, ankle-brachial index (ABI), and carotid ultrasonographic data on intima-media thickness (IMT) and occurrence of carotid stenosis. The relations between GDF-15 concentrations (transformed with the natural logarithm) and the different determinants were evaluated using uni- and multivariable linear regression models. All pre-specified variables were included in the multivariable models. Results The multivariable analysis identified independent relations of GDF-15 with several of the included variables (adjusted R 2 = 0.48). Diabetes (beta coefficient [β] of 0.37, 95% confidence interval [95% CI] 0.25 to 0.50), low-density lipoprotein (LDL) cholesterol (β = -0.22, 95% confidence interval [CI]: -0.34 to -0.09), and physical activity (β = -0.16, 95% CI: -0.25 to -0.06) had the strongest associations. In contrast, no significant independent associations with GDF-15 level were observed for cardiac geometry and function, ABI, IMT, or carotid stenosis. Conclusions Circulating GDF-15 is more strongly associated with traditional CV risk factors, especially diabetes, LDL cholesterol, and physical activity than with specific indicators of atherosclerotic burden or cardiac dysfunction. To better understand the pathophysiological role of GDF-15 and its link to clinical outcomes in patients with PAD, future studies should focus on the metabolic processes involved in atherosclerotic disease.
Collapse
Affiliation(s)
- Emma Skau
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
- Department of Cardiology, Danderyd University Hospital, Stockholm, Sweden
| | - Philippe Wagner
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
| | - Jerzy Leppert
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
| | - Johan Ärnlöv
- School of Health and Social Studies, Dalarna University, Falun, Sweden
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Huddinge, Sweden
| | - Pär Hedberg
- Centre for Clinical Research, Uppsala University, Västmanland County Hospital, Västerås, Sweden
- Department of Clinical Physiology, Västmanland County Hospital, Västerås, Sweden
| |
Collapse
|
22
|
Liu Y, Lu K, Zhang R, Hu D, Yang Z, Zeng J, Cai W. Advancements in the Treatment of Atherosclerosis: From Conventional Therapies to Cutting-Edge Innovations. ACS Pharmacol Transl Sci 2024; 7:3804-3826. [PMID: 39698263 PMCID: PMC11651175 DOI: 10.1021/acsptsci.4c00574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Atherosclerosis is a leading cause of morbidity and mortality worldwide, driven by a complex interplay of lipid dysregulation, inflammation, and vascular pathology. Despite advancements in understanding the multifactorial nature of atherosclerosis and improvements in clinical management, existing therapies often fall short in reversing the disease, focusing instead on symptom alleviation and risk reduction. This review highlights recent strides in identifying genetic markers, elucidating inflammatory pathways, and understanding environmental contributors to atherosclerosis. It also evaluates the efficacy and limitations of current pharmacological treatments, revascularization techniques, and the impact of these interventions on patient outcomes. Furthermore, we explore innovative therapeutic strategies, including the promising fields of nanomedicine, nucleic acid-based therapies, and immunomodulation, which offer potential for targeted and effective treatment modalities. However, integrating these advances into clinical practice is challenged by regulatory, economic, and logistical barriers. This review synthesizes the latest research and clinical advancements to provide a comprehensive roadmap for future therapeutic strategies and emphasize the critical need for innovative approaches to fundamentally change the course of atherosclerosis management.
Collapse
Affiliation(s)
- Yan Liu
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Center
for Molecular Imaging and Nuclear Medicine, State Key Laboratory of
Radiation Medicine and Protection, School for Radiological and Interdisciplinary
Sciences (RAD-X), Collaborative Innovation Center of Radiological
Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Kuan Lu
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Ruru Zhang
- Center
for Molecular Imaging and Nuclear Medicine, State Key Laboratory of
Radiation Medicine and Protection, School for Radiological and Interdisciplinary
Sciences (RAD-X), Collaborative Innovation Center of Radiological
Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Dongliang Hu
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
- Center
for Molecular Imaging and Nuclear Medicine, State Key Laboratory of
Radiation Medicine and Protection, School for Radiological and Interdisciplinary
Sciences (RAD-X), Collaborative Innovation Center of Radiological
Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Zhe Yang
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Jianfeng Zeng
- Center
for Molecular Imaging and Nuclear Medicine, State Key Laboratory of
Radiation Medicine and Protection, School for Radiological and Interdisciplinary
Sciences (RAD-X), Collaborative Innovation Center of Radiological
Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Wu Cai
- The
Second Affiliated Hospital of Soochow University, Suzhou, 215000, China
| |
Collapse
|
23
|
Thangasparan S, Kamisah Y, Ugusman A, Mohamad Anuar NN, Ibrahim N‘I. Unravelling the Mechanisms of Oxidised Low-Density Lipoprotein in Cardiovascular Health: Current Evidence from In Vitro and In Vivo Studies. Int J Mol Sci 2024; 25:13292. [PMID: 39769058 PMCID: PMC11676878 DOI: 10.3390/ijms252413292] [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: 11/13/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Cardiovascular diseases (CVD) are the number one cause of death worldwide, with atherosclerosis, which is the formation of fatty plaques in the arteries, being the most common underlying cause. The activation of inflammatory events and endothelium dysfunction are crucial for the development and pathophysiology of atherosclerosis. Elevated circulating levels of low-density lipoprotein (LDL) have been associated with severity of atherosclerosis. LDL can undergo oxidative modifications, resulting in oxidised LDL (oxLDL). OxLDL has been found to have antigenic potential and contribute significantly to atherosclerosis-associated inflammation by activating innate and adaptive immunity. Various inflammatory stimuli such as interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α) and intercellular adhesion molecule 1 (ICAM-1) play major roles in atherosclerosis. To date, studies have provided valuable insights into the role of oxLDL in the development of atherosclerosis. However, there remains a gap in understanding the specific pathways involved in this process. This review aims to provide and discuss the mechanisms by which oxLDL modulates signalling pathways that cause cardiovascular diseases by providing in vitro and in vivo experimental evidence. Its critical role in triggering and sustaining endothelial dysfunction highlights its potential as a therapeutic target. Advancing the understanding of its atherogenic role and associated signalling pathways could pave the way for novel targeted therapeutic strategies to combat atherosclerosis more effectively.
Collapse
Affiliation(s)
- Sahsikala Thangasparan
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
| | - Azizah Ugusman
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia
| | - Nur Najmi Mohamad Anuar
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
- Programme of Biomedical Science, Center for Toxicology & Health Risk Studies (CORE), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Nurul ‘Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras 56000, Kuala Lumpur, Malaysia; (S.T.); (Y.K.)
- Cardiovascular and Pulmonary Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia; (A.U.); (N.N.M.A.)
