1
|
Shi S, Zheng Z, Chen W, Song Y, Dou K. Low density lipoprotein cholesterol but not statins is the direct cause of intracerebral hemorrhage. Eur J Pharmacol 2025; 998:177443. [PMID: 40023359 DOI: 10.1016/j.ejphar.2025.177443] [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: 11/12/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE To investigate the direct and indirect relationships between statin use, low-density lipoprotein cholesterol (LDL-C) levels, and intracerebral hemorrhage (ICH), providing new insights into this complex scientific question. METHODS In this cohort study, UK Biobank data from 2006 to 2010 were used to construct Structural Equation Models of statin use, LDL-C, and ICH, including 414,253 participants with LDL-C data. Published Genome-Wide Association Studies data were used for drug-target Mendelian Randomization analysis. RESULTS The study included 414,253 participants, comprising 225,454 women (54.4%) with a mean age of 56.07 (8.11) years. During a median follow-up of 14.01 years, 2973 patients experienced ICH. Structural Equation Modelling showed the indirect effect (path a∗b) of statin on ICH was 0.003 (P < 0.001), the direct effect (path c') was -0.001 (P = 0.568), the total effect (path c) was 0.002 (P = 0.391), and the mediation proportion of LDL-C (a∗b/c) was 150.0%. Mendelian Randomization showed a negative association between LDL-C levels and ICH (β: -0.663, SE: 0.229, P = 0.004), with no causal relationship between statin use and ICH (β: -1.454, SE: 3.133, P = 0.643). Drug-targeted Mendelian Randomization revealed LDL-C levels, predicted by variants in or near HMGCR, PCSK9, CETP, ABCG8/5, and LAP, were negatively associated with ICH risk. CONCLUSIONS This study confirmed that statins increase the risk of ICH primarily through their LDL-C-lowering effects, rather than the direct effects of the statins themselves. LDL-C is negatively associated with ICH, an association not confined to the effects of the HMGCR loci. This advance provides evidence for the controversy between statin use, LDL-C levels, and ICH risk.
Collapse
Affiliation(s)
- Shanshan Shi
- Cardiometabolic Medicine Center, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Zhihao Zheng
- Cardiometabolic Medicine Center, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Weihua Chen
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100053, China
| | - Yanjun Song
- Cardiometabolic Medicine Center, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Kefei Dou
- Cardiometabolic Medicine Center, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Cardiovascular Disease, 167, Beilishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
2
|
Derobertmasure A, Toh LS, Wotring VE, Williams PM, Morbidelli L, Stingl JC, Vinken M, Ramadan R, Chhun S, Boutouyrie P. Pharmacological countermeasures for long-duration space missions: addressing cardiovascular challenges and advancing space-adapted healthcare. Eur J Pharm Sci 2025; 209:107063. [PMID: 40064402 DOI: 10.1016/j.ejps.2025.107063] [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: 11/29/2024] [Revised: 02/10/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Future long-duration crewed space missions beyond Low Earth Orbit (LEO) will bring new healthcare challenges for astronauts for which pharmacological countermeasures (pharmacological countermeasures) are crucial. This paper highlights current pharmacological countermeasures challenges described in the ESA SciSpacE Roadmap, with a focus on the cardiovascular system as a model to demonstrate the potential implication of the challenges and recommendations. New pharmacological approaches and procedures need to be adapted to spaceflight (spaceflight) conditions, including ethical and reglementary considerations. Potential strategies include combining pharmacological biomarkers such as pharmacogenomics with therapeutic drug monitoring, advancing microsampling techniques, and implementing a pharmacovigilance system to gain deep insights into pharmacokinetics/pharmacodynamics (PK/PD) spaceflight alteration on drug exposure. Emerging therapeutic approaches (such as long-term regimens) or manufacturing drugs in the space environment, can address specific issues related to drug storage and stability. The integration of biobanks and innovative technologies like organoids and organ-on-a-chip, artificial intelligence (AI), including machine learning will further enhance PK modelling leading to personalized treatments. These innovative pharmaceutical tools will also enable reciprocal game-changing healthcare developments to be made on Earth as well as in space and are essential to ensure space explorers receive safe effective pharmaceutical care.
Collapse
Affiliation(s)
- Audrey Derobertmasure
- Faculty of Medicine, Paris Cité University, INSERM PARCC, Service de Pharmacologie Clinique, Hôpital Européen Georges Pompidou Hospital (AP-HP), Paris, France
| | - Li Shean Toh
- School of Pharmacy, Faculty of Science, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Virginia E Wotring
- International Space University, 1 rue Jean-Dominique Cassini, Parc d'Innovation, 6700 Illkirch-Graffenstaden, France
| | - Philip M Williams
- School of Pharmacy, Faculty of Science, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Julia C Stingl
- Institute of Clinical Pharmacology, University Hospital of RWTH Aachen, Wendlingweg 2, 52064, Aachen, Germany
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Raghda Ramadan
- Interdisciplinary Biosciences Group, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Stephanie Chhun
- Faculty of Medicine, Paris Cité University, Paris, France; Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253; AP-HP, Laboratory of Immunology, Necker-Enfants Malades Hospital, Paris, France
| | - Pierre Boutouyrie
- Faculty of Medicine, Paris Cité University, INSERM PARCC, Service de Pharmacologie Clinique, Hôpital Européen Georges Pompidou Hospital (AP-HP), Paris, France.
| |
Collapse
|
3
|
Mthembu SXH, Mazibuko-Mbeje SE, Silvestri S, Orlando P, Nkambule BB, Muller CJF, Tiano L, Dludla PV. Prolonged exposure to simvastatin affects coenzyme Q 9/10 status leading to impaired mitochondrial respiratory capacity and reduced viability of cultured cardiac cells. Toxicol In Vitro 2025; 106:106052. [PMID: 40089196 DOI: 10.1016/j.tiv.2025.106052] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
This study investigates the effects of prolonged simvastatin exposure on coenzyme Q9/10 (CoQ9/10) levels, an essential component of antioxidant defense, in cultured cardiac cells. Statins, commonly used to manage dyslipidemia and reduce cardiovascular risk, may impair mitochondrial function, but their impact on CoQ10 depletion and oxidative stress is not well understood. We examined the influence of simvastatin on mitochondrial oxidative capacity, reactive oxygen species (ROS) production, and CoQ9/10 status at concentrations of 0.3, 0.6, 1.25, 2.5, 5, 10, and 20 μM, over durations of 24, 48, and 72 h. Using an in vitro model of cultured H9c2 cardiomyoblasts, our results showed that short-term exposure (24 h) at lower concentrations (<5 μM) enhanced cytosolic and mitochondrial ROS levels without affecting mitochondrial function or CoQ9/10 status. However, prolonged exposure to higher concentrations (≥10 μM for >48 h) resulted in impaired mitochondrial oxidative capacity, indicated by increased proton leak and elevated ROS levels, which were followed by significantly reduced cell viability. These findings suggest that prolonged, high-dose simvastatin exposure may disrupt the oxidative balance of CoQ9/10, leading to myocardial injury. This research addresses a gap in understanding the long-term effects of statins on mitochondrial health and underscores the need for further studies to optimize statin therapy and minimize adverse effects on myocardial function.
Collapse
Affiliation(s)
- Sinenhlanhla X H Mthembu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry, Mafikeng Campus, Northwest University, Mmabatho 2735, South Africa.
| | | | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Centre for Cardiometabolic Research Africa (CARMA), Division of Medical Physiology, Stellenbosch University, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Phiwayinkosi V Dludla
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| |
Collapse
|
4
|
Magurova M, Bacova M, Papcunova S, Kiss Bimbova K, Kuruc T, Kisucka A, Ihnatova L, Kucharova K, Lukacova N, Galik J. Exploring synergistic effects: Atorvastatin and electrical stimulation in spinal cord injury therapy. IBRO Neurosci Rep 2025; 18:389-399. [PMID: 40124115 PMCID: PMC11927724 DOI: 10.1016/j.ibneur.2025.02.012] [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: 10/31/2024] [Accepted: 02/23/2025] [Indexed: 03/25/2025] Open
Abstract
Spinal cord trauma represents a significant clinical challenge, and improving patient outcomes is a main priority for many scientific teams globally. Despite advances in the understanding its pathogenesis, the overall mechanisms occurring in the spinal cord after traumatic injury remain unclear. This study explores the possible synergistic effects of a regenerative therapy that combines electrical stimulation with the anti-inflammatory drug Atorvastatin (ATR) after spinal cord injury (SCI). SCI was induced at the T9 segment under isoflurane anesthesia and applying a compression force of 40 g for 15 minutes. An oscillating field stimulator (OFS) was implanted subcutaneously, delivering a weak electric current (50 µA) that changed polarity every 15 minutes for six weeks to promote axonal growth at the injury site. Female Wistar albino rats were divided into four groups: SCI with non-functional stimulator (SCI + nOFS), SCI with functional stimulator (SCI+OFS), and two groups that received ATR together with stimulator for 7 days after injury (SCI+OFS+ATR, SCI+nOFS+ATR). Behavioral tests (hot-plate test and BBB scale) showed improvement in sensory and motor performance in animals treated with the combination therapy. The protein levels of astrocytes (GFAP), neurofilaments (NF-L), newly sprouting axons (GAP-43), and oligodendrocytes (PLP -1, CNPase) were analysed by Western blot. The results showed increased neurofilaments, newly sprouting axons and oligodendrocytes in groups receiving both individual and combination therapies, with a decrease in their concentrations in the following order: SCI+OFS+ATR, SCI+nOFS+ATR, SCI+OFS, SCI+nOFS. In addition, astrocyte protein levels were lower in the SCI+OFS+ATR group compared with others. Histological analysis showed a significant reduction in white and gray matter after SCI, but less white and gray matter volume loss was found in the groups receiving therapies (SCI+OFS+ATR, SCI+nOFS+ATR, SCI+OFS). These results suggest that the combination of Atorvastatin with OFS stimulation promotes neural recovery after SCI, highlighting the potential of combination therapies in enhancing regenerative outcomes.
Collapse
Affiliation(s)
- Martina Magurova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Maria Bacova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Stefania Papcunova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Katarina Kiss Bimbova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Tomas Kuruc
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Alexandra Kisucka
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Lenka Ihnatova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Karolina Kucharova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Nadezda Lukacova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Jan Galik
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| |
Collapse
|
5
|
Li X, Liu Y, Song T, Wu X, Xing Y. Statins and the risk of dementia: a meta-analysis. Acta Neurol Belg 2025; 125:699-705. [PMID: 40199829 DOI: 10.1007/s13760-025-02758-3] [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: 10/17/2023] [Accepted: 02/28/2025] [Indexed: 04/10/2025]
Abstract
Clarify the relationship between the use of statins and the risk of dementia, and further guide the clinical application of statins. This paper uses computer search Chinese and foreign language databases Pubmed, Medline, Cochrane, Embase, Web of Science, Weipu, CBM, Wanfang, CNKI, etc., to include the literature that meets the standards from the establishment of the library to December 2020, and uses Revman 5.1 software and Stata15.0 software for meta-analysis. Of the 34 included observational studies, stratified subgroup analyses were performed based on the type of clinical trial, sex of participants, presence or absence of apolipoprotein E-4 (ApoEε4) allele, and duration of use. The conclusions showed that (1) the use of statins can significantly reduce the risk of dementia; (2) There is no significant sex difference in reducing the risk of dementia; (3) Continuous use of statins for ≥ 1 year can significantly reduce the risk of dementia compared with the time of taking the drug for < 1 year; (4) Compared with subjects with negative ApoEε4 allele, the use of statins has a more significant effect on reducing the risk of dementia in subjects carrying the ApoEε4 allele.
Collapse
Affiliation(s)
- Xue Li
- Department of Neurology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, China
| | - Yuzhen Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, China
| | - Tiantian Song
- Department of Neurology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, China
| | - Xiaoxuan Wu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, China
| | - Ying Xing
- Department of Neurology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, 130033, China.
| |
Collapse
|
6
|
Sabeel S, Motaung B, Nguyen KA, Ozturk M, Mukasa SL, Wolmarans K, Blom DJ, Sliwa K, Nepolo E, Günther G, Wilkinson RJ, Schacht C, Kengne AP, Thienemann F, Guler R. Impact of statins as immune-modulatory agents on inflammatory markers in adults with chronic diseases: A systematic review and meta-analysis. PLoS One 2025; 20:e0323749. [PMID: 40440323 PMCID: PMC12121830 DOI: 10.1371/journal.pone.0323749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/15/2025] [Indexed: 06/02/2025] Open
Abstract
While numerous studies have extensively documented the pleiotropic effects of statins, including their capacity to reduce inflammation, there is a lack of research estimating the anti-inflammatory effectiveness of statins among individuals with chronic diseases. This meta-analysis evaluates the effect of statin therapy on inflammatory markers and the lipid profile in patients with chronic diseases by analysing evidence from randomized controlled trials (RCTs). We conducted a systematic review and searched articles published between 1st January 1999 and 31st December 2023 in databases including PubMed, Web of Science, Scopus, and Cochrane. The meta-analysis was performed using random effects models and inverse variance. Effect measures were mean differences (MD) and 95% confidence intervals (CI). Collectively, statins significantly reduced IL-6 (MD = -0.24 ng/dL [95% CI, -0.36 to -0.13], I2 = 98.3%, p < 0.001), TNF-α (MD = -0.74 ng/dL [95% CI, -1.08 to -0.40], I2 = 98.8%, p < 0.001); and CRP (MD = -1.58 mg/L [95% CI, -2.22 to -0.94], I2 = 86.5%, p < 0.001). Notably, atorvastatin demonstrated the most significant reduction in IL-6 and TNF-α levels, while fluvastatin and rosuvastatin displayed the greatest impact on decreasing CRP and LDL-C levels, respectively. Stratification by a longer treatment duration of more than four months revealed that atorvastatin achieved the most significant reduction in IL-6 and TNF-α. In conclusion, statin therapy not only regulates the lipid profile but also reduces systemic inflammatory biomarkers. Prolonged administration of statins led to a more substantial reduction in IL-6 and TNF-α, with atorvastatin exhibiting the greatest effect in our analysis.
