1
|
Zhang Z, Zheng H, Zhang L, Su P, Chen X, Xiang A, Yang J, Guan H, Fan J, Yu Q. Atorvastatin enhances LDL receptor-mediated LDL-C uptake and modulates PCSK9 protein expression in pancreatic β-cells. Islets 2025; 17:2479906. [PMID: 40090035 PMCID: PMC11913382 DOI: 10.1080/19382014.2025.2479906] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/28/2025] [Accepted: 03/10/2025] [Indexed: 03/18/2025] Open
Abstract
Statins are widely used to treat hyperlipidemia and atherosclerotic cardiovascular diseases (ACVD) by significantly lowering low-density lipoprotein cholesterol (LDL-C) levels. However, their use has been associated with an increased risk of type 2 diabetes (T2D), a paradox given their lipid-lowering benefits. This study investigates the role of LDL receptors (LDLR) and proprotein convertase subtilisin/kexin type 9 (PCSK9) in the diabetogenic effects of atorvastatin on pancreatic β-cells. Using the MIN6 pancreatic β-cell line, we assessed the impact of atorvastatin on LDL-C uptake, PCSK9 expression, glucose-stimulated insulin release (GSIR), and cell proliferation. Cellular cholesterol assays, EdU labeling, Dil-LDL uptake, western blot analysis, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and ELISA, were employed to measure relevant biomarkers and cellular responses. Male C57BL/6j mice were treated with atorvastatin to validate in vitro findings. Atorvastatin enhances LDL-C uptake by upregulating LDLR on the cell surface, without causing excess cholesterol accumulation. Additionally, atorvastatin suppresses PCSK9 expression, which is crucial for LDLR degradation. Interestingly, atorvastatin, combined with exogenous LDL-C, impairs glucose-stimulated insulin release (GSIR) but promotes cell proliferation, highlighting a potential mechanism for statin-associated diabetes. Oral administration of atorvastatin in mice reduced plasma PCSK9 and insulin levels, supporting the in vitro findings. These results indicate that while atorvastatin effectively lowers circulating cholesterol, it may adversely affect pancreatic β-cell function by modulating LDLR and LDL-C uptake, thereby increasing the risk of T2D. This study highlights the importance of further research to develop strategies mitigating the diabetogenic effects of statins while maintaining their cardiovascular benefits.
Collapse
Affiliation(s)
- Zhiyan Zhang
- Engineering Research Center of Brain Health Industry of Chinese Medicine, Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang, China
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Huadong Zheng
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
- Department of Gerontology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lusha Zhang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Peihong Su
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Xiaochang Chen
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Aoqi Xiang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Juan Yang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Hua Guan
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Jianglin Fan
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Qi Yu
- Engineering Research Center of Brain Health Industry of Chinese Medicine, Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine of Shaanxi Administration of Traditional Chinese Medicine, Department of Pharmacology, Shaanxi University of Chinese Medicine, Xianyang, China
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases and Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| |
Collapse
|
2
|
Tramontano D, Bini S, Maiorca C, Di Costanzo A, Carosi M, Castellese J, Arizaj I, Commodari D, Covino S, Sansone G, Minicocci I, Arca M, D'Erasmo L. Renal Safety Assessment of Lipid-Lowering Drugs: Between Old Certainties and New Questions. Drugs 2025; 85:755-775. [PMID: 40106181 PMCID: PMC12098426 DOI: 10.1007/s40265-025-02158-0] [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] [Accepted: 02/09/2025] [Indexed: 03/22/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in patients with chronic kidney disease (CKD). Quantitative and qualitative changes in plasma lipoprotein profiles are frequently associated with CKD and represent a significant risk factor for CVD in patients with CKD. Guidelines from the European Society of Cardiology and the European Atherosclerosis Society classify CKD as a condition with high or very high cardiovascular risk and set specific low-density lipoprotein cholesterol targets. Conventional lipid-lowering therapies (LLTs), such as statins, ezetimibe, and fibrates, can control CKD-associated dyslipidemia and, to some extent, prevent major atherosclerotic events in patients with CKD, but their use in clinical practice presents challenges because of the potential renal safety concerns. In recent years, novel therapies with the ability to lower both low-density lipoprotein cholesterol and triglycerides have been introduced to the market (e.g., proprotein convertase subtilisin/kexin type 9 inhibitors, bempedoic acid, lomitapide, volanesorsen) to improve our ability to control lipid abnormalities. However, their impact on kidney functionality has not been fully elucidated. The aim of this review was to examine the renal safety profiles of various LLTs, with special reference to novel medications, and to highlight important considerations and guidance for the use of these medications in overt CKD or in patients with some degree of renal function impairment. We underscore the lack of a comprehensive understanding of kidney safety, particularly for novel LLT therapies, and strongly emphasize the importance of future dedicated research to fully assess the safety and efficacy of these agents in patients with kidney abnormalities.
Collapse
Affiliation(s)
- Daniele Tramontano
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| | - Simone Bini
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Carlo Maiorca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessia Di Costanzo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Carosi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Jacopo Castellese
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ina Arizaj
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Commodari
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Stella Covino
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giorgia Sansone
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ilenia Minicocci
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marcello Arca
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Laura D'Erasmo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
3
|
Peng H, Ren J, Zhao Y, Fang X, Wang X, Liu C, Wan Z. Unraveling the Connection between PCOS and renal Complications: Current insights and Future Directions. Diabetes Res Clin Pract 2025; 224:112235. [PMID: 40334925 DOI: 10.1016/j.diabres.2025.112235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/09/2025]
Abstract
Polycystic ovary syndrome (PCOS) represents the most prevalent endocrine disorder among women of reproductive age, affecting approximately 5-18% of females worldwide. Characterized by irregular ovulation, hyperandrogenism, and polycystic ovaries, hyperandrogenism is the defining feature. Recent evidence highlights that, in addition to its notable reproductive and metabolic consequences, PCOS may also contribute to an elevated risk of renal complications. This increased risk is attributed to chronic low-grade inflammation, hormonal dysregulation, and disturbances in lipid metabolism inherent to the condition. However, the pathological mechanisms, clinical manifestations, and progression of secondary renal damage in this cohort remain insufficiently studied. This review consolidates current understanding of the relationship between PCOS and chronic kidney disease (CKD), aiming to clarify potential mechanisms by which PCOS may induce secondary renal dysfunction, encompassing both direct renal impairment and indirect damage mediated through systemic alterations. Furthermore, it advocates for comprehensive management strategies to mitigate renal risks in patients with PCOS, emphasizing the necessity of multidisciplinary approaches and further research to address these critical gaps.
Collapse
Affiliation(s)
- Haoyu Peng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Junyi Ren
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Zhao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinyi Fang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiao Wang
- Department of Organ Transplantation, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chi Liu
- Department of Nephrology, Sichuan Clinical Research Center for Kidney Disease, Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, China.
| | - Zhengwei Wan
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
4
|
Mohammad FK, Al-Shalchi RF. Mini meta-analysis of anticholinesterase actions of atorvastatin, simvastatin and rosuvastatin, and in silico identification of their protein targets in Mus musculus. Toxicol Rep 2025; 14:101958. [PMID: 40026475 PMCID: PMC11869536 DOI: 10.1016/j.toxrep.2025.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
Dyslipidemic statins reduce blood and brain cholinesterase (ChE) activities in mice, with scarce information on other protein/enzyme targets. The study aims at conducting a mini meta-analysis on in vivo and in vitro adverse anti-ChE effects of atorvastatin, simvastatin and rosuvastatin in mice, and using the SwissPrediction to identify in silico body target proteins. The data comprised 72 records of plasma, erythrocytes and brain ChE activities, expressed as percent mean ± SD of respective controls. We conducted a randomized effects size single-arm meta-analysis. The risk of bias scoring was according to those of animal experiments. The effect size (% ChE activity) of statin treatments was significantly decreased by 25.85 % (combined effect size=74.15, p = 0.0001), with significant heterogeneity (Q=1133.19, p < 0.0001, I2=93.73 %). Subgroup analysis was significantly dose and concentration-dependent. The funnel plot showed non-symmetrical data distribution, with no imputed points. The risk of bias was moderate. In silico mouse body protein targets for the statins were mainly classes of Family AG protein- coupled receptor (20.0 %-33.3 %), Oxidoreductase (6.7-13.3 %) and Eraser (13.3 % each), with others at 0-26.7 %. The findings highlight statin effects in mice by reducing blood and brain ChE activities, in a dose/concentration-dependent manner, that would potentially modulate the cholinergic system. This anti-ChE effect together with in silico protein targets recognized could be the basis of further experimental explorations of adverse effects of statins.
Collapse
Affiliation(s)
- Fouad Kasim Mohammad
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Mosul, Mosul, Iraq
- College of Nursing, The American University of Kurdistan, Duhok, Iraq
| | - Rawnaq Faris Al-Shalchi
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Mosul, Mosul, Iraq
| |
Collapse
|
5
|
Ansari R, Khalili H, Mohammadi K. Optimizing statin therapy in HIV-infected patients: a review of pharmacotherapy considerations. BMC Cardiovasc Disord 2025; 25:421. [PMID: 40450213 DOI: 10.1186/s12872-025-04887-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 05/26/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND People living with HIV (PLWH) are more vulnerable to cerebrovascular disease, including coronary artery disease. Dyslipidemia is a risk factor for major adverse cardiovascular events in this population as a whole. Dyslipidemia might result from general risk factors, HIV itself, or the adverse effects of the antiretroviral drug, which have different effects on lipid profile. The present study aims to review the latest studies regarding the role of statin initiation in this population and clinically significant drug interactions in the field. METHODS Databases, including Scopus, PubMed, Google Scholar, EMBASE, and Web of Science, were searched for relevant literature on the role of statins in primary/secondary prevention of CVD in PLWH based on the PICO search strategy. Moreover, ARTs-statin drug interactions were investigated and summarized based on the University of Liverpool's website. RESULTS Nearly 70 studies were found and summarized. Guidelines recommendations for using statins in PLWH and current practice, as well as the role and potential mechanism of statins in PLWH, were investigated. Based on the available data, we developed a practical algorithm that clinicians can use to optimize statin therapy in PLWH. CONCLUSION More studies are required to fully define the role of statins in HIV patients, including time to initiate and proper dosing. Moreover, a marked inconsistency exists between clinical guidelines and actual practice, mainly due to irrational concerns regarding antiretroviral-statin drug interactions.
Collapse
Affiliation(s)
- Ramin Ansari
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Khalili
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Keyhan Mohammadi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Li X, Zhao X, Wang J, Xu B, Feng J, Huang W. High-Pressure Microfluidic Homogenization Improves the Stability and Antioxidant Properties of Coenzyme Q10 Nanoliposomes. BIOLOGY 2025; 14:568. [PMID: 40427757 PMCID: PMC12108558 DOI: 10.3390/biology14050568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Revised: 05/12/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025]
Abstract
Coenzyme Q10 is a natural antioxidant with anti-tumor and mitochondrial protective effects. However, its unstable physicochemical properties and large molecular weight result in low bioavailability. This study aimed to develop an effective technique for constructing nanoliposomes to improve the physicochemical properties of CoQ10 by using high-pressure microfluidic homogenization. Liposomes were prepared using the ethanol injection method and homogenized by high-pressure microfluidics to optimize their physicochemical properties. Liposome morphology and microstructure were observed via transmission electron microscopy (TEM). The particle size distribution, polydispersity index (PDI), and encapsulation efficiency were assessed, while effects on cell viability and antioxidant properties were investigated in HepG2 cells. The results indicate that the prepared liposomes exhibit favorable characteristics, including high encapsulation efficiency (>96%) and low PDI (<0.3), indicating uniform particle size distribution and good stability. The storage stability of liposomes at room temperature was significantly enhanced compared to liposomes not subjected to high pressure homogenization. In vitro cell experiments confirmed the liposomes' non-cytotoxicity and substantial antioxidant activity, ensuring their safety for biomedical applications. This study introduced a liposome preparation method combining ethanol injection and high-pressure microfluidic homogenization, offering a novel approach for liposome modification with potential for development and application in innovative drug delivery systems and antioxidant therapy.
Collapse
Affiliation(s)
- Xinyu Li
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Xingyu Zhao
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Jing Wang
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China;
| | - Baoshun Xu
- Kangcare Bioindustry Co., Ltd., Nanjing 210006, China;
| | - Jin Feng
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
| | - Wuyang Huang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China;
- Institute of Agro-Product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China;
- College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China;
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| |
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
|
Verma N, Bajiya M, Dolhey R, Surabhi, Yadav AS, Chaudhary C, Meena D, Arya H, Bhatt TK, Yadav JK, Shukla JN, Swaroop S, Pandey J. Mechanistic Insights into the Antibiofilm Activity of Simvastatin and Lovastatin against Bacillus subtilis. Mol Pharm 2025; 22:2703-2722. [PMID: 40100146 DOI: 10.1021/acs.molpharmaceut.5c00191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Statins have been reported for diverse pleiotropic activities, including antimicrobial and antibiofilm. However, due to the limited understanding of their mode of action, none of the statins have gained approval for antimicrobial or antibiofilm applications. In a recent drug repurposing study, we observed that two statins (i.e., Simvastatin and Lovastatin) interact stably with TasA(28-261), the principal extracellular matrix protein of Bacillus subtilis, and also induce inhibition of biofilm formation. Nevertheless, the underlying mechanism remained elusive. In the present study, we examined the impact of these statins on the physiological activity of TasA(28-261), specifically its interaction with TapA(33-253) and aggregation into the amyloid-like structure using purified recombinant TasA(28-261) and TapA(33-253) in amyloid detection-specific in vitro assays (i.e., CR binding and ThT staining assays). Results revealed that both statins interfered with amyloid formation by the TasA(28-261)-TapA(33-253) complex, while neither statin inhibited amyloid formation by lysozyme, a model amyloid-forming protein. Moreover, neither statin significantly altered the expressions of terminal regulatory genes (viz, sinR, sinI) and terminal effector genes (viz, tasA, tapA, and bslA) involved in biofilm formation by B. subtilis. While the intricate interplay between Simvastatin and Lovastatin with the diverse molecular constituents of B. subtilis biofilm remains to be elucidated conclusively, the findings obtained during the present study suggest that the underlying mechanism for Simvastatin- and Lovastatin-mediated inhibition of B. subtilis biofilm formation is manifested by interfering with the aggregation and amyloid formation by TasA(28-261)-TapA(33-253). These results represent one of the first experimental evidence for the underlying mechanism of antibiofilm activity of statins and offer valuable directions for future research to harness statins as antibiofilm therapeutics.