| |
Collapse
|
24
|
Prasad K. Atherogenic Effect of Homocysteine, a Biomarker of Inflammation and Its Treatment. Int J Angiol 2024; 33:262-270. [PMID: 39502352 PMCID: PMC11534477 DOI: 10.1055/s-0044-1788280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
Hyperhomocysteinemia (HHcy) is an independent risk factor for atherosclerosis. Ischemic stroke and heart disease, coronary heart disease, and cardiovascular disease are events resulting from long-lasting and silent atherosclerosis. This paper deals with the synthesis of homocysteine (Hcy), causes of HHcy, mechanism of HHcy-induced atherosclerosis, and treatment of HHcy. Synthesis and metabolism of Hcy involves demethylation, transmethylation, and transsulfuration, and these processes require vitamin B 6 and vitamin B 12 folic acid (vitamin B 9 ). Causes of HHcy include deficiency of vitamins B 6 , B 9 , and B 12 , genetic defects, use of smokeless tobacco, cigarette smoking, alcohol consumption, diabetes, rheumatoid arthritis, low thyroid hormone, consumption of caffeine, folic acid antagonist, cholesterol-lowering drugs (niacin), folic acid antagonist (phenytoin), prolonged use of proton pump inhibitors, metformin, and hypertension. HHcy-induced atherosclerosis may be mediated through oxidative stress, decreased availability of nitric oxide (NO), increased expression of monocyte chemoattractant protein-1, smooth muscle cell proliferation, increased thrombogenicity, and induction of arterial connective tissue. HHcy increases the generation of atherogenic biomolecules such as nuclear factor-kappa B, proinflammatory cytokines (IL-1β, IL-6, and IL-8), cell adhesion molecules (intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selection), growth factors (IGF-1 and TGF-β), and monocyte colony-stimulating factor which lead to the development of atherosclerosis. NO which is protective against the development of atherosclerosis is reduced by HHcy. Therapy with folic acid, vitamin B 6 , and vitamin B 12 lowers the levels of Hcy, with folic acid being the most effective. Dietary sources of folic acid, vitamin B 6 , vitamin B 12 , omega-3 fatty acid, and green coffee extract reduce Hcy. Abstaining from drinking coffee and alcohol, and smoking also reduces blood levels of Hcy. In conclusion, HHcy induces atherosclerosis by generating atherogenic biomolecules, and treatment of atherosclerosis-induced diseases may be by reducing the levels of Hcy.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
25
|
Prasad K. Role of C-Reactive Protein, An Inflammatory Biomarker in The Development of Atherosclerosis and Its Treatment. Int J Angiol 2024; 33:271-281. [PMID: 39502349 PMCID: PMC11534478 DOI: 10.1055/s-0044-1788296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
This article deals with the role of c-reactive protein (CRP) in the development of atherosclerosis and its treatment. CRP has a predictive value in ischemic heart disease, restenosis, coronary artery disease, aortic atherosclerosis, and cerebrovascular disease. This article deals with the synthesis and mechanism of CRP-induced atherosclerosis and its treatment. CRP increases the formation of numerous atherogenic biomolecules such as reactive oxygen species (ROS), cytokines (interleukin [IL]-1β and IL-6), cell adhesion molecules (intercellular adhesion molecule-1, vascular cell adhesion molecule-1, monocyte chemoattractant protein-1, activated complement C 5 , monocyte colony-stimulating factor, and numerous growth factors [insulin-like growth factor, platelet-derived growth factor, and transforming growth factor-β]). ROS mildly oxidizes low-density lipoprotein (LDL)-cholesterol to form minimally modified LDL which is further oxidized to form oxidized LDL. The above atherogenic biomolecules are involved in the development of atherosclerosis and has been described in detail in the text. This paper also deals with the treatment modalities for CRP-induced atherosclerosis which includes lipid-lowering drugs, antihypertensive drugs, antioxidants, aspirin, antidiabetic drugs, angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, regular physical activity, weight reduction, and stoppage of cigarette smoking. In conclusion, CRP induces atherosclerosis through increases in atherogenic biomolecules and the treatment modalities would prevent, regress, and slow the progression of CRP-induced atherosclerosis.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
26
|
Abrego-Guandique DM, Galmés S, García-Rodríguez A, Cannataro R, Caroleo MC, Ribot J, Bonet ML, Cione E. β-Carotene Impacts the Liver MicroRNA Profile in a Sex-Specific Manner in Mouse Offspring of Western Diet-Fed Mothers: Results from Microarray Analysis by Direct Hybridization. Int J Mol Sci 2024; 25:12899. [PMID: 39684610 DOI: 10.3390/ijms252312899] [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: 10/18/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Maternal unbalanced diets cause adverse metabolic programming and affect the offspring's liver microRNA (miRNA) profile. The liver is a site of β-carotene (BC) metabolism and a target of BC action. We studied the interaction of maternal Western diet (WD) and early-life BC supplementation on the epigenetic remodeling of offspring's liver microRNAs. Mouse offspring of WD-fed mothers were given a daily placebo (controls) or BC during suckling. Biometric parameters and liver miRNAome by microarray hybridization were analyzed in newly weaned animals. BC sex-dependently impacted the liver triacylglycerol content. The liver miRNAome was also differently affected in male and female offspring, with no overlap in differentially expressed (DE) miRNAs between sexes and more impact in females. Bioinformatic analysis of DE miRNA predicted target genes revealed enrichment in biological processes/pathways related to metabolic processes, regulation of developmental growth and circadian rhythm, liver homeostasis and metabolism, insulin resistance, and neurodegeneration, among others, with differences between sexes. Fifty-five percent of the overlapping target genes in both sexes identified were targeted by DE miRNAs changed in opposite directions in males and females. The results identify sex-dependent responses of the liver miRNA expression profile to BC supplementation during suckling and may sustain further investigations regarding the long-term impact of early postnatal life BC supplementation on top of an unbalanced maternal diet.
Collapse
Affiliation(s)
| | - Sebastià Galmés
- Laboratory of Molecular Biology, Nutrition, and Biotechnology (LBNB), Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Research Group, Universitat de les Illes Balears, 07122 Palma, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Adrián García-Rodríguez
- Laboratory of Molecular Biology, Nutrition, and Biotechnology (LBNB), Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Research Group, Universitat de les Illes Balears, 07122 Palma, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Roberto Cannataro
- Galascreen Laboratories, University of Calabria, 87036 Rende, Italy
- Research Division, Dynamical Business & Science Society-DBSS International SAS, Bogotá 110311, Colombia
| | - Maria Cristina Caroleo
- Department of Health Sciences, University of Magna Graecia Catanzaro, 88100 Catanzaro, Italy
- Galascreen Laboratories, University of Calabria, 87036 Rende, Italy
| | - Joan Ribot
- Laboratory of Molecular Biology, Nutrition, and Biotechnology (LBNB), Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Research Group, Universitat de les Illes Balears, 07122 Palma, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria Luisa Bonet
- Laboratory of Molecular Biology, Nutrition, and Biotechnology (LBNB), Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Research Group, Universitat de les Illes Balears, 07122 Palma, Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa), 07120 Palma, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Artificial Intelligence Research Institute of the Balearic Islands (IAIB), University of the Balearic Islands, 07122 Palma, Spain
| | - Erika Cione
- Galascreen Laboratories, University of Calabria, 87036 Rende, Italy
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
27
|
Marchán-Figueroa Y, Tepec-Casarrubias B, de la Cruz-Mosso U, Astudillo-López CC, Matia-García I, Salgado-Goytia L, Espinoza-Rojo M, Castro-Alarcón N, Flores-Alfaro E, Parra-Rojas I. Relationship Between Serum Levels of Oxidized Lipoproteins, Circulating Levels of Myeloperoxidase and Paraoxonase 1, and Diet in Young Subjects with Insulin Resistance. Nutrients 2024; 16:3930. [PMID: 39599716 PMCID: PMC11597308 DOI: 10.3390/nu16223930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/03/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Oxidized low-density lipoproteins (ox-LDLs) are involved in atherosclerotic plaque formation and progression and have been linked to insulin resistance (IR). Myeloperoxidase is a potent oxidant of lipoproteins related to atherogenic risk. High-density lipoproteins (HDLs) are considered antioxidants due to their association with paraoxonase 1 (PON1). However, HDL can also be oxidized (ox-HDL), and its relationship with IR has not been described. This study evaluated the relationship between circulating levels of myeloperoxidase and paraoxonase 1, diet, and serum levels of ox-LDL and ox-HDL in young people with IR. This cross-sectional study examined 136 young subjects (67 and 69 with and without insulin resistance, respectively). Serum levels of ox-LDL, ox-HDL, myeloperoxidase, and PON1 were quantified using an enzyme-linked immunosorbent assay. The nutritional dietary content of the foods was determined with a food frequency questionnaire, which was analyzed with Nutrimind 2013 software. Serum ox-HDL levels were higher in young subjects without IR than those with IR (p = 0.031). Women with IR presented increased ox-LDL levels compared with women without IR (p = 0.012) and men with IR (p < 0.001). In the IR group, serum ox-LDL levels were negatively correlated with total cholesterol, triglycerides, and LDL-C, whereas the correlation was positive in the insulin-sensitive group. Consumption of vitamins B1 and B2 was related to increased HDL-C levels, while higher ox-LDL levels were related to vitamin K intake. In addition, low energy consumption and phosphorus increased PON1 levels. The results suggest that insulin resistance in young women may promote lipoprotein oxidation, and the intake of B complex vitamins may have an antiatherogenic effect.
Collapse
Affiliation(s)
- Yaquelin Marchán-Figueroa
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico; (Y.M.-F.); (B.T.-C.); (C.C.A.-L.); (I.M.-G.); (L.S.-G.)
| | - Brenda Tepec-Casarrubias
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico; (Y.M.-F.); (B.T.-C.); (C.C.A.-L.); (I.M.-G.); (L.S.-G.)
| | - Ulises de la Cruz-Mosso
- Instituto de Neurociencias Traslacionales, Departamento de Neurociencias, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
| | - Constanza Cecilia Astudillo-López
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico; (Y.M.-F.); (B.T.-C.); (C.C.A.-L.); (I.M.-G.); (L.S.-G.)
| | - Inés Matia-García
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico; (Y.M.-F.); (B.T.-C.); (C.C.A.-L.); (I.M.-G.); (L.S.-G.)
| | - Lorenzo Salgado-Goytia
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico; (Y.M.-F.); (B.T.-C.); (C.C.A.-L.); (I.M.-G.); (L.S.-G.)
| | - Mónica Espinoza-Rojo
- Laboratorio de Biología Molecular y Genómica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico;
| | - Natividad Castro-Alarcón
- Laboratorio de Investigación en Microbiología, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico;
| | - Eugenia Flores-Alfaro
- Laboratorio de Investigación en Epidemiología Clínica y Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico;
| | - Isela Parra-Rojas
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39087, Guerrero, Mexico; (Y.M.-F.); (B.T.-C.); (C.C.A.-L.); (I.M.-G.); (L.S.-G.)
