1
|
Fawaz A, Mohammed MM, Ismail A, Rani KGA, Samsudin AR. The influence of simvastatin on osteoblast functionality in the presence of titanium dioxide particles In-vitro. Arch Oral Biol 2024; 167:106065. [PMID: 39146660 DOI: 10.1016/j.archoralbio.2024.106065] [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: 03/17/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVE Leaching of particles from dental titanium implant surfaces into preimplant microenvironment causes detrimental effects on bone cells. The current study investigated influence of simvastatin in mitigating adverse pro-inflammatory effects of titanium dioxide (TiO2) micro (MP) and nano (NP) particles on hFOB 1.19 cells in vitro. DESIGN Viability of hFOB 1.19 cells following exposure to varying concentrations of TiO2 MPs and NPs and simvastatin were measured by XTT assay. hFOB 1.19 cells were treated with 100 µg/mL of TiO2 MPs, 100 µg/mL of TiO2 NPs, 0.1 µM simvastatin, 100 µg/mL of TiO2 MPs+ 0.1 µM simvastatin and 100 µg/mL of TiO2 NPs+ 0.1 µM simvastatin. After 24 h, ROS was measured by flow cytometry. On day 14, real-time PCR analysis for pro-inflammatory cytokines and bone formation markers was done for TNFα, IL1β, osteocalcin, ALP, and Col1 markers; while ALP and RANKL/OPG ratio were determined by colorimetric and ELISA assays respectively. Further, mineralization study using Alizarin Red S staining (ARS) and calcium quantification were performed. RESULTS Exposure of hFOB to TiO2 MPs and NPs generated ROS and reduced cell viability significantly, with upregulation of pro-inflammatory markers TNFα and IL1β and downregulation of bone formation markers OC and increased RANKL/OPG ratio and lowered degree of mineralization. Treatment with 0.1 µM of simvastatin treatment reversed the effects by mitigating oxidative stress, dampening pro-inflammatory markers, upregulation of bone formation markers, lowering RANKL/OPG ratio and increasing degree of mineralization. CONCLUSION Simvastatin possesses antioxidant, anti-inflammatory, and pro-osteogenic properties that may support bone healing around titanium implants.
Collapse
Affiliation(s)
- Ahmad Fawaz
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Marwan Mansoor Mohammed
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Asmaa Ismail
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - K G Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - A R Samsudin
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|
2
|
Davuluri KS, Singh AK, Singh AV, Chaudhary P, Raman SK, Kushwaha S, Singh SV, Chauhan DS. Atorvastatin Potentially Reduces Mycobacterial Severity through Its Action on Lipoarabinomannan and Drug Permeability in Granulomas. Microbiol Spectr 2023; 11:e0319722. [PMID: 36719189 PMCID: PMC10100658 DOI: 10.1128/spectrum.03197-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 11/05/2022] [Indexed: 02/01/2023] Open
Abstract
The majority of preclinical research has shown that Mycobacterium tuberculosis can modify host lipids in various ways. To boost its intramacrophage survival, M. tuberculosis causes host lipids to build up, resulting in the development of lipid-laden foam cells. M. tuberculosis binds to and enters the macrophage via the cell membrane cholesterol. Aggregation of cholesterol in the cell wall of M. tuberculosis and an increase in vascularity at the granuloma site reduce the permeability of rifampicin and isoniazid concentrations. However, very few studies have assessed the effect of statins on drug penetration. Here, we used atorvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, to observe its effect on the bacterial burden by increasing the drug concentration at the infection site. We looked into how atorvastatin could be used in conjunction with first-line drugs to promote drug permeation. In this study, we detected an accumulation of drugs at the