Collapse
Affiliation(s)
- Nidhi Verma
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Mamta Bajiya
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Ragini Dolhey
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Surabhi
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Abhishek Singh Yadav
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Chhavi Chaudhary
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Dhankesh Meena
- Department of Biochemistry, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Hemant Arya
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Tarun K Bhatt
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Jay Kant Yadav
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Jayendra Nath Shukla
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Shiv Swaroop
- Department of Biochemistry, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| | - Janmejay Pandey
- Department of Biotechnology, Central University of Rajasthan, Kishangarh, Ajmer 305801, Rajasthan, India
| |
Collapse
|
9
|
Li S, Wang L, Han M, Fan H, Tang H, Gao H, Li G, Xu Z, Zhou Z, Du J, Peng C, Peng F. Combination of Sodium Butyrate and Immunotherapy in Glioma: regulation of immunologically hot and cold tumors via gut microbiota and metabolites. Front Immunol 2025; 16:1532528. [PMID: 40297576 PMCID: PMC12035444 DOI: 10.3389/fimmu.2025.1532528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Background Recent studies have highlighted the importance of cross-talk along the gut-brain axis in regulating inflammatory nociception, inflammatory responses, and immune homeostasis. The gut microbiota, particularly its bacterial composition, plays a crucial role in the development and function of the immune system. Moreover, metabolites produced by the gut microbiota can significantly impact both systemic immune responses and central nervous system (CNS) immunity. Sodium butyrate is a key metabolite produced by the gut microbiota and, as a histone deacetylase inhibitor, can enhance the anti-tumor immunity of cytotoxic CD8+ T cells. However, it remains unclear whether sodium butyrate treatment can enhance the efficacy of PD-1 blockade in glioma therapy. In this research, the effect and underlying mechanism of combination of gut microbiota metabolites and anti-mouse PD-1 mAb on glioma has been investigated. Methods RNA-seq assay in glioma cell and biomedical databases, including ONCOMINE, GEPIA and TCGA were incorporated. Subsequently, the inhibitory effect of sodium butyrate on glioma cells and its related mechanisms were assessed through Counting Kit-8 (CCK-8), Flow Cytometry, Western blot (WB), reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and other in vitro experiments. In vitro, an orthotopic mouse glioma model was established. MRI imaging, Immunohistochemistry, and Immune cell flow cytometry were used to investigate the therapeutic effects of combined sodium butyrate and PD-1 inhibitor treatment on glioma-bearing mice. Results We discovered that deacetylation-associated gene expression is significantly increased in glioma patients and affects patient survival time. Moreover, we found sodium butyrate promoted glioma cell apoptosis, disrupted the cell cycle, and inhibited tumor growth. Additionally, sodium butyrate may upregulate PD-L1 expression in glioma cells by modulating the PI3K/AKT pathway. The experimental results demonstrated that this combination therapy significantly reduced tumor volume and prolonged survival in an orthotopic murine glioma model. Moreover, combination therapy led to an increase in the proportion of probiotic bacteria in the mouse gut microbiota, resulting in elevated levels of antitumor metabolites and a decrease in metabolites that affect immune cell function.
Collapse
Affiliation(s)
- Sui Li
- West China School of Pharmacy, Sichuan University, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Wang
- Bioinformatics Department, Jiangsu Sanshu Biotechnology Co., Ltd., Nantong, China
| | - MingYu Han
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Huali Fan
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangdong, China
| | - Huile Gao
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Guobo Li
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zheng Xu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Department of Clinical Medicine, Zhengzhou University, Henan, China
| | - JunRong Du
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Song Y, Chen Y, Li J, Sun W, Jin F. Manual acupuncture enhanced therapeutic efficacy in vascular dementia rat model: systematic review and network meta-analysis. Syst Rev 2025; 14:80. [PMID: 40186311 PMCID: PMC11971794 DOI: 10.1186/s13643-025-02821-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/12/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE This study aimed to systematically evaluate the efficacy of electroacupuncture and manual acupuncture for treating vascular dementia and to determine the optimal acupuncture point combination scheme for efficacy. METHODS The PubMed, Embase, Web of Science, Cochrane, CNKI, VIP, and Wanfang electronic databases were searched up to July 2024 to identify relevant randomized controlled trials. RevMan 5.4 software and Addis software were used to assess the risk of bias for each study, determine subgroup classifications, and conduct meta-analyses. RESULTS A total of 29 RCTs involving 659 animals were ultimately included. The meta-analysis results revealed that acupuncture treatment had a significant effect compared with the vascular dementia model group [mean difference (MD) = - 21.68, 95% confidence interval (CI) (- 25.77, - 17.59), P < 0.00001]. Manual acupuncture demonstrated better efficacy than electroacupuncture did [MD = - 0.42, 95% CI (- 12.72, 12.27)]. Among the different acupuncture point combinations, the Baihui (GV20) + Dazhui (GV14) combination yielded the best efficacy [MD = - 23.03, 95% CI (- 30.02, - 16.04), P < 0.00001]. Compared with other acupuncture protocols, the experiment conducted by Caiyu Peng et al. exhibited superior efficacy [MD = - 24.96, 95% CI (- 92.68, - 40.76)]. CONCLUSION Acupuncture significantly improves cognitive function in rats with vascular dementia. Manual acupuncture is more effective than electroacupuncture. Among the different acupuncture point combinations, manual acupuncture at GV20 and GV14 yields the best results. Compared with other acupuncture protocols, the best efficacy was observed when the two-vessel occlusion (2VO) model was used in 230 ± 10 g SD rats; when the Mingmen (GV4), Dazhui (GV14), Fengfu (GV16), Baihui (GV20), Shenting (GV24), Shuigou (GV26), Neiguan (PC6), Dalin (PC7), and Laogong (PC8) acupoints were selected; and when manual acupuncture with reinforcing and reducing methods was used for 30 min per day for 14 days. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42024551402.
Collapse
Affiliation(s)
- Yuanyu Song
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Yinghua Chen
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150006, China.
| | - Junfeng Li
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Wei Sun
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| | - Fangfang Jin
- Heilongjiang University of Chinese Medicine, Harbin, 150006, China
| |
Collapse
|
11
|
Marzioni D, Piani F, Di Simone N, Giannubilo SR, Ciavattini A, Tossetta G. Importance of STAT3 signaling in preeclampsia (Review). Int J Mol Med 2025; 55:58. [PMID: 39918020 PMCID: PMC11878484 DOI: 10.3892/ijmm.2025.5499] [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: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 03/06/2025] Open
Abstract
Placentation is a key process that is tightly regulated that ensures the normal placenta and fetal development. Preeclampsia (PE) is a hypertensive pregnancy‑associated disorder characterized by increased oxidative stress and inflammation. STAT3 signaling plays a key role in modulating important processes such as cell proliferation, differentiation, invasion and apoptosis. The present review aimed to analyse the role of STAT3 signaling in PE pregnancies, discuss the main natural and synthetic compounds involved in modulation of this signaling both in vivo and in vitro and summarize the main cellular modulators of this signaling to identify possible therapeutic targets and treatments to improve the outcome of PE pregnancies.
Collapse
Affiliation(s)
- Daniela Marzioni
- Department of Experimental and Clinical Medicine, Polytechnic University of Marche, I-60126 Ancona, Italy
| | - Federica Piani
- Hypertension and Cardiovascular Risk Research Center, Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, I-40126 Bologna, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, I-20072 Milan, Italy
- Scientific Institutes for Hospitalization and Care (IRCCS), Humanitas Research Hospital, I-20089 Rozzano, Italy
| | | | - Andrea Ciavattini
- Department of Clinical Sciences, Polytechnic University of Marche, I-60123 Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Polytechnic University of Marche, I-60126 Ancona, Italy
| |
Collapse
|
12
|
Zheng Y, Gu H, Kong Y. Statin is associated with higher cortical thickness in early Alzheimer's disease. Exp Gerontol 2025; 202:112698. [PMID: 39900257 DOI: 10.1016/j.exger.2025.112698] [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: 08/16/2024] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND The brain is the most cholesterol-rich organ, essential for myelination and neuronal function. Statins, widely used to lower cholesterol, cross the blood-brain barrier and may impact brain cholesterol synthesis. Despite their widespread use, the effects of statins on cortical regions relevant to Alzheimer's disease (AD) are not well understood. This study aimed to compare cortical thickness between statin-exposed and statin-unexposed older adults and evaluate the potential neuroprotective effects of statins. METHODS Data were obtained from the Alzheimer's Disease Neuroimaging Initiative (ADNI). The sample included 193 healthy controls (HC), 485 individuals with mild cognitive impairment (MCI), and 169 individuals with Alzheimer's disease (AD). Participants were categorized as statin users if they had used statins for at least two years. MRI data were processed using FreeSurfer software to estimate cortical thickness in 64 regions of interest. ANCOVA models assessed the association between statin use and cortical thickness at baseline, and linear mixed models evaluated longitudinal changes. RESULTS Statin use was associated with increased cortical thickness in multiple brain regions across HC, MCI, and AD participants. In HC, statin users had greater thickness in the right lateral occipital, left middle temporal, and left parahippocampal regions. MCI participants exhibited additional increases in the right cuneus, right posterior cingulate, and left superior temporal cortex. In AD, statin users had higher thickness in the right cuneus and right superior parietal lobule. Longitudinal analysis revealed no statin-related differences in cortical thickness changes among HC and AD groups, but in MCI, statins slowed cortical thinning in the left medial orbitofrontal cortex. CONCLUSION Statin use is associated with greater cortical thickness in older adults, particularly in those with MCI. These findings suggest that statins may have neuroprotective effects, potentially mitigating neurodegenerative changes in early cognitive decline. Further research with larger cohorts and longer follow-up periods is needed to confirm these findings and understand the mechanisms involved.
Collapse
Affiliation(s)
- Yane Zheng
- Department of Neurology, Shanghai Jiangong Hospital, Shanghai 200083, China.
| | - Huiying Gu
- Department of Internal Medicine, Tangqiao Community Health Service Center, Shanghai 200127, China
| | - Yuming Kong
- Department of Neurology, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200438, China
| |
Collapse
|
13
|
Chu Z, Yue W, Mu Q, Xu D, Chang Z, Liang M, Geng Y, Ding P. Effects of statin use on blood pressure and other hypertension-related outcome indicators in hypertensive patients: A systematic review and meta-analysis. Prostaglandins Other Lipid Mediat 2025; 178:106991. [PMID: 40174858 DOI: 10.1016/j.prostaglandins.2025.106991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/19/2024] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
AIMS This meta-analysis delved into the impact of statin therapy, both as a monotherapy and in conjunction with antihypertensive medications, on blood pressure levels and outcomes pertinent to hypertension. METHODS We searched the PubMed, EMBASE, and Cochrane databases for studies published before October 1 2023. Studies designed as cohort studies or randomized controlled trials and investigating the effects of single use of statin or its combined use with other antihypertensive therapy were included. Authors extracted the data independently and differences were decided through discussion. Random-effects model was used to evaluate the merged outcomes. Due to the high heterogeneity of the HDL-C group, we performed subgroup analysis according to the type of statin. To assess the robustness and potential publication bias of our findings, we utilized sensitivity analysis, Egger's test, and funnel plots. RESULTS 23 trials were included in this meta-analysis. The primary outcomes revealed that administering statins did not significantly impact the systolic pressure (SBP) of hypertensive patients (MD, -1.77; 95 % CI, -4.82-1.27). -The promoted effect of statin treatment on diastolic pressure (DBP) in hypertensive patients was found (MD, -1.87; 95 % CI, -3.72 --0.01). The secondary outcomes revealed that the use of statins resulted in a significant reduction in low-density lipoprotein (LDL-C), while significantly increasing high-density lipoprotein (HDL-C) in hypertensive patients. CONCLUSION Statin use did not modulate SBP and DBP of patients with hypertension, but SBP was decreased in the rosuvastatin or pravastatin subgroup, while DBP was decreased in the simvastatin or pravastatin subgroup. Statin treatment reduced LDL-C, increased HDL-C, and reduced the incidence of cardiovascular events and mortality compared to control groups.