Collapse
Affiliation(s)
- Solima Sabeel
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Sciences, University of Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Bongani Motaung
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Sciences, University of Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Kim A. Nguyen
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Mumin Ozturk
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Sciences, University of Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
| | - Sandra L. Mukasa
- General Medicine & Global Health (GMGH), Department of Medicine and Cape Heart Institute, Faculty of Health Science, University of Cape Town, South Africa
| | - Karen Wolmarans
- General Medicine & Global Health (GMGH), Department of Medicine and Cape Heart Institute, Faculty of Health Science, University of Cape Town, South Africa
| | - Dirk J. Blom
- Division of Lipidology, Department of Medicine and Cape Heart Institute, University of Cape Town, South Africa
| | - Karen Sliwa
- Cape Heart Institute and Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Emmanuel Nepolo
- Department of Human, Biological & Translational Sciences, School of Medicine, University of Namibia, Windhoek, Namibia
| | - Gunar Günther
- Department of Human, Biological & Translational Sciences, School of Medicine, University of Namibia, Windhoek, Namibia
- Department of Pulmonary Medicine and Allergology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Robert J. Wilkinson
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, South Africa
- Francis Crick Institute, London, United Kingdom
- Department of Infectious Diseases, Imperial College London, United Kingdom
| | | | - Andre Pascal Kengne
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Friedrich Thienemann
- General Medicine & Global Health (GMGH), Department of Medicine and Cape Heart Institute, Faculty of Health Science, University of Cape Town, South Africa
- Department of Internal Medicine, University Hospital Zurich, University of Zurich, Switzerland
- Cape Universities Body Imaging Centre (CUBIC), University of Cape Town, South Africa
| | - Reto Guler
- Institute of Infectious Diseases and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Sciences, University of Cape Town, South Africa
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, South Africa
| |
Collapse
|
7
|
Arsha S, Tripathi A, Kangarlu J, Rehman B, Frishman WH, Aronow WS. Chemotherapy-Induced Cardiomyopathy: A Focus on the Utility of Statins. Cardiol Rev 2025:00045415-990000000-00492. [PMID: 40358411 DOI: 10.1097/crd.0000000000000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Chemotherapy-induced cardiomyopathy (CICM) is a critical adverse consequence associated with chemotherapeutic treatments such as anthracyclines, taxanes, and alkylating agents. Cardiac dysfunction, characterized by left ventricular systolic dysfunction, is the primary effect found in these patients. This may result in heart failure, with heart failure related to chemotherapy resulting in a 3.5-fold increased risk of mortality compared with idiopathic cardiomyopathy alone. Multiple factors, including oxidative stress, inflammation, and disruption of key cellular pathways, are involved in cardiomyocyte damage and influence CICM pathophysiology. So far, dexrazoxane is the sole FDA-approved preventive therapy, but alternative interventions, such as beta-blockers, angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, and statins, have been studied for their cardioprotective potential. Statins, beyond their cholesterol-lowering capabilities, stand out for their pleiotropic effects, including antioxidant, anti-inflammatory, and endothelial-protective actions, which counteract inflammatory effects. Multiple studies and meta-analyses suggest that statin therapy may decrease both the incidence and severity of chemotherapy-related cardiotoxicity (CTX), as evidenced by smaller declines in left ventricular ejection fraction and lower rates of heart failure in statin-treated patients. However, not all investigations confirm these protective benefits; for instance, some trials, including SPARE-HF, reported no significant differences in cardiac outcomes. While these conflicting findings underscore the need for larger randomized trials, they also reflect the heterogeneity of cancer types, chemotherapy regimens, and patient profiles. Statins show promise as a cardioprotective strategy for individuals at risk of CICM. Enhancing patient selection and specifying the timing and duration of statin therapy are essential steps for incorporating these agents into standard care. Optimizing these parameters may reduce chemotherapy-related cardiac damage, improve long-term cardiac function, and enhance overall survival in cancer survivors.
Collapse
Affiliation(s)
- Sanjana Arsha
- From the Department of Internal Medicine, Westchester Medical Center, Vallhala, NY
| | - Ashish Tripathi
- From the Department of Internal Medicine, Westchester Medical Center, Vallhala, NY
| | - John Kangarlu
- From the Department of Internal Medicine, Westchester Medical Center, Vallhala, NY
| | - Bilal Rehman
- From the Department of Internal Medicine, Westchester Medical Center, Vallhala, NY
| | - William H Frishman
- Department of Cardiology, Westchester Medical Center, Valhalla, NY
- Department of Medicine, New York Medical College, Valhalla, NY
| | - Wilbert S Aronow
- Department of Cardiology, Westchester Medical Center, Valhalla, NY
- Department of Medicine, New York Medical College, Valhalla, NY
| |
Collapse
|
8
|
Zerin F, Hoque N, Menon SN, Ezewudo E, Simon NP, Sooreni S, Shahid MS, Jones M, Pandey A, Gökçe Y, Rahman T, Hasan R. Nanomolar therapeutic concentrations of statins rapidly induce cerebral artery vasoconstriction by stimulating L-type calcium channels. Biochem Pharmacol 2025; 238:116970. [PMID: 40320051 DOI: 10.1016/j.bcp.2025.116970] [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: 02/16/2025] [Revised: 04/03/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
All commonly prescribed statins have been reported to cause reversible memory loss within weeks of therapy, though the exact molecular mechanism remains unknown. However, whether therapeutic concentrations of statins can directly regulate the contractility of resistance cerebral arteries that control cerebrovascular perfusion remains unexplored. Here, we examined the acute vascular effects of statins on rat cerebral arteries and the underlying molecular mechanisms. Our pressure myography data demonstrate that, at therapeutically-relevant nanomolar concentrations, statins produced a robust and rapid vasoconstriction, appearing within 2-3 min of drug application. Interestingly, such vasoconstriction was largely absent in female rat cerebral arteries. Endothelial denudation or mevalonate supplementation did not alter statin-induced vasoconstriction, suggesting an endothelium- and cholesterol-independent mechanism. In contrast, such vasoconstriction was abolished upon removal of extracellular Ca2+, pharmacological blockade of the smooth muscle cell voltage-gated Ca2+ channel, CaV1.2, or siRNA knockdown of CaV1.2 - all of which reduced [Ca2+]i, indicating that Ca2+ entry through CaV1.2 plays a critical role in cerebral artery vasoconstriction. Arterial biotinylation revealed that acute statin exposure did not alter the surface expression, distribution, or function of CaV1.2 channels. Altogether, our data unveil an unexpected role of statins in rapidly inducing constriction of resistance cerebral arteries by directly stimulating CaV1.2 in smooth muscle cells. These findings offer a plausible explanation for statin-associated reversible memory impairment, its mitigation by calcium channel blockers, and why such effects may not be observed in all subjects, particularly those concurrently taking antihypertensive agents.
Collapse
Affiliation(s)
- Farzana Zerin
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Nazia Hoque
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA; Department of Pharmacy, East West University, Dhaka, Bangladesh
| | - Sreelakshmi N Menon
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Emmanuella Ezewudo
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Nimi P Simon
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Samira Sooreni
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Mashmum S Shahid
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Morgan Jones
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA
| | - Ajay Pandey
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA; Department of Biological Sciences, Augusta University, Augusta, GA, USA
| | - Yasin Gökçe
- Department of Biophysics, School of Medicine, Harran University, Sanlıurfa 63300, Turkey
| | - Taufiq Rahman
- Department of Pharmacology, University of Cambridge, UK
| | - Raquibul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, USA.
| |
Collapse
|
9
|
Apalama ML, Begue F, Tanaka S, Cournot M, Couret D, Meilhac O, Pokeerbux MR. High-density lipoproteins and COVID-19: preparing the next pandemic. J Lipid Res 2025; 66:100779. [PMID: 40090619 PMCID: PMC12141899 DOI: 10.1016/j.jlr.2025.100779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/18/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025] Open
Abstract
High-density lipoproteins (HDLs) are heterogeneous particles with pleiotropic functions including anti-inflammatory and anti-infectious effects. In clinical studies, lower HDL-associated cholesterol (HDL-C) concentration has been associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, severity, and mortality. A reduction in the number of HDL particles, particularly small ones has been observed with alterations in their protein and lipid composition impairing their functions. These observations have supported HDL supplementation with promising results in small preliminary studies. This review summarizes available evidence to better understand the two-way interaction between HDLs and Coronavirus disease 2019 (COVID-19) and guide future HDL-based therapies for preparing for the next pandemic.
Collapse
Affiliation(s)
- Marie Laurine Apalama
- Université de La Réunion, UMR Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM U1188, Saint-Pierre, France
| | - Floran Begue
- Université de La Réunion, UMR Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM U1188, Saint-Pierre, France; USMD, Délégation de la Recherche Clinique et de l'Innovation, CHU de La Réunion, Saint-Pierre, France
| | - Sébastien Tanaka
- Université de La Réunion, UMR Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM U1188, Saint-Pierre, France; AP-HP, Service d'Anesthésie-Réanimation, CHU Bichat-Claude Bernard, Paris, France
| | - Maxime Cournot
- Université de La Réunion, UMR Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM U1188, Saint-Pierre, France; Clinique Les Orchidées, Groupe de santé Clinifutur, Le Port, France
| | - David Couret
- Université de La Réunion, UMR Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM U1188, Saint-Pierre, France; Service de Neuroréanimation, CHU de la Réunion, Saint-Pierre, France
| | - Olivier Meilhac
- Université de La Réunion, UMR Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM U1188, Saint-Pierre, France; INSERM CIC1410, Plateforme de Recherche Clinique et Translationnelle, CHU de La Réunion, Saint-Pierre, France.
| | - Mohammad Ryadh Pokeerbux
- Université de La Réunion, UMR Diabète Athérothrombose Réunion Océan Indien (DéTROI), INSERM U1188, Saint-Pierre, France; Service de Médecine Interne et Polyvalente, CHU de la Réunion, Saint-Pierre, France
| |
Collapse
|
10
|
Veedu AP, Kunhipurayil D, Beegum F, George KT, Kanwal A, Shenoy RR, Nandakumar K. Biochanin‑A as SIRT‑1 modulator in preventing statin‑associated diabetogenesis: An in vitro study. Biomed Rep 2025; 22:91. [PMID: 40171401 PMCID: PMC11959223 DOI: 10.3892/br.2025.1969] [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: 07/02/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025] Open
Abstract
The widespread use of statin therapy for hypercholesterolemia has raised concerns due to its associated risk of inducing diabetes. Biochanin-A (BA), an isoflavone, exhibits potential in preventing diabetes and hyperlipidemia, yet its efficacy in mitigating statin-induced diabetes remains unexplored. This gap prompts a crucial inquiry: Can BA reduce the risk of diabetes associated with statin therapy? The present study investigated the molecular mechanisms behind atorvastatin's diabetogenic nature and evaluated the potential of BA to counteract these effects. Insulin resistance was assessed using L6 skeletal muscle cells and pancreatic beta cell apoptosis in MIN-6 cells. Our hypothesis posits that atorvastatin exacerbates free fatty acid accumulation, leading to the downregulation of sirtuin-1 (SIRT-1) and decreased uncoupling protein (UCP) 3 expression, culminating in insulin resistance. Conversely, BA is assumed to positively modulate SIRT-1 and downregulate UCP2, thus offering a protective effect. In vitro studies using L6 and MIN-6 cells revealed that BA has increased cell viability and shown optimal protection against the toxicity induced by atorvastatin in both cell lines at different concentrations. BA effectively inhibited the reduction in glucose uptake caused by atorvastatin. Pre-treatment with BA upregulated proteins that are involved in the insulin-signaling pathway and reversed the expression levels of UCPs induced by atorvastatin. BA also enhanced insulin release, preserved mitochondrial function, and prevented atorvastatin-induced apoptosis. Furthermore, BA improved SIRT-1 expression, potentially through the nicotinamide phospho-ribosyl-transferase-nicotinamide adenine dinucleotide + SIRT1-pathway, revealing that BA may play a role in modulating cellular processes in statin-associated SIRT-1 downregulation. BA can be considered a promising molecule to counteract statin-induced diabetes, suggesting a prospective therapeutic role in enhancing the safety profile of statin therapy. This research lays the groundwork for future clinical evaluations of BA as an adjunctive treatment for patients at risk of statin-induced diabetes.
Collapse
Affiliation(s)
- Anuranjana Putiya Veedu
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Divya Kunhipurayil
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab 151005, India
| | - Rekha Raghuveer Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Co-ordinator, Center for Animal Research, Ethics and Training (CARET), Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
11
|
Zolio L, Cohen H, Isenberg D. Challenges of anticoagulation in patients with systemic lupus erythematosus. Expert Opin Pharmacother 2025; 26:849-862. [PMID: 40253682 DOI: 10.1080/14656566.2025.2491509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Anticoagulation is frequently required for patients with systemic lupus erythematosus (SLE), given their high prevalence of cardiovascular disease and thrombosis, due to the complexity of disease pathophysiology, some overlap with antiphospholipid syndrome (APS), comorbidities, prevalent cardiovascular risk factors and treatment complications. AREAS COVERED This article outlines the epidemiology and pathophysiology of cardiovascular disease and arterial and/or venous thrombosis in SLE, with/without APS. We discuss common cardiovascular comorbidities and thrombotic disorders that may present as a complication of SLE and/or APS and highlight recommendations in current guidelines for anticoagulation management, alongside relevant disease-specific considerations. We specifically comment on the use of direct oral anticoagulants (DOACs) for venous thromboembolism (VTE) in these patients. EXPERT OPINION Assessment of cardiovascular risk and aPL profile is paramount in SLE patients. While warfarin is preferred in high-risk APS patients, DOACs can be used in a selected group of SLE and/or APS patients with VTE and no prior history of arterial thrombosis. Initiating anticoagulation in the setting of Catastrophic APS (CAPS) can be extremely challenging. Knowledge gaps remain regarding the management of patients with recurrent arterial and/or venous thrombosis despite anticoagulation. Research is needed to optimize strategies to reduce thrombotic events in APS patients.
Collapse
Affiliation(s)
- Luigi Zolio
- Department of Ageing, Rheumatology and Regenerative Medicine, Division of Medicine, University College London, London, UK
- Department of Medicine, University of Melbourne at St Vincent's Hospital, Fitzroy, Australia
| | - Hannah Cohen
- Department of Haematology, University College London Hospitals NHS Foundation Trust and University College London, London, UK
| | - David Isenberg
- Department of Ageing, Rheumatology and Regenerative Medicine, Division of Medicine, University College London, London, UK
| |
Collapse
|
12
|
Kumar K, Aggarwal S, Kandpal A, Kaur R, Jaggi AS, Yadav HN, Singh D, Chopra D, Singh N. Unraveling the role of brain renin angiotensin system in vascular dementia: mechanisms and therapeutic perspectives. Exp Brain Res 2025; 243:130. [PMID: 40285869 DOI: 10.1007/s00221-025-07072-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/29/2025] [Indexed: 04/29/2025]
Abstract
Dementia is a heterogeneous syndrome characterized by the progressive deterioration of various brain functions, severely impacting cognitive, emotional, and social abilities. According to a World Health Organization (WHO) report, dementia represents a pressing global health concern, with the number of affected individuals projected to triple by 2050. Among its various subtypes, vascular dementia (VD) stands as the second most common form, following Alzheimer's disease (AD). Despite ongoing efforts in drug development, no pharmaceutical entity has yet received approval from the U.S. Food and Drug Administration (FDA) for the treatment of VD. Emerging evidence underscores the critical involvement of the brain's Renin-Angiotensin System (RAS) in the pathogenesis of multiple neurodegenerative disorders, including VD. The intricate roles of RAS components include regulating vascular tone, neuronal growth and survival, regulating cerebral blood flow and endothelial dysfunction, increasing neuroinflammation (by increasing release of IL-1, IL-6, TNF-α, microglial activation), oxidative stress and destruction of BBB integrity, mainly through Angiotensin II type 1 (AT1) and type 2 (AT2) receptors, are of significant interest in the pathophysiology of VD. However, disruptions in these signaling pathways are believed to contribute substantially to the progression of VD. This review addresses the limitations of current therapeutic approaches for VD while emphasizing the untapped potential of RAS-targeted interventions. We systematically explore the neurophysiological mechanisms of brain RAS, their role in promoting neuronal health, and the factors that compromise these pathways, ultimately leading to cognitive decline. By elucidating these mechanisms and challenges, the review offers novel insights into designing innovative RAS-based therapeutic strategies, paving the way for effective clinical management of VD. This work aspires to stimulate further research and development in this underexplored yet promising domain.