| |
Collapse
|
28
|
Montoro-Alonso S, Duque-Soto C, Rueda-Robles A, Reina-Manuel J, Quirantes-Piné R, Borrás-Linares I, Lozano-Sánchez J. Functional Olive Oil Production via Emulsions: Evaluation of Phenolic Encapsulation Efficiency, Storage Stability, and Bioavailability. Nutrients 2024; 16:3909. [PMID: 39599696 PMCID: PMC11597669 DOI: 10.3390/nu16223909] [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: 10/24/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND/OBJECTIVES Olive oil is valued for its health benefits, largely due to its bioactive compounds, including hydroxytyrosol (HTyr) and oleuropein (OLE), which have antioxidant, anti-inflammatory, and cardioprotective properties. However, many of these compounds are lost during the production process. This study developed a functional olive oil-derived product using water-in-oil emulsions (W/O) to incorporate commercial extracts rich in HTyr and OLE. METHODS HTyr and OLE were encapsulated in a W/O emulsion to preserve their bioactivity. The encapsulation efficiency (EE) was evaluated, and the performance of the emulsion was tested using an in vitro gastrointestinal digestion model. Bioaccessibility was measured by calculating the recovery percentage of HTyr and OLE during the digestion stages. RESULTS The results showed that OLE exhibited higher EE (88%) than HTyr (65%). During digestion, HTyr exhibited a gradual and controlled release, with bioaccessibility exceeding 80% in the gastric phase and a maintained stability throughout the intestinal phase. In contrast, OLE displayed high bioaccessibility in the gastric phase but experienced a notable decrease during the intestinal phase. Overall, the W/O emulsion provided superior protection and stability for both compounds, particularly for the secoiridoids, compared to the non-emulsified oil. CONCLUSIONS The W/O emulsion improved the encapsulation and bioaccessibility of HTyr and OLE, constituting a promising method for enriching olive oil with bioactive phenolic compounds. Therefore, this method could enhance olive oil's health benefits by increasing the availability of these bioactive compounds during digestion, offering the potential for the development of fortified foods.
Collapse
Affiliation(s)
- Sandra Montoro-Alonso
- Department of Food Science and Nutrition, University of Granada, Campus Universitario Cartuja s/n, 18071 Granada, Spain; (S.M.-A.); (J.R.-M.); (J.L.-S.)
| | - Carmen Duque-Soto
- Department of Food Science and Nutrition, University of Granada, Campus Universitario Cartuja s/n, 18071 Granada, Spain; (S.M.-A.); (J.R.-M.); (J.L.-S.)
| | - Ascensión Rueda-Robles
- Department of Food Science and Nutrition, University of Granada, Campus Universitario Cartuja s/n, 18071 Granada, Spain; (S.M.-A.); (J.R.-M.); (J.L.-S.)
| | - José Reina-Manuel
- Department of Food Science and Nutrition, University of Granada, Campus Universitario Cartuja s/n, 18071 Granada, Spain; (S.M.-A.); (J.R.-M.); (J.L.-S.)
| | - Rosa Quirantes-Piné
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Campus Fuentenueva s/n, 18071 Granada, Spain; (R.Q.-P.); (I.B.-L.)
| | - Isabel Borrás-Linares
- Department of Analytical Chemistry, Faculty of Sciences, University of Granada, Campus Fuentenueva s/n, 18071 Granada, Spain; (R.Q.-P.); (I.B.-L.)
| | - Jesús Lozano-Sánchez
- Department of Food Science and Nutrition, University of Granada, Campus Universitario Cartuja s/n, 18071 Granada, Spain; (S.M.-A.); (J.R.-M.); (J.L.-S.)
| |
Collapse
|
29
|
Wojtacha JJ, Morawin B, Wawrzyniak-Gramacka E, Tylutka A, de Freitas AKE, Zembron-Lacny A. Endothelial Dysfunction with Aging: Does Sex Matter? Int J Mol Sci 2024; 25:12203. [PMID: 39596269 PMCID: PMC11594464 DOI: 10.3390/ijms252212203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress and inflammation accompany endothelial dysfunction that results from the excessive or uncontrolled production of reactive oxygen and nitrogen species (RONS) in older adults. This study was designed to assess the usefulness of serum oxi-inflammatory component combinations in vascular disease prediction and prevention with regard to sex. Women (n = 145) and men (n = 50) aged 72.2 ± 7.8 years participated in this project. The females demonstrated the elevated production of hydrogen peroxide (H2O2) and nitric oxide (NO) responsible for intravascular low-density lipoprotein oxidation. NO generation was enhanced in the women, but its bioavailability was reduced, which was expressed by a high 3-nitrotyrosine (3-NitroT) concentration. The relation of NO/3-NitroT (rs = 0.811, p < 0.001) in the women and NO/3-NitroT (rs = -0.611, p < 0.001) in the men showed that sex determines endothelial dysfunction. RONS generation in the women simultaneously promoted endothelial regeneration, as demonstrated by a ~1.5-fold increase in circulating progenitor cells. Inflammation-specific variables, such as the neutrophil-to-lymphocyte ratio, the systemic immune inflammation index, and the neutrophil-to-high-density lipoprotein (HDL) ratio, were reduced in the women and showed their diagnostic utility for clinical prognosis in vascular dysfunction, especially the C-reactive-protein-to-HDL ratio (AUC = 0.980, specificity 94.7%, sensitivity 93.3%, OR = 252, 95% CI 65-967, p < 0.001). This study is the first to have revealed sex-specific changes in the oxi-inflammatory response, which can generate the risk of cardiovascular events at an older age.
Collapse
Affiliation(s)
- Jakub Jozue Wojtacha
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Barbara Morawin
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Edyta Wawrzyniak-Gramacka
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Anna Tylutka
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| | - Ana Karyn Ehrenfried de Freitas
- School of Health Science, Positivo University, 5300 Professor Pedro Viriato Parigot de Souza Street, Campo Comprido, Curitiba 81280-330, PR, Brazil;
- Department of Cardiology, Hospital da Cruz Vermelha Brasileira Filial do Paraná, Av. Vicente Machado, 1280, R. Cap. Souza Franco, 50-Batel, Curitiba 80420-011, PR, Brazil
| | - Agnieszka Zembron-Lacny
- Department of Applied and Clinical Physiology, University of Zielona Gora, 28 Zyty Str., 65-417 Zielona Gora, Poland; (J.J.W.); (B.M.); (E.W.-G.); (A.T.)
| |
Collapse
|
30
|
Lusta KA, Churov AV, Beloyartsev DF, Golovyuk AL, Lee AA, Sukhorukov VN, Orekhov AN. The two coin sides of bacterial extracellular membrane nanovesicles: atherosclerosis trigger or remedy. DISCOVER NANO 2024; 19:179. [PMID: 39532781 PMCID: PMC11557815 DOI: 10.1186/s11671-024-04149-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Among the numerous driving forces that cause the atherosclerotic cardiovascular disease (ASCVD), pathogenic bacterial extracellular membrane nanovesicles (BEMNs) containing toxins and virulence factors appear to be the key trigger of inflammation and atherogenesis, the major processes involved in the pathogenesis of ASCVD. Since BEMNs are the carriers of nanosized biomolecules to distant sites, they are now being considered as a novel drug delivery system. Nowadays, many therapeutic strategies are used to treat ASCVD. However, the conventional anti-atherosclerotic therapies are not effective enough. This primarily due to the inefficiency of non-targeted drug delivery systems to tissue affected areas, which, in turn, leads to numerous side effects, as well as faulty pharmacokinetics. In this regard, nanomedicine methods using nanoparticles (NPs) as targeted drug delivery vehicles proved to be extremely useful. Bioengineered BEMNs equipped with disease-specific ligand moieties and loaded with corresponding drugs represent a promising tool in nanomedicine, which can be used as a novel drug delivery system for a successful therapy of ASCVD. In this review, we outline the involvement of pathogenic BEMNs in the triggering of ASCVD, the conventional therapeutic strategies for the treatment of ASCVD, and the recent trends in nanomedicine using BEMNs and NPs as a vehicle for targeted drug delivery.