Collapse
Affiliation(s)
- Zhaohan Chu
- Department of Cardiology Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| | - Wei Yue
- Department of Critical Care Medicine, 988 hospital of the PLA Joint Logistic Support Force (PLAJLSF), Zhengzhou 450000, China.
| | - Qingqing Mu
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Dong Xu
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Zexu Chang
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Mengke Liang
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Yixiao Geng
- School of Clinical Medicine, Zhengzhou University, Zhengzhou 450000, China.
| | - Ping Ding
- Department of Critical Care Medicine, 988 hospital of the PLA Joint Logistic Support Force (PLAJLSF), Zhengzhou 450000, China.
| |
Collapse
|
14
|
Altuwaijri N, Fitaihi R, Alkathiri FA, Bukhari SI, Altalal AM, Alsalhi A, Alsulaiman L, Alomran AO, Aldosari NS, Alqhafi SA, Alhamdan M, Alfaraj R. Assessing the Antibacterial Potential and Biofilm Inhibition Capability of Atorvastatin-Loaded Nanostructured Lipid Carriers via Crystal Violet Assay. Pharmaceuticals (Basel) 2025; 18:417. [PMID: 40143193 PMCID: PMC11944405 DOI: 10.3390/ph18030417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Atorvastatin (ATR), an antihyperlipidemic drug with a potential antibacterial effect, was investigated in this study. Like other statins, ATR has been repurposed for several uses, ranging from anti-inflammatory to antimicrobial applications, and has demonstrated successful results. However, the efficacy of ATR is limited by its low solubility, indicating an opportunity for its encapsulation in a nanotechnology-based drug delivery system. Methods: Nanostructured lipid carrier (NLC) formulations were prepared using high-pressure homogenization and ultrasonication. The formulations were characterized, including their particle size, polydispersity index, zeta potential, encapsulation efficiency, and in vitro release. Antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA), Escherichia coli (E. coli), and Staphylococcus aureus (S. aureus) was evaluated using the growth curve (bacterial growth over time) and well diffusion methods (zone of inhibition and minimum inhibitory concentration (MIC) determination). The crystal violet assay was employed to assess biofilm inhibition. Results: The NLC formulations were optimized, and the size and zeta potential of the blank nanoparticles were 130 ± 8.39 nm and -35 ± 0.5 mV, respectively. In comparison, the encapsulated NLCs had a size of 142 ± 52.20 nm and a zeta potential of -31 ± 1.41 mV. The average encapsulation efficiency was 94%, and 70% of the drug was released after 24 h. The ATR-loaded NLCs showed significantly enhanced antibacterial activity by reducing the minimum inhibitory concentration by 2.5-fold for E. coli, 1.8-fold for S. aureus, and 1.4-fold for MRSA, and promoting more effective bacterial growth inhibition. Notably, biofilm inhibition was significantly improved with ATR-NLCs, achieving 80% inhibition for S. aureus, 40% for E. coli, and 30% for MRSA, compared to free ATR (p < 0.001). These findings suggest that NLC encapsulation enhances ATR's antimicrobial efficacy and biofilm suppression. Conclusions: This study identified NLCs as successful carriers of ATR, significantly enhancing its antibacterial efficacy and biofilm inhibition capabilities. This formulation, which shows antimicrobial potential against both Gram-positive and Gram-negative bacteria, should be further studied and developed against different resistant microbial strains.
Collapse
Affiliation(s)
- Njoud Altuwaijri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Rawan Fitaihi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Fai A. Alkathiri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Sarah I. Bukhari
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Alanoud M. Altalal
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Alyaa Alsalhi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Lama Alsulaiman
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Aljawhara O. Alomran
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Noura S. Aldosari
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Safa A. Alqhafi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Majd Alhamdan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| | - Rihaf Alfaraj
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.); (F.A.A.); (S.I.B.); (A.M.A.); (A.A.); (L.A.); (A.O.A.); (S.A.A.); (M.A.); (R.A.)
| |
Collapse
|
15
|
Cheng WM, Li PC, Nguyen MTB, Lin YT, Huang YT, Cheng TS, Nguyen TH, Tran TH, Huang TY, Hoang TH, Chen SY, Chu YC, Wu CW, Lee MF, Chiou YS, Liu HS, Hong YR, Chang PMH, Hu YF, Chang YC, Lai JM, Huang CYF. Repurposing pitavastatin and atorvastatin to overcome chemoresistance of metastatic colorectal cancer under high glucose conditions. Cancer Cell Int 2025; 25:79. [PMID: 40050889 PMCID: PMC11887183 DOI: 10.1186/s12935-025-03712-2] [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: 04/04/2024] [Accepted: 02/22/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) poses a significant clinical challenge because of drug resistance, which can adversely impact patient outcomes. Recent research has shown that abnormalities within the tumor microenvironment, especially hyperglycemia, play a crucial role in promoting metastasis and chemoresistance, and thereby determine the overall prognosis of patients with advanced CRC. METHODS This study employs data mining and consensus molecular subtype (CMS) techniques to identify pitavastatin and atorvastatin as potential agents for targeting high glucose-induced drug resistance in advanced CRC cells. CRC cells maintained under either low or high glucose conditions were established and utilized to assess the cytotoxic effects of pitavastatin and atorvastatin, both with and without 5-fluorouracil (5-FU). CRC 3D spheroids cultured were also included to demonstrate the anti-drug resistance of pitavastatin and atorvastatin. RESULTS A bioinformatics analysis identified pitavastatin and atorvastatin as promising drug candidates. The CMS4 CRC cell line SW480 (SW480-HG) was established and cultured under high glucose conditions to simulate hyperglycemia-induced drug resistance and metastasis in CRC patients. Pitavastatin and atorvastatin could inhibit cell proliferation and 3D spheroid formation of CMS4 CRC cells under high glucose conditions. In addition, both pitavastatin and atorvastatin can synergistically promote the 5-FU-mediated cytotoxic effect and inhibit the growth of 5-FU-resistant CRC cells. Mechanistically, pitavastatin and atorvastatin can induce apoptosis and synergistically promote the 5-FU-mediated cytotoxic effect by activating autophagy, as well as the PERK/ATF4/CHOP signaling pathway while decreasing YAP expression. CONCLUSION This study highlights the biomarker-guided precision medicine strategy for drug repurposing. Pitavastatin and atorvastatin could be used to assist in the treatment of advanced CRC, particularly with CMS4 subtype CRC patients who also suffer from hyperglycemia. Pitavastatin, with an achievable dosage used for clinical interventions, is highly recommended for a novel CRC therapeutic strategy.
Collapse
Affiliation(s)
- Wei-Ming Cheng
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Urology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Division of Urology, Department of Surgery, Zhongxiao Branch, Taipei City Hospital, Taipei, 115, Taiwan
| | - Po-Chen Li
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Minh Tran-Binh Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam
| | - Yu-Teng Lin
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yu-Tang Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Tai-Shan Cheng
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City, 220, Taiwan
| | - Thi-Huong Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Institute of Biotechnology and Food Technology, Thai Nguyen University of Agriculture and Forestry, Thai Nguyen, Vietnam
| | - Thu-Ha Tran
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 112, Taiwan
| | - Tzu-Yi Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Thu-Huyen Hoang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Sin-Yu Chen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yu-Chieh Chu
- Taipei First Girls High School, Taipei, 110, Taiwan
| | - Chih-Wei Wu
- Taipei First Girls High School, Taipei, 110, Taiwan
| | - Ming-Fen Lee
- Department of Nutrition, China Medical University, Taichung, 406, Taiwan
| | - Yi-Shiou Chiou
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hsiao-Sheng Liu
- Medical Research Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- 13 M.Sc. Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Center for Cancer Research, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 807, Taiwan
| | - Yi-Ren Hong
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Peter Mu-Hsin Chang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Yu-Feng Hu
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, 112, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, 115, Taipei, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Jin-Mei Lai
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City, 242, Taiwan.
| | - Chi-Ying F Huang
- Program in Molecular Medicine, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- Chong Hin Loon Memorial Cancer and Biotherapy Research Center, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
| |
Collapse
|
16
|
Wan M, Pan S, Shan B, Diao H, Jin H, Wang Z, Wang W, Han S, Liu W, He J, Zheng Z, Pan Y, Han X, Zhang J. Lipid metabolic reprograming: the unsung hero in breast cancer progression and tumor microenvironment. Mol Cancer 2025; 24:61. [PMID: 40025508 PMCID: PMC11874147 DOI: 10.1186/s12943-025-02258-1] [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: 12/03/2024] [Accepted: 02/02/2025] [Indexed: 03/04/2025] Open
Abstract
Aberrant lipid metabolism is a well-recognized hallmark of cancer. Notably, breast cancer (BC) arises from a lipid-rich microenvironment and depends significantly on lipid metabolic reprogramming to fulfill its developmental requirements. In this review, we revisit the pivotal role of lipid metabolism in BC, underscoring its impact on the progression and tumor microenvironment. Firstly, we delineate the overall landscape of lipid metabolism in BC, highlighting its roles in tumor progression and patient prognosis. Given that lipids can also act as signaling molecules, we next describe the lipid signaling exchanges between BC cells and other cellular components in the tumor microenvironment. Additionally, we summarize the therapeutic potential of targeting lipid metabolism from the aspects of lipid metabolism processes, lipid-related transcription factors and immunotherapy in BC. Finally, we discuss the possibilities and problems associated with clinical applications of lipid‑targeted therapy in BC, and propose new research directions with advances in spatiotemporal multi-omics.
Collapse
Affiliation(s)
- Mengting Wan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Benjie Shan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Haizhou Diao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Ziqi Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Wan Nan Medical College, Wuhu, Anhui, China
| | - Shuya Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Wan Liu
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jiaying He
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Graduate School of Bengbu Medical University, Bengbu, Anhui Province, China
| | - Zihan Zheng
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- School of Medical Oncology, Anhui Medical University, Hefei, China
| | - Yueyin Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
17
|
Millichap L, Turton N, Alomosh R, Heaton RA, Bateman A, Al-Shanti N, Lightfoot AP, Damiani E, Marcheggiani F, Orlando P, Silvestri S, Tiano L, Hargreaves IP. The effect of simvastatin induced neurotoxicity on mitochondrial function in human neuronal cells. Toxicol Mech Methods 2025:1-12. [PMID: 40028788 DOI: 10.1080/15376516.2025.2471807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase (HMGR) inhibitors, commonly known as statins, are drugs frequently used in the treatment of hypercholesterolemia and hyperlipidemia. However, the current study has demonstrated that simvastatin induces neurotoxicity and is associated with cellular coenzyme Q10 (CoQ10) depletion. CoQ10 has a significant role in the mitochondrial electron transport chain (ETC), in addition to being a fundamental lipid-soluble antioxidant. Depletion of CoQ10 is frequently associated with impaired mitochondrial function and increased oxidative stress. The aim of this study was to investigate the potential mechanisms of simvastatin-induced neurotoxicity assessing mitochondrial function and evidence of oxidative stress in an in vitro SH-SY5Y human neuronal cell line. Fluorescence studies assessed via flow cytometry determined significant increases in intracellular and mitochondrial reactive oxygen species production following SH-SY5Y treatment with simvastatin compared to control cells. Additionally, spectrophotometric enzyme studies determined a significant (p < 0.0001) inhibition of ETC complex I and II-III activities which accompanied a significant decrease in neuronal CoQ10 content (p < 0.005) and cell viability (p < 0.0001). The results of the present study have indicated evidence of mitochondrial dysfunction and increased oxidative stress, resulting in increased loss of neuronal viability following simvastatin treatment. Thus, these results demonstrate evidence of neurotoxicity associated with statin therapy.
Collapse
Affiliation(s)
- Lauren Millichap
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Nadia Turton
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Razan Alomosh
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Robert A Heaton
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Amy Bateman
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Nasser Al-Shanti
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Adam P Lightfoot
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Elisabetta Damiani
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Fabio Marcheggiani
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Patrick Orlando
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Iain P Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
18
|
Yang B, Yao B, Zou Q, Li S, Yang S, Yang M. Causal Association Between Cholesterol-Lowering Drugs and Diabetic Microvascular Complications: A Drug-Target Mendelian Randomization Study. J Diabetes Res 2025; 2025:3661739. [PMID: 40225015 PMCID: PMC11986941 DOI: 10.1155/jdr/3661739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 01/10/2025] [Accepted: 02/01/2025] [Indexed: 04/15/2025] Open
Abstract
Background: It remains unclear whether cholesterol-lowering therapy can reduce the incidence of microvascular complications in patients with diabetes. We aim to explore the potential causal relationship between three common types of cholesterol-lowering drugs and diabetic microvascular complications through drug-target Mendelian randomization (MR) study, laying the groundwork for the development of new medications. Methods: In this study, we collected single nucleotide polymorphisms (SNPs) associated with HMGCR (3-hydroxy-3-methylglutaryl-CoA reductase) inhibitors, PCSK9 (proprotein convertase subtilisin/kexin type 9) inhibitors, and NPC1L1 (Niemann-Pick C1-Like 1) inhibitors from published genome-wide association study statistics. Subsequently, drug-target MR analyses were performed to investigate the effects of these inhibitors on low-density lipoprotein cholesterol (LDL-C) level-mediated microvascular complications in diabetes mellitus. Coronary atherosclerosis as a positive control. Primary outcomes included diabetic nephropathy, diabetic retinopathy, and diabetic neuropathy from the FinnGen Consortium. Results: The MR analysis revealed significant associations between HMGCR inhibition and increased risks of diabetic nephropathy (OR [95%confidence interval (CI)] = 1.88 [1.50, 2.36], p = 5.55 × 10-8), retinopathy (OR [95%CI] = 1.86 [1.54, 2.24], p = 6.28 × 10-11), and neuropathy (OR [95%CI] = 2.63 [1.84, 3.75], p = 1.14 × 10-7) using the inverse variance weighted method. PCSK9 inhibitors have been associated with an increased risk of diabetic nephropathy (OR [95%CI] = 1.30 [1.07, 1.58], p = 0.009) and diabetic neuropathy (OR [95%CI] = 1.40 [1.15, 1.72], p = 0.001); NPC1L1 inhibitors significantly reduce the incidence of diabetic retinopathy (OR [95%CI] = 0.48 [0.28, 0.85], p = 0.01). The coronary heart disease as positive control. Conclusions: The findings show that HMGCR inhibitors and PCSK9 inhibitors may significantly increase the risk of diabetic microvascular complications. However, NPC1L1 inhibitors may provide protection against diabetic retinopathy.