Collapse
Affiliation(s)
- Kuldeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Sonal Aggarwal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Ayush Kandpal
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Ramanpreet Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Amteshwar S Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Harlokesh Narayan Yadav
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Dhandeep Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Dimple Chopra
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
13
|
Li X, Wang SW, Zhang ZJ, Luo ZY, Tang JF, Tao T. Real-world pharmacovigilance analysis of drug-related cataracts using the FDA adverse event reporting system database. Front Pharmacol 2025; 16:1498191. [PMID: 40343006 PMCID: PMC12058479 DOI: 10.3389/fphar.2025.1498191] [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: 09/18/2024] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
Objective Although numerous drugs have been associated with cataracts, the risk for most drugs remains unclear. This study aimed to investigate the risk factors for drug-induced cataracts by analyzing large-scale data from the U.S. Food and Drug Administration Adverse Event Reporting System (FAERS). Methods We used the reporting odds ratio (ROR) to evaluate reports of drug-induced cataracts in FAERS from the first quarter of 2004 to the third quarter of 2024. A univariate analysis, LASSO (least absolute shrinkage and selection operator) regression, and a multivariate regression analysis were performed to identify drug-related risk factors for cataracts, and Bonferroni correction was applied for multiple comparisons. Results Multivariate logistic regression ultimately identified 15 drugs as independent risk factors, including immunomodulators (6/15), antineoplastic drugs (3/15), psychotropic drugs (1/15), respiratory drugs (1/15), gastrointestinal drugs (1/15), orthopedic drugs (1/15), metabolic regulators (1/15), and ophthalmic drugs (1/15). The median time to onset of drug-induced cataracts was 449 days (interquartile range [IQR]: 150-901 days), with approximately 75% of adverse events occurring within 747 days. Conclusion These findings may help clinicians detect drug-related cataracts at an early stage and provide valuable insights for future research on the mechanisms of drug-induced cataracts.
Collapse
Affiliation(s)
- Xiang Li
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, Wan Zhou, China
| | - Shu Wen Wang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhi-Jie Zhang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhan Yang Luo
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jia Feng Tang
- Chongqing Key Laboratory of Development and Utilization of Genuine Medicinal Materials in Three Gorges Reservoir Area, Chongqing Three Gorges Medical College, Wan Zhou, China
| | - Tao Tao
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
14
|
Borgquist S, Jensen MB, Bendorff CL, Christiansen P, Offersen BV, Kodahl AR, Ewertz M, Jensen AB, Ahern TP, Cronin-Fenton D, Ejlertsen B, On behalf of the Danish Breast Cancer Group. Statin Therapy in Early Breast Cancer: The MASTER Trial; A Randomized Phase III, Placebo-Controlled Comparison of Standard (Neo)Adjuvant Therapy Plus Atorvastatin versus Standard (Neo)Adjuvant Therapy Plus Placebo. Clin Epidemiol 2025; 17:409-419. [PMID: 40260427 PMCID: PMC12011050 DOI: 10.2147/clep.s509873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/06/2025] [Indexed: 04/23/2025] Open
Abstract
Purpose Statin use has been consistently associated with improved clinical outcomes (especially recurrence) in breast cancer in multiple observational studies backed by compelling preclinical evidence. The strength of this evidence warrants a clinical trial to test the efficacy of statin exposure on breast cancer recurrence. Patients and Methods The double-blind, phase III, randomized, placebo-controlled MASTER (MAmmary cancer STatins in ER positive breast cancer) trial includes women diagnosed with early-stage, estrogen receptor-positive (ER+) breast cancer who are candidates for systemic (neo)adjuvant therapy. Enrolled patients are given standard (neo)adjuvant therapy and additionally randomized to either atorvastatin (80 mg/day) or placebo for two years. The trial's primary outcome is invasive disease-free survival (IDFS), with a target accrual of 3360 patients in total to achieve 80% power (two-sided alpha=0.05) to detect a 25% reduction in the risk of an IDFS event comparing the statin and placebo arms. At 3-, 6-, 12-, and 24-month follow-up time points, patients will have blood drawn for biomarker studies, answer patient-reported outcome (PRO) questionnaires, and control for adverse events. Subsequently, patients will receive annual PRO-criteria for Adverse Events (CTCAE) questionnaires until the completion of their 10 years of follow-up. Secondary endpoints include additional clinical endpoints; pathological response (neo-adjuvant treated patients), recurrence-free survival, distant-recurrence-free interval, overall survival and cardiac death-free interval, co-morbidity, and health-related quality-of-life measured by PRO-CTCAE questionnaires during and beyond study medication. Translational endpoints are evaluated in collected blood- and tumor samples. Discussion If a protective effect of statins on breast cancer recurrence is supported by evidence from the MASTER trial, then the indications for a safe, well-tolerated, and inexpensive treatment can be expanded towards improved clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Signe Borgquist
- Department of Oncology, Aarhus University Hospital/Aarhus University, Aarhus, Denmark
- Division of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Maj-Britt Jensen
- Danish Breast Cancer Cooperative Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Peer Christiansen
- Department of Plastic and Breast Surgery, Aarhus University Hospital/Aarhus University, Aarhus, Denmark
| | | | - Annette Raskov Kodahl
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Marianne Ewertz
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Anders Bonde Jensen
- Department of Oncology, Aarhus University Hospital/Aarhus University, Aarhus, Denmark
| | - Thomas P Ahern
- Department of Oncology, Aarhus University Hospital/Aarhus University, Aarhus, Denmark
- Department of Surgery, The Robert Larner, M.D. College of Medicine at The University of Vermont, Burlington, VT, USA
| | - Deirdre Cronin-Fenton
- Department of Clinical Epidemiology, Department of Clinical Medicine, Aarhus University Hospital/Aarhus University, Aarhus, Denmark
| | - Bent Ejlertsen
- Danish Breast Cancer Cooperative Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - On behalf of the Danish Breast Cancer Group
- Department of Oncology, Aarhus University Hospital/Aarhus University, Aarhus, Denmark
- Division of Oncology, Clinical Sciences, Lund University, Lund, Sweden
- Danish Breast Cancer Cooperative Group, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Plastic and Breast Surgery, Aarhus University Hospital/Aarhus University, Aarhus, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Surgery, The Robert Larner, M.D. College of Medicine at The University of Vermont, Burlington, VT, USA
- Department of Clinical Epidemiology, Department of Clinical Medicine, Aarhus University Hospital/Aarhus University, Aarhus, Denmark
| |
Collapse
|
15
|
Kambalapalli S, Bhandari M, Punnanithinont N, Iskander B, Khan MA, Budoff M. Bridging Prevention and Imaging: The Influence of Statins on CAC and CCTA Findings. Curr Atheroscler Rep 2025; 27:50. [PMID: 40198377 DOI: 10.1007/s11883-025-01287-x] [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] [Accepted: 03/08/2025] [Indexed: 04/10/2025]
Abstract
To evaluate the impact of statins on CHD prevention, role of CAC scoring and CCTA in guiding statin therapy for both primary and secondary prevention in ASCVD. Coronary artery calcium (CAC) scoring and coronary computed tomography angiography (CCTA) have emerged as vital non-invasive imaging tools for refining cardiovascular risk assessment and guiding statin therapy in patients with atherosclerotic cardiovascular disease (ASCVD). CAC scoring helps stratify patients based on subclinical atherosclerosis burden, while CCTA provides detailed insights into plaque composition and distribution. Multiple studies, including the Multi-Ethnic Study of Atherosclerosis (MESA) and the CONFIRM registry, have demonstrated the utility of CAC scoring in identifying individuals at risk of major adverse cardiovascular events (MACE) and guiding personalized statin therapy. CAC scores, categorized into risk-based thresholds, enable clinicians to determine when statins should be initiated or deferred. CCTA complements CAC scoring by assessing plaque characteristics, including non-calcified plaque (NCP), calcified plaque, and high-risk features such as low-attenuation plaques, spotty calcifications, and positive remodeling. Serial CCTA imaging has further highlighted the effect of high-intensity statin therapy on plaque progression, demonstrating reductions in NCP and stabilization through increased calcification. CAC scoring effectively identifies patients with subclinical atherosclerosis who would benefit from statin therapy, particularly those with CAC scores > 100 or in the ≥ 75th percentile for age and sex. Statin therapy has been shown to promote plaque stabilization by increasing calcified plaque volume while reducing the progression of non-calcified plaques, thereby mitigating the risk of plaque rupture. CCTA provides additional value by identifying vulnerable plaque features and monitoring the impact of statin therapy over time. Studies have demonstrated significant reductions in total plaque volume and low-attenuation plaques in patients undergoing intensive lipid-lowering therapy, reinforcing the role of CCTA in guiding statin decisions for patients with established ASCVD. CAC scoring serves as a powerful tool to refine risk stratification and guide statin therapy initiation, particularly in asymptomatic individuals. CCTA enhances this approach by providing comprehensive plaque assessment and monitoring the response to statin therapy. Integrating CAC scoring and CCTA into clinical practice allows for a personalized approach to ASCVD management, improving patient outcomes through optimized statin therapy and targeted risk reduction.
Collapse
Affiliation(s)
- Soumya Kambalapalli
- Department of Advanced Cardiovascular Imaging, UCLA-Harbor, the Lundquist Institute, Torrance, CA, USA.
| | - Mrinal Bhandari
- Department of Advanced Cardiovascular Imaging, UCLA-Harbor, the Lundquist Institute, Torrance, CA, USA
| | - Natdanai Punnanithinont
- Department of Advanced Cardiovascular Imaging, UCLA-Harbor, the Lundquist Institute, Torrance, CA, USA
| | - Beshoy Iskander
- Department of Advanced Cardiovascular Imaging, UCLA-Harbor, the Lundquist Institute, Torrance, CA, USA
| | - Muneeb A Khan
- Department of Advanced Cardiovascular Imaging, UCLA-Harbor, the Lundquist Institute, Torrance, CA, USA
| | - Matthew Budoff
- Department of Cardiology, UCLA-Harbor, the Lundquist Institute, Torrance, CA, USA
| |
Collapse
|
16
|
Blázquez AB, Mingo-Casas P, Quesada E, Priego EM, Pérez-Perez MJ, Martín-Acebes MA. Lipid-targeting antiviral strategies: Current state and future perspectives. Antiviral Res 2025; 236:106103. [PMID: 39947433 DOI: 10.1016/j.antiviral.2025.106103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/26/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
There is an urgent need for antiviral compounds effective against currently known and future viral threats. The development of host-targeting antivirals (HTAs) appears as an alternative strategy to fight viral infections minimizing the potential of resistant mutant development and potentially leading to the identification of broad-spectrum antiviral agents. Among the host factors explored for HTA strategy, lipids constitute an attractive target as many viruses, even genetically diverse, hijack specific lipids during their lifecycle. Multiple repurposing efforts have been performed to analyze the antiviral properties of lipid-targeting compounds. These studies include the analysis of the effects of cholesterol lowering drugs such as statins, cholesterol transport inhibitors, sphingolipid modulators, de novo lipogenesis inhibitors blocking fatty acid synthesis, compounds targeting glycerophospholipids or drugs interfering with lipid droplet metabolism. This review is focused on the current status of lipid-based or lipid-targeting antiviral strategies and their potential for the development of antiviral therapies, with special emphasis on those studies that have reached advanced stages of development such as efficacy studies in animal models or clinical trials. Whereas there is still a long way to go, multiple proof-of-concept studies and clinical evidence reinforce the therapeutic potential of these strategies warranting their further development into effective antiviral therapies.
Collapse
Affiliation(s)
- Ana-Belén Blázquez
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain.
| | - Patricia Mingo-Casas
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain; Universidad Autónoma de Madrid (UAM, Escuela de Doctorado), Spain
| | | | | | | | - Miguel A Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA-CSIC), Madrid, Spain.
| |
Collapse
|
17
|
Cheng EY, Mirzaei A. Differential risk of autoimmune disorders in non-traumatic osteonecrosis: clue to pathogenesis. Expert Rev Clin Immunol 2025; 21:413-424. [PMID: 40035487 DOI: 10.1080/1744666x.2025.2475982] [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: 10/09/2024] [Revised: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Non-traumatic osteonecrosis is a frequent complication in patients with autoimmune disorders, though its prevalence varies markedly depending upon the type of disorder. Understanding the causes of this difference can help uncover the underlying pathophysiology of osteonecrosis and guide the development of effective preventive and therapeutic strategies. AREAS COVERED In this perspective study, we reviewed available databases, including PubMed, Cochrane Library, Scopus, and Web of Science, to explore why the risk of osteonecrosis varies among different autoimmune disorders. Is this variation primarily due to the disease's pathophysiology, the use of medications such as corticosteroids, or a combination of both? If both factors are involved, what is the extent of each contribution in this context? EXPERT OPINION Non-traumatic osteonecrosis is often induced by an interaction between disease pathophysiology and corticosteroid use. In patients with different autoimmune disorders but an identical history of corticosteroid use, the risk of osteonecrosis is influenced by how the underlying pathophysiology compromises bone health. In autoimmune disorders with multiple adverse effects on bone, such as SLE (systemic lupus erythematosus), there is a much higher risk of osteonecrosis compared to disorders with minimal impact on bone health, such as celiac disease and MS (multiple sclerosis).
Collapse
Affiliation(s)
- Edward Y Cheng
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Alireza Mirzaei
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
18
|
Fravel MA, Ernst ME, Woods RL, Orchard SG, Polkinghorne KR, Wolfe R, Wetmore JB, Nelson MR, Bongetti E, Murray AM, Zoungas S, Zhou Z. Effects of statins on kidney function in older adults. J Am Geriatr Soc 2025; 73:1082-1093. [PMID: 39696786 PMCID: PMC11970218 DOI: 10.1111/jgs.19319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 11/24/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The effect of statin therapy on kidney function among older adults is unclear. OBJECTIVES To examine the association between statin use and changes in estimated glomerular filtration rate (eGFR) and urine albumin-to-creatinine ratio (UACR), positive or negative, in an older adult cohort with versus without chronic kidney disease (CKD) at baseline. METHODS This analysis included 18,056 participants aged ≥65 years with versus without CKD at baseline in a randomized trial of low-dose aspirin, who had no prior cardiovascular events, major physical disability, or dementia initially. Outcome measures included eGFR and UACR. Linear mixed-effects models were used to estimate the associations of baseline statin use versus no use with eGFR and UACR changes over time. The inverse-probability of treatment-weighting technique was used for all analyses to address confounding by indication due to the lack of randomization in treatment assignment. RESULTS Statin use was not associated with change in eGFR, UACR, or incident CKD in participants with or without CKD at baseline (p > 0.05 for all associations). Subgroup analyses found no significant interactions between statin and age, sex, diabetes, country, and frailty status on any of the study outcomes. CONCLUSIONS Among adults ≥65 years of age, with and without CKD, statin therapy was not associated with improved or worsened kidney function. This data suggests that the decision to use versus not use statins in this population may be ideally guided by factors other than kidney health.
