1
|
Targas ABA, Victoriano PHM, Garcia MBB, Alexandre-Silva V, Cominetti MR. Exploring the connection between dementia and cardiovascular risk with a focus on ADAM10. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167825. [PMID: 40174790 DOI: 10.1016/j.bbadis.2025.167825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Alzheimer's disease (AD) represents a leading cause of dementia, characterized by progressive cognitive and functional decline. Although extensive research has unraveled critical aspects of AD pathology, its etiology remains incompletely understood, urging further exploration into potential risk factors. Growing evidence underscores a significant link between cardiovascular disease (CVD) risk factors and AD, with modifiable lifestyle elements - such as physical inactivity, high low-density lipoprotein (LDL) levels, obesity, hypertension, atherosclerosis, and diabetes - emerging as contributors to cerebrovascular damage and neurodegeneration. ADAM10, a disintegrin and metalloproteinase involved in the non-amyloidogenic processing of amyloid precursor protein (APP), has garnered interest for its dual role in cardiovascular and neurodegenerative processes. ADAM10's regulation of neuroinflammation, endothelial function, and proteolytic cleavage of APP potentially moderates amyloid-β (Aβ) peptide formation, thus influencing both cardiovascular and brain health. Given these interconnected roles, this narrative review investigates whether ADAM10-driven vascular dysfunction accelerates neurodegeneration, how lipid metabolism influences ADAM10 activity in CVD and AD, and whether targeting ADAM10 could offer a dual-benefit therapeutic strategy to mitigate disease burden. By exploring epidemiological data, clinical studies, and molecular pathways, we aim to clarify ADAM10's bridging function between AD and cardiovascular risk, offering a new perspective into therapeutic opportunities to alleviate the dual burden of these interrelated conditions.
Collapse
Affiliation(s)
| | | | | | | | - Marcia Regina Cominetti
- Department of Gerontology, Federal University of São Carlos, São Carlos, SP, Brazil; Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
2
|
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 PMCID: PMC12016489 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
|
3
|
Khowdiary MM, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Elhenawy AA, Rashwan EK, Alexiou A, Papadakis M, Fetoh MEAE, Batiha GES. The Peripheral Amyloid-β Nexus: Connecting Alzheimer's Disease with Atherosclerosis through Shared Pathophysiological Mechanisms. Neuromolecular Med 2025; 27:20. [PMID: 40032716 DOI: 10.1007/s12017-025-08836-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025]
Abstract
Alzheimer's disease (AD) and atherosclerosis (AS) are two chronic diseases with seemingly distinct pathologies. However, emerging research points to a bidirectional relationship driven by common mechanisms, such as inflammation, oxidative stress, and dysregulation of Amyloid-Beta (Aβ). This review focuses on the role of Aβ as a critical molecular link between AD and AS, emphasizing its contribution to neuronal impairment and vascular damage. Specifically, peripheral Aβ produced in the pancreas and skeletal muscle tissues exacerbates AS by promoting endothelial dysfunction and insulin resistance (IR). Furthermore, AS accelerates AD progression by impairing cerebral blood flow and inducing chronic hypoxia, causing Aβ accumulation. This review critically evaluates recent findings, highlighting inconsistencies in clinical studies and suggesting future research directions. Understanding the bidirectional influence of AD and AS could pave the way for novel therapeutic approaches targeting shared molecular pathways, particularly emphasizing Aβ clearance and inflammation.