Collapse
Affiliation(s)
- Konstantin A Lusta
- Institute for Atherosclerosis Research, Ltd, Osennyaya Street 4-1-207, Moscow, Russia, 121609.
| | - Alexey V Churov
- Institute on Aging Research, Russian Gerontology Clinical Research Center, Pirogov Russian National Research Medical University, Moscow, Russia, 129226
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Alexander L Golovyuk
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, Moscow, Russia, 117997
| | - Arthur A Lee
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
| | - Vasily N Sukhorukov
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| | - Alexander N Orekhov
- Insitute of Human Morphology, Petrovsky Russian National Center of Surgery, 2 Abrikosovsky Lane, Moscow, Russia, 119991
- Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315
| |
Collapse
|
31
|
Cuevas-Sierra A, de la O V, Higuera-Gómez A, Chero-Sandoval L, de Cuevillas B, Martínez-Urbistondo M, Moreno-Torres V, Pintos-Pascual I, Castejón R, Martínez JA. Mediterranean Diet and Olive Oil Redox Interactions on Lactate Dehydrogenase Mediated by Gut Oscillibacter in Patients with Long-COVID-19 Syndrome. Antioxidants (Basel) 2024; 13:1358. [PMID: 39594500 PMCID: PMC11591431 DOI: 10.3390/antiox13111358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Chronic viral inflammation is associated with oxidative stress and changes in gut microbiota. The Mediterranean diet (MD), with recognized anti-inflammatory and antioxidant properties, modulates gut microorganisms, specifically on the interaction between extra virgin olive oil, a key component of the MD with well-documented antioxidant effects. This study investigated the influence of adherence to MD and antioxidant-rich foods (extra virgin olive oil) on biochemical, inflammatory, and microbiota profiles in patients with chronic inflammation defined as a prolonged inflammatory response due to immune dysregulation following the acute phase of the viral infection. Participants were classified into low (n = 54) and high (n = 134) MD adherence groups (cut-off of 7 points based on previous studies utilizing the same threshold in the assessment of MD adherence). Gut microbiota was sequenced using the 16S technique, and the adherence to MD was assessed using a validated questionnaire for a Spanish population. High adherence to the MD was linked to significant improvements in inflammatory and oxidative stress markers, including reductions in LDL-cholesterol, glucose, and lactate dehydrogenase (LDH) levels, an indicative of redox balance, as well as a significant higher consumption of antioxidant foods. Moreover, gut microbiota analysis revealed distinct compositional shifts and a lower abundance of the Oscillibacter genus in the high adherence group. Notably, a significant interaction was observed between MD adherence and extra virgin olive oil consumption, with Oscillibacter abundance influencing LDH levels, suggesting that the MD antioxidant properties may modulate inflammation through gut microbiota-mediated mechanisms. These findings provide new evidence that adherence to the Mediterranean diet can reduce inflammatory markers in patients with long-COVID-19, a population that has not been extensively studied, while also highlighting the potential role of the bacterial genus Oscillibacter in modulating this effect.
Collapse
Affiliation(s)
- Amanda Cuevas-Sierra
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain; (V.d.l.O.); (A.H.-G.); (L.C.-S.); (B.d.C.); (J.A.M.)
- Faculty of Health Sciences, International University of La Rioja (UNIR), 26006 Logroño, Spain;
| | - Victor de la O
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain; (V.d.l.O.); (A.H.-G.); (L.C.-S.); (B.d.C.); (J.A.M.)
- Faculty of Health Sciences, International University of La Rioja (UNIR), 26006 Logroño, Spain;
| | - Andrea Higuera-Gómez
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain; (V.d.l.O.); (A.H.-G.); (L.C.-S.); (B.d.C.); (J.A.M.)
| | - Lourdes Chero-Sandoval
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain; (V.d.l.O.); (A.H.-G.); (L.C.-S.); (B.d.C.); (J.A.M.)
- Department of Endocrinology and Nutrition, University Clinical Hospital, University of Valladolid, 47002 Valladolid, Spain
| | - Begoña de Cuevillas
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain; (V.d.l.O.); (A.H.-G.); (L.C.-S.); (B.d.C.); (J.A.M.)
| | - María Martínez-Urbistondo
- Internal Medicine Service, Puerta de Hierro Majadahonda University Hospital, 28222 Madrid, Spain; (M.M.-U.); (I.P.-P.)
| | - Victor Moreno-Torres
- Faculty of Health Sciences, International University of La Rioja (UNIR), 26006 Logroño, Spain;
- Internal Medicine Service, Puerta de Hierro Majadahonda University Hospital, 28222 Madrid, Spain; (M.M.-U.); (I.P.-P.)
| | - Ilduara Pintos-Pascual
- Internal Medicine Service, Puerta de Hierro Majadahonda University Hospital, 28222 Madrid, Spain; (M.M.-U.); (I.P.-P.)
| | - Raquel Castejón
- Internal Medicine Service, Puerta de Hierro Majadahonda University Hospital, 28222 Madrid, Spain; (M.M.-U.); (I.P.-P.)
| | - J. Alfredo Martínez
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Campus of International Excellence (CEI) UAM+CSIC, 28049 Madrid, Spain; (V.d.l.O.); (A.H.-G.); (L.C.-S.); (B.d.C.); (J.A.M.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Centro de Medicina y Endocrinología, Universidad de Valladolid, 47005 Valladolid, Spain
| |
Collapse
|
32
|
Le MPT, Marasinghe CK, Je JY. Chitosan oligosaccharides: A potential therapeutic agent for inhibiting foam cell formation in atherosclerosis. Int J Biol Macromol 2024; 282:137186. [PMID: 39491693 DOI: 10.1016/j.ijbiomac.2024.137186] [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: 01/02/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Foam cell formation is a key hallmark in atherosclerosis and associated cardiovascular diseases (CVDs). The potential anti-atherosclerotic potential of chitosan oligosaccharides (COS) was investigated using oxLDL-treated RAW264.7 murine cells. COS treatment led to a significant inhibition of lipid accumulation, as demonstrated by Oil Red O staining, and reduced levels of total cholesterol, free cholesterol, cholesterol esters, and triglycerides in.oxLDL-treated RAW264.7 cells. COS blocked cholesterol influx through down-regulating class A1 scavenger receptors (SR-A1) and cluster of differentiation 36 (CD36) expression and stimulated cholesterol efflux through up-regulating ABC transporters ABCA-1 and ABCG-1 expressions. Additionally, COS treatment stimulated nuclear signaling pathways involving peroxisome proliferator-activated receptor-γ (PPAR-γ) and liver X receptor α (LXR-α), and also led to the phosphorylation of AMP-activated protein kinase (AMPK). COS further demonstrated anti-inflammatory effects by inhibiting the production of pro-inflammatory cytokines and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in oxLDL-treated RAW264.7 cells, through suppression of NF-κB signaling. Furthermore, COS alleviated oxidative stress induced by oxLDL by activating Nrf2 signaling and enhancing the expression of antioxidant genes, including heme oxygenase-1 (HO-1), superoxide dismutase (SOD), glutathione peroxidase (Gpx), and catalase (CAT). In conclusion, COS can be beneficial in preventing atherosclerosis and related diseases.
Collapse
Affiliation(s)
- My Phuong Thi Le
- Department of Food and Nutrition, Pukyong National University, Busan 48513, Republic of Korea
| | | | - Jae-Young Je
- Major of Human Bioconvergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
33
|
Atcha H, Kulkarni D, Meli VS, Veerasubramanian PK, Wang Y, Cahalan MD, Pathak MM, Liu WF. Piezo1-mediated mechanotransduction enhances macrophage oxidized low-density lipoprotein uptake and atherogenesis. PNAS NEXUS 2024; 3:pgae436. [PMID: 39544498 PMCID: PMC11563038 DOI: 10.1093/pnasnexus/pgae436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/21/2024] [Indexed: 11/17/2024]
Abstract
Macrophages in the vascular wall ingest and clear lipids, but abundant lipid accumulation leads to foam cell formation and atherosclerosis, a pathological condition often characterized by tissue stiffening. While the role of biochemical stimuli in the modulation of macrophage function is well studied, the role of biophysical cues and the molecules involved in mechanosensation are less well understood. Here, we use genetic and pharmacological tools to show extracellular oxidized low-density lipoproteins (oxLDLs) stimulate Ca2+ signaling through activation of the mechanically gated ion channel Piezo1. Moreover, macrophage Piezo1 expression is critical in the transduction of environmental stiffness and channel deletion suppresses, whereas a gain-of-function mutation exacerbates oxLDL uptake. Additionally, we find that depletion of myeloid Piezo1 protects from atherosclerotic plaque formation in vivo. Together, our study highlights an important role for Piezo1 and its respective mutations in macrophage mechanosensing, lipid uptake, and cardiovascular disease.