Collapse
Affiliation(s)
- Michelle A. Fravel
- Department of Pharmacy Practice and Science, College of PharmacyThe University of IowaIowa CityIowaUSA
| | - Michael E. Ernst
- Department of Pharmacy Practice and Science, College of PharmacyThe University of IowaIowa CityIowaUSA
- Department of Family Medicine, Carver College of MedicineThe University of IowaIowa CityIowaUSA
| | - Robyn L. Woods
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Suzanne G. Orchard
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Kevan R. Polkinghorne
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
- Department of Nephrology, Monash Medical CentreMonash HealthMelbourneVictoriaAustralia
- Department of MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Rory Wolfe
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - James B. Wetmore
- Department of MedicineHennepin HealthcareMinneapolisMinnesotaUSA
- Nephrology DepartmentHennepin HealthcareMinneapolisMinnesotaUSA
- Hennepin Healthcare Research InstituteMinneapolisMinnesotaUSA
| | - Mark R. Nelson
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
- Menzies Institute for Medical ResearchUniversity of TasmaniaHobartTasmaniaAustralia
| | - Elisa Bongetti
- Department of Nephrology, Monash Medical CentreMonash HealthMelbourneVictoriaAustralia
- Department of MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Anne M. Murray
- Berman Center for Outcomes and Clinical ResearchHennepin‐Health Research InstituteMinneapolisMinnesotaUSA
- Division of Geriatrics, Department of MedicineHennepin HealthcareMinneapolisMinnesotaUSA
| | - Sophia Zoungas
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| | - Zhen Zhou
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
19
|
Nakashima R, Ikeda S, Shinohara K, Matsumoto S, Yoshida D, Ono Y, Nakashima H, Miyamoto R, Matsushima S, Kishimoto J, Itoh H, Komuro I, Tsutsui H, Abe K. Triglyceride/high density lipoprotein cholesterol index and future cardiovascular events in diabetic patients without known cardiovascular disease. Sci Rep 2025; 15:9217. [PMID: 40097497 PMCID: PMC11914472 DOI: 10.1038/s41598-025-92933-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
The triglyceride/high-density lipoprotein cholesterol (TG/HDL-C) index, calculated as TG divided by HDL-C, has been suggested as a predictor of cardiovascular disease (CVD). We investigated the association between the TG/HDL-C index and CVD events in type 2 diabetes mellitus (T2DM) patients with retinopathy and hyperlipidemia but no known CVD, enrolled in the EMPATHY study, which compared intensive and standard statin therapy (targeting LDL-C levels < 70 mg/dL and ≥ 100 to < 120 mg/dL, respectively). A total of 4665 patients were divided into high (TG/HDL-C ≥ 2.5, n = 2013) and low (TG/HDL-C < 2.5, n = 2652) TG/HDL-C index groups. During a median follow-up of 36.8 months, 260 CVD events occurred. The high TG/HDL-C index group had higher CVD risk than the low group (HR 1.89, 95% CI 1.45-2.47, p < 0.001). This association remained consistent across subgroups. A trend toward interaction between TG/HDL-C index and statin treatment allocation for CVD risk was observed (p for interaction = 0.062). Intensive statin treatment reduced CVD risk in the high TG/HDL-C group but not in the low group. In conclusion, a TG/HDL-C index ≥ 2.5 was associated with higher CVD risk in T2DM patients with retinopathy and hyperlipidemia without a history of CVD. The TG/HDL-C index may identify patients who benefit from intensive statin treatment.
Collapse
Affiliation(s)
- Ryosuke Nakashima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Shota Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Keisuke Shinohara
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Sho Matsumoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Daisuke Yoshida
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yoshiyasu Ono
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hiroka Nakashima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Ryohei Miyamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Junji Kishimoto
- Center for Clinical and Translational Research, Kyushu University Hospital, Fukuoka, Japan
| | - Hiroshi Itoh
- Center for Preventive Medicine, School of Medicine, Keio University, Tokyo, Japan
| | - Issei Komuro
- Department of Frontier Cardiovascular Science, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- International University of Health and Welfare, Tokyo, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- International University of Health and Welfare, Fukuoka, Japan
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
20
|
Caiati C, Arrigoni R, Stanca A, Lepera ME. Kidney Toxicity of Drugs for the Heart: An Updated Perspective. Metabolites 2025; 15:191. [PMID: 40137155 PMCID: PMC11943962 DOI: 10.3390/metabo15030191] [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: 01/11/2025] [Revised: 02/14/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Cardiovascular drugs are widely used for the prevention and treatment of various cardiac and vascular disorders. However, some of these drugs can also cause adverse effects on the kidney, leading to acute or chronic renal dysfunction, electrolyte imbalances, and increased mortality. The mechanisms of drug-induced renal toxicity vary depending on the type and class of the drug, the dose and duration of exposure, and the patient's characteristics and comorbidities. In this review, we summarize the current knowledge on the renal effects of some common cardiovascular drugs, such as diuretics, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, calcium channel blockers, beta-blockers, antiplatelet agents, anticoagulants, and statins and proton-pump inhibitors. We also discuss the clinical implications and management strategies for preventing or minimizing drug-induced nephrotoxicity, as well as the potential role of oxidative stress in its pathogenesis.
Collapse
Affiliation(s)
- Carlo Caiati
- Unit of Cardiovascular Diseases, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.); (M.E.L.)
| | - Roberto Arrigoni
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70124 Bari, Italy;
| | - Alessandro Stanca
- Unit of Cardiovascular Diseases, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.); (M.E.L.)
| | - Mario Erminio Lepera
- Unit of Cardiovascular Diseases, Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.); (M.E.L.)
| |
Collapse
|
21
|
Kang I, Potluri V, Khan N, Abdullatef R, Liu A, Babrowski T, Blecha M. Risk factors contributing to 30-day and 1-year mortality event scores following major lower extremity amputation for limb ischemia. J Vasc Surg 2025:S0741-5214(25)00355-6. [PMID: 40054791 DOI: 10.1016/j.jvs.2025.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/28/2025] [Accepted: 02/05/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE The purpose of this study was to create a risk score for 30-day and 1-year mortality after major lower extremity amputation to facilitate clinical expectations and the identification of patients in need of heightened vigilance in longitudinal care. METHODS In the Vascular Quality Initiative, 25,150 patients were identified who underwent lower extremity amputation. Two primary outcomes were investigated: 30-day mortality after major lower extremity amputation and 1-year mortality after amputation. Univariable analysis for the 30-day and 1-year mortality analysis was conducted with χ2 analysis. Significant (P < .05) univariable factors were then included in binary logistic regression analysis to perform multivariable investigation toward the outcomes. Variables that achieved multivariable significance (P < .05) for the outcomes were then used in the respective risk scores with the regression beta coefficient being used to weigh the variables. RESULTS Overall, 7.2% of patients experienced 30-day mortality and 22.4% suffered mortality within 1 year. Variables with a significant multivariable association (P < .05) with 1-year mortality were female sex, advancing age, body mass index less than 20 kg/m2, coronary artery disease, history of coronary revascularization, congestive heart failure (CHF), chronic obstructive pulmonary disease, dialysis status at presentation, baseline renal insufficiency, anemia, lack of statin medication at presentation, being on anticoagulation at presentation, need for above-knee amputation, and need for emergent amputation. Variables that were protective vs mortality on multivariable analysis (P < .05) were body mass index greater than 30 kg/m2, history of ipsilateral amputation at a lower level, and history of ipsilateral infrainguinal bypass. Pertinent negatives included all socio-demographic variables including rural living status, insurance status, and area deprivation index home area. Variables with a statistically significant (P < .05) multivariable association with 30-day mortality were female sex, advancing age, history of coronary artery disease, history of prior coronary revascularization, history of CHF, class 3 or 4 CHF, chronic obstructive pulmonary artery disease, dialysis requirement at presentation, baseline renal insufficiency, lack of antiplatelet medication at time of presentation, lack of statin medication at time of presentation, need for above-knee amputation, acute ischemia indication, and need for emergent amputation. Variables that were protective (P < .05) vs 30-day mortality on multivariable analysis were diabetes, prior ipsilateral amputation at a lower level, prior infrainguinal bypass ipsilateral to amputation, prior ipsilateral infrainguinal endovascular revascularization (endovascular or bypass), and prior ipsilateral inflow arterial bypass. For receiver operating curve analysis, the 30-day risk score had an area under the curve of 0.715 and the 1-year risk score analysis 0.722. Hosmer-Lemeshow investigation of the multivariable regressions resulted in an overall accuracy of 92.8% for the 30-day mortality investigation (99% accurate for survival) and 78% overall accuracy on the 1-year mortality risk score (96.8% in predicting survival accurately). This indicates that these models perform far better in determining which patients will survive rather than precisely determining who will experience 1-year mortality. CONCLUSIONS Risk scores for mortality at 30 days and 1 year after major lower extremity amputation have been created that have good accuracy and steep escalation with advancing comorbidity. Variables with the most potent deleterious effect on survival were renal insufficiency, dialysis requirement, CHF, and need for emergent amputation. Patients who have already been receiving care from vascular specialists at the time of amputation have improved survival vs those without prior arterial interventions or a lower level of amputation. Social determinants of health do not impact survival among patients undergoing major lower extremity amputation at Vascular Quality Initiative centers.
Collapse
Affiliation(s)
- Ian Kang
- Division of Vascular Surgery and Endovascular Therapy, Stritch School of Medicine, Loyola University Health System, Loyola University Chicago, Maywood, IL
| | - Vamsi Potluri
- Division of Vascular Surgery and Endovascular Therapy, Stritch School of Medicine, Loyola University Health System, Loyola University Chicago, Maywood, IL
| | - Nabeeha Khan
- Division of Vascular Surgery and Endovascular Therapy, Stritch School of Medicine, Loyola University Health System, Loyola University Chicago, Maywood, IL
| | - Rand Abdullatef
- Division of Vascular Surgery and Endovascular Therapy, Stritch School of Medicine, Loyola University Health System, Loyola University Chicago, Maywood, IL
| | - Amy Liu
- Division of Vascular Surgery and Endovascular Therapy, Stritch School of Medicine, Loyola University Health System, Loyola University Chicago, Maywood, IL
| | - Trissa Babrowski
- Section of Vascular Surgery and Endovascular Therapy, University of Chicago Medical Center, Chicago, IL
| | - Matthew Blecha
- Division of Vascular Surgery and Endovascular Therapy, Stritch School of Medicine, Loyola University Health System, Loyola University Chicago, Maywood, IL.
| |
Collapse
|
22
|
Karakasis P, Theofilis P, Patoulias D, Vlachakis PK, Antoniadis AP, Fragakis N. Diabetes-Driven Atherosclerosis: Updated Mechanistic Insights and Novel Therapeutic Strategies. Int J Mol Sci 2025; 26:2196. [PMID: 40076813 PMCID: PMC11900163 DOI: 10.3390/ijms26052196] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The global rise in diabetes prevalence has significantly contributed to the increasing burden of atherosclerotic cardiovascular disease (ASCVD), a leading cause of morbidity and mortality in this population. Diabetes accelerates atherosclerosis through mechanisms such as hyperglycemia, oxidative stress, chronic inflammation, and epigenetic dysregulation, leading to unstable plaques and an elevated risk of cardiovascular events. Despite advancements in controlling traditional risk factors like dyslipidemia and hypertension, a considerable residual cardiovascular risk persists, highlighting the need for innovative therapeutic approaches. Emerging treatments, including sodium-glucose cotransporter 2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, epigenetic modulators, and RNA-based therapies, are showing promise in addressing the unique challenges of diabetes-associated ASCVD. Precision medicine strategies, such as nanoparticle-based drug delivery and cell-specific therapies, offer further potential for mitigating cardiovascular complications. Advances in multiomics and systems biology continue to deepen our understanding of the molecular mechanisms driving diabetes-associated atherosclerosis. This review synthesizes recent advances in understanding the pathophysiology and treatment of diabetes-related atherosclerosis, offering a roadmap for future research and precision medicine approaches to mitigate cardiovascular risk in this growing population.
Collapse
Affiliation(s)
- Paschalis Karakasis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| | - Panagiotis Theofilis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (P.K.V.)
| | - Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Faculty of Medicine, School of Health Sciences Aristotle, University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Panayotis K. Vlachakis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (P.K.V.)
| | - Antonios P. Antoniadis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| | - Nikolaos Fragakis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| |
Collapse
|
23
|
DURMAN DKALELİ, ARAS M, AKTAŞ GR, DOĞAN BSUYDEŞ. Comparative vasorelaxant effects of atorvastatin and rosuvastatin in rat aorta: investigating the role of perivascular adipose tissue. Turk J Med Sci 2025; 55:518-524. [PMID: 40342323 PMCID: PMC12058016 DOI: 10.55730/1300-0144.5996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/17/2025] [Accepted: 02/26/2025] [Indexed: 05/11/2025] Open
Abstract
Background/aim Statins are known to display pleiotropic effects on the vascular system, beyond their lipid-lowering properties. Studies on isolated vascular preparations have demonstrated their acute relaxant effects on vascular tone. Considering the increased evidence in regard to the contribution of perivascular adipose tissue (PVAT) in the regulation of vascular homeostasis, we aimed to investigate the possible modulatory role of PVAT in the vascular effects of atorvastatin and rosuvastatin in isolated rat aorta. Materials and methods Thoracic aortas isolated from young male Wistar rats were divided into two groups: rings with intact PVAT (+) and rings without PVAT (-), and then mounted in an isolated organ bath system. Rat aortic rings were standardized with potassium chloride (KCl, 40 mM), and then endothelium-dependent relaxation responses were checked by acetylcholine (Ach, 10-7-10-4 M). The concentration-dependent (10-7-10-4 M) effects of atorvastatin and rosuvastatin were studied on rat aortic rings precontracted submaximally with phenylephrine (Phe, 10-6-3 × 10-5 M). In addition, endothelium-independent relaxation responses were evaluated by sodium nitroprusside (SNP, 10-6 M) at the end of each experiment. Results Rat aortic rings with intact PVAT (+) and without PVAT (-) displayed similar endothelium-dependent and -independent relaxations to Ach and SNP, respectively. Increasing concentrations (10-7-10-4 M) of atorvastatin and rosuvastatin directly relaxed the aortic rings with and without PVAT. The maximum relaxant effects of rosuvastatin was found significantly greater than atorvastatin. Conclusion The current study demonstrated that atorvastatin and rosuvastatin displayed prominent relaxations in rat aortic rings with intact PVAT (+) and without PVAT (-). Notably, rosuvastatin produced a greater vasorelaxant effect compared to atorvastatin in rat aortic rings with and without PVAT. Current study provides a novel evidence that PVAT does not significantly influence statin-mediated vasorelaxation under physiological conditions.