Collapse
Affiliation(s)
- Bo Yang
- Department of Endocrinology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Bo Yao
- Department of General Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Qu Zou
- Department of General Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Sicheng Li
- Department of Endocrinology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Shun Yang
- Department of General Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Mengxue Yang
- Department of Endocrinology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Lu C, Chen J, Zhang S, Tian Y, Ying X, Zhang D, Luo Z, Si X, Li M. Development of a vitrified CRISPR/Cas12b-based assay for rapid genotyping of SLCO1B1 SNPs without DNA amplification. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:2083-2093. [PMID: 39936934 DOI: 10.1039/d4ay02015d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Two single nucleotide polymorphisms (SNPs) in the human SLCO1B1 gene, c.388A>G (rs2306283) and c.521T>C (rs4149056), are independent determinants of the efficacy and side effects of statin drugs. Multinational clinical guidelines recommend testing for SLCO1B1 genotypes before the initial use of statins. Current SLCO1B1 SNP identification methods, primarily based on quantitative fluorescence PCR, rely on expensive equipment, are time-consuming, and require cold-chain storage for reagents, making them unsuitable for use in resource-limited healthcare settings. In this study, we developed a CRISPR/Cas12b-based amplification-free genotyping technique for SLCO1B1 SNPs. Within 30 minutes of the isothermal reaction, genotyping of the c.388A>G and c.521T>C SNPs in the SLCO1B1 gene can be observed by the naked eyes under blue light. Additionally, maltodextrin was identified as an effective vitrification stabilizer for the CRISPR/Cas12b premix. A low-cost vitrification process was optimized to prepare a glass like solid reagent via room-temperature vacuum drying. The vitrified CRISPR/Cas12b reagent retained approximately 88% of its activity after 30 days of storage at 37 °C, eliminating the need for cold-chain storage and allowing for long-term preservation at room temperature. This vitrified CRISPR/Cas12b based rapid SNP detection technique is especially suitable for genotyping drug metabolism genes in primary healthcare settings, providing effective guidance for precision medicine in clinical practice.
Collapse
Affiliation(s)
- Chen Lu
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Institute of Marine Resources Development, Lianyungang 222005, China
| | - Jiayue Chen
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Shengsheng Zhang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu BestEnzymes Biotech Co., Ltd., Lianyungang 222005, China
| | - Yaru Tian
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xinyu Ying
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Dan Zhang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zhidan Luo
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Institute of Marine Resources Development, Lianyungang 222005, China
| | - Xinxin Si
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, China.
- Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
| | - Ming Li
- Department of Pharmacy, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, China.
| |
Collapse
|
20
|
Yu F, Chen J, Wang X, Hou S, Li H, Yao Y, He Y, Chen K. Metabolic reprogramming of peritoneal mesothelial cells in peritoneal dialysis-associated fibrosis: therapeutic targets and strategies. Cell Commun Signal 2025; 23:114. [PMID: 40016825 PMCID: PMC11866825 DOI: 10.1186/s12964-025-02113-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
Peritoneal dialysis (PD) is considered a life-saving treatment for end-stage renal disease. However, prolonged PD use can lead to the development of peritoneal fibrosis (PF), diminishing its efficacy. Peritoneal mesothelial cells (PMCs) are key initiators of PF when they become damaged. Exposure to high glucose‑based peritoneal dialysis fluids (PDFs) contributes to PF development by directly affecting highly metabolically active PMCs. Recent research indicates that PMCs undergo metabolic reprogramming when exposed to high-glucose PDFs, including enhanced glycolysis, impaired oxidative phosphorylation, abnormal lipid metabolism, and mitochondrial dysfunction. Although this metabolic transition temporarily compensates for the cellular damage and maintains energy levels, its long-term impact on peritoneal tissue is concerning. Multiple studies have identified a close association between this shift in energy metabolism and PF, and may promote the progression of PF through various molecular mechanisms. This review explores recent findings regarding the role and mechanism of PMC metabolic reprogramming in PF progression. Moreover, it provides a summary of potential therapeutic strategies aimed at various metabolic processes, including glucose metabolism, lipid metabolism, and mitochondrial function. The review establishes that targeting metabolic reprogramming in PMCs may be a novel strategy for preventing and treating PD-associated fibrosis.
Collapse
Affiliation(s)
- Fang Yu
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Jia Chen
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Xiaoyue Wang
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Shihui Hou
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Hong Li
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Yaru Yao
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China
| | - Yani He
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- State Key Laboratory of Trauma and Chemical poisoning, Burns and Combined Injury, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
| | - Kehong Chen
- Department of Nephrology, Daping Hospital, Army Medical Center, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
- State Key Laboratory of Trauma and Chemical poisoning, Burns and Combined Injury, Army Medical University, NO. 10 Changjiang Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
21
|
Zhou J, Feng Z, Qiu H, Li T, Huang X, Ye L, Huang L, Guo C, Guo C, He L. Association between statin administration and outcome in patients with sepsis: A retrospective study. Open Med (Wars) 2025; 20:20241112. [PMID: 39927168 PMCID: PMC11806238 DOI: 10.1515/med-2024-1112] [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: 03/21/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 02/11/2025] Open
Abstract
Background & aims There was considerable debate regarding the effect of statins administration on the outcome of septic patients. This retrospective study aimed to assess the association between statins administration and mortality in sepsis patients and investigate whether this association differed according to the types of statins. Methods We performed a retrospective study based on the electronic ICU Collaborative Research Database, Medical Information Mart for Intensive Care Database, and the Amsterdam University Medical Centers Database. The participants with sepsis were divided as two groups, statins group and non-statins group. The primary endpoint was the all-cause mortality. We utilized logistic regression, propensity score matching (PSM), and sub-analysis to assess the association between statins administration and outcome in patients with sepsis. Results A total of 19,327 sepsis patients were enrolled. Among these, 3,721 patients were prescribed statins. Pooled analyses of three databases showed that statin users had a decreased risk of mortality in sepsis as compared with nonusers (OR 0.73, 95% CI 0.66-0.80, P < 0.001). Sub-analysis of statin showed that atorvastatin had the most distinct effectiveness in decreasing mortality (OR 0.67, 95% CI 0.59-0.76, P = 0.035), whereas pravastatin, simvastatin, and rosuvastatin were not. PSM analysis confirmed these findings for statins (OR 0.75, 95% CI 0.67-0.84, P < 0.001) and atorvastatin (OR 0.70, 95% CI 0.59-0.82, P < 0.001). Conclusions The use of statins could decrease the risk of mortality in patients with sepsis during the hospital period. Among different types of statins, atorvastatin showed the most significant trend to reduce the risk of mortality in patients with sepsis.
Collapse
Affiliation(s)
- Jianzhu Zhou
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Zeying Feng
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Hui Qiu
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Tong Li
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xin Huang
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Ling Ye
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Longjian Huang
- West Guangxi Key Laboratory for Prevention and Treatment of High-Incidence Diseases, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Chengjun Guo
- School of Applied Mathematics, Guangdong University of Technology, Guangzhou, 510006, Guangdong, China
| | - Chengxian Guo
- Center of Clinical Pharmacology, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Li He
- Department of Pediatrics, the Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| |
Collapse
|
22
|
da Silva Pereira ENG, Franco RLC, Santos RDCD, Daliry A. Statins and non-alcoholic fatty liver disease: A concise review. Biomed Pharmacother 2025; 183:117805. [PMID: 39755024 DOI: 10.1016/j.biopha.2024.117805] [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: 10/16/2024] [Revised: 12/12/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common hepatic manifestation of metabolic syndrome affecting 20-30 % of the adult population worldwide. This disease, which includes simple steatosis and non-alcoholic steatohepatitis, poses a significant risk for cardiovascular and metabolic diseases. Lifestyle modifications are crucial in the treatment of NAFLD; however, patient adherence remains challenging. As there is no specific treatment, drug repositioning is being researched as an alternative strategy. Statins, which are known for their cholesterol-lowering effects, are considered potential interventions for NAFLD. This review aimed to present the current understanding of the effects of statins on liver physiology in the context of NAFLD. The pathophysiology of NAFLD includes steatosis, inflammation, and fibrosis, which are exacerbated by dyslipidemia and insulin resistance. Statins, which inhibit 3-hydroxy-3-methylglutaryl-CoA reductase, have pleiotropic effects beyond cholesterol-lowering and affect pathways related to inflammation, fibrogenesis, oxidative stress, and microcirculation. Although clinical guidelines support the use of statins for dyslipidemia in patients with NAFLD, more studies are needed to demonstrate their efficacy in liver disease. This comprehensive review serves as a foundation for future studies on the therapeutic potential of statins in NAFLD.
Collapse
Affiliation(s)
| | - Rafaela Luiza Costa Franco
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Rafaele Dantas Cruz Dos Santos
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Anissa Daliry
- Laboratory of Clinical and Experimental Physiopathology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
23
|
Mikhael S, Daoud G. Navigating Metabolic Challenges in Ovarian Cancer: Insights and Innovations in Drug Repurposing. Cancer Med 2025; 14:e70681. [PMID: 39969135 PMCID: PMC11837049 DOI: 10.1002/cam4.70681] [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: 10/29/2024] [Revised: 01/16/2025] [Accepted: 01/30/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the most lethal gynecological malignancy and a major global health concern, often diagnosed at advanced stages with poor survival rates. Despite advancements in treatment, resistance to standard chemotherapy remains a critical challenge with limited treatment options available. In recent years, the role of metabolic reprogramming in OC has emerged as a key factor driving tumor progression, therapy resistance, and poor clinical outcomes. METHODS This review explores the intricate connections between metabolic syndrome, enhanced glycolysis, and altered lipid metabolism within OC cells, which fuel the aggressive nature of the disease. We discuss how metabolic pathways are rewired in OC to support uncontrolled cell proliferation, survival under hypoxic conditions, and evasion of cell death mechanisms, positioning metabolic alterations as central to disease progression. The review also highlights the potential of repurposed metabolic-targeting drugs, such as metformin and statins, which have shown promise in preclinical studies for their ability to disrupt these altered metabolic pathways. CONCLUSION Drug repurposing offers a promising strategy to overcome chemoresistance and improve patient outcomes. Future research should focus on unraveling the complex metabolic networks in OC to develop innovative, targeted therapies that can enhance treatment efficacy and patient survival.
Collapse
Affiliation(s)
- Sara Mikhael
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of MedicineAmerican University of BeirutBeirutLebanon
| | - Georges Daoud
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of MedicineAmerican University of BeirutBeirutLebanon
| |
Collapse
|
24
|
AlJunaydil NA, Lambarte RNA, Sumague TS, Alghamdi OG, Niazy AA. Lovastatin and Resveratrol Synergistically Improve Wound Healing and Inhibit Bacterial Growth. Int J Mol Sci 2025; 26:851. [PMID: 39859566 PMCID: PMC11766293 DOI: 10.3390/ijms26020851] [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: 12/05/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Wound healing is a complex physiological process, with scarring and infection caused by Staphylococcus aureus and Pseudomonas aeruginosa being the most common complications. The reutilization of known medications has received increased attention for their role in cell function as small molecules. Examples of these include lovastatin, a cholesterol-lowering agent, and resveratrol, which have multiple biological properties. Both molecules have been reported to improve wound healing and possess antibacterial properties, with conflicting results. The wound-healing capabilities of human mesenchymal stem cells were evaluated after exposure to lovastatin, resveratrol, and their combination through scratch test, migrations assay, and qPCR. Protein docking was performed to assess the lovastatin/resveratrol combination as potential wound-healing targets. AlamarBlue assay was used to determine cell viability. Additionally, the impact of lovastatin and resveratrol combination to inhibit the growth of S. aureus and P. aeruginosa was tested using broth microdilution test and checkerboard assay to determine synergism. The combination of lovastatin 0.1 μM and resveratrol 0.1 μM synergistically improved wound healing and demonstrated an additive effect against S. aureus and P. aeruginosa, presenting potential antibacterial applications.
Collapse
Affiliation(s)
- Norah A. AlJunaydil
- Department of Oral and Maxillofacial Surgery, College of Dentistry, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.A.); (O.G.A.)
| | - Rhodanne Nicole A. Lambarte
- Molecular and Cell Biology Laboratory, Prince Naif bin Abdulaziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.)
| | - Terrence S. Sumague
- Molecular and Cell Biology Laboratory, Prince Naif bin Abdulaziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.)
| | - Osama G. Alghamdi
- Department of Oral and Maxillofacial Surgery, College of Dentistry, King Saud University, Riyadh 11451, Saudi Arabia; (N.A.A.); (O.G.A.)
| | - Abdurahman A. Niazy
- Molecular and Cell Biology Laboratory, Prince Naif bin Abdulaziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.)
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, King Saud University, Riyadh 11545, Saudi Arabia
| |
Collapse
|
25
|
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
|
26
|
Pietrzko MM, Pietrzko M, Niemczyk W, Skaba D, Wiench R. Subgingival Delivery of Statins as an Adjunct in the Non-Surgical Treatment of Periodontitis: A Systematic Review. Biomedicines 2025; 13:182. [PMID: 39857766 PMCID: PMC11761576 DOI: 10.3390/biomedicines13010182] [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: 12/10/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES The gold standard in the non-surgical treatment of periodontitis is scaling and root planning (SRP). In recent years, studies have emerged suggesting additional clinical benefits from the use of statins as an adjunct to classical periodontal disease treatment. The aim of the present study was to review the relevant literature relating to the subgingival use of statins as an adjunctive treatment to the classical, non-surgical treatment of periodontitis, with a particular focus on groups with general factors that may affect the outcome of treatment. METHODS The authors conducted a systematic review following the PRISMA 2020 guidelines. The electronic literature search conducted included the MEDLINE (PubMed) database, Web of Science, Scopus, and Google Scholar from 1 January 2012 to 14 June 2024. The keywords used for the PubMed search were determined with the help of the MeSH Browser Tool and were as follows: Periodontitis [Mesh] AND Statin [Mesh] OR Simvastatin [Mesh] OR Atorvastatin [Mesh] or Rosuvastatin Calcium [Mesh]. Based on the authors' inclusion and exclusion criteria, 20 results were included in the review, out of 937. RESULTS The improvement was more pronounced in patients without systematic diseases compared to those with type II diabetes and in non-smokers compared to smoking patients. Greater improvements in clinical and radiological parameters were seen in patients diagnosed with aggressive periodontitis compared to patients with chronic periodontitis. CONCLUSIONS This literature review led the authors to the conclusion that statins applied locally might be competent agents for improving the therapeutic outcomes of SRP.