Collapse
Affiliation(s)
- Hamza Atcha
- Department of Bioengineering, University of California, San Diego, La Jolla 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla 92037, USA
| | - Daanish Kulkarni
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
| | - Vijaykumar S Meli
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine 92697, USA
| | - Praveen Krishna Veerasubramanian
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
| | - Yuchun Wang
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California Irvine, Irvine 92697, USA
| | - Medha M Pathak
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- Department of Physiology and Biophysics, University of California Irvine, Irvine 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine 92697, USA
| | - Wendy F Liu
- Department of Biomedical Engineering, University of California, Irvine, Irvine 92697, USA
- The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine 92697, USA
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine 92697, USA
| |
Collapse
|
34
|
Alharbi HOA, Khan A, Rahmani AH. Investigating the Role of Hub Calcification Proteins in Atherosclerosis via Integrated Transcriptomics and Network-Based Approach. BIOLOGY 2024; 13:867. [PMID: 39596822 PMCID: PMC11592380 DOI: 10.3390/biology13110867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory condition of the arteries, characterized by plaque formation that can restrict blood flow and lead to potentially fatal cardiovascular events. Given that AS is responsible for a quarter of global deaths, this study aimed to develop a systematic bioinformatics approach to identify biomarkers and regulatory targets involved in plaque development, with the goal of reducing cardiovascular disease risk. AS-specific mRNA expression profiles were retrieved from a publicly accessible database, followed by differentially expressed genes (DEGs) identification and AS-specific weighted gene co-expression network (WGCN) construction. Thereafter, calcification and atherosclerosis-specific (CASS) DEGs were utilized for protein-protein interaction network (PPIN) formation, followed by gene ontology (GO) term and pathway enrichment analyses. Lastly, AS-specific 3-node miRNA feed-forward loop (FFL) construction and analysis was performed. Microarray datasets GSE43292 and GSE28829 were obtained from gene expression omnibus (GEO). A total of 3785 and 6176 DEGs were obtained in case of GSE28829 and GSE43292; 3256 and 5962 module DEGs corresponding to GSE28829 and GSE43292 were obtained from WGCN. From a total of 54 vascular calcification (VC) genes, 20 and 29 CASS-DEGs corresponding to GSE28829 and GSE43292 were overlapped. As observed from FFL centrality measures, the highest-order subnetwork motif comprised one TF (SOX7), one miRNA (miR-484), and one mRNA (SPARC) in the case of GSE28829. Also, in the case of GSE43292, the highest-order subnetwork motif comprised one TF (ESR2), one miRNA (miR-214-3p), and one mRNA (MEF2C). These findings have important implications for developing new therapeutic strategies for AS. The identified TFs and miRNAs may serve as potential therapeutic targets for treating atherosclerotic plaques, offering insights into the molecular mechanisms underlying the pathogenesis and highlighting new avenues for research and treatment.
Collapse
Affiliation(s)
- Hajed Obaid A. Alharbi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Asifa Khan
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA;
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
35
|
Mosalmanzadeh N, Pence BD. Oxidized Low-Density Lipoprotein and Its Role in Immunometabolism. Int J Mol Sci 2024; 25:11386. [PMID: 39518939 PMCID: PMC11545486 DOI: 10.3390/ijms252111386] [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: 08/19/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Modified cholesterols such as oxidized low-density lipoprotein (OxLDL) contribute to atherosclerosis and other disorders through the promotion of foam cell formation and inflammation. In recent years, it has become evident that immune cell responses to inflammatory molecules such as OxLDLs depend on cellular metabolic functions. This review examines the known effects of OxLDL on immunometabolism and immune cell responses in atherosclerosis and several other diseases. We additionally provide context on the relationship between OxLDL and aging/senescence and identify gaps in the literature and our current understanding in these areas.
Collapse
Affiliation(s)
| | - Brandt D. Pence
- College of Health Sciences and Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN 38111, USA
| |
Collapse
|
36
|
Lara-Guzmán OJ, Arango-González Á, Rivera DA, Muñoz-Durango K, Sierra JA. The colonic polyphenol catabolite dihydroferulic acid (DHFA) regulates macrophages activated by oxidized LDL, 7-ketocholesterol, and LPS switching from pro- to anti-inflammatory mediators. Food Funct 2024; 15:10399-10413. [PMID: 39320081 DOI: 10.1039/d4fo02114b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Macrophage activation plays a central role in the development of atherosclerotic plaques. Interaction with oxidized low-density lipoprotein (oxLDL) leads to macrophage differentiation into foam cells and oxylipin production, contributing to plaque formation. 7-Ketocholesterol (7KC) is an oxidative byproduct of cholesterol found in oxLDL particles and is considered a factor contributing to plaque progression. During atherosclerotic lesion regression or stabilization, macrophages undergo a transformation from a pro-inflammatory phenotype to a reparative anti-inflammatory state. Interleukin-10 (IL-10) and PGE1 appear to be crucial in resolving both acute and chronic inflammatory processes. After coffee consumption, the gut microbiota processes non-absorbed chlorogenic acids producing various lower size phenolic acids. These colonic catabolites, including dihydroferulic acid (DHFA), may exert various local and systemic effects. We focused on DHFA's impact on inflammation and oxidative stress in THP-1 macrophages exposed to oxLDL, 7KC, and lipopolysaccharides (LPS). Our findings reveal that DHFA inhibits the release of several pro-inflammatory mediators induced by LPS in macrophages, such as CCL-2, CCL-3, CCL-5, TNF-α, IL-6, and IL-17. Furthermore, DHFA reduces IL-18 and IL-1β secretion in an inflammasome-like model. DHFA demonstrated additional benefits: it decreased oxLDL uptake and CD36 expression induced by oxLDL, regulated reactive oxygen species (ROS) and 8-isoprostane secretion (indicating oxidative stress modulation), and selectively increased IL-10 and PGE1 levels in the presence of inflammatory stimuli (LPS and 7KC). Finally, our study highlights the pivotal role of PGE1 in foam cell inhibition and inflammation regulation within activated macrophages. This study highlights DHFA's potential as an antioxidant and anti-inflammatory agent, particularly due to its ability to induce PGE1 and IL-10.
Collapse
Affiliation(s)
- Oscar J Lara-Guzmán
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Ángela Arango-González
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Diego A Rivera
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Katalina Muñoz-Durango
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| | - Jelver A Sierra
- Vidarium - Nutrition, Health and Wellness Research Center, Nutresa Business Group, Calle 8 Sur No. 50-67, Medellin, Colombia.
| |
Collapse
|
37
|
Zheng H, Vidili G, Casu G, Navarese EP, Sechi LA, Chen Y. Microplastics and nanoplastics in cardiovascular disease-a narrative review with worrying links. FRONTIERS IN TOXICOLOGY 2024; 6:1479292. [PMID: 39449982 PMCID: PMC11499192 DOI: 10.3389/ftox.2024.1479292] [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: 08/13/2024] [Accepted: 09/30/2024] [Indexed: 10/26/2024] Open
Abstract
With the widespread use of plastic products and the increase in waste, microplastics and nanoplastics (MNPs) have become an important issue in global environmental pollution. In recent years, an increasing number of studies have shown that MNPs may have negative impacts on human health. This review aimed to explore the association between MNPs and cardiovascular disease and provide an outlook for future research. Research has shown that there may be a link between MNPs exposure and cardiovascular disease. Laboratory studies have shown that animals exposed to MNPs often exhibit abnormalities in the cardiovascular system, such as increased blood pressure, vascular inflammation, and myocardial damage. Epidemiological surveys have also revealed that people exposed to MNPs are more likely to suffer from cardiovascular diseases, such as hypertension and myocardial infarction. Although the specific impact mechanism is not fully understood, there are several possible pathways of action, including the effects of toxic substances on MNPs and interference with the endocrine system. In summary, MNPs exposure may have a negative impact on cardiovascular health, but further research is needed to confirm its specific mechanism and extent of impact to guide relevant public health and environmental policies.
Collapse
Affiliation(s)
- Haixiang Zheng
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Gianpaolo Vidili
- Department of Medicine, Surgery, and Pharmacy, University of Sassari, Azienda Ospedaliero, Sassari, Italy
| | - Gavino Casu
- Clinical and Experimental Cardiology, Clinical and Interventional Cardiology, University of Sassari, Sassari, Italy
| | - Eliano Pio Navarese
- Clinical and Experimental Cardiology, Clinical and Interventional Cardiology, University of Sassari, Sassari, Italy
| | - Leonardo A. Sechi
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
- Complex Structure of Microbiology and Virology, AOU Sassari, Sassari, Italy
| | - Youren Chen
- Department of Cardiology, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
38
|
Mokhtari I, Moumou M, Mokhtari C, Harnafi M, Milenkovic D, Amrani S, Harnafi H. Nutritional Composition and Effect of Loquat Fruit ( Eriobotrya japonica L. var. Navela) on Lipid Metabolism and Liver Steatosis in High-Fat High-Sucrose Diet-Fed Mice. Prev Nutr Food Sci 2024; 29:256-269. [PMID: 39371510 PMCID: PMC11450286 DOI: 10.3746/pnf.2024.29.3.256] [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: 04/29/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 10/08/2024] Open
Abstract
Loquat (Eriobotrya japonica L.) is a popular fruit known for its sweet and slightly tangy flavor, which is widely consumed both fresh and in various processed forms. This study aimed to analyze the biochemical composition of loquat juice and investigate its metabolic benefits in mice fed a high-fat/high-sucrose diet (HFSD). Mice were fed either a standard diet or an HFSD and received or not the loquat juice at 4 or 8 mL/kg body weight for 8 weeks. Body weight, food efficiency ratio, plasma lipoprotein profile, plasma glucose, and lipid indices were monitored throughout the experiment. At the end of the experiment, additional assessments were performed, including lipid content measurements in liver, adipose tissue, bile, and feces; hepatic antioxidant enzyme activities (superoxide dismutase and catalase); hepatic malondialdehyde content; plasma biomarkers of liver injury; liver histology; and organ relative weight. Feeding mice with the HFSD resulted in a significant perturbation in lipid and glucose metabolism, obesity, liver steatosis, and oxidative stress-related enzymes. However, the concomitant administration of loquat juice significantly corrected this imbalance. Fresh loquat juice is low in fat and protein, moderately sugary, and energetically light; however, it is rich in minerals, vitamin C, and various phytochemicals compounds, such as phenolic acids, flavonoids, and carotenoids. The loquat juice could be considered a functional food and could be valorized through the extraction of active substances and their use as food supplements to prevent lipid metabolism disorders and the resulting health complications.