Collapse
Affiliation(s)
- Deniz KALELİ DURMAN
- Department of Pharmacology, Faculty of Pharmacy, İstanbul University, İstanbul,
Turkiye
| | - Meryem ARAS
- Department of Pharmacology, Faculty of Pharmacy, İstanbul University, İstanbul,
Turkiye
- Graduate School of Health Sciences, İstanbul University, İstanbul,
Turkiye
- Department of Pharmacology, Faculty of Pharmacy, Biruni University, İstanbul,
Turkiye
| | - G. Ruveyda AKTAŞ
- Department of Pharmacology, Faculty of Pharmacy, İstanbul University, İstanbul,
Turkiye
- Graduate School of Health Sciences, İstanbul University, İstanbul,
Turkiye
| | - B. Sönmez UYDEŞ DOĞAN
- Department of Pharmacology, Faculty of Pharmacy, İstanbul University, İstanbul,
Turkiye
| |
Collapse
|
24
|
Dickerson AG, Joseph CA, Kashfi K. Current Approaches and Innovations in Managing Preeclampsia: Highlighting Maternal Health Disparities. J Clin Med 2025; 14:1190. [PMID: 40004721 PMCID: PMC11856135 DOI: 10.3390/jcm14041190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Preeclampsia (PE) is a major cause of maternal mortality and morbidity, affecting 3-6% of pregnancies worldwide and ranking among the top six causes of maternal deaths in the U.S. PE typically develops after 20 weeks of gestation and is characterized by new-onset hypertension and/or end-organ dysfunction, with or without proteinuria. Current management strategies for PE emphasize early diagnosis, blood pressure control, and timely delivery. For prevention, low-dose aspirin (81 mg/day) is recommended for high-risk women between 12 and 28 weeks of gestation. Magnesium sulfate is also advised to prevent seizures in preeclamptic women at risk of eclampsia. Emerging management approaches include antiangiogenic therapies, hypoxia-inducible factor suppression, statins, and supplementation with CoQ10, nitric oxide, and hydrogen sulfide donors. Black women are at particularly high risk for PE, potentially due to higher rates of hypertension and cholesterol, compounded by healthcare disparities and possible genetic factors, such as the APOL1 gene. This review explores current and emerging strategies for managing PE and addresses the underlying causes of health disparities, offering potential solutions to improve outcomes.
Collapse
Affiliation(s)
- Alexis G. Dickerson
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; (A.G.D.); (C.A.J.)
| | - Christiana A. Joseph
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; (A.G.D.); (C.A.J.)
- Department of Chemistry and Physics, State University of New York at Old Westbury, Old Westbury, NY 11568, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY 10031, USA; (A.G.D.); (C.A.J.)
- Department of Chemistry and Physics, State University of New York at Old Westbury, Old Westbury, NY 11568, USA
- Graduate Program in Biology, City University of New York Graduate Center, New York, NY 10091, USA
| |
Collapse
|
25
|
Kumar R, Atluri SN, Achanta A, Bogishetty C, Chunduri TR, Pss T, Ravi R. Efficacy of Simvastatin in Inhibiting Bone Resorption and Promoting Healing in Delayed Tooth Avulsion: A Case Series. Cureus 2025; 17:e79139. [PMID: 40109815 PMCID: PMC11921055 DOI: 10.7759/cureus.79139] [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] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
An avulsion is a severe dental injury characterized by the complete displacement of a tooth from its socket, often resulting in a compromised prognosis. One of the key factors influencing the success of reimplantation is the extraoral dry time, which refers to the duration the tooth remains outside of the socket. Prolonged dry time significantly impairs the viability of the periodontal ligament cells, crucial for successful healing and reattachment. Despite various protocols and treatment strategies developed to manage avulsed teeth, no single approach addresses all treatment needs effectively, particularly in cases of delayed reimplantation. Simvastatin, an anti-lipidemic drug, has demonstrated pleiotropic effects that extend beyond cholesterol lowering. These effects include anti-inflammatory properties, promotion of bone regeneration, and enhancement of periodontal ligament cell survival. Such actions suggest that simvastatin may have a beneficial role in improving outcomes following the delayed reimplantation of avulsed teeth. This case series proposes the use of simvastatin as an adjunctive treatment for avulsed teeth along with platelet-rich fibrin and hydroxyapatite, particularly in situations where reimplantation is delayed. By mitigating inflammation and stimulating tissue regeneration, simvastatin may help counteract the damage caused by prolonged extraoral dry time. Its potential to promote periodontal ligament cell survival and enhance healing processes could improve the prognosis of reimplantation, even in cases where traditional treatment alone would be less effective. Given these promising properties, simvastatin may represent a valuable addition to the treatment protocol for avulsed teeth with extended dry times. However, further clinical studies and trials are necessary to validate its efficacy and establish a clear role in the management of delayed reimplantation.
Collapse
Affiliation(s)
- Rajesh Kumar
- Paediatric Dentistry, Malla Reddy Institute of Dental Sciences, Hyderabad, IND
| | - Supraja N Atluri
- Paediatric Dentistry, Malla Reddy Institute of Dental Sciences, Hyderabad, IND
| | - Alekhya Achanta
- Paediatric Dentistry, Malla Reddy Institute of Dental Sciences, Hyderabad, IND
| | | | | | - Tejaswini Pss
- Oral and Maxillofacial Surgery, Malla Reddy Institute of Dental Sciences, Hyderabad, IND
| | - Ramakrishna Ravi
- Conservative Dentistry and Endodontics, Malla Reddy Institute of Dental Sciences, Hyderabad, IND
| |
Collapse
|
26
|
Shao H, Xu C, Zhang C, Li L, Wu P, Chen Z, Guan R. Genetic Insights Into Lipid Traits and Lipid-Modifying Drug Targets in Pregnancy Complications: A Two-Sample Mendelian Randomization Study. Int J Womens Health 2025; 17:221-234. [PMID: 39911358 PMCID: PMC11794394 DOI: 10.2147/ijwh.s496268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Background Dyslipidemia is linked to pregnancy complications, but its causal role remains uncertain. This two-sample Mendelian Randomization (MR) study investigated the causal relationship between lipid traits and pregnancy complications and evaluated the impact of lipid-modifying drug targets. Methods Genetic instruments for lipid traits and targets for lipid-modifying drugs were obtained from the Global Lipids Genetics Consortium. Three pregnancy complications' summary statistics came from the FinnGen R9 database. Significant drug targets underwent further analysis using Expression Quantitative Trait Loci data, and mediation analysis identified potential mediators. Results Increased high-density lipoprotein cholesterol (HDL-C) reduced the incidence of preeclampsia (OR: 0.755, 95% CI: 0.639-0.891, p=0.001, FDR=0.012) and gestational diabetes mellitus (GDM) (OR: 0.835, 95% CI: 0.741-0.942, p=0.003, FDR=0.018). Genetic proxies for cholesteryl ester transfer protein (CETP) inhibition correlated with a decreased risk of preeclampsia (OR: 0.863, 95% CI: 0.786-0.947, p=0.002, FDR=0.027), while genetic inhibition of HMG-CoA reductase (HMGCR) increased preeclampsia risk (OR: 1.700, 95% CI: 1.189-2.431, p=0.004, FDR=0.036). Genetically mimicking the enhancement of lipoprotein lipase (LPL) related to a reduced risk of GDM (OR: 0.681, 95% CI: 0.560-0.829, p=1.29×10-4, FDR=0.004). Higher LPL expression in subcutaneous adipose tissue also reduced GDM risk (OR: 0.642, 95% CI: 0.454-0.909, p=0.013). Waist circumference (4.2%) and waist-to-hip ratio adjusted by BMI (5.7%) partially mediated LPL's effect on GDM risk. Conclusion Elevated HDL-C levels help prevent preeclampsia and GDM. CETP and LPL could be therapeutic targets for preeclampsia and GDM, respectively. However, caution is advised with HMGCR-targeting drugs, as they may increase the preeclampsia risk.
Collapse
Affiliation(s)
- Huijing Shao
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, People’s Republic of China
| | - Chang Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Caihong Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| | - Lirong Li
- Department of Traditional Chinese Gynecology, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Pengfei Wu
- Department of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People’s Republic of China
| | - Zixi Chen
- Department of Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, People’s Republic of China
| | - Rui Guan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
27
|
Zhang J, Zhou Y, Guo J, Yan M, Liu C, Du B. Core-Shell Nanoparticles with Sequential Drug Release Depleting Cholesterol for Reverse Tumor Multidrug Resistance. ACS APPLIED MATERIALS & INTERFACES 2025; 17:6689-6702. [PMID: 39813326 DOI: 10.1021/acsami.4c17858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Multidrug resistance (MDR) facilitates tumor recurrence and metastasis, which has become a main cause of chemotherapy failure in clinical. However, the current therapeutic effects against MDR remain unsatisfactory, mainly hampered by the rigid structure of drug-resistant cell membranes and the uncontrolled drug release. In this study, based on a sequential drug release strategy, we engineered a core-shell nanoparticle (DOX-M@CaP@ATV@HA) depleting cholesterol for reverse tumor MDR. DOX-M@CaP@ATV@HA could accurately target tumor cells due to the active targetability of hyaluronic acid (HA) toward CD44 receptors. The calcium phosphate (CaP) shell was cleaved in the lysosomal acidic environment so that the cholesterol-lowering drug atorvastatin (ATV) was rapidly released to diminish cholesterol and P-glycoprotein (P-gp) level on the membrane, thereby boosting tumor cell drug uptake. Next, doxorubicin (DOX) was gradually released from the hydrophobic core of the mPEG-DSPE micelle, inflicting irreversible DNA damage and triggering apoptosis. The nanosystem was proven both in vitro and in vivo to reverse MDR effectively and exhibited a remarkable therapeutic efficacy on drug-resistant tumors with high biosafety. In conclusion, DOX-M@CaP@ATV@HA effectively reverses MDR via cholesterol depletion, which provides an innovative strategy for tumor MDR treatment.
Collapse
Affiliation(s)
- Jieke Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yingying Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jialing Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Mei Yan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Chenxin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Du
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, Henan, China
| |
Collapse
|
28
|
Shirahata T, Enzer NA, Castro V, Chiles J, McDonald ML, Choi B, Diaz AA, Washko GR, San José Estépar R, Ash SY, Rahaghi FN. Effect of Common Medications on Longitudinal Pectoralis Muscle Area in Smokers. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2025; 12:23-32. [PMID: 39636057 PMCID: PMC11925068 DOI: 10.15326/jcopdf.2024.0557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Background Cigarette smoke contributes to skeletal muscle wasting. While exercise and nutritional therapies are effective in improving skeletal muscle quantity and quality, the effect of medications on longitudinal muscle loss is unclear. We investigated whether long-term use of common medications affects longitudinal skeletal muscle changes in current and former smokers. Methods Using quantitative computed tomography imaging, we measured the 5-year changes in pectoralis muscle area (delta-PMA) and pectoralis muscle density (delta-PMD) of 4191 participants in the COPD Genetic Epidemiology (COPDGene®) study. We tested whether specific medications were associated with delta-PMA and/or delta-PMD using regression analyses. Propensity score matching (PSM) analysis was performed to determine the effect of the medications on longitudinal changes on delta-PMA. Results Over the study period, the median delta-PMA for the entire population showed a decrease of 2.23cm2 (interquartile range: -6.52, 1.54). Regression analyses demonstrated statin use was associated with less loss of PMA, whereas, aspirin use was associated with a greater loss of PMA. Specifically, in the PSM-adjusted analysis, statin use was associated with attenuated loss of PMA (median; -1.5 versus -2.5cm2, p=0.017), while aspirin use was associated with increased loss of PMA (median; -2.5 versus -1.9cm2, p=0.040). Conclusion In current and former smokers, statin use was associated with reduced pectoralis muscle wasting, while aspirin use was associated with increased muscle loss. Additional research is needed to verify these findings. (Clinicaltrials.gov identifier NCT00608764).
Collapse
Affiliation(s)
- Toru Shirahata
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan
| | - Nicholas A Enzer
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Victor Castro
- Boston University School of Medicine, Boston, Massachusetts, United States
| | - Joe Chiles
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
| | - Merry-Lynn McDonald
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
- Department of Genetics, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, United States
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama, United States
| | - Bina Choi
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Alejandro A Diaz
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - George R Washko
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Raúl San José Estépar
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Samuel Y Ash
- Department of Critical Care Medicine, South Shore Hospital, South Weymouth, Massachusetts, United States
- Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Farbod N Rahaghi
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
29
|
Marom H, Khan MA, Darvish N, Tornetta III P, Khoury A, Weil YA, Skelley NWM, Allison DC, Meiron S, Ehrmann Barr T. β-Caryophyllene and Statins in Bone Fracture Healing - A Narrative Review. Orthop Res Rev 2025; 17:31-42. [PMID: 39872403 PMCID: PMC11771162 DOI: 10.2147/orr.s506427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/09/2025] [Indexed: 01/30/2025] Open
Abstract
Bone fractures are a leading cause of morbidity and healthcare expenditure globally. The complex healing process involves inflammation, cartilage formation, mineralization, and bone remodeling. Current treatments like immobilization, surgery, and bone grafting, though effective, pose significant challenges, such as prolonged recovery and high costs. Emerging therapies such as biological agents, pharmacological treatments, and physical stimulation techniques are also associated with high costs, side effects, and practical applicability limitations. There is a critical need for alternative therapies that are cost-effective, safe, and easy to use. Recent studies suggest the potential of β-caryophyllene (BCP) and statins in promoting bone healing. BCP, a naturally occurring anti-inflammatory and antioxidant compound found in essential oils, enhances osteoblast activity and inhibits osteoclastogenesis. Statins, known for their cholesterol-lowering effects, also promote bone formation and reduce bone resorption through multiple biochemical pathways. Both BCP and statins have shown promising results in preclinical studies, enhancing bone density and promoting fracture healing. This review explores the individual and potential synergistic effects of BCP and statins on bone fracture healing. It highlights the complementary mechanisms of these agents: BCP's anti-inflammatory and osteogenic properties and statins' ability to inhibit osteoclast activity and promote angiogenesis. Combining BCP and statins could offer a multifaceted approach to enhance fracture healing, reduce complications, and improve patient outcomes. While individual effects are supported preclinically, further studies investigating synergies, formulations, and clinical translation are needed to develop this promising novel therapeutic approach for improving fracture repair outcomes.
Collapse
Affiliation(s)
| | | | - Nissim Darvish
- Corporate Office, OrthoTreat Ltd, Tel Aviv-Jaffa, Israel
| | - Paul Tornetta III
- Department of Orthopaedic Surgery and Orthopaedic Trauma, Chobanian and Avedisian School of Medicine, Boston Medical Center, Boston, MA, USA
| | - Amal Khoury
- Division of Orthopaedic Surgery, Orthopaedic, and Reconstructive Trauma Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yoram A Weil
- Department of Orthopaedics, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | | | - Daniel C Allison
- Department of Orthopaedic Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sahar Meiron
- Corporate Office, OrthoTreat Ltd, Tel Aviv-Jaffa, Israel
| | | |
Collapse
|
30
|
Shtroblia V, Petakh P, Kamyshna I, Halabitska I, Kamyshnyi O. Recent advances in the management of knee osteoarthritis: a narrative review. Front Med (Lausanne) 2025; 12:1523027. [PMID: 39906596 PMCID: PMC11790583 DOI: 10.3389/fmed.2025.1523027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025] Open
Abstract
Knee osteoarthritis (OA) is a common condition that causes pain and reduces the quality of life for many people. It also leads to high health and financial costs. Managing knee OA pain requires using different methods together for the best results. This review overviews current therapeutic options for knee OA pain, focusing on their efficacy, safety, and potential roles in clinical practice. Topical treatments, such as NSAIDs and capsaicin, offer significant pain relief with minimal systemic side effects and are suitable for initial therapy, together with nonpharmacologic interventions like exercise and, when relevant, weight loss. Oral analgesics, including acetaminophen and opioids, have limited efficacy and serious side effects, making them appropriate only for short-term or rescue therapy. Intra-articular injections, such as corticosteroids, hyaluronic acid, and platelet rich plasma, demonstrate varying levels of efficacy and safety. Nutritional supplements, including curcumin, Boswellia serrata, and glucosaminechondroitin combinations, offer modest benefits and are best used as adjuncts to standart treatment. Nonpharmacological treatments, such as transcutaneous electrical nerve stimulation (TENS), acupuncture, and local heat therapy, provide variable pain relief and should be customized based on individual patient responses. Targeted biologic agents, such as antibodies to TNF-α, IL-1, and NGF, hold promise for more precise pain relief; however, further research is required to establish their routine use. Treating knee OA pain should be personalized, combining several methods. Research must continue to improve treatments and make them safer.