Collapse
Affiliation(s)
| | - Maciej Pietrzko
- Pietrzko Stomatology, Budowlanych 1 Street, 43-300 Bielsko-Biała, Poland;
| | - Wojciech Niemczyk
- Department of Periodontal Diseases and Oral Mucosa Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Pl. Traugutta 2, 41-800 Zabrze, Poland; (W.N.); (D.S.); (R.W.)
| | - Dariusz Skaba
- Department of Periodontal Diseases and Oral Mucosa Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Pl. Traugutta 2, 41-800 Zabrze, Poland; (W.N.); (D.S.); (R.W.)
| | - Rafał Wiench
- Department of Periodontal Diseases and Oral Mucosa Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Pl. Traugutta 2, 41-800 Zabrze, Poland; (W.N.); (D.S.); (R.W.)
| |
Collapse
|
27
|
Deng H, Zhou J, Liu Z, Huang L, Gu Y, Chen P, Xiao H. Concomitant medication effects on patients with lung cancer taking immune checkpoint inhibitors a review. Med Oncol 2025; 42:40. [PMID: 39762456 DOI: 10.1007/s12032-024-02591-3] [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: 10/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
In the past decade, a variety of immune checkpoint inhibitors (ICIs) are currently approved for lung cancer in the world. As a new therapeutic approach, ICIs have shown significant clinical benefits in the first-line or second-line treatment for advanced lung cancer, improving the survival and quality of life of patients. Patients need to take multiple drugs in the meantime due to their own disease or side effects during treatment. In view of drug interactions, concomitant medications have a positive or negative impact on the prognosis of lung cancer patients. In this review, we reviewed the effects of multiple drugs on the prognosis of patients with lung cancer taking ICIs. Several studies indicate that antibiotics, proton pump inhibitors (PPIs), corticosteroids, and opioid analgesics can decrease the efficacy of ICIs. Aspirin and bone-targeting drugs can enhance the efficacy of ICIs and improve the survival rate. The effects of metformin (MET), renin-angiotensin-aldosterone system inhibitors (RASI), nonsteroidal anti-inflammatory drug (NSAIDS) (except aspirin), and statins on ICIs are controversial. Future research should further explore the effects of these concomitant medications on ICIs and develop personalized prescriptions based on the specific needs of patients.
Collapse
Affiliation(s)
- Han Deng
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Junxiang Zhou
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Zhixi Liu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Lu Huang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Yanru Gu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Peng Chen
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China
| | - Hongtao Xiao
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55, Section 4, South Renmin Road, Chengdu, Sichuan, China.
| |
Collapse
|
28
|
Wang B, Huang S, Li S, Deng Y, Li Z, Wang Y, Shi X, Zhang W, Shi L, Wang X, Tang X. Hepatotoxicity of statins: a real-world study based on the US Food and Drug Administration Adverse Event Reporting System database. Front Pharmacol 2025; 15:1502791. [PMID: 39840096 PMCID: PMC11747658 DOI: 10.3389/fphar.2024.1502791] [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: 09/27/2024] [Accepted: 12/13/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Statins, as an important class of lipid-lowering drugs, play a key role in the prevention and treatment of cardiovascular diseases. However, with their widespread use in clinical practice, some adverse events have gradually emerged. In particular, the hepatotoxicity associated with statins use has become one of the clinical concerns that require sufficient attention. METHODS In this study, we conducted a comprehensive and detailed analysis of the hepatotoxicity of statins based on the data of the US Food and Drug Administration Adverse Event Reporting System database from the first quarter (Q1) of 2004 to the Q1 of 2024 and used Reporting Odds Ratios and Empirical Bayes Geometric Mean to mine the signal of adverse events. RESULTS In this study, hepatic disorder related seven statins all exhibited positive signals. Through signal mining, we identified a total of 14,511 cases of adverse events associated with hepatic disorder caused by these statin drugs, with atorvastatin, simvastatin, and rosuvastatin occurring at a higher rate. A total of 148 positive signals related to adverse events of hepatic disorder were captured. Autoimmune hepatitis and drug-induced liver injury both presented positive signals across multiple statin drugs. Notably, atorvastatin had the most significant signal strength in cholestatic pruritus and bilirubin conjugation abnormal. Fluvastatin also showed notable signal strength in autoimmune hepatitis, while simvastatin had a relatively weaker signal strength for hepatic enzyme increased. CONCLUSION This study discovered specific adverse event signal values, revealing potential hepatotoxic risks associated with the use of statin drugs. The results provide an important reference for the safe clinical use of drugs, help to improve the understanding of the safety of statins, and also provide a scientific basis for clinicians to make more accurate and safe decisions when making treatment plans.
Collapse
Affiliation(s)
- Bojing Wang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shu Huang
- Department of Gastroenterology, Lianshui County People’ Hospital, Huaian, China
- Department of Gastroenterology, Lianshui People’ Hospital of Kangda College Affiliated to Nanjing Medical University, Huaian, China
| | - Shiqi Li
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yaqi Deng
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ziyan Li
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yizhou Wang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaomin Shi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Zhang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lei Shi
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaohong Wang
- Department of Gastroenterology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Xiaowei Tang
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
29
|
Cortês IT, Silva KDP, Cogo-Müller K. Effects of simvastatin on the mevalonate pathway and cell wall integrity of Staphylococcus aureus. J Appl Microbiol 2025; 136:lxaf012. [PMID: 39788721 DOI: 10.1093/jambio/lxaf012] [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: 10/16/2024] [Revised: 12/21/2024] [Accepted: 01/08/2025] [Indexed: 01/12/2025]
Abstract
AIMS To investigate the effects of simvastatin as an antimicrobial, considering its influence on the mevalonate pathway and the bacterial cell wall of S. aureus. METHODS AND RESULTS S. aureus ATCC 29213 and 33591 were exposed to simvastatin in the presence of exogenous mevalonate to determine whether mevalonate could reverse the inhibition. S. aureus was also treated with simvastatin and gene expression analysis assays were performed to evaluate genes associated with the mevalonate pathway (mvaA, mvaS, mvaK1, and mvaK2), peptidoglycan synthesis (uppS, uppP, and murG), and cell wall stress (vraX, sgtB, and tcaA). Transmission electron microscopy was used to identify the presence of morphological changes. The data were compared using two-way ANOVA and Bonferroni post-test, or the Mann-Whitney test. Addition of exogenous mevalonate was able to partially or completely reverse the inhibition caused by simvastatin. A significant increase of the vraX gene and a reduction of the mvaA gene were observed, together with changes in bacterial morphology. CONCLUSION Simvastatin can exert its antimicrobial effect by means of changes in the cell wall associated with the mevalonate pathway.
Collapse
Affiliation(s)
- Iago Torres Cortês
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Avenida Limeira, 901, Areião, Piracicaba, SP 13414-903, Brazil
| | - Kátia de Pádua Silva
- Universidade Estadual de Campinas, Faculdade de Ciências Farmacêuticas, Rua Cândido Portinari, 200, Cidade Universitária, Campinas, SP 13083-871, Brazil
| | - Karina Cogo-Müller
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Avenida Limeira, 901, Areião, Piracicaba, SP 13414-903, Brazil
- Universidade Estadual de Campinas, Faculdade de Ciências Farmacêuticas, Rua Cândido Portinari, 200, Cidade Universitária, Campinas, SP 13083-871, Brazil
| |
Collapse
|
30
|
Gama F, Meirinho S, Pires PC, Tinoco J, Martins Gaspar MC, Baltazar G, Alves G, Santos AO. Simvastatin is delivered to the brain by high-strength intranasal cationic SMEDDS and nanoemulsions. Drug Deliv Transl Res 2025:10.1007/s13346-024-01769-6. [PMID: 39747745 DOI: 10.1007/s13346-024-01769-6] [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] [Accepted: 12/11/2024] [Indexed: 01/04/2025]
Abstract
The repurposing of statins as neuroprotective agents and/or anti-brain tumor drugs is limited by challenges in brain bioavailability and systemic off-target effects. Therefore, improved and targeted delivery of statins to the brain is necessary. This study aimed to develop a high-strength liquid formulation of the poorly soluble prodrug simvastatin for intranasal administration, as a strategy to achieve high brain concentrations of the prodrug and/or its active form, tenivastatin. Cationic simvastatin nanoemulsions (c-NE) and self-microemulsifying drug delivery systems (c-SMEDDS) were screened for composition, extensively characterized, and the viscosity of the nanoemulsion was further optimized. The optimized c-NE and c-SMEDDS formulations achieved high drug strengths, approximately 5.5% and 9% (w/w), respectively. They formed highly homogeneous aqueous dispersions (polydispersity index < 0.1) with small droplet sizes (< 120 nm and ~ 25 nm, respectively) and remained stable for at least four months under refrigeration. Neither the c-NE nor the c-SMEDDS induced hemolysis up to concentrations of 40 µg/mL and 450 µg/mL of simvastatin, respectively. The zero-shear viscosity of the c-NE was increased to 186 mPa·s by incorporating 0.25% (w/w) polyvinylpyrrolidone, which approached the viscosity of the c-SMEDDS (~ 126 mPa·s). Following intranasal administration of the optimized formulations to Wistar rats at a dose of 10 mg/kg, simvastatin levels in the brain reached 50 to 150 ng/g between 15 and 60 min post-administration. These findings indicate that the developed c-NE and c-SMEDDS formulations hold promise for simvastatin intranasal delivery and brain targeting, potentially paving the way for the realization of simvastatin's neuroprotective potential.
Collapse
Affiliation(s)
- Francisco Gama
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Sara Meirinho
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Patrícia C Pires
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- RISE-Health - Health Research & Innovation, University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal
- Group of Pharmaceutical Technology, Faculty of Pharmacy, REQUIMTE/LAQV, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Johann Tinoco
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Maria Carolina Martins Gaspar
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Graça Baltazar
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- RISE-Health - Health Research & Innovation, University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Gilberto Alves
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
- RISE-Health - Health Research & Innovation, University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal
| | - Adriana O Santos
- Faculty of Health Sciences (FCS), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal.
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal.
- RISE-Health - Health Research & Innovation, University of Beira Interior, Av. Infante D. Henrique, Covilhã, 6200-506, Portugal.
| |
Collapse
|
31
|
Shen HC, Tseng CH, Lin YH, Yeh HY, Tsai HC, Hong SY, Li TH, Su CW, Perng DW, Yang YY, Hou MC. Protective effects of statins on pulmonary function in patients with persistent hyperlipidemia: a retrospective cohort study. Ther Adv Respir Dis 2025; 19:17534666251320875. [PMID: 39989019 PMCID: PMC11848893 DOI: 10.1177/17534666251320875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 01/29/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Pulmonary function tests offer crucial parameters for evaluating lung health and predicting clinical outcomes. Hyperlipidemia, a prevalent metabolic disorder, has been linked to declining pulmonary function. Statins are an essential therapy for lowering lipid levels in hyperlipidemia. OBJECTIVES This study aims to investigate the therapeutic potential of statins in mitigating the decline in pulmonary function. DESIGN This is a retrospective cohort study. METHODS Out of 8286 patients who underwent spirometry testing from January 2018 to December 2020, 492 patients were included in the final analysis. The relationship between statin usage, dosage, along with other biometric indices and spirometry parameters were evaluated. Multivariate logistic regression analyses were employed to assess the association between statin use and the decline in pulmonary function. RESULTS In patients with persistent hyperlipidemia, the use of statins was associated with a higher predicted percentage of forced expiratory volume in 1 second (FEV1) compared to non-users (84.0% vs 78.0%, p = 0.015). Logistic regression models further revealed that statin use independently prevented FEV1 decline, irrespective of dosage (adjusted OR 0.036, 95% CI: 0.002-0.618 in lower statins dose group and adjusted OR 0.170, 95% CI: 0.019-1.552 in higher statins dose group). CONCLUSION The findings suggested that statin usage, regardless of dosage, independently mitigated the decline in pulmonary function among patients with persistent hyperlipidemia. Early initiation of statin therapy may hold promise for individuals experiencing hyperlipidemia and declining pulmonary function.
Collapse
Affiliation(s)
- Hsiao-Chin Shen
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Chest, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Che-Hao Tseng
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Hsuan Lin
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Yun Yeh
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hung-Cheng Tsai
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Allergy, Immunology, Rheumatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shiao-Ya Hong
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-Hao Li
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Shin Kong Wu Ho-Su Memorial Foundation Hospital
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien-Wei Su
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Diahn-Warng Perng
- Department of Chest, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Ying Yang
- Department of Medical Education, Taipei Veterans General Hospital, National Yang Ming Chiao Tung University, #201, Sec. Shih-Pai Road, Taipei 11217, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Chih Hou
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
32
|
Asmar V, Bergman E, Lindhagen E, Sherwood K, Westman G, Gaugaz FZ. Towards streamlined product information: reporting of transporter-mediated drug interactions. Eur J Clin Pharmacol 2025; 81:151-161. [PMID: 39545952 PMCID: PMC11695577 DOI: 10.1007/s00228-024-03772-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE The purpose of this study is to investigate the reporting of risks associated with transporter-mediated drug-drug interactions (DDIs) in medicinal product information and to identify suitable wording for future standardisation of summaries of product characteristics (SmPCs). METHODS The SmPCs of medicinal products approved in the European Union from 2012 to 2023 were screened for warnings on Organic Anion Transporting Polypeptide 1B1 and 1B3 (OATP1B1 and OATP1B3), and Breast Cancer Resistance Protein (BCRP). An in-house search engine for product information was used. Warnings were categorised into different DDI scenarios based on the SmPC texts. RESULTS A total of 192 out of 859 approved medicinal products had SmPC text pertaining to OATP1B1, 1B3 and/or BCRP. The majority of products had text for all three transporters Most texts were located in SmPC Sect. 5.2, followed by Sect. 4.5. Numerous interaction-texts either concluded that the interaction lacked clinical relevance or lacked information on the clinical relevance of the finding. The highest number of SmPC texts indicating a clinically relevant interaction with outlined clinical consequences was found for BCRP. The article also presents SmPC texts for each DDI scenario, which the authors consider as examples of explicit wordings with actionable recommendations. CONCLUSION A potential for improvement of SmPC text for transporter-mediated DDI was identified: Warnings without clinical relevance could be omitted, and some warnings with clinical relevance could be updated to provide actionable recommendations to the prescribers. A selection of unambiguous texts was identified as starting point to generate standard texts.