Collapse
Affiliation(s)
- Imane Mokhtari
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Oujda 60000, Morocco
| | - Mohammadine Moumou
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Oujda 60000, Morocco
| | - Chakib Mokhtari
- Laboratory of Applied Chemistry and Environment, Faculty of Sciences, Mohammed First University, Oujda 60000, Morocco
| | - Mohamed Harnafi
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Oujda 60000, Morocco
| | - Dragan Milenkovic
- Department of Nutrition, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Souliman Amrani
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Oujda 60000, Morocco
| | - Hicham Harnafi
- Laboratory of Bioresources, Biotechnologies, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Oujda 60000, Morocco
| |
Collapse
|
39
|
Aswani SS, Jayan SG, Mohan MS, Aparna NS, Boban PT, Saja K. Chrysin downregulates the expression of ADAMTS-4 in foam cells. Mol Biol Rep 2024; 51:968. [PMID: 39249599 DOI: 10.1007/s11033-024-09896-6] [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: 06/28/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Chrysin, a polyphenolic compound, possesses antioxidant and anti-inflammatory properties. In this study, we investigated the effect of chrysin on the expression of A disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS-4), a protease enzyme involved in degrading extracellular matrix associated with atherosclerosis. METHODS AND RESULTS We have studied the cell viability by MTT assay and foam cell formation by oil red O staining. The mRNA and protein expression of ADAMTS-4 was studied using quantitative polymerase chain reaction (qPCR) and Western blotting, respectively. Our study showed that chrysin significantly downregulates the expression of ADAMTS-4 in foam cells. CONCLUSION Chrysin's ability to downregulate the expression of ADAMTS-4, a protease involved in degrading the extracellular matrix, bestows upon it a new therapeutic potential for managing atherosclerosis.
Collapse
Affiliation(s)
- S S Aswani
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - Sreelekshmi G Jayan
- Department of Biotechnology, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - Mithra S Mohan
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - N S Aparna
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - P T Boban
- Department of Biochemistry, Government College Kariavattom, Thiruvananthapuram, Kerala, 695581, India
| | - K Saja
- Department of Biochemistry, University of Kerala, Kariavattom, Thiruvananthapuram, Kerala, 695581, India.
| |
Collapse
|
40
|
Peretz E, Musa S. Design, Synthesis, and Characterization of Novel Cannabidiol-Based Derivatives with Potent Antioxidant Activities. Int J Mol Sci 2024; 25:9579. [PMID: 39273525 PMCID: PMC11395037 DOI: 10.3390/ijms25179579] [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: 08/02/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
In recent years, extensive research has focused on cannabidiol (CBD), a well-studied non-psychoactive component of the plant-derived cannabinoids. CBD has shown significant therapeutic potential for treating various diseases and disorders, including antioxidants and anti-inflammatory effects. Due to the promising therapeutic effect of CBD in a wide variety of diseases, synthetic derivatization of this compound has attracted the attention of drug discovery in both industry and academia. In the current research, we focused on the derivatization of CBD by introducing Schiff base moieties, particularly (thio)-semicarbazide and aminoguanidine motifs, at the 3-position of the olivetolic ring. We have designed, synthesized, and characterized new derivatives based on CBD's framework, specifically aminoguanylhydrazone- and (thio)-semicarbazones-CBD-aldehyde compounds. Their antioxidant potential was assessed using FRAP and DPPH assays, alongside an evaluation of their effect on LDL oxidation induced by Cu2+ and AAPH. Our findings suggest that incorporating the thiosemicarbazide motif into the CBD framework produces a potent antioxidant, warranting further investigation.
Collapse
Affiliation(s)
- Eliav Peretz
- Department of Biotechnology, Tel-Hai Academic College, Kiryat Shmona 11016, Israel
- Natural Compounds and Organic Synthesis Laboratory, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
| | - Sanaa Musa
- Department of Biotechnology, Tel-Hai Academic College, Kiryat Shmona 11016, Israel
- Natural Compounds and Organic Synthesis Laboratory, Migal-Galilee Research Institute, Kiryat Shmona 11016, Israel
| |
Collapse
|
41
|
Wei S, Liu N, Fu Y, Sun M. Novel insights into modifiable risk factors for arteriovenous fistula failure and the importance of CKD lipid profile: A meta-analysis. J Vasc Access 2024; 25:1416-1431. [PMID: 36951426 DOI: 10.1177/11297298221115557] [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: 03/24/2023] Open
Abstract
BACKGROUND Arteriovenous fistula (AVF) failure can occur in patients undergoing hemodialysis (HD). In this study, we explored the correlation between hyperlipidemia and AVF failure in patients undergoing HD. Moreover, we compared the lipid profiles of patients with chronic kidney disease (CKD) with those of healthy people to provide a basis for lipid-lowering in patients undergoing HD. METHOD AND ANALYSIS We searched PubMed, Web of Science, Embase, the Cochrane library, CNKI, CBM, the China Science Periodical Database, and the China Science and Technology Journal Database. The final search was conducted on August 31, 2021, and the search period was restricted between 2000 and August 31, 2021, without publication restrictions. All studies met the inclusion criteria, and the influences of sex, age, geographical location, diagnosis method, and publication year were excluded. The data were analyzed using the random-effects model and the fixed-effects model. RESULTS Twenty-eight studies were included in the meta-analysis with 121,666 patients in the CKD group and 1714 patients in the AVF failure group. Triglyceride concentration in patients with CKD was higher than in healthy subjects (MD: -31.56, 95% CI: -41.23 to -21.90, p < 0.00001). A high total cholesterol (TC) concentration (MD: 6.97, 95% CI: 2.19-11.74, p = 0.004) and a high low-density lipoprotein cholesterol (LDL-C) concentration (MD: 23.83, 95% CI: 18.48-29.18, p < 0.00001) were associated with AVF failure. Furthermore, HDL-C was lower in the AVF failure group than in the AVF patency group (MD: -2.68, 95% CI: -4.60 to -0.76, p = 0.006). CONCLUSION Our analysis indicates that the AVF failure may be related to the increase of TC/LDL-C and the decrease of HDL-C. Although current guidelines do not consider intensive lipid-lowering therapy as necessary in patients undergoing HD, our research indicates that patients with AVF undergoing HD may need regular TC/LDL-C-lowering therapy to prevent AVF failure. However, this issue still needs well designed prospective trials.
Collapse
Affiliation(s)
- Shizhuo Wei
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Naimeng Liu
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yingli Fu
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Mindan Sun
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
42
|
Pihtili Taş N, Aydogan Baykara R, Kamanli A, Gürbüz A, Cure E, Cumhur Cüre M, Erdem M, Tasar Yildirim T. Proprotein convertase subtilisin/kexin type 9 and apelin in fibromyalgia syndrome. Arch Rheumatol 2024; 39:375-383. [PMID: 39507838 PMCID: PMC11537681 DOI: 10.46497/archrheumatol.2024.10462] [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: 09/04/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2024] Open
Abstract
OBJECTIVES This study aimed to investigate the potential roles of proprotein convertase subtilisin/ kexin type 9 (PCSK9) and apelin in the etiology of fibromyalgia syndrome (FS). PATIENTS AND METHODS The retrospective study was conducted between May 2022 and February 2023. Fifty-eight female FS patients (mean age: 45.2±9.9 years; range, 25 to 66 years) and 30 age- and body mass index-matched control subjects (mean age: 43.1±9.9 years; range, 26 to 67 years) were included in the study. Apelin and PCSK9 levels of all individuals were measured using appropriate methods. RESULTS The levels of PCSK9 (173.2±62.2 vs. 75.1±44.1, p<0.001) and apelin (354.6±195.5 vs. 229.0±83.2, p<0.001) were significantly higher in patients with FS compared to the control group. A positive correlation was found between PCSK9 and apelin levels and various measures, including the Fibromyalgia Impact Questionnaire (FIQ), Symptom Severity Scale (SSS), Pittsburgh Sleep Quality Index (PSQI), and Beck Depression Inventory (BDI). Additionally, there was a positive correlation between apelin levels and FIQ, SSS, PSQI, Beck Anxiety Inventory, and BDI scores. The optimal cutoff value for PCSK9 in predicting FS was 110.0 ng/mL, with a sensitivity of 84.5% and specificity of 83.9% (area under the curve [AUC]=0.920, 95% confidence interval [CI]: 0.852-0.987, p<0.001). For apelin, the optimal cutoff value for predicting FS was 258.8 ng/L, with a sensitivity of 63.8% and specificity of 64.5% (AUC=0.732, 95% CI: 0.623-0.840, p<0.001). CONCLUSION Our findings suggest that PCSK9 may play a role in FS etiology and potentially contribute to oxidative stress. Increased apelin levels may be a compensatory response to high oxidative stress, possibly leading to hyperalgesia. Both PCSK9 and apelin can be predictive markers for FS.