Collapse
Affiliation(s)
- Manal M Khowdiary
- Department of Chemistry, Faculty of Applied Science, Lieth Collage, Umm Al-Qura University, 24382, Makkah, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu./Najaf-Iraq, PO. Box13, Kufa, Iraq
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, 11884, Egypt
- Chemistry Department, Faculty of Science, AlBaha University, 65731, Al Bahah, Saudi Arabia
| | - Eman K Rashwan
- Department of Physiology, College of Medicine, Jouf University, Akaka, Saudi Arabia
| | - Athanasios Alexiou
- Department of Research & Development, Funogen, 11741, Athens, Attiki, Greece
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
| | - Marios Papadakis
- University Hospital, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Mohammed E Abo-El Fetoh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Badr City, 11829, Cairo, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
4
|
Kosinski J, Sechi A, Hain J, Villwock S, Ha SA, Hauschulz M, Rose M, Steib F, Ortiz‐Brüchle N, Heij L, Maas SL, van der Vorst EPC, Knoesel T, Altendorf‐Hofmann A, Simon R, Sauter G, Bednarsch J, Jonigk D, Dahl E. ITIH5 as a multifaceted player in pancreatic cancer suppression, impairing tyrosine kinase signaling, cell adhesion and migration. Mol Oncol 2024; 18:1486-1509. [PMID: 38375974 PMCID: PMC11161730 DOI: 10.1002/1878-0261.13609] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 02/21/2024] Open
Abstract
Inter-alpha-trypsin inhibitor heavy chain 5 (ITIH5) has been identified as a metastasis suppressor gene in pancreatic cancer. Here, we analyzed ITIH5 promoter methylation and protein expression in The Cancer Genome Atlas (TCGA) dataset and three tissue microarray cohorts (n = 618), respectively. Cellular effects, including cell migration, focal adhesion formation and protein tyrosine kinase activity, induced by forced ITIH5 expression in pancreatic cancer cell lines were studied in stable transfectants. ITIH5 promoter hypermethylation was associated with unfavorable prognosis, while immunohistochemistry demonstrated loss of ITIH5 in the metastatic setting and worsened overall survival. Gain-of-function models showed a significant reduction in migration capacity, but no alteration in proliferation. Focal adhesions in cells re-expressing ITIH5 exhibited a smaller and more rounded phenotype, typical for slow-moving cells. An impressive increase of acetylated alpha-tubulin was observed in ITIH5-positive cells, indicating more stable microtubules. In addition, we found significantly decreased activities of kinases related to focal adhesion. Our results indicate that loss of ITIH5 in pancreatic cancer profoundly affects its molecular profile: ITIH5 potentially interferes with a variety of oncogenic signaling pathways, including the PI3K/AKT pathway. This may lead to altered cell migration and focal adhesion formation. These cellular alterations may contribute to the metastasis-inhibiting properties of ITIH5 in pancreatic cancer.
Collapse
Affiliation(s)
- Jennifer Kosinski
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Antonio Sechi
- Department of Cell and Tumor BiologyRWTH Aachen UniversityGermany
| | - Johanna Hain
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Sophia Villwock
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Stefanie Anh Ha
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Maximilian Hauschulz
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Michael Rose
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Florian Steib
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Nadina Ortiz‐Brüchle
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Lara Heij
- Institute of PathologyUniversity Hospital EssenGermany
- Department of Surgery and Transplantation, Medical FacultyRWTH Aachen UniversityGermany
- Department of PathologyErasmus Medical Center RotterdamThe Netherlands
- NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityThe Netherlands
| | - Sanne L. Maas
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR)Medical Faculty of RWTH Aachen UniversityGermany
| | - Emiel P. C. van der Vorst
- Interdisciplinary Center for Clinical Research (IZKF), Institute for Molecular Cardiovascular Research (IMCAR)Medical Faculty of RWTH Aachen UniversityGermany
- Institute for Cardiovascular Prevention (IPEK)Ludwig‐Maximilians‐University MunichGermany
| | - Thomas Knoesel
- Institute of PathologyLudwig‐Maximilians‐University MunichGermany
| | | | - Ronald Simon
- Institute of PathologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Guido Sauter
- Institute of PathologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Jan Bednarsch
- Department of Surgery and Transplantation, Medical FacultyRWTH Aachen UniversityGermany
| | - Danny Jonigk
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- RWTH centralized Biomaterial Bank (RWTH cBMB)Medical Faculty of the RWTH Aachen UniversityGermany
- German Center for Lung Research (DZL), BREATHHanoverGermany
| | - Edgar Dahl
- Institute of PathologyMedical Faculty of RWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- RWTH centralized Biomaterial Bank (RWTH cBMB)Medical Faculty of the RWTH Aachen UniversityGermany
| |
Collapse
|
5
|
Hong JG, Zheng HL, Wang P, Huang P, Gong DP, Zeng ZY. Hsa_ circ_0006867 regulates ox-LDL-induced endothelial injury via the miR-499a-3p/ADAM10 axis. Clin Hemorheol Microcirc 2024; 88:115-127. [PMID: 37694359 PMCID: PMC11491994 DOI: 10.3233/ch-231895] [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: 09/12/2023]
Abstract
Circular RNAs (circRNAs) have been reported to participate in the development of various diseases. In this study, we investigated the potential mechanism underlying the role of circRNAs in atherosclerosis. Human umbilical vein endothelial cells (HUVECs) were treated with 100 μg/mL oxidized low-density lipoprotein (ox-LDL) to simulate atherosclerosis. We observed that hsa_circ_0006867 (circ_0006867), a circRNA markedly increased in ox-LDL-treated endothelial cells, acted as a molecular sponge of miR-499a-3p and regulated its expression. This interaction led to changes in the downstream target gene ADAM10, thus affecting cell apoptosis and migration. Thus, our study suggests that circ_0006867 regulates ox-LDL-induced endothelial injury via the circ_0006867/miR-499a-3p/ADAM10 axis, indicating its potential as an exploitable therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Ji-Ge Hong
- Department of Geriatric Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, Guangxi, China
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| | - Hui-Lei Zheng
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, Guangxi, China
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
- Department of Health Management, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Peng Wang
- Department of Health Management, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ping Huang
- Department of Health Management, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Dan-Ping Gong
- Department of Geriatric Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhi-Yu Zeng
- Department of Geriatric Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, Guangxi, China
- Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, Guangxi, China
| |
Collapse
|
6
|
Khan MA, Mohammad I, Banerjee S, Tomar A, Varughese KI, Mehta JL, Chandele A, Arockiasamy A. Oxidized LDL receptors: a recent update. Curr Opin Lipidol 2023:00041433-990000000-00037. [PMID: 37171285 DOI: 10.1097/mol.0000000000000884] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
PURPOSE OF REVIEW LDL in its oxidized form, or 'oxLDL', is now generally acknowledged to be highly proatherogenic and to play a significant role in atherosclerotic plaque formation. Therefore, there has been increasing interest in understanding the significance of oxLDL and its receptors in different phases of atherosclerosis, leading to the accumulation of additional data at the cellular, structural, and physiological levels. This review focuses on the most recent discoveries about these receptors and how they influence lipid absorption, metabolism, and inflammation in various cell types. RECENT FINDINGS Two crystal structures of lectin-like oxLDL receptor-1 (LOX-1), one with a small molecule inhibitor and the other with a monoclonal antibody have been published. We recently demonstrated that the 'surface site' of LOX1, adjacent to the positively charged 'basic spine region' that facilitates oxLDL binding, is a targetable site for drug development. Further, recent human studies showed that soluble LOX-1 holds potential as a biomarker for cardiovascular disease diagnosis, prognosis, and assessing the efficacy of therapy. SUMMARY Receptor-mediated oxLDL uptake results in cellular dysfunction of various cell types involved in atherogenesis and plaque development. The current advancements clearly demonstrate that targeting oxLDL-LOX-1 axis may lead to development of future therapeutics for the treatment of atherosclerotic cardiovascular and cerebrovascular diseases.
Collapse
Affiliation(s)
- Mohd Azeem Khan
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Irshad Mohammad
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Sohom Banerjee
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Akanksha Tomar
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Kottayil I Varughese
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences
| | - Jawahar L Mehta
- Division of Cardiology, University of Arkansas for Medical Sciences and the VA Medical Center, Little Rock, Arkansas, USA
| | - Anmol Chandele
- ICGEB-Emory Vaccine Centre, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| | - Arulandu Arockiasamy
- Membrane Protein Biology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi, India
| |
Collapse
|
7
|
Maas SL, Donners MMPC, van der Vorst EPC. ADAM10 and ADAM17, Major Regulators of Chronic Kidney Disease Induced Atherosclerosis? Int J Mol Sci 2023; 24:ijms24087309. [PMID: 37108478 PMCID: PMC10139114 DOI: 10.3390/ijms24087309] [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: 03/15/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Chronic kidney disease (CKD) is a major health problem, affecting millions of people worldwide, in particular hypertensive and diabetic patients. CKD patients suffer from significantly increased cardiovascular disease (CVD) morbidity and mortality, mainly due to accelerated atherosclerosis development. Indeed, CKD not only affects the kidneys, in which injury and maladaptive repair processes lead to local inflammation and fibrosis, but also causes systemic inflammation and altered mineral bone metabolism leading to vascular dysfunction, calcification, and thus, accelerated atherosclerosis. Although CKD and CVD individually have been extensively studied, relatively little research has studied the link between both diseases. This narrative review focuses on the role of a disintegrin and metalloproteases (ADAM) 10 and ADAM17 in CKD and CVD and will for the first time shed light on their role in CKD-induced CVD. By cleaving cell surface molecules, these enzymes regulate not only cellular sensitivity to their micro-environment (in case of receptor cleavage), but also release soluble ectodomains that can exert agonistic or antagonistic functions, both locally and systemically. Although the cell-specific roles of ADAM10 and ADAM17 in CVD, and to a lesser extent in CKD, have been explored, their impact on CKD-induced CVD is likely, yet remains to be elucidated.
Collapse
Affiliation(s)
- Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Marjo M P C Donners
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), 80336 Munich, Germany
| |
Collapse
|