Collapse
Affiliation(s)
| | - Erik Bergman
- Swedish Medical Products Agency, Uppsala, Sweden
| | | | - Kim Sherwood
- Swedish Medical Products Agency, Uppsala, Sweden
| | | | | |
Collapse
|
33
|
Iturbe-Rey S, Maccali C, Arrese M, Aspichueta P, Oliveira CP, Castro RE, Lapitz A, Izquierdo-Sanchez L, Bujanda L, Perugorria MJ, Banales JM, Rodrigues PM. Lipotoxicity-driven metabolic dysfunction-associated steatotic liver disease (MASLD). Atherosclerosis 2025; 400:119053. [PMID: 39581063 DOI: 10.1016/j.atherosclerosis.2024.119053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/19/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a spectrum of liver lesions, ranging from simple steatosis to metabolic dysfunction-associated steatohepatitis (MASH), that may further progress to cirrhosis. MASLD is estimated to affect more than one third of the general population and it represents a risk factor for end-stage liver failure and liver cancer, substantially contributing to liver-related morbidity and mortality. Although the pathogenesis of MASLD is incompletely understood, it is known to consist of a multifactorial process influenced by extrinsic and intrinsic factors such as metabolic, environmental and demographic features, gut microbiota and genetics. Dysregulation of both extracellular and intracellular lipid composition is known to promote the generation of toxic lipid species, thereby triggering lipotoxicity and cellular stress. These events ultimately lead to the activation of distinct cell death pathways, resulting in inflammation, fibrogenesis and, eventually, carcinogenesis. In this manuscript, we provide a comprehensive review of the role of lipotoxicity during MASLD pathogenesis, discussing the most relevant lipid species and related molecular mechanisms, summarizing the cell type-specific effects and highlighting the most promising putative therapeutic strategies for modulating lipotoxicity and lipid metabolism in MASLD.
Collapse
Affiliation(s)
- Santiago Iturbe-Rey
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain
| | - Claudia Maccali
- Clinical and Experimental Gastroenterology Laboratory LIM-07, Department of Gastroenterology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marco Arrese
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile Santiago, 8330077, Chile
| | - Patricia Aspichueta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain; Biobizkaia Health Research Institute, Cruces University Hospital, 48903, Barakaldo, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain
| | - Claudia P Oliveira
- Clinical and Experimental Gastroenterology Laboratory LIM-07, Department of Gastroenterology, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Division of Clinical Gastroenterology and Hepatology, Hospital das Clínicas de São Paulo, HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Rui E Castro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ainhoa Lapitz
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; Department of Medicine, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940, Leioa, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| | - Pedro M Rodrigues
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, "Instituto de Salud Carlos III"), Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
34
|
El-Said KS, Attia MS, Abdelmoaty BE, Salim EI. Synergistic antitumor effects of atorvastatin and chemotherapies: In vitro and in vivo studies. Biochem Biophys Res Commun 2025; 742:151078. [PMID: 39632292 DOI: 10.1016/j.bbrc.2024.151078] [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: 06/14/2024] [Revised: 10/23/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Atorvastatin (ATOR) acts on certain antitumor pathways; the consequences of chemotherapies continue to be a major concern, notwithstanding the increased efficacy provided by contemporary therapies. This study investigated the synergistic effects and underlying mechanisms of different treatment protocols using ATOR on the THP-1 cell line and on lung cancer in mice. For the in vitro study, an MTT assay was performed, and then different combinations against the THP-1 cell line were used as follows: non-treated cells, THP-1/ATOR IC50, THP-1/cytarabine (CYT) IC50, THP-1/doxorubicin (DOX) IC50, THP-1/DOX/CYT, THP-1/ATOR/CYT, THP-1/ATOR/DOX, and THP-1/ATOR/CYT/DOX. For the in vivo study, CD-1 male mice were used; G1 was the normal control. Gs2-5 were administered with urethane (Ure) and butylated hydroxytoluene (BHT). G2 was the positive control. G3 was treated with ATOR (20 mg/kg). G4 was treated with Bevacizumab (Bev) (5 mg/kg). G5 was co-treated with ATOR/Bev. Histopathological and immunohistochemical investigations, flow cytometry and molecular analysis of PI3K, Akt, and mTOR genes were performed after different treatment protocols. The results showed that different combinatorial treatment settings of ATOR in vitro increase the apoptotic-inducing capacity and cell cycle arrest. Co-treatment with ATOR and Bev led to a significant decrease in S-phase and G2/M percentages. Furthermore, in vivo co-treatment with ATOR/Bev decreased tumor incidence and size with a significant reduction of the immunohistochemical PCNA (LI%) in lung parenchyma, targeting PI3K/Akt/mTOR, and VEGF-A signaling pathways. Co-treatment with ATOR and chemotherapies led to cell cycle arrest, modulation of the PI3K/Akt/mTOR, and VEGF-A signaling pathways in tumor cells.
Collapse
Affiliation(s)
- Karim Samy El-Said
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Merna Saied Attia
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Bassant Ezzat Abdelmoaty
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Elsayed Ibrahim Salim
- Research Lab. of Molecular Carcinogenesis, Zoology Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
35
|
Geng X, Tian W, Zhuang M, Shang H, Gong Z, Li J. Green Radish Polysaccharides Ameliorate Hyperlipidemia in High-Fat-Diet-Induced Mice via Short-Chain Fatty Acids Production and Gut Microbiota Regulation. Foods 2024; 13:4113. [PMID: 39767053 PMCID: PMC11675633 DOI: 10.3390/foods13244113] [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: 11/22/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
The objective of this study was to examine the hypolipidemic effect and potential mechanism of action of green radish polysaccharide (GRP) in hyperlipidemic mice. We found that in mice fed a high-fat diet, supplementing with GRP reduced body weight and liver index, significantly improved serum lipid levels and markers of liver damage, and mitigated oxidative stress and inflammation. Mechanistically, in these hyperlipidemic mice, the size of fat cells was reduced by GRP, and the abnormal accumulation of lipid droplets was reduced. We also found that GRP regulates the composition of the intestinal microbiota, including the ratio of Firmicutes to Mycobacteria F/B and the levels of Blautia spp., which have been shown to alleviate liver damage and treat hyperlipidemia. Metabolite pathway analysis using the Kyoto Encyclopedia of Genes and Genomes identified the glycolysis/glycolytic metabolism and propionate metabolism pathways as potential targets for GRP in the amelioration of hyperlipidemia.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianrong Li
- Department of Food Science and Engineering, College of Food Science and Engineering, Bohai University, Jinzhou 121013, China; (X.G.); (W.T.); (M.Z.); (H.S.); (Z.G.)
| |
Collapse
|
36
|
Gholamalizadeh H, Ensan B, Karav S, Jamialahmadi T, Sahebkar A. Regulatory effects of statins on CCL2/CCR2 axis in cardiovascular diseases: new insight into pleiotropic effects of statins. J Inflamm (Lond) 2024; 21:51. [PMID: 39696507 DOI: 10.1186/s12950-024-00420-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND HMG-CoA reductase inhibitors are well-known medications in the treatment of cardiovascular disorders due to their pleiotropic and lipid-lowering properties. Herein, we reviewed the effects of statins on the CCL2/CCR2 axis. METHOD Scopus and Pubmed databases were systematically searched using the following keywords:" Hydroxymethylglutaryl CoA Reductase Inhibitors"," HMG-CoA Reductase Inhibitors"," Statins", "CCL2, Chemokine", "Monocyte Chemoattractant Protein-1" and "Chemokine (C-C Motif) Ligand 2". Evidence investigating the role of statin on MCP-1 in CVD was identified and bibliographies were completely evaluated to gather further related studies. RESULTS The anti-inflammatory effects of statins on the CCL2/CCR2 pathway have been widely investigated. Despite inconclusive results, a great body of research supports the regulatory roles of statins on this pathway due to their pleiotropic effects. By disrupting the CCL2/CCR2 axis, statins attenuate the infiltration of monocytes and macrophages into the zone of inflammation and hence down-regulate the inflammatory cascades in various CVDs including atherosclerosis, cardiac remodeling, and stroke, among others. CONCLUSION CCL2 plays a major role in the pathogenesis of cardiovascular disorders. Down-regulation of CCL2 is proposed as one of the pleiotropic properties of statins. However, more investigations are required to elucidate which statin in what dose exerts a more potent effect on CCL2/CCR2 pathway.
Collapse
Affiliation(s)
- Hanieh Gholamalizadeh
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Behzad Ensan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Tannaz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Polpichai N, Saowapa S, Jaroenlapnopparat A, Sierra L, Danpanichkul P, Fangsaard P, Wattanachayakul P, Kaewdech A. Statin Use in Metabolic Dysfunction-Associated Steatotic Liver Disease and Effects on Vibration-Controlled Transient Elastography-Derived Scores—A Population-Based Inverse Probability Treatment Weighting Analysis. LIVERS 2024; 4:677-687. [DOI: 10.3390/livers4040046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading cause of chronic liver disease globally. The impact of statins on liver fibrosis severity in MASLD individuals remains uncertain, despite their known cardiovascular benefits. Methods: A cross-sectional study was performed utilizing the National Health and Nutrition Examination Survey (NHANES) database from 2017 to 2018. MASLD was defined by hepatic steatosis (controlled attenuation parameter [CAP] score ≥ 288 dB/m) without other etiologies. Using inverse probability treatment weighting to minimize confounding, we examined the association between statin use and MASLD outcomes, including at-risk steatohepatitis (FibroScan-aspartate aminotransferase [AST] [FAST] score ≥ 0.67), significant and advanced fibrosis (liver stiffness measurement [LSM] ≥ 8.8 kilopascals [kPa] and ≥ 11.7 kPa), and advanced fibrosis (AGILE 3+ score ≥ 0.68). Results: Of 1283 MASLD patients, 376 were prescribed statins within the past 30 days. After adjustment for confounders, statin use was significantly associated with reduced risks of at-risk steatohepatitis, significant fibrosis, and high AGILE 3+ scores, with odds ratios (ORs) of 0.29 (95% CI: 0.01 to 0.87), 0.54 (95% CI: 0.31 to 0.95), and 0.41 (95% CI: 0.22 to 0.75), respectively. However, a subgroup analysis showed this effect persisted only with lipophilic statins. Conclusions: Statin use was associated with reduced steatohepatitis and fibrosis in patients with MASLD, supported by robust causal inference and vibration-controlled transient elastography-derived scores.
Collapse
Affiliation(s)
| | - Sakditad Saowapa
- Department of Medicine, Texas Tech University Health Science Center, Lubbock, TX 79430, USA
| | | | - Leandro Sierra
- Department of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Pojsakorn Danpanichkul
- Department of Medicine, Texas Tech University Health Science Center, Lubbock, TX 79430, USA
| | - Panisara Fangsaard
- Department of Internal Medicine, Bassett Medical Center, Cooperstown, NY 13326, USA
| | | | - Apichat Kaewdech
- Gastroenterology and Hepatology Unit, Division of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
38
|
Singh H, Kaur S, Kaushal P, Sharma J, Singla M. Risk of new onset diabetes mellitus with pitavastatin as compared to atorvastatin and rosuvastatin: a systematic review and meta-analysis. Expert Rev Clin Pharmacol 2024; 17:1173-1181. [PMID: 39587804 DOI: 10.1080/17512433.2024.2433603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Statins are linked to the risk of new-onset diabetes mellitus (NODM). While atorvastatin and rosuvastatin are often associated with NODM, pitavastatin may carry a lower risk. This systematic review and meta-analysis (SRMA) evaluated the impact of pitavastatin on NODM compared to atorvastatin and rosuvastatin. METHODS We conducted a systematic literature search using PubMed, CENTRAL, EMBASE, and ClinicalTrials.gov. Two authors independently screened studies, assessed the risk of bias using Joanna Briggs Institute, Newcastle-Ottawa, and Scottish Intercollegiate Guidelines Network checklists, and extracted data. The analysis was performed using RevMan 5.4.1, and results were represented as risk ratios (RR) with 95% confidence intervals (CI) and heterogeneity was evaluated using the I2 statistic. RESULTS Of 517 records screened, 13 studies were included, comprising observational studies, and randomized controlled trials. Most of the studies showed pitavastatin to be associated with a lower or no risk of NODM. Meta-analysis revealed that pitavastatin had a lower risk of NODM compared to atorvastatin (RR = 0.86, 95% CI = 0.79-0.93, p = 0.0002) and rosuvastatin (RR = 0.77, 95% CI = 0.71-0.84, p < 0.00001). CONCLUSION Pitavastatin poses a lower risk of NODM than other statins, making it a potentially safer option for patients requiring long-term statin therapy. PROTOCOL REGISTRATION www.crd.york.ac.uk/prospero identifier is CRD42022371741.