Collapse
Affiliation(s)
- Nevsun Pihtili Taş
- Department of Physical Medicine and Rehabilitation, Health Sciences University, Elazığ Fethi Sekin City Health Application and Research Center, Elazığ, Türkiye
| | - Rabia Aydogan Baykara
- Department of Physical Medicine and Rehabilitation, Malatya Turgut Özal University, Trainnig and Research Hospital, Malatya, Türkiye
| | - Ayhan Kamanli
- Department of Physical Medicine and Rehabilitation, Sakarya University Faculty of Medicine, Sakarya, Türkiye
| | - Ali Gürbüz
- Department of Physical Medicine and Rehabilitation, Elazığ Fethi Sekin City Hospital, Elazığ, Türkiye
| | - Erkan Cure
- Department of Internal Medicine, Bağcılar Medilife Hospital, İstanbul, Türkiye
| | | | - Mehmet Erdem
- Department of Biochemistry, Malatya Turgut Özal University, Malatya, Türkiye
| | | |
Collapse
|
43
|
Ugusman A, Hisam NSN, Othman NS, Anuar NNM, Hamid AA, Kumar J, Razmi MM, Aminuddin A. Pharmacological interventions for intraplaque neovascularization in atherosclerosis. Pharmacol Ther 2024; 261:108685. [PMID: 38977083 DOI: 10.1016/j.pharmthera.2024.108685] [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: 03/02/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Advanced atherosclerosis is linked to plaque instability, which can result in rupture and the onset of a heart attack. Evidence gathered from human atheroma plaques indicates that intraplaque neovascularization poses a risk to plaque stability and may lead to plaque hemorrhage. Hence, targeting the neovascularization within the atheroma plaque has the potential to mitigate the plaque's vulnerability. While neovascularization has been extensively explored in the context of cancer, research on pharmacological inhibition of this phenomenon in atherosclerosis remains limited. This systematic review aimed to comprehensively assess current and emerging pharmacological interventions for inhibiting intraplaque neovascularization in preclinical settings. Electronic databases (Web of Science, PubMed, Scopus, and Ovid) were searched from January 2013 until February 1, 2024. Preclinical studies reporting the effect of any pharmacological interventions targeting intraplaque neovascularization were included. A total of 10 articles involving in vivo animal studies were eligible for inclusion, with five of them incorporating in vitro experiments to complement their in vivo findings. The pharmacological interventions studied were axitinib, ghrelin, K5, rosuvastatin, atorvastatin, 3PO, everolimus, melatonin, Si-Miao-Yong-A, and protocatechuic aldehyde. All the interventions showed a positive impact in inhibiting intraplaque neovascularization in various atherosclerotic animal models through various signaling pathways. This review provides valuable insights into pharmacological approaches to attenuate intraplaque neovascularization that could serve as a promising therapeutic avenue to enhance plaque stability.
Collapse
Affiliation(s)
- Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia.
| | - Nur Syahidah Nor Hisam
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia; Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Nur Syakirah Othman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Nur Najmi Mohamad Anuar
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Adila A Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Maisarah Md Razmi
- Programme of Biomedical Science, Centre for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
44
|
Annink ME, Kraaijenhof JM, Stroes ESG, Kroon J. Moving from lipids to leukocytes: inflammation and immune cells in atherosclerosis. Front Cell Dev Biol 2024; 12:1446758. [PMID: 39161593 PMCID: PMC11330886 DOI: 10.3389/fcell.2024.1446758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the most important cause of morbidity and mortality worldwide. While it is traditionally attributed to lipid accumulation in the vascular endothelium, recent research has shown that plaque inflammation is an important additional driver of atherogenesis. Though clinical outcome trials utilizing anti-inflammatory agents have proven promising in terms of reducing ASCVD risk, it is imperative to identify novel actionable targets that are more specific to atherosclerosis to mitigate adverse effects associated with systemic immune suppression. To that end, this review explores the contributions of various immune cells from the innate and adaptive immune system in promoting and mitigating atherosclerosis by integrating findings from experimental studies, high-throughput multi-omics technologies, and epidemiological research.
Collapse
Affiliation(s)
- Maxim E. Annink
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jordan M. Kraaijenhof
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Erik S. G. Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, Netherlands
| |
Collapse
|
45
|
Stringham NT, Green M, Roche W, Prado-Cabrero A, Mulcahy R, Nolan J. Lutein, zeaxanthin, and meso-zeaxanthin supplementation attenuates inflammatory cytokines and markers of oxidative cardiovascular processes in humans. Nutr Metab Cardiovasc Dis 2024; 34:1976-1983. [PMID: 38890092 DOI: 10.1016/j.numecd.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIMS Systemic inflammation and oxidation are primary contributors to the development of atherosclerosis. Oxidation of low-density lipoprotein (LDL) particles within the vascular endothelium has been hypothesized to be an initial step in the formation of atherosclerotic plaques, with inflammatory cytokines serving as the signaling mechanism for concomitant macrophage activation. Supplementation with the antioxidative macular xanthophylls (lutein [L], zeaxanthin [Z], and meso-zeaxanthin [MZ]) has been shown to aid in the reduction of inflammatory physiologic responses; therefore, we hypothesized that in our study population, supplementation with these xanthophylls would facilitate a systemic reduction in markers of inflammation and cardiovascular lipid oxidation. METHODS AND RESULTS In this double-blind placebo-controlled supplementation study, participants were randomly allocated to receive the active intervention containing L (10 mg) + MZ (10 mg) + Z (2 mg) or placebo (containing sunflower oil). Serum concentrations of carotenoids (assessed by HPLC), inflammatory cytokines (IL-6, IL-1β, TNF-α) and oxidized LDL (OxLDL; by solid-phase sandwich ELISA) were measured at baseline and at 6-months. Results showed that over the supplementation period, compared to placebo, the active group demonstrated statistically significant increases in serum concentrations of L, Z, & MZ (p < 0.05), reductions in inflammatory cytokines IL-1β (p < 0.001) and TNF-α (p = 0.003), as well as a corresponding reduction in serum OxLDL (p = 0.009). CONCLUSIONS Our data show that L, Z, & MZ supplementation results in decreased serum IL-1β, TNF-α, and OxLDL. This suggests that these carotenoids are acting systemically to attenuate oxidative lipid products and inflammation, thus reducing their contribution to atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Nicole T Stringham
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland; Northern Arizona University, Flagstaff, AZ, USA.
| | - Marina Green
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Warren Roche
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Alfonso Prado-Cabrero
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - Riona Mulcahy
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| | - John Nolan
- Nutrition Research Centre Ireland (NRCI), Southeast Technical University, Waterford, Ireland
| |
Collapse
|
46
|
Kozina N, Jukić I, Mihaljević Z, Matić A, Dobrivojević Radmilović M, Barić A, Drenjančević I. The Effect of High-Salt Diet on Oxidative Stress Production and Vascular Function in Tff3-/-/C57BL/6N Knockout and Wild Type (C57BL/6N) Mice. J Vasc Res 2024; 61:214-224. [PMID: 39074455 DOI: 10.1159/000539614] [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: 09/05/2023] [Accepted: 05/25/2024] [Indexed: 07/31/2024] Open
Abstract
INTRODUCTION It is well documented that high-salt (HS) diet increases systemic and vascular oxidative stress in various animal models and in humans, leading to impairment of vascular reactivity. The present study examined the interaction of genotype and HS diet intake and the potential effects of oxidative stress - antioxidative system balance on the flow-induced dilation (FID) in pressurized carotid arteries of normotensive Tff3-/-/C57BL/6N knockout mice and their wild-type (WT) controls. METHODS Male, ten-week-old transgenic Tff3-/-/C57BL/6N (Tff3-/-) knockout mice and WT/C57BL/6N (WT) (parental strain) healthy mice were divided in LS (0.4% NaCl in rodent chow) and HS (4% NaCl in rodent chow fed for 1 week) groups. Additionally, LS and HS groups were treated with 1 mmol/L 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (TEMPOL) dissolved in the drinking water. After anesthesia with ketamine chloride (100 mg/kg) and midazolam (5 mg/kg), blood pressure was measured, carotid arteries and aortas were isolated, and blood samples were collected. RESULTS FID was decreased in WT_HS mice and restored by superoxide scavenger TEMPOL in vivo. On the other hand, attenuated FID of Tff3-/- mice was not further affected by HS diet or TEMPOL in vivo treatment. Vascular superoxide/reactive oxygen species levels were increased with HS diet in both strains and restored by TEMPOL. HS upregulated glutathione peroxidase 1 (GPx1) gene expression in WT_HS and Tff3-/-_HS mice, while GPx activity was significantly decreased only in WT_HS group. Systemic (serum) markers of oxidative stress (oxLDL and AOPP) and arterial blood pressure were similar among groups. CONCLUSION HS diet increases vascular oxidative stress and impairs vasodilation in WT mice. Tff3 gene deficiency attenuates vasodilation per se, without further effects of HS intake. This can be attributed to vascular upregulation of antioxidative enzyme GPx1 in Tff3-/-/C57BL/6N mice conferring protection from oxidative stress.