Collapse
Affiliation(s)
- Viktor Shtroblia
- Department of General Surgery, Uzhhorod National University, Uzhhorod, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
31
|
Shi Z, Han S. Personalized statin therapy: Targeting metabolic processes to modulate the therapeutic and adverse effects of statins. Heliyon 2025; 11:e41629. [PMID: 39866414 PMCID: PMC11761934 DOI: 10.1016/j.heliyon.2025.e41629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/28/2025] Open
Abstract
Statins are widely used for treating lipid disorders and cardiovascular diseases. However, the therapeutic efficiency and adverse effects of statins vary among different patients, which numerous clinical and epidemiological studies have attributed to genetic polymorphisms in statin-metabolizing enzymes and transport proteins. The metabolic processes of statins are relatively complex, involving spontaneous or enzyme-catalyzed interconversion between more toxic lactone metabolites and active acid forms in the liver and bloodstream, influenced by multiple factors, including the expression levels of many metabolic enzymes and transporters. Addressing the variable statin therapeutic outcomes is a pressing clinical challenge. Transcription factors and epigenetic modifications regulate the metabolic enzymes and transporters involved in statin metabolism and disposition and, therefore, hold promise as 'personalized' targets for achieving optimized statin therapy. In this review, we explore the potential for customizing therapy by targeting the metabolism of statin medications. The biochemical bases of adverse reactions to statin drugs and their correlation with polymorphisms in metabolic enzymes and transporters are summarized. Next, we mainly focus on the regulatory roles of transcription factors and epigenetic modifications in regulating the gene expression of statin biochemical machinery. The recommendations for future therapies are finally proposed by targeting the central regulatory factors of statin metabolism.
Collapse
Affiliation(s)
- Zhuangqi Shi
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, 830046, China
| | - Shuxin Han
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, 830046, China
| |
Collapse
|
32
|
Khan AW, Jandeleit-Dahm KAM. Atherosclerosis in diabetes mellitus: novel mechanisms and mechanism-based therapeutic approaches. Nat Rev Cardiol 2025:10.1038/s41569-024-01115-w. [PMID: 39805949 DOI: 10.1038/s41569-024-01115-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Atherosclerosis is a disease of large and medium arteries that can lead to life-threatening cardiovascular and cerebrovascular consequences, such as myocardial infarction and stroke. Moreover, atherosclerosis is a major contributor to cardiovascular-related mortality in individuals with diabetes mellitus. Diabetes aggravates the pathobiological mechanisms that underlie the development of atherosclerosis. Currently available anti-atherosclerotic drugs or strategies solely focus on optimal control of systemic risk factors, including hyperglycaemia and dyslipidaemia, but do not adequately target the diabetes-exacerbated mechanisms of atherosclerotic cardiovascular disease, highlighting the need for targeted, mechanism-based therapies. This Review focuses on emerging pathological mechanisms and related novel therapeutic targets in atherosclerotic cardiovascular disease in patients with diabetes.
Collapse
Affiliation(s)
- Abdul Waheed Khan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Karin A M Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- German Diabetes Centre, Leibniz Centre for Diabetes Research at the Heinrich Heine University, Dusseldorf, Germany
| |
Collapse
|
33
|
Seth B, Okello D, Saad Ullah S, Yousaf R, Alfalasi SB, Hafeez M, Rasool N, Bhullar G, Ian Gidley TN, Abdi SAH, Murtaza K. Role of Statins in Reducing Cardiovascular Mortality: A Systematic Review of Long-Term Outcomes. Cureus 2025; 17:e78137. [PMID: 40018472 PMCID: PMC11867218 DOI: 10.7759/cureus.78137] [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] [Accepted: 01/26/2025] [Indexed: 03/01/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of mortality worldwide, highlighting the critical need for effective preventive therapies. Statins, or HMG-CoA reductase inhibitors, are widely prescribed for their ability to lower low-density lipoprotein (LDL) cholesterol and reduce CV risk. This systematic review evaluates the long-term impact of statins on CV and all-cause mortality across diverse populations, including those with chronic kidney disease, chronic heart failure, and other comorbid conditions. A comprehensive search of major databases identified randomized controlled trials and large observational cohort studies with follow-up periods exceeding one year. Findings demonstrated significant reductions in CV mortality (hazard ratio (HR) range: 0.38-0.76) and all-cause mortality (HR range: 0.55-0.80) with statin therapy, particularly among high-risk groups, such as individuals with elevated LDL-C and moderate chronic kidney disease. Additional benefits were observed in preventing major adverse cardiovascular events (MACEs). Subgroup analyses revealed variations in efficacy based on age, sex, comorbidities, and statin type or dosage, with some populations, such as those with chronic heart failure and chronic obstructive pulmonary disease, showing limited benefit. Geographic and ethnic diversity were underrepresented in the included studies, and data on long-term effects in populations with advanced renal impairment or inflammatory conditions remain insufficient. These gaps underscore the need for methodologically robust studies and tailored approaches to statin therapy that account for individual patient profiles, including comorbidities and demographic factors. Practical steps include integrating statins with newer lipid-lowering agents and developing personalized treatment protocols to maximize their benefits and minimize risks. This review reinforces the critical role of statins in reducing the global burden of CVDs while emphasizing areas for future research.
Collapse
Affiliation(s)
- Bipasha Seth
- Intensive Care Unit, Niraj Intensive and Anesthesia Care Private Limited, Delhi, IND
| | - David Okello
- Internal Medicine, Ministry of Health, Lusaka, ZMB
| | - Syed Saad Ullah
- Pulmonology, Jinnah Postgraduate Medical Center, Karachi, PAK
| | - Rabia Yousaf
- Internal Medicine, Shifa College of Medicine, Islamabad, PAK
| | | | - Muhammad Hafeez
- Pharmacology, Quetta Institute of Medical Sciences, Quetta, PAK
| | - Naveed Rasool
- Internal Medicine, East and North Hertfordshire NHS Trust, London, GBR
| | - Gurman Bhullar
- Internal Medicine, Sri Guru Ram Das University of Health Sciences and Research, Amritsar, IND
| | | | | | - Khakan Murtaza
- Internal Medicine, Nishtar Medical University, Multan, PAK
| |
Collapse
|
34
|
Sastri KT, Gupta NV, Kannan A, Dutta S, Ali M Osmani R, V B, Ramkishan A, S S. The next frontier in multiple sclerosis therapies: Current advances and evolving targets. Eur J Pharmacol 2024; 985:177080. [PMID: 39491741 DOI: 10.1016/j.ejphar.2024.177080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Recent advancements in research have significantly enhanced our comprehension of the intricate immune components that contribute to multiple sclerosis (MS) pathogenesis. By conducting an in-depth analysis of complex molecular interactions involved in the immunological cascade of the disease, researchers have successfully identified novel therapeutic targets, leading to the development of innovative therapies. Leveraging pioneering technologies in proteomics, genomics, and the assessment of environmental factors has expedited our understanding of the vulnerability and impact of these factors on the progression of MS. Furthermore, these advances have facilitated the detection of significant biomarkers for evaluating disease activity. By integrating these findings, researchers can design novel molecules to identify new targets, paving the way for improved treatments and enhanced patient care. Our review presents recent discoveries regarding the pathogenesis of MS, highlights their genetic implications, and proposes an insightful approach for engaging with newer therapeutic targets in effectively managing this debilitating condition.
Collapse
Affiliation(s)
- K Trideva Sastri
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India.
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India.
| | - Anbarasu Kannan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - Balamuralidhara V
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - A Ramkishan
- Deputy Drugs Controller (India), Central Drugs Standard Control Organization, Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| | | |
Collapse
|
35
|
Mizuta H, Ishii M, Ikebe S, Otsuka Y, Yamanouchi Y, Nakamura T, Tsujita K. Triglycerides and the Risk of Atherosclerotic Cardiovascular Events Across Different Risk Categories. J Atheroscler Thromb 2024:65334. [PMID: 39675972 DOI: 10.5551/jat.65334] [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: 12/17/2024] Open
Abstract
AIMS To investigate the association between triglyceride levels and major adverse cardiovascular events (MACE) in primary and secondary prevention cohorts. METHODS This retrospective study was conducted with a nationwide health insurance claims database, which included approximately 3.8 million participants with medical checkups between January 2005 and August 2020 in Japan. The participants were classified into primary prevention (n=3,415,522) and secondary prevention (n=29,806) cohorts based on cardiovascular or cerebrovascular disease history. Each participant was categorized as having very low (triglyceride <50 mg/dL), low normal (50-99), high normal (100-149), or hypertriglyceridemia (≥ 150). The primary prevention cohort was further stratified into low-, intermediate-, and high-risk groups according to atherosclerotic cardiovascular diseases risk. Outcome was MACE, including acute myocardial infarction (AMI), unstable angina, ischemic stroke, and cardiac death. RESULTS Over a mean follow-up of 3.25 years, 0.3% and 2.6% MACE occurred in primary and secondary prevention, respectively. Hypertriglyceridemia was associated with high risk of MACE in the primary prevention, but not in the secondary prevention. A significant interaction was observed between prevention categories and the association of TG levels with MACE in those with TG <150 mg/dL and ischemic stroke in those with TG ≥ 150 mg/dL. The population-attributable fraction for hypertriglyceridemia in primary prevention was 4.1% for MACE. In primary prevention, lower risks of AMI were observed in the lower TG category compared to the current threshold. CONCLUSIONS This study suggests distinct triglyceride thresholds for MACE risk in primary and secondary prevention cohorts, requiring further prospective validation for clinical implementation.
Collapse
Affiliation(s)
- Hiroyuki Mizuta
- Tokushukai Isen Clinic
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
- Department of Medical Information Science, Graduate School of Medical Sciences, Kumamoto University
| | - So Ikebe
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | - Yasuhiro Otsuka
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
| | | | - Taishi Nakamura
- Department of Medical Information Science, Graduate School of Medical Sciences, Kumamoto University
- Department of Clinical Investigation, Kumamoto University Hospital
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University
- Department of Clinical Investigation, Kumamoto University Hospital
| |
Collapse
|
36
|
Hashemizadeh S, Alaee E, Aghajani N, Azizi H, Semnanian S. Atorvastatin facilitates extinction and prevents reinstatement of morphine-induced conditioned place preference in rats. Biomed Pharmacother 2024; 181:117639. [PMID: 39520913 DOI: 10.1016/j.biopha.2024.117639] [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: 08/10/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Opioid addiction is known as a chronic relapsing disorder associated with long-lasting molecular and cellular neuroadaptations that lead to compulsive behavior. Current pharmacotherapies target the modulation of mu-opioid receptors (MOR); however, the relapse rate remains high. In this study, we evaluated the potential effect of atorvastatin, a blood-brain barrier-permeable statin, on preventing morphine relapse through both extinction-reinstatement and abstinence-reinstatement models using conditioned place preference (CPP). Adult male Wistar rats were used to establish morphine-induced CPP (5 mg/kg), followed by extinction training and subsequent priming injection of morphine (2 mg/kg, i.p.) to induce relapse-like behavior. Extinguished rats significantly reinstated their morphine-seeking behavior. In contrast, rats that received different doses of atorvastatin (0.1, 0.5, 1 mg/kg) 1 hour before each extinction training session did not show a preference for the morphine-paired chamber. Moreover, acute atorvastatin injection (1 mg/kg, i.p.) 1 h before the reinstatement test significantly prevented reinstated morphine-seeking behavior. We found that atorvastatin 1 mg/kg attenuated morphine-seeking behaviors, and this attenuation of reinstatement was partly mediated by the upregulation of brain-derived neurotrophic factor (BDNF) in the prefrontal cortex (PFC) and hippocampus (Hipp). Furthermore, atorvastatin reversed Oprm1 upregulation (mu-opioid receptor gene) induced by relapse in the nucleus accumbens and Hipp. Moreover, treatment with atorvastatin during the extinction period alters the electrophysiological properties of the mPFC neurons following morphine priming and enhances neuronal excitability. We conclude that atorvastatin was effective in decreasing reinstatement.
Collapse
Affiliation(s)
- Shiva Hashemizadeh
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Elham Alaee
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Niloofar Aghajani
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran; Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Saeed Semnanian
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
37
|
Zhou S, Chen R, Liu J, Guo Z, Su L, Li Y, Zhang X, Luo F, Gao Q, Lin Y, Pang M, Cao L, Xu X, Nie S. Comparative Effectiveness and Safety of Atorvastatin Versus Rosuvastatin : A Multi-database Cohort Study. Ann Intern Med 2024; 177:1641-1651. [PMID: 39467290 DOI: 10.7326/m24-0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Rosuvastatin and atorvastatin are the most widely prescribed moderate- to high-intensity statins. However, evidence on their efficacy and safety during actual use is limited. OBJECTIVE To compare the real-world effectiveness and safety of rosuvastatin and atorvastatin. DESIGN Active comparator cohort study using target trial emulation. SETTING The China Renal Data System (CRDS) and UK Biobank (UKB) databases. PARTICIPANTS Adults newly prescribed rosuvastatin or atorvastatin. MEASUREMENTS The primary outcome was all-cause mortality. Cox proportional hazards regressions were used after 1:1 multilevel propensity score matching. RESULTS Among the 285 680 eligible participants in both databases, 6-year all-cause mortality was lower for rosuvastatin than for atorvastatin (2.57 vs. 2.83 per 100 person-years in the CRDS database and 0.66 vs. 0.90 per 100 person-years in the UKB database), with differences in cumulative incidence of -1.03% (95% CI, -1.44% to -0.46%) in the CRDS database and -1.38% (CI, -2.50% to -0.21%) in the UKB database. For secondary outcomes in both databases, rosuvastatin conferred lower risks for major adverse cardiovascular events and major adverse liver outcomes. In the UKB database, the risk for development of type 2 diabetes mellitus was higher with rosuvastatin, and the 2 medications carried similar risks for development of chronic kidney disease and other statin-related adverse effects. LIMITATION Possible residual confounding. CONCLUSION This study found differences in risks for some important outcomes associated with rosuvastatin and atorvastatin. The differences were relatively small, and many did not meet traditional standards for statistical significance. Further research is needed to understand whether these findings can be used with confidence in clinical practice. PRIMARY FUNDING SOURCE National Key R&D Program of China and National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Shiyu Zhou
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Ruixuan Chen
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Jiao Liu
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Zhixin Guo
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Licong Su
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Yanqin Li
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Xiaodong Zhang
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Fan Luo
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Qi Gao
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Yuxin Lin
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Mingzhen Pang
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Lisha Cao
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Xin Xu
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| | - Sheng Nie
- Division of Nephrology, National Clinical Research Center for Kidney Disease, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China (S.Z., R.C., J.L., Z.G., L.S., Y.Li, X.Z., F.L., Q.G., Y.Lin, M.P., L.C., X.X., S.N.)
| |
Collapse
|
38
|
Karamat M, Hussain B, Ahmed MM, Hamza M, Mir J, Alamri A, Shafiq A, Sattar Y, Khan MZ, Thyagaturu H, Gonuguntla K, Patel BD. Deciphering the cardioprotective effects of statins in anthracycline-related cardiac dysfunction: A systematic review and meta-analysis. Am J Prev Cardiol 2024; 20:100874. [PMID: 39524983 PMCID: PMC11543556 DOI: 10.1016/j.ajpc.2024.100874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 09/07/2024] [Accepted: 09/22/2024] [Indexed: 11/16/2024] Open
Abstract
Background Cancer induced chronic inflammation and cancer drugs effects on the vascular system can lead to rapidly progressing atherosclerotic burden. Statins drugs are known to reduce atherosclerotic plaque burden and inflammation. We studied outcomes of statins for anthracycline-related cardiac dysfunction (ARCD). Methods We conducted an online systematic search on PubMed and Embase to identify studies assessing statins' role in alleviating ARCD. We selected 9 studies that had patients with ARCD and use of statins. We primarily focused on outcomes including incidence of heart failure (HF), mean changes in left ventricular ejection fraction (LVEF), end-diastolic volume (LVEDV), and end-systolic volume (LVESV) from baseline. Odds ratios (OR) were calculated using a random effect model in R-statistics. Results Among 9 studies with a total of 2784 patients we noticed a significant reduction in the incidence of HF among patients administered statins, with an OR of 0.52 (95 % CI 0.37-0.74, p < 0.0003), indicating a substantial protective effect. However, the mean changes in EF, LVEDV, and LVESV from baseline, represented by Hedges's g of 1.09 (95 % CI -0.40 to 2.57, p = 0.15), 0.91 (95 % CI -1.69 to 3.51, p = 0.47), and 1.32 (95 % CI -2.30 to 4.94, p = 0.49) respectively, were not statistically significant. (Figure 1). Conclusion Our meta-analysis confirms statins' effectiveness in reducing risk of ARCD. However, their influence on EF, LVEDV, and LVESV remains uncertain, warranting further study.