Collapse
Affiliation(s)
- Harmanjit Singh
- Department of Pharmacology, Government Medical College & Hospital, Chandigarh, India
| | - Sangambir Kaur
- Department of Community Medicine, Government Medical College & Hospital, Chandigarh, India
| | | | - Jatin Sharma
- Department of Pharmacology, AIIMS, New Delhi, India
| | - Mandeep Singla
- Department of Medicine, Government Medical College & Hospital, Chandigarh, India
| |
Collapse
|
39
|
Yaacoub S, Boudaka A, AlKhatib A, Pintus G, Sahebkar A, Kobeissy F, Eid AH. The pharmaco-epigenetics of hypertension: a focus on microRNA. Mol Cell Biochem 2024; 479:3255-3271. [PMID: 38424404 PMCID: PMC11511726 DOI: 10.1007/s11010-024-04947-9] [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: 11/26/2023] [Accepted: 01/20/2024] [Indexed: 03/02/2024]
Abstract
Hypertension is a major harbinger of cardiovascular morbidity and mortality. It predisposes to higher rates of myocardial infarction, chronic kidney failure, stroke, and heart failure than most other risk factors. By 2025, the prevalence of hypertension is projected to reach 1.5 billion people. The pathophysiology of this disease is multifaceted, as it involves nitric oxide and endothelin dysregulation, reactive oxygen species, vascular smooth muscle proliferation, and vessel wall calcification, among others. With the advent of new biomolecular techniques, various studies have elucidated a gaping hole in the etiology and mechanisms of hypertension. Indeed, epigenetics, DNA methylation, histone modification, and microRNA-mediated translational silencing appear to play crucial roles in altering the molecular phenotype into a hypertensive profile. Here, we critically review the experimentally determined associations between microRNA (miRNA) molecules and hypertension pharmacotherapy. Particular attention is given to the epigenetic mechanisms underlying the physiological responses to antihypertensive drugs like candesartan, and other relevant drugs like clopidogrel, aspirin, and statins among others. Furthermore, how miRNA affects the pharmaco-epigenetics of hypertension is especially highlighted.
Collapse
Affiliation(s)
- Serge Yaacoub
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ammar Boudaka
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ali AlKhatib
- Department of Nutrition and Food Sciences, Lebanese International University, Beirut, Lebanon
| | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro, 07100, Sassari, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics and Biomarkers (CNMB), Morehouse School of Medicine, Neuroscience Institute, Atlanta, GA, USA
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
40
|
Millar JE, Craven TH, Shankar-Hari M. Steroids and Immunomodulatory Therapies for Acute Respiratory Distress Syndrome. Clin Chest Med 2024; 45:885-894. [PMID: 39443005 DOI: 10.1016/j.ccm.2024.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by a dysregulated immune response to infection or injury. This framework has driven long-standing interest in immunomodulatory therapies as treatments for ARDS. In this narrative review, we first define what constitutes a dysregulated immune response in ARDS. In this context, we describe the rationale and available evidence for immunomodulatory therapies studied in randomized controlled trials of ARDS patients to date. Finally, we address factors that have contributed to the failure to develop therapies in the past and highlight current and future developments designed to address them.
Collapse
Affiliation(s)
- Jonathan E Millar
- Centre for Inflammation Research, Institute for Repair and Regeneration, University of Edinburgh, Edinburgh EH16 4UU, UK; Department of Critical Care, Intensive Care Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - Thomas H Craven
- Centre for Inflammation Research, Institute for Repair and Regeneration, University of Edinburgh, Edinburgh EH16 4UU, UK; Department of Critical Care, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Manu Shankar-Hari
- Centre for Inflammation Research, Institute for Repair and Regeneration, University of Edinburgh, Edinburgh EH16 4UU, UK; Department of Critical Care, Royal Infirmary of Edinburgh, Edinburgh, UK.
| |
Collapse
|
41
|
Roule V, Alexandre J, Lemaitre A, Chrétien B, Sassier M, Fedrizzi S, Beygui F, Dolladille C. Rhabdomyolysis with Co-Administration of Statins and Antiplatelet Therapies-Analysis of the WHO Pharmacovigilance Database. Cardiovasc Drugs Ther 2024; 38:1191-1199. [PMID: 37115431 DOI: 10.1007/s10557-023-07459-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 04/29/2023]
Abstract
PURPOSE While statins and antiplatelet therapies are largely prescribed together worldwide, limited information is available on the safety of their association regarding rhabdomyolysis occurrence. We aimed to assess the reporting of rhabdomyolysis in patients treated with a combination of statin and antiplatelet therapy, compared to statin alone. METHODS We used the World Health Organization pharmacovigilance database (VigiBase®) to compare the rhabdomyolysis reporting between statin (atorvastatin, fluvastatin, pravastatin, rosuvastatin, and simvastatin) plus antiplatelet therapy (acetylsalicylic acid, clopidogrel, prasugrel and ticagrelor) groups versus statin alone groups, for each statin and antiplatelet therapy. Study setting was restricted to patients aged 45 or older, including reports up until 1st September, 2021. We computed reporting Odds-Ratio (ROR) and their 95% confidence interval (CI) to quantify the disproportionality between groups, adjusted on age and sex. RESULTS Among the 11,431,708 reports of adverse reactions, we extracted 9,489 cases of rhabdomyolysis in patients treated with statins, of whom 2,464 (26%) were also treated with antiplatelet therapy. The reporting of rhabdomyolysis was increased when ticagrelor was associated with atorvastatin (ROR 1.30 [1.02-1.65]) or rosuvastatin (ROR 1.90 [1.42-2.54]) compared to the respective statin alone but did not change when aspirin, clopidogrel or prasugrel were considered. CONCLUSION Rhabdomyolysis reporting was increased when ticagrelor -but not other antiplatelet agents- was notified with the most prescribed statins in practice. This finding needs to be considered by physicians especially in high-risk patients.
Collapse
Affiliation(s)
- Vincent Roule
- Service de Cardiologie, CHU de Caen Normandie, 14000, Caen, France.
- UMRS 1237, INSERM, GIP Cyceron, 14000, Caen, France.
- Cardiology department, Caen University Hospital, Avenue Cote de Nacre, 14033, Caen, France.
| | - Joachim Alexandre
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| | - Adrien Lemaitre
- Service de Cardiologie, CHU de Caen Normandie, 14000, Caen, France
| | - Basile Chrétien
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| | - Marion Sassier
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| | - Sophie Fedrizzi
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| | - Farzin Beygui
- Service de Cardiologie, CHU de Caen Normandie, 14000, Caen, France
- UMRS 1237, INSERM, GIP Cyceron, 14000, Caen, France
- ACTION Study Group, Pitié-Salpêtrière University Hospital, Cardiology Department, Paris, France
| | - Charles Dolladille
- Service de Pharmacologie, CHU de Caen Normandie, PICARO Cardio-Oncology Program, 14000, Caen, France
| |
Collapse
|
42
|
Srinivasan V, Prabu S, Sfeir JG, Muthusamy K. Statin-Induced Necrotizing Autoimmune Myopathy: Diagnosis and Treatment Approach. JCEM CASE REPORTS 2024; 2:luae227. [PMID: 39659392 PMCID: PMC11630794 DOI: 10.1210/jcemcr/luae227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Indexed: 12/12/2024]
Abstract
The widespread use of statins for cardiovascular diseases has unveiled a new subset of inflammatory myopathy, immune-mediated necrotizing myopathy (IMNM). We describe below an unusual case of anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase (anti-HMGCR) myopathy. A 64-year-old male individual with type 2 diabetes, hyperlipidemia, and coronary artery disease presented with progressive proximal muscle weakness and pain for 3 months. He took atorvastatin 40 mg for 4 years, which was discontinued due to elevated liver enzymes and resumed treatment with rosuvastatin 5 mg later due to worsening hyperlipidemia. Physical examination showed significant weakness of the hip, shoulder girdle, and biceps/triceps. Creatinine kinase (CK) was found to be 232.48 µkat/L (13 921 IU/L) (normal: 0.833-5.133 µkat/L; 50-308 IU/L). Electromyography and left vastus lateralis muscle biopsy showed findings of myonecrosis. Anti-HMGCR assay was strongly positive with antibodies > 200 chemiluminescent units (CU) (normal: 0-20 CU). He was started on prednisone followed by human-immunoglobulin (IVIG) which led to a decline in CK. Statin-induced necrotizing autoimmune myopathy (SINAM) is an exceptionally rare side effect of statins. Although statins come with a good side-effect profile, one should be aware of marked, persistent elevations in muscle enzyme levels. Prompt confirmation with antibody levels, drug discontinuation, and early initiation of immunosuppression can lead to good outcomes.
Collapse
Affiliation(s)
- Varshini Srinivasan
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
| | - Samyuktha Prabu
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
| | - Jad G Sfeir
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Kalpana Muthusamy
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
43
|
Spina A, Amone F, Zaccaria V, Insolia V, Perri A, Lofaro D, Puoci F, Nobile V. Citrus bergamia Extract, a Natural Approach for Cholesterol and Lipid Metabolism Management: A Randomized, Double-Blind Placebo-Controlled Clinical Trial. Foods 2024; 13:3883. [PMID: 39682955 DOI: 10.3390/foods13233883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Strategies for controlling cholesterol and lipid metabolism, including the use of food supplements, are part of the non-pharmacological intervention to ameliorate cardiovascular health. To demonstrate the efficacy of a standardized flavonoids (150 mg/day) extract from Citrus bergamia on cholesterol and lipid management, a placebo-controlled clinical trial on 64 subjects with high cholesterol was carried out. The total study duration was 4 months, with intermediate checkpoints at 1-month intervals. Low-density lipoprotein (LDL-C), high-density lipoprotein (HDL-C) cholesterol, total cholesterol (TC) levels, oxidized low-density lipoprotein (ox-LDL), and paraoxonase activity (PON1) were measured as primary endpoints (efficacy evaluation), while weight, blood pressure, hepatic and renal function blood markers were measured as secondary endpoints (safety evaluation). After 4 months, both TC and LDL-C significantly decreased by 8.8% and 11.5%, respectively, along with a 5.5% increase in HDL-C which was trending towards significance. In addition, ox-LDL was significantly reduced by 2.0%, while PON1 was significantly increased by 6.5%. The secondary endpoints were not changed during the study, demonstrating a good tolerability of the test product. Our findings demonstrate the efficacy of the extract as a natural approach for cholesterol and lipid metabolism management.
Collapse
Affiliation(s)
- Amelia Spina
- Nutratech S.r.l., Spin-Off of University of Calabria, 87036 Rende, Italy
| | - Fabio Amone
- Nutratech S.r.l., Spin-Off of University of Calabria, 87036 Rende, Italy
| | | | | | - Anna Perri
- Department of Experimental and Clinical Medicine, Magna Grecia University of Catanzaro, 88100 Catanzaro, Italy
| | - Danilo Lofaro
- Department of Mechanical, Energy, Management Engineering, University of Calabria, 87036 Rende, Italy
| | - Francesco Puoci
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87100 Cosenza, Italy
| | - Vincenzo Nobile
- R&D Department, Complife Italia S.r.l., 27028 San Martino Siccomario, Italy
| |
Collapse
|
44
|
von Känel-Cordoba I, Wirnitzer K, Weiss K, Nikolaidis PT, Devrim-Lanpir A, Hill L, Rosemann T, Knechtle B. Efficacy, side effects, adherence, affordability, and procurement of dietary supplements for treating hypercholesterolemia: a narrative review. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:189. [PMID: 39563466 PMCID: PMC11575005 DOI: 10.1186/s41043-024-00679-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/03/2024] [Indexed: 11/21/2024]
Abstract
INTRODUCTION Statins are effective in reducing high cholesterol levels; however, due to associated side effects, many patients actively seek alternative medications. This review evaluates the efficacy, side effects, patient adherence, cost-effectiveness, and accessibility of dietary supplements (DS) as a treatment option for hypercholesterolemia. METHODS This narrative review compares red yeast rice (RYR), flaxseed, artichokes, bergamot, Ayurvedic mixtures (with garlic as a prominent ingredient), and statins for treating hypercholesterolemia. We searched PubMed, Scopus, and Cochrane databases for studies published between 2012 and 2024 using "hypercholesterolemia" in combination with a dietary supplement (red yeast rice, flaxseed, artichokes, garlic, or bergamot). The selected articles were published until 28th January 2024 with no language restrictions. RESULTS Study results suggest that alternative treatments using dietary supplements such as flaxseed, bergamot, or red yeast rice may effectively reduce cholesterol levels, with the specific value varying based on the study. CONCLUSION The following natural ingredients - red yeast rice, artichoke extract, bergamot, garlic, and flaxseed - have been specifically selected for their cholesterol-lowering properties. Based on consistent usage, except for aged garlic extract, these ingredients appear to have a beneficial impact on cholesterol levels. (1) It is advisable to conduct a comprehensive cohort study to assess the efficacy of relevant dietary supplements, particularly red yeast rice, bergamot, and flaxseed, in treating hypercholesterolemia. This is important due to the varying effectiveness of alternative treatments. However, there are lingering concerns regarding the lack of supervision and quality control that require attention. (2) Further research into the specific molecular composition and the underlying mechanisms by which it reduces cholesterol levels is warranted.
Collapse
Affiliation(s)
| | - Katharina Wirnitzer
- Department of Pediatric Oncology and Hematology, Otto-Heubner Centre for Paediatric and Adolescent Medicine (OHC), Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Sport Science, Leopold-Franzens University of Innsbruck, Fürstenweg 185, Innsbruck, 6020, Austria
- Research Center Medical Humanities, Leopold-Franzens University of Innsbruck, Innrain 52, Innsbruck, 6020, Austria
- Department of Research and Development in Teacher Education, University College of Teacher Education Tyrol, Pastorstraße 7, Innsbruck, 6010, Austria
| | - Katja Weiss
- Institute of Primary Care, University of Zurich, Zurich, Switzerland
| | | | - Asli Devrim-Lanpir
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
- Department of Nutrition and Dietetics, Istanbul Medeniyet University, Istanbul, Turkey
| | - Lee Hill
- Dept. of Pediatrics, Research Institute, McGill University Health Centre, Montreal, Canada
| | - Thomas Rosemann
- Institute of Primary Care, University of Zurich, Zurich, Switzerland
| | - Beat Knechtle
- Institute of Primary Care, University of Zurich, Zurich, Switzerland.