Collapse
Affiliation(s)
- Nataša Kozina
- Institute and Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia,
- Scientific Centre of Excellence for Personalized Health Care University of Osijek, Osijek, Croatia,
| | - Ivana Jukić
- Institute and Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Centre of Excellence for Personalized Health Care University of Osijek, Osijek, Croatia
| | - Zrinka Mihaljević
- Institute and Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Centre of Excellence for Personalized Health Care University of Osijek, Osijek, Croatia
| | - Anita Matić
- Institute and Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Institute for Integrative Medicine, Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | | | - Anja Barić
- Croatian Institute for Brain Research, University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Ines Drenjančević
- Institute and Department of Physiology and Immunology, Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
- Scientific Centre of Excellence for Personalized Health Care University of Osijek, Osijek, Croatia
| |
Collapse
|
47
|
Sarkar S, Brown TT. CROI 2024: Metabolic and Other Complications of HIV Infection. TOPICS IN ANTIVIRAL MEDICINE 2024; 32:504-510. [PMID: 39142292 PMCID: PMC11293603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Comorbid conditions have a major impact on the health, quality of life, and survival of people with HIV (PWH), particularly as they age. The 2024 Conference on Retroviruses and Opportunistic Infections (CROI) featured many excellent reports related to specific comorbidities, most notably cardiovascular disease, cancer, fatty liver disease, and hypertension. Major themes included hypertension management strategies used in low- and middle-income countries, important insights from the REPRIEVE (Randomized Trial to Prevent Vascular Events in HIV) study that focused on cardiometabolic outcomes, studies investigating metabolic-associated fatty liver disease, and the use of glucagon-like peptide-1 receptor agonists in PWH. This review focuses on the abstracts presented at CROI 2024 that discussed these areas, highlighting those with the most clinical impact.
Collapse
|
48
|
Piergentili R, Sechi S. Non-Coding RNAs of Mitochondrial Origin: Roles in Cell Division and Implications in Cancer. Int J Mol Sci 2024; 25:7498. [PMID: 39000605 PMCID: PMC11242419 DOI: 10.3390/ijms25137498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024] Open
Abstract
Non-coding RNAs (ncRNAs) are a heterogeneous group, in terms of structure and sequence length, consisting of RNA molecules that do not code for proteins. These ncRNAs have a central role in the regulation of gene expression and are virtually involved in every process analyzed, ensuring cellular homeostasis. Although, over the years, much research has focused on the characterization of non-coding transcripts of nuclear origin, improved bioinformatic tools and next-generation sequencing (NGS) platforms have allowed the identification of hundreds of ncRNAs transcribed from the mitochondrial genome (mt-ncRNA), including long non-coding RNA (lncRNA), circular RNA (circRNA), and microRNA (miR). Mt-ncRNAs have been described in diverse cellular processes such as mitochondrial proteome homeostasis and retrograde signaling; however, the function of the majority of mt-ncRNAs remains unknown. This review focuses on a subgroup of human mt-ncRNAs whose dysfunction is associated with both failures in cell cycle regulation, leading to defects in cell growth, cell proliferation, and apoptosis, and the development of tumor hallmarks, such as cell migration and metastasis formation, thus contributing to carcinogenesis and tumor development. Here we provide an overview of the mt-ncRNAs/cancer relationship that could help the future development of new biomedical applications in the field of oncology.
Collapse
Affiliation(s)
| | - Stefano Sechi
- Istituto di Biologia e Patologia Molecolari del Consiglio Nazionale delle Ricerche, Dipartimento di Biologia e Biotecnologie, Università Sapienza di Roma, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| |
Collapse
|
49
|
Sabayan B, Akhavan Sigari A, Modir R, Meyer BC, Hemmen T, Meyer D, Bavarsad Shahripour R. Statin treatment intensity and cerebral vasomotor reactivity response in patients with ischemic stroke. J Neuroimaging 2024; 34:445-450. [PMID: 38590021 DOI: 10.1111/jon.13200] [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: 02/13/2024] [Revised: 03/08/2024] [Accepted: 03/25/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Cerebral vasomotor reactivity (VMR) is vital for regulating brain blood flow and maintaining neurological function. Impaired cerebral VMR is linked to a higher risk of stroke and poor post-stroke outcomes. This study explores the relationship between statin treatment intensity and VMR in patients with ischemic stroke. METHODS Seventy-four consecutive patients (mean age 69.3 years, 59.4% male) with recent ischemic stroke were included. VMR levels were assessed 4 weeks after the index stroke using transcranial Doppler, measuring the breath-holding index (BHI) as an indicator of the percentage increase in middle cerebral artery blood flow (higher BHI signifies higher VMR). Multistep multivariable regression models, adjusted for demographic and cerebrovascular risk factors, were employed to examine the association between statin intensity treatment and BHI levels. RESULTS Forty-one patients (55%) received high-intensity statins. Patients receiving high-intensity statins exhibited a mean BHI of 0.85, whereas those on low-intensity statins had a mean BHI of 0.67 (mean difference 0.18, 95% confidence interval: 0.13-0.22, p-value<.001). This significant difference persisted in the fully adjusted model (adjusted mean values: 0.84 vs. 0.68, p-value: .008). No significant differences were observed in BHI values within patient groups on high-intensity or low-intensity statin therapy (all p-values>.05). Furthermore, no significant association was found between baseline low-density lipoprotein (LDL) levels and BHI. CONCLUSIONS High-intensity statin treatment post-ischemic stroke is linked to elevated VMR independent of demographic and clinical characteristics, including baseline LDL level. Further research is needed to explore statin therapy's impact on preserving brain vascular function beyond lipid-lowering effects.
Collapse
Affiliation(s)
- Behnam Sabayan
- Department of Neurology, Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Royya Modir
- UCSD Comprehensive Stroke Center, Department of Neurosciences, University of California, San Diego, California, USA
| | - Brett C Meyer
- UCSD Comprehensive Stroke Center, Department of Neurosciences, University of California, San Diego, California, USA
| | - Thomas Hemmen
- UCSD Comprehensive Stroke Center, Department of Neurosciences, University of California, San Diego, California, USA
| | - Dawn Meyer
- UCSD Comprehensive Stroke Center, Department of Neurosciences, University of California, San Diego, California, USA
| | - Reza Bavarsad Shahripour
- UCSD Comprehensive Stroke Center, Department of Neurosciences, University of California, San Diego, California, USA
| |
Collapse
|
50
|
Tan C, Chen J, Tu T, Chen L, Zou J. Lycopene inhibits pyroptosis of endothelial progenitor cells induced by ox-LDL through the AMPK/mTOR/NLRP3 pathway. Open Med (Wars) 2024; 19:20240973. [PMID: 38919547 PMCID: PMC11197008 DOI: 10.1515/med-2024-0973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/16/2024] [Accepted: 05/03/2024] [Indexed: 06/27/2024] Open
Abstract
The malfunction of endothelial progenitor cells (EPCs) due to ox-LDL is a risk contributor for arteriosclerotic disease. Meanwhile, lycopene possesses anti-inflammatory and antioxidative qualities. This investigation aimed to determine if lycopene can protect EPCs from ox-LDL-induced damage and to elucidate the underlying mechanism. The effects of lycopene on the survival, migration, and tube-forming capacity of EPCs were determined via in vitro assays. Expression of proteins related to pyroptosis and cellular proteins related to AMPK/mTOR/NLRP3 signaling was determined by western blot/flow cytometry. Our results demonstrated that lycopene treatment significantly enhanced proliferation, tube formation, and migration of EPCs stimulated by ox-LDL. Additionally, lycopene was found to suppress pyroptosis in ox-LDL-induced EPCs through the activation of AMPK, which led to the inhibition of mTOR phosphorylation and subsequent downregulation of the downstream NLRP3 inflammasome. In summary, our study suggests that lycopene mitigates ox-LDL-induced dysfunction in EPCs and inhibits pyroptosis via AMPK/mTOR/NLRP3 signaling. Our study suggests that lycopene may act as promising therapies for preventing atherosclerosis.
Collapse
Affiliation(s)
- Chujun Tan
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Junqiu Chen
- Department of Cardiology, Fuwai Hospital, Chinese Academy of Medical Sciences, (Shenzhen Sun Yat-sen Cardiovascular Hospital), Shenzhen, 518057, China
| | - Tengcan Tu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Lifang Chen
- Department of Cardiology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 528200, China
| | - Jun Zou
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528200, China
| |
Collapse
|