Collapse
Affiliation(s)
- Mubashar Karamat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL, USA
| | - Bilal Hussain
- Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, USA
| | | | - Mohammad Hamza
- Department of Internal Medicine, Albany Medical Center, Albany, NY
| | - Junaid Mir
- Department of Internal Medicine, University of Missouri, Columbia, MO, USA
| | - Ayedh Alamri
- Department of Cardiovascular Department, Tufts Medical Center, Boston, USA
| | - Aimen Shafiq
- Department of Internal Medicine, Dow University of Health Sciences, Pakistan
| | - Yasar Sattar
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | - Muhammad Zia Khan
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| | | | | | - Birjesh D Patel
- Department of Cardiology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
39
|
Kobayashi K, Murakami K, Baba K. Effects of Lipophilic Statins on Cell Viability and Tissue Factor Expression in Canine Haemangiosarcoma Cells. Vet Comp Oncol 2024; 22:581-591. [PMID: 39319370 DOI: 10.1111/vco.13012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/18/2024] [Accepted: 09/06/2024] [Indexed: 09/26/2024]
Abstract
Canine haemangiosarcoma (HSA) is a highly aggressive cancer often associated with coagulation abnormalities. Statins, inhibitors of 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGCR) clinically prescribed for hypercholesterolemia, are also believed to possess antitumour and anticoagulant properties by inhibiting downstream Akt activation. Akt phosphorylation is involved in the mechanism of the antitumour and tissue factor (TF)-lowering effects of statins. In the present study, we aimed to investigate whether statins could inhibit cell viability while concurrently inducing anticoagulant properties by regulating the expression of TFs in canine HSA cells. Using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), we initially exclusively detected HMGCR mRNA expression in canine HSA tissues and cell lines but not in normal cephalic vein and spleen tissues. Moreover, treatment with lipophilic statins, including atorvastatin, fluvastatin, and simvastatin, inhibited cell viability in a concentration-dependent manner and decreased TF expression both at the mRNA and protein levels, as evidenced by cell viability assays, RT-qPCR, and immunoblotting, respectively. Further investigation using cell viability assays and flow cytometry revealed that simvastatin decreased Akt phosphorylation, and MK-2206, a specific Akt inhibitor, mirrored the effect of simvastatin on cell viability and cell cycle arrest. However, MK-2206 exhibited different effects on TF expression depending on the cell type, indicating that Akt phosphorylation may not consistently regulate TF expression. Overall, this study provides insights into the potential therapeutic use of statins in targeting tumour growth and coagulation abnormalities in canine HSA. Further research is warranted to fully elucidate the underlying mechanisms and clinical applications of statins in canine HSA treatment.
Collapse
Affiliation(s)
- Kosuke Kobayashi
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Kohei Murakami
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Kenji Baba
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
40
|
Roque SM, Furian AC, Takemoto MK, Duarte MCT, Parolina RD, Roque AL, Duran N, Sardi JDCO, Duarte RMT, Muller KC. Biosynthesis and Characterization of Silver Nanoparticles and Simvastatin Association in Titanium Biofilms. Pharmaceuticals (Basel) 2024; 17:1612. [PMID: 39770455 PMCID: PMC11678683 DOI: 10.3390/ph17121612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Simvastatin is an antilipidemic drug that has already demonstrated antibacterial activities on oral and non-oral microorganisms. Silver nanoparticles also exhibit antimicrobial properties, particularly for coating implant surfaces. In this study, we evaluated the effects of combining simvastatin with silver nanoparticles on the formation and viability of biofilms consolidated on titanium discs. METHODS Silver nanoparticles were first biosynthesized using the fungus Fusarium oxysporum and then characterized using Dynamic Light Scattering, X-ray Diffraction, Transmission Electron Microscopy, and energy dispersive spectroscopy. Species of Streptococcus oralis, Streptococcus mutans, Porphyromonas gingivalis, Methicillin-sensitive Staphylococcus aureus, and Methicillin-resistant Staphylococcus aureus were used and tested using Minimum Inhibitory Concentration assays with concentrations of silver nanoparticles and simvastatin alone and in combination. Biofilm inhibition and viability tests were performed on titanium surfaces. Toxicity tests were also performed on Galleria mellonella moth larvae. RESULTS The silver nanoparticles had a spherical shape without the formation of aggregates as confirmed by Transmission Electron Microscopy. Dynamic Light Scattering revealed nanoparticles with an average diameter of 53.8 nm (±1.23 nm), a polydispersity index of 0.23 and a zeta potential of -25 mV (±2.19 mV). The silver nanoparticles inhibited the growth of the strains tested in the range of 0.001592 and 63.75, while simvastatin alone inhibited the growth of the same strains in the range of 3.125-62.5 µg/mL. The antibacterial activity test of the combination of the two substances showed a reduction in the Minimum Inhibitory Concentration of about two to eight times, showing synergistic effects on Staphylococcus aureus and additive effects on Streptococcus oralis and Porphyromonas gingivalis. As for biofilm, sub-inhibitory concentrations of the combination of substances showed better antibacterial activity in inhibiting the formation of Streptococcus oralis biofilm, and this combination also proved effective in eradicating already established biofilms compared to the substances alone. The combination of silver nanoparticles and simvastatin showed low toxicity to Galleria mellonella moth larvae. CONCLUSIONS The results presented indicate that the combination of the two substances could be an alternative for the prevention and reduction of biofilms on implants. These findings open up new possibilities in the search for alternatives for the treatment of peri-implant infections, as well as the possibility of using lower doses compared to single drugs, achieving the same results and reducing potential toxic effects.
Collapse
Affiliation(s)
- Sindy Magri Roque
- Laboratório de Farmacologia de Antimicrobianos e Microbiologia, Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, Brazil; (S.M.R.); (A.C.F.)
- Departamento de Biociências, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba 13414-903, Brazil; (M.K.T.); (M.C.T.D.); (R.D.P.)
| | - Ana Carolina Furian
- Laboratório de Farmacologia de Antimicrobianos e Microbiologia, Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, Brazil; (S.M.R.); (A.C.F.)
| | - Marcela Kim Takemoto
- Departamento de Biociências, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba 13414-903, Brazil; (M.K.T.); (M.C.T.D.); (R.D.P.)
| | - Marta Cristina Teixeira Duarte
- Departamento de Biociências, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba 13414-903, Brazil; (M.K.T.); (M.C.T.D.); (R.D.P.)
- Centro Pluridisciplinar de Pesquisas Químicas, Biológicas e Agrícolas—CPQBA, Universidade Estadual de Campinas (UNICAMP), Paulínia 13148-218, Brazil;
| | - Rafaela Durrer Parolina
- Departamento de Biociências, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba 13414-903, Brazil; (M.K.T.); (M.C.T.D.); (R.D.P.)
| | - Adriano Luís Roque
- Programa de Pós Graduação em Medicina (Cardiologia), Universidade Federal de São Paulo (UNIFESP), São Paulo 04021-001, Brazil;
| | - Nelson Duran
- Laboratório de Carcinogenese Urogenital e Imunoterapia, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-862, Brazil;
| | | | - Renata Maria Teixeira Duarte
- Centro Pluridisciplinar de Pesquisas Químicas, Biológicas e Agrícolas—CPQBA, Universidade Estadual de Campinas (UNICAMP), Paulínia 13148-218, Brazil;
| | - Karina Cogo Muller
- Laboratório de Farmacologia de Antimicrobianos e Microbiologia, Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas (UNICAMP), Campinas 13083-970, Brazil; (S.M.R.); (A.C.F.)
- Departamento de Biociências, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas (UNICAMP), Piracicaba 13414-903, Brazil; (M.K.T.); (M.C.T.D.); (R.D.P.)
| |
Collapse
|
41
|
Oh S, Kim JH, Cho KH, Kim MC, Sim DS, Hong YJ, Lee SW, Ahn Y, Jeong MH. Differential statin intensity and outcomes in patients following myocardial infarction with very low low-density lipoprotein cholesterol. Cardiol J 2024; 31:802-813. [PMID: 39564956 PMCID: PMC11706264 DOI: 10.5603/cj.99136] [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: 01/27/2024] [Revised: 10/05/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Despite increasing evidence on the benefits of statin therapy for acute myocardial infarction (AMI), differential outcomes in accordance with statin intensity have not been evaluated in patients with AMI and low-density lipoprotein cholesterol (LDL-C) levels < 55 mg/dL. Therefore, this study aimed to compare the clinical outcomes of high- and moderate-intensity statin therapy in this population. METHODS A total of 752 participants with AMI and LDL-C levels < 55 mg/dL from a Korean nationwide multicenter observational cohort (2016-2020) were included and categorized into two groups: high-intensity statin group (n = 384) and moderate-intensity statin group (n = 368). The primary outcome was 1-year major adverse cardiac and cerebrovascular events (MACCEs). Propensity score matching (PSM) and Cox models were used to determine whether statin intensity independently influenced the primary outcome. RESULTS Compared to the moderate-intensity statin group, the high-intensity statin group had a comparable risk of MACCE in all Cox models and PSM-adjusted analyses. The cumulative incidence of MACCE was comparable between the two groups. CONCLUSIONS Statin intensity appeared to have no significant impact on clinical outcomes in AMI patients with LDL-C levels < 55 mg/dL. These results underscore the need for further investigations aimed at refining treatment strategies for this specific patient cohort, potentially reducing treatment-related burdens without compromising clinical effectiveness.
Collapse
Affiliation(s)
- Seok Oh
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Ju Han Kim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea.
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea.
| | - Kyung Hoon Cho
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| | - Min Chul Kim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| | - Doo Sun Sim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| | - Young Joon Hong
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| | - Seung-Won Lee
- Department of Anatomy, Chonnam National University Medical School, Gwangju, Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| | - Myung Ho Jeong
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- Department of Cardiology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
42
|
Menendez-Gonzalez M. Therapeutic Challenges Derived from the Interaction Among Apolipoprotein E, Cholesterol, and Amyloid in Alzheimer's Disease. Int J Mol Sci 2024; 25:12029. [PMID: 39596098 PMCID: PMC11593474 DOI: 10.3390/ijms252212029] [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: 10/19/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The isoform E4 of the Apolipoprotein E (ApoE) represents one of the strongest genetic risk factors for late-onset Alzheimer's disease (AD). ApoE has key roles in cholesterol transport and amyloid-β (Aβ) metabolism, which are both central to AD pathogenesis. The E4 isoform has been implicated in reduced cholesterol homeostasis, increased Aβ aggregation, and heightened tau phosphorylation, contributing to amyloid plaques and neurodegeneration. This manuscript examines the complex interactions among ApoE isoforms, cholesterol metabolism, and amyloid pathology. Moreover, the therapeutic challenges associated with lipid-lowering agents (e.g., statins, PCSK9 inhibitors), anti-amyloid immunotherapies, and anticoagulants are described, focusing on ApoE4 carriers. Decision-making challenges are discussed by analyzing the pros and cons of these therapies.
Collapse
Affiliation(s)
- Manuel Menendez-Gonzalez
- Departamento de Medicina, Facultad de Ciencias de la Salud, Universidad de Oviedo, ES-33006 Oviedo, Spain;
- Servicio de Neurología, Hospital Universitario Central de Asturias, ES-33011 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), ES-33011 Oviedo, Spain
| |
Collapse
|
43
|
Cheng EY, Mirzaei A. Potential molecular targets for the pharmacologic management of non-traumatic osteonecrosis. Expert Opin Ther Targets 2024; 28:991-1000. [PMID: 39469902 DOI: 10.1080/14728222.2024.2421755] [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: 08/11/2024] [Accepted: 10/23/2024] [Indexed: 10/30/2024]
Abstract
INTRODUCTION Non-traumatic osteonecrosis is a debilitating condition marked by bone death, primarily due to reduced blood supply. Currently, no effective pharmacologic intervention is available to manage this condition effectively. AREAS COVERED Lipid metabolic disorders, chronic inflammation, vascular dysfunction, coagulopathy, and impaired bone homeostasis are suggested as the key pathogenic mechanisms involved in the development of non-traumatic osteonecrosis. Targeting any of these dysfunctions offers a potential avenue for pharmacologic intervention. However, the potential molecular targets for pharmacologic treatment of non-traumatic osteonecrosis remain underexplored. In this study, we reviewed available databases to compile a comprehensive set of pathogenic mechanisms and corresponding therapeutic targets for non-traumatic osteonecrosis. EXPERT OPINION Evidence suggests that a single pathogenic mechanism cannot fully explain the development of osteonecrosis, supporting the adoption of a multi-pathogenic theory. This theory implies that effective management of non-traumatic osteonecrosis requires targeting multiple pathogenic mechanisms simultaneously. Moreover, the same pathogenic mechanisms are unlikely to explain osteonecrosis development in patients with different etiologies. Consequently, a one-size-fits-all approach to medication is unlikely to be effective across all types of non-traumatic osteonecrosis. Future research should, therefore, focus on developing multi-target pharmacologic treatments tailored to the specific etiology of non-traumatic osteonecrosis.