- Medbase St. Gallen Am Vadianplatz, St. Gallen, Switzerland.
| |
Collapse
|
45
|
Ghosh R, Herberg S. The role of YAP/TAZ mechanosignaling in trabecular meshwork and Schlemm's canal cell dysfunction. Vision Res 2024; 224:108477. [PMID: 39208753 PMCID: PMC11470804 DOI: 10.1016/j.visres.2024.108477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
This focused review highlights the importance of yes-associated protein (YAP)/transcriptional coactivator with PDZ binding motif (TAZ) mechanosignaling in human trabecular meshwork and Schlemm's canal cells in response to glaucoma-associated extracellular matrix stiffening and cyclic mechanical stretch, as well as biochemical pathway modulators (with signaling crosstalk) including transforming growth factor beta 2, glucocorticoids, Wnt, lysophosphatidic acid, vascular endothelial growth factor, and oxidative stress. We provide a comprehensive overview of relevant literature from the last decade, highlight intriguing research avenues with translational potential, and close with an outlook on future directions.
Collapse
Affiliation(s)
- Rajanya Ghosh
- Department of Ophthalmology and Visual Sciences, Center for Vision Research, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences, Center for Vision Research, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA; BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY 13244, USA.
| |
Collapse
|
46
|
Keshavarz Shahbaz S, Koushki K, Keshavarz Hedayati S, McCloskey AP, Kesharwani P, Naderi Y, Sahebkar A. Polymer nanotherapeutics: A promising approach toward microglial inhibition in neurodegenerative diseases. Med Res Rev 2024; 44:2793-2824. [PMID: 39031446 DOI: 10.1002/med.22064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 01/30/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
Nanoparticles (NPs) that target multiple transport mechanisms facilitate targeted delivery of active therapeutic agents to the central nervous system (CNS) and improve therapeutic transport and efficacy across the blood-brain barrier (BBB). CNS nanotherapeutics mostly target neurons and endothelial cells, however, microglial immune cells are the first line of defense against neuronal damage and brain infections. Through triggering release of inflammatory cytokines, chemokines and proteases, microglia can however precipitate neurological damage-a significant factor in neurodegenerative diseases. Thus, microglial inhibitory agents are attracting much attention among those researching and developing novel treatments for neurodegenerative disorders. The most established inhibitors of microglia investigated to date are resveratrol, curcumin, quercetin, and minocycline. Thus, there is great interest in developing novel agents that can bypass or easily cross the BBB. One such approach is the use of modified-nanocarriers as, or for, delivery of, therapeutic agents to the brain and wider CNS. For microglial inhibition, polymeric NPs are the preferred vehicles for choice. Here, we summarize the immunologic and neuroinflammatory role of microglia, established microglia inhibitor agents, challenges of CNS drug delivery, and the nanotherapeutics explored for microglia inhibition to date. We also discuss applications of the currently considered "most useful" polymeric NPs for microglial-inhibitor drug delivery in CNS-related diseases.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Khadije Koushki
- Department of Neurosurgery, University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | | | - Alice P McCloskey
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Yazdan Naderi
- Department of Pharmacology, Faculty of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
47
|
Gomes Daré R, Beatriz Chieco Costa A, Silva Martins T, Lopes LB. Simvastatin and adenosine-co-loaded nanostructured lipid carriers for wound healing: Development, characterization and cell-based investigation. Eur J Pharm Biopharm 2024:114533. [PMID: 39414092 DOI: 10.1016/j.ejpb.2024.114533] [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: 08/13/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/18/2024]
Abstract
Chronic wounds represent a significant global health burden, characterized by delayed skin healing and associated comorbidities. The present study aimed to develop nanostructured lipid carriers (NLCs) as a topical delivery system for the co-administration of simvastatin and adenosine to address chronic wound management. The rationale behind the co-delivery approach was to mitigate the cytotoxicity associated with high-dose simvastatin, while preserving its therapeutic benefits through a potential synergistic or additive effect. A significant challenge in the development of these NLCs was the encapsulation of the highly hydrophilic adenosine within the hydrophobic lipid matrix. The NLCs were prepared using a hot homogenization-sonication method with a double emulsion technique and optimized through a series of formulation trials, employing various surfactants, solid and liquid lipids, to achieve efficient drug encapsulation, particularly for the hydrophilic adenosine. Optimized formulations F5- and F10-S/A 0.6 %/2 % (containing 0.6 % simvastatin and 2 % adenosine), exhibited promising physicochemical properties. The main difference was the liquid lipid used: F5 containing Miglyol 810 N, while F10 Capmul MCM C-8. Both formulations displayed a mean particle size below 230 nm, a polydispersity index (PDI) of approximately 0.2, and a zeta potential of around -22 mV. While simvastatin association efficiency (AE) was nearly 100 %, adenosine AE was higher for F10 (24 %), compared to F5 (13.5 %). F5 demonstrated superior stability compared to F10, maintaining consistent particle size and PDI over a 60-day period. Formulation F5 also demonstrated superior cell-based in vitro performance compared to F10, with higher cell viability (MTT assay), greater cell proliferation induction (SRB assay), and enhanced cell proliferation and migration in the wound-scratch assay. While F10 displayed higher adenosine AE, F5 excelled in terms of stability and biological activity. The slightly increase in intracellular reactive oxygen species levels observed with F5 may contribute to its enhanced proliferative effects. In-depth characterization revealed that F5 comprised spherical nanoparticles, and thermal analysis indicated no significant changes in the nanocarrier structure upon drug encapsulation. Additionally, ex vivo permeability study demonstrated superior skin retention of both simvastatin and adenosine for F5 compared to an emulsion control. Overall, the F5 nanocarrier demonstrated suitable physicochemical properties, cellular biocompatibility, induction of cell proliferation and migration events, and drug retention capacity in the skin layers, indicating its potential as a promising topical treatment for difficult-to-heal wounds.
Collapse
Affiliation(s)
- Regina Gomes Daré
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil.
| | - Ana Beatriz Chieco Costa
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil
| | - Tereza Silva Martins
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, 210 São Nicolau Street, 09913-030 Diadema, SP, Brazil
| | - Luciana B Lopes
- Institute of Biomedical Sciences, University of São Paulo, 1524 Professor Lineu Prestes Avenue, 05508-000 São Paulo, SP, Brazil.
| |
Collapse
|
48
|
Henry JP, Gabriel L, Luchian ML, Higny J, Benoit M, Xhaët O, Blommaert D, Telbis AM, Robaye B, Guedes A, Demeure F. Evaluating the Efficacy of a Pre-Established Lipid-Lowering Algorithm in Managing Hypercholesterolemia in Patients at Very High Cardiovascular Risk. J Pers Med 2024; 14:1044. [PMID: 39452551 PMCID: PMC11509033 DOI: 10.3390/jpm14101044] [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: 09/13/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Recent data from European studies (EUROASPIRE V, DA VINCI, SANTORINI) indicate that achieving the LDL cholesterol (LDL-C) target in patients at very high cardiovascular risk is uncommon. Additionally, using a combination therapy involving statins and ezetimibe remains infrequent. METHODS A single-center assessment of a pre-defined lipid lowering treatment algorithm's effectiveness at achieving the LDL-C target in patients at very high cardiovascular risk one month and one year after hospitalization. RESULTS 81 patients were included, all in secondary prevention. The average age of the patient was 66.9 years, and the main cardiovascular risk factors included hypertension, diabetes mellitus, and smoking history. Following the predefined lipid-lowering algorithm specific to our study, which involves initiating high-intensity statin therapy or a combination of statin and ezetimibe depending on initial LDL-C levels and patient history; 30 (37%) patients initiated high-intensity statin therapy (Atorvastatin (40 mg, 80 mg) or Rosuvastatin (20 mg, 40 mg)), while 51 (63%) started combination therapy with high-intensity statin and ezetimibe 10 mg. After one year, 57 (70.4%) remained adherent to their initial treatment, achieving a mean LDL-C of 49.5 ± 16.9 mg/dL, with 36 (63.2%) of them reaching the LDL-C target of <55 mg/dL. A total of 13 patients discontinued treatment, and 9 were lost to follow-up, withdrew from the study, or died. CONCLUSION Initiating dual statin and ezetimibe therapy or high-intensity statin therapy early, based on the expected treatment efficacy, holds the potential to more rapidly and effectively achieve LDL-C targets in a larger proportion of very high-risk cardiovascular patients.
Collapse
Affiliation(s)
- Jean Philippe Henry
- Department of Cardiology, Université Catholique de Louvain, CHU UCL Namur, 5530 Yvoir, Belgium; (L.G.); (M.-L.L.); (J.H.); (M.B.); (O.X.); (D.B.); (A.-M.T.); (B.R.); (A.G.); (F.D.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lin YC, Lai TS, Chen YT, Chou YH, Chen YM, Hung KY, Tu YK. Comparative efficacy and choice of lipid-lowering drugs for cardiovascular and kidney outcomes in patients with chronic kidney disease: A systematic review and network meta-analysis. J Formos Med Assoc 2024:S0929-6646(24)00474-1. [PMID: 39389802 DOI: 10.1016/j.jfma.2024.09.037] [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: 08/08/2024] [Revised: 09/07/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND The effect of exact classes of lipid-lowering drugs (LLDs) on preventing major adverse cardiovascular events (MACEs) and poor renal outcomes is not well characterized in the chronic kidney disease (CKD) population. METHODS We performed a frequentist random-effects network meta-analysis of randomized controlled trials (RCTs) to evaluate the protective effect of the LLDs in non-dialysis CKD patients. The PubMed, Embase, Web of Science, and Cochrane Library databases were systematically searched for relevant trials published before March 31, 2024. The primary outcome was the incidence of MACEs. The secondary outcomes comprised all-cause mortality, end-stage kidney disease, changes in estimated glomerular filtration rate (eGFR) and proteinuria, and safety. RESULTS Forty-nine eligible RCTs with 77,826 participants with non-dialysis CKD were included. With moderate confidence in the evidence, rosuvastatin and atorvastatin showed statistically significantly more efficacy in reducing the risk of MACE, with a pooled risk ratio of 0.55 (95% CI 0.33-0.91) for rosuvastatin and 0.67 (0.49-0.90) for atorvastatin, respectively, compared with the control group. For the change in the eGFR, atorvastatin (mean difference [MD], 1.40; 95% CI, 0.61 to 2.18), rosuvastatin (MD, 1.73; 95% CI, 0.63 to 2.83), and statin plus ezetimibe (MD, 2.35; 95% CI, 0.44 to 4.26) showed statistically significant increases in the mean eGFR. CONCLUSION In patients with non-dialysis CKD, there is sufficient evidence to show that rosuvastatin and atorvastatin were statistically significantly more effective and preferable in reducing the risk of MACE and increasing the mean eGFR compared with the control group.
Collapse
Affiliation(s)
- Yi-Chih Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Medicine, National Taiwan University Hospital Jinshan Branch, New Taipei City, Taiwan
| | - Tai-Shuan Lai
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Yi-Ting Chen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Hsiang Chou
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yung-Ming Chen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuan-Yu Hung
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Kang Tu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan; Health Data Research Center, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
50
|
Wu J, Chen T, Zhang M, Li X, Fu R, Xu J, Nüssler A, Gu C. Atorvastatin exerts a preventive effect against steroid-induced necrosis of the femoral head by modulating Wnt5a release. Arch Toxicol 2024; 98:3365-3380. [PMID: 38971901 DOI: 10.1007/s00204-024-03817-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 06/27/2024] [Indexed: 07/08/2024]
Abstract
Steroid-induced osteonecrosis of the femoral head (SONFH) is a prevalent form of osteonecrosis in young individuals. More efficacious clinical strategies must be used to prevent and treat this condition. One of the mechanisms through which SONFH operates is the disruption of normal differentiation in bone marrow adipocytes and osteoblasts due to prolonged and extensive use of glucocorticoids (GCs). In vitro, it was observed that atorvastatin (ATO) effectively suppressed the impact of dexamethasone (DEX) on bone marrow mesenchymal stem cells (BMSCs), specifically by augmenting their lipogenic differentiation while impeding their osteogenic differentiation. To investigate the underlying mechanisms further, we conducted transcriptome sequencing of BMSCs subjected to different treatments, leading to the identification of Wnt5a as a crucial gene regulated by ATO. The analyses showed that ATO exhibited the ability to enhance the expression of Wnt5a and modulate the MAPK pathway while regulating the Wnt canonical signaling pathway via the WNT5A/LRP5 pathway. Our experimental findings provide further evidence that the combined treatment of ATO and DEX effectively mitigates the effects of DEX, resulting in the upregulation of osteogenic genes (Runx2, Alpl, Tnfrsf11b, Ctnnb1, Col1a) and the downregulation of adipogenic genes (Pparg, Cebpb, Lpl), meanwhile leading to the upregulation of Wnt5a expression. So, this study offers valuable insights into the potential mechanism by which ATO can be utilized in the prevention of SONFH, thereby holding significant implications for the prevention and treatment of SONFH in clinical settings.
Collapse
Affiliation(s)
- Junfeng Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tao Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Minghang Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xing Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou, China
| | - Rongkun Fu
- Department of Zhengzhou University Clinical Medicine, Zhengzhou, China
| | - Jianzhong Xu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Andreas Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Schnarrenbergstr. 95, 72076, Tübingen, Germany
| | - Chenxi Gu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|