Collapse
Affiliation(s)
- Edward Y Cheng
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Alireza Mirzaei
- Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
44
|
Paciullo F, Falcinelli E, Fierro T, Gresele P, Del Pinto M. High - but not standard-dose atorvastatin prevents the increase of plasma matrix metalloproteinase-2 triggered by acute coronary syndromes. Coron Artery Dis 2024; 35:622-624. [PMID: 39087641 PMCID: PMC11426972 DOI: 10.1097/mca.0000000000001414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/13/2024] [Indexed: 08/02/2024]
Affiliation(s)
- Francesco Paciullo
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia
| | - Emanuela Falcinelli
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia
| | - Tiziana Fierro
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia
| | - Paolo Gresele
- Section of Internal and Cardiovascular Medicine, Department of Medicine and Surgery, University of Perugia
| | | |
Collapse
|
45
|
Lee Y, Hung M, Chen T, Mao C, Yeh C, Kounis NG, Chen IY, Hu P, Hung M. Effects of statins in patients with coronary artery spasm: A nationwide population-based study. Clin Transl Sci 2024; 17:e70087. [PMID: 39568301 PMCID: PMC11579374 DOI: 10.1111/cts.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Controversies regarding the benefits of statin treatment on clinical outcomes in coronary artery spasm (CAS) without obstructive coronary artery disease (CAD) persist due to limited data. In this retrospective nationwide population-based cohort study from the Taiwan National Health Insurance Research Database during the period 2000-2012, the matched cohorts consisted of 12,000 patients with CAS. After propensity score matching with 1:1 ratio, 2216 patients were eligible for outcome analysis in either statin or nonstatin group, with the mean follow-up duration of 4.8 and 4.6 years, respectively. Statin users versus nonusers had a significantly reduced risk of major adverse cardiovascular events (MACEs) (6.7% vs. 9.5%, hazard ratio [HR] 0.68; 95% confidence interval [CI] 0.55-0.84) and all-cause mortality (6.0% vs. 7.6%; HR 0.77; 95% CI 0.61-0.96). While the results of MACEs were mainly contributed by cardiovascular death (1.9% vs. 3.2%; HR 0.56; 95% CI 0.38-0.83) and ischemic stroke (3.8% vs. 5.4%; subdistribution HR 0.69; 95% CI 0.52-0.91), they were primarily driven by reductions in ischemic but not hemorrhagic stroke. The benefit of statins was significantly pronounced in patients with hypertension and diabetes. Nevertheless, the effect on MACEs was consistent irrespective of age, sex, dyslipidemia, and mental disorder. Statins significantly reduced the risk of MACEs and all-cause mortality in CAS patients. The benefit of statin therapy in reducing MACEs appeared to be linear, with greater risk reduction with higher doses and longer duration without upper threshold, reflecting the dose-dependent relationship of statins with MACEs in CAS patients.
Collapse
Affiliation(s)
- Yu‐Ching Lee
- TMU Research Center of Cancer Translational MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Ming‐Jui Hung
- Division of Cardiology, Department of Medicine and Community Medicine Research Center, Chang Gung Memorial Hospital, KeelungChang Gung University College of MedicineKeelung CityTaiwan
| | - Tien‐Hsing Chen
- Division of Cardiology, Department of Medicine and Community Medicine Research Center, Chang Gung Memorial Hospital, KeelungChang Gung University College of MedicineKeelung CityTaiwan
| | - Chun‐Tai Mao
- Division of Cardiology, Department of Medicine and Community Medicine Research Center, Chang Gung Memorial Hospital, KeelungChang Gung University College of MedicineKeelung CityTaiwan
| | - Chi‐Tai Yeh
- Department of Medical Research and Education, Shuang Ho HospitalTaipei Medical UniversityNew Taipei CityTaiwan
- Department of Medical Laboratory Science and BiotechnologyYuanpei University of Medical TechnologyHsinchu CityTaiwan
| | | | - Ian Y. Chen
- Division of Cardiovascular Medicine, Department of Medicine, Department of Radiology, Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCaliforniaUSA
- Cardiology Section, Medical Service, Veterans Affairs Palo Alto Health Care SystemPalo AltoCaliforniaUSA
| | - Patrick Hu
- University of California, RiversideRiversideCaliforniaUSA
- Department of CardiologyRiverside Medical ClinicRiversideCaliforniaUSA
| | - Ming‐Yow Hung
- Division of Cardiology, Department of Internal Medicine, Shuang Ho HospitalTaipei Medical UniversityNew Taipei CityTaiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Taipei Heart InstituteTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
46
|
de la Riva P, Marta-Enguita J, Rodríguez-Antigüedad J, Bergareche A, de Munain AL. Understanding Endothelial Dysfunction and Its Role in Ischemic Stroke After the Outbreak of Recanalization Therapies. Int J Mol Sci 2024; 25:11631. [PMID: 39519182 PMCID: PMC11546609 DOI: 10.3390/ijms252111631] [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: 08/22/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Despite recent advances in treatment options, stroke remains a highly prevalent and devastating condition with significant socioeconomic impact. Recanalization therapies, including intravenous thrombolysis and endovascular treatments, have revolutionized stroke management and prognosis, providing a promising framework for exploring new therapeutic strategies. Endothelial dysfunction plays a critical role in the pathophysiology, progression, and prognosis of stroke. This review aims to synthesize the current evidence regarding the involvement of the nitric oxide (NO)/endothelium pathway in ischemic stroke, with a particular focus on aging, response to recanalization therapies, and therapeutic approaches. While significant progress has been made in recent years in understanding the relationship between endothelial dysfunction and stroke, many uncertainties persist, and although treatments targeting this pathway are promising, they have yet to demonstrate clear clinical benefits.
Collapse
Affiliation(s)
- Patricia de la Riva
- Department of Neurology, Donostia University Hospital, Dr Begiristain sn., 20003 San Sebastian, Spain; (P.d.l.R.); (A.B.); (A.L.d.M.)
- Ictus, Biogipuzkoa Institute, Doctor Begiristain sn., 20003 San Sebastian, Spain
- Facultad de Ciencias de la Salud, Deusto University, Mundaiz 50, 20003 San Sebastian, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS)-Ictus, Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Juan Marta-Enguita
- Department of Neurology, Donostia University Hospital, Dr Begiristain sn., 20003 San Sebastian, Spain; (P.d.l.R.); (A.B.); (A.L.d.M.)
- Ictus, Biogipuzkoa Institute, Doctor Begiristain sn., 20003 San Sebastian, Spain
- Facultad de Ciencias de la Salud, Deusto University, Mundaiz 50, 20003 San Sebastian, Spain
- Red de Investigación Cooperativa Orientada a Resultados en Salud (RICORS)-Ictus, Instituto Salud Carlos III, 28029 Madrid, Spain
| | - Jon Rodríguez-Antigüedad
- Movement Disorders Unit and Institut d’Investigacions Biomediques-Sant Pau, Hospital Sant Pau, 08025 Barcelona, Spain;
| | - Alberto Bergareche
- Department of Neurology, Donostia University Hospital, Dr Begiristain sn., 20003 San Sebastian, Spain; (P.d.l.R.); (A.B.); (A.L.d.M.)
| | - Adolfo López de Munain
- Department of Neurology, Donostia University Hospital, Dr Begiristain sn., 20003 San Sebastian, Spain; (P.d.l.R.); (A.B.); (A.L.d.M.)
- Ictus, Biogipuzkoa Institute, Doctor Begiristain sn., 20003 San Sebastian, Spain
| |
Collapse
|
47
|
Kanda M, Kumasawa K, Nemoto K, Miyatake R, Inaba K, Sayama S, Seyama T, Iriyama T, Nagamatsu T, Fujii T, Hirota Y, Osuga Y, Kimura T. The Effects of Low Concentrations of Pravastatin on Placental Cells. Reprod Sci 2024; 31:3139-3147. [PMID: 38836966 PMCID: PMC11438613 DOI: 10.1007/s43032-024-01611-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Pravastatin is a promising medication to treat preeclampsia. However, the appropriate dose of pravastatin for managing preeclampsia has not been established. In this in vitro study, we examined the effects of low concentrations of pravastatin (0.01 to 10 µM) under hypoxic conditions on two types of placental cells and found that pravastatin decreased sFlt-1 levels up to 34% in cytotrophoblast cells isolated from human term placentas. Furthermore, we showed that sFlt-1 levels in HTR-8/SVneo cells, a cell line derived from first trimester trophoblast cells, decreased after exposure to very low concentrations of pravastatin (0.01, 0.1 µM). We also examined the effects of pravastatin on uterine spiral artery remodeling-related events and showed in wound healing and tube formation assays that low concentrations of pravastatin upregulated cell migration and invasion in HTR-8/SVneo cells. These results demonstrated that a low dose of pravastatin has in vitro effects that suggest a potential for anti-preeclamptic effects in vivo.
Collapse
Affiliation(s)
- Masako Kanda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Keiichi Kumasawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Kazunari Nemoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Risa Miyatake
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kei Inaba
- Department of Obstetrics and Gynecology, Tokyo Metropolitan Toshima Hospital of the Tokyo Metropolitan Hospital Organization, Itabashi-ku, Tokyo, Japan
| | - Seisuke Sayama
- Department of Obstetrics and Gynecology, Tokyo-Kita Medical Center, Kita-ku, Tokyo, Japan
| | - Takahiro Seyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Sanno Hospital, Minato-ku, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
48
|
Scaletti ER, Unterlass JE, Almlöf I, Koolmeister T, Vallin KS, Kapsitidou D, Tsuber V, Helleday T, Stenmark P, Jemth AS. Kinetic and structural characterization of NUDT15 and NUDT18 as catalysts of isoprene pyrophosphate hydrolysis. FEBS J 2024; 291:4301-4322. [PMID: 38944687 DOI: 10.1111/febs.17202] [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: 01/12/2024] [Revised: 04/19/2024] [Accepted: 06/06/2024] [Indexed: 07/01/2024]
Abstract
Isoprene pyrophosphates play a crucial role in the synthesis of a diverse array of essential nonsterol and sterol biomolecules and serve as substrates for posttranslational isoprenylation of proteins, enabling specific anchoring to cellular membranes. Hydrolysis of isoprene pyrophosphates would be a means to modulate their levels, downstream products, and protein isoprenylation. While NUDIX hydrolases from plants have been described to catalyze the hydrolysis of isoprene pyrophosphates, homologous enzymes with this function in animals have not yet been reported. In this study, we screened an extensive panel of human NUDIX hydrolases for activity in hydrolyzing isoprene pyrophosphates. We found that human nucleotide triphosphate diphosphatase NUDT15 and 8-oxo-dGDP phosphatase NUDT18 efficiently catalyze the hydrolysis of several physiologically relevant isoprene pyrophosphates. Notably, we demonstrate that geranyl pyrophosphate is an excellent substrate for NUDT18, with a catalytic efficiency of 2.1 × 105 m-1·s-1, thus making it the best substrate identified for NUDT18 to date. Similarly, geranyl pyrophosphate proved to be the best isoprene pyrophosphate substrate for NUDT15, with a catalytic efficiency of 4.0 × 104 M-1·s-1. LC-MS analysis of NUDT15 and NUDT18 catalyzed isoprene pyrophosphate hydrolysis revealed the generation of the corresponding monophosphates and inorganic phosphate. Furthermore, we solved the crystal structure of NUDT15 in complex with the hydrolysis product geranyl phosphate at a resolution of 1.70 Å. This structure revealed that the active site nicely accommodates the hydrophobic isoprenoid moiety and helped identify key binding residues. Our findings imply that isoprene pyrophosphates are endogenous substrates of NUDT15 and NUDT18, suggesting they are involved in animal isoprene pyrophosphate metabolism.
Collapse
Affiliation(s)
- Emma R Scaletti
- Department of Biochemistry and Biophysics, Stockholm University, Sweden
| | - Judith E Unterlass
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Ingrid Almlöf
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Tobias Koolmeister
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Karl S Vallin
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Despina Kapsitidou
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Viktoriia Tsuber
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Thomas Helleday
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, Sweden
| | - Ann-Sofie Jemth
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Stockholm, 171 77, Sweden
| |
Collapse
|
49
|
Yang J, Lee KB, Kim H, Kim SW, Kim YH, Sung SA, Kim J, Oh KH, Jung JY, Hyun YY. Statin Use and the Progression of Coronary Artery Calcification in CKD: Findings From the KNOW-CKD Study. Kidney Int Rep 2024; 9:3027-3034. [PMID: 39430188 PMCID: PMC11489501 DOI: 10.1016/j.ekir.2024.07.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 10/22/2024] Open
Abstract
Introduction Statin treatment can reduce the risk of cardiovascular disease (CVD). Paradoxically, previous studies have shown that the use of statin is associated with the progression coronary artery calcification (CAC), a well-known predictor of CVD, in individuals with preserved renal function or in patients on dialysis. However, little is known about the association in patients with predialysis chronic kidney disease (CKD). The aim of this study was to characterize the relationship between statin use and progression of CAC in a CKD cohort of Korean adults. Methods We analyzed 1177 participants registered in the Korean Cohort Study for Outcome in Patients with Chronic Kidney Disease (KNOW-CKD) cohort. Coronary artery calcium score (CACS) was assessed using cardiac computed tomography at baseline and 4 years after enrollment. CAC progression was defined using the Sevrukov method. Statin users were defined as those who used statins for 50% or more of the follow-up period. Results The median (interquartile range) of CACS was 0 (0-30.33), and 318 (44.2%) participants had CACS above 0 at baseline. There were 447 (38.0%) statin users and 730 (62.0%) statin nonusers. After 4 years, 374 patients (52.0%) demonstrated CAC progression, which was significantly more frequent in statin users than in statin nonusers (218 [58.3%] vs. 156 [41.7%], P < 0.001). The multivariate-adjusted odds ratio for CAC progression in statin users compared to statin nonusers was 1.78 (1.26-2.50). Conclusion Statin use, significantly and independently, is associated with CAC progression in Korean patients with predialysis CKD. Further research is warranted to verify the prognosis of statin-related CAC progression.
Collapse
Affiliation(s)
- Jihyun Yang
- Division of Nephrology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyu-Beck Lee
- Division of Nephrology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyang Kim
- Division of Nephrology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yeong Hoon Kim
- Department of Internal Medicine, Inje University Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Su Ah Sung
- Department of Internal Medicine, Nowon Eulji Medical Center, Eulji University, Seoul, Republic of Korea
| | - Jayoun Kim
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Yong Jung
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Young Youl Hyun
- Division of Nephrology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
50
|
Arachchillage DJ, Platton S, Hickey K, Chu J, Pickering M, Sommerville P, MacCallum P, Breen K. Guidelines on the investigation and management of antiphospholipid syndrome. Br J Haematol 2024; 205:855-880. [PMID: 39031476 DOI: 10.1111/bjh.19635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 06/26/2024] [Indexed: 07/22/2024]
Affiliation(s)
- Deepa J Arachchillage
- Department of Haematology, Imperial College Healthcare NHS Trust, London, UK
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Sean Platton
- The Royal London Hospital Haemophilia Centre, Barts Health NHS Trust, London, UK
| | - Kieron Hickey
- Sheffield Laboratory Medicine, Department of Coagulation, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Justin Chu
- Department of Obstetrics and Gynaecology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Matthew Pickering
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
- Department of Rheumatology, Imperial College Healthcare NHS Trust, London, UK
| | - Peter Sommerville
- Department of Stroke Medicine, Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Peter MacCallum
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Clinical Haematology, Barts Health NHS Trust, London, UK
| | - Karen Breen
- Department of Haematology, Guy's & St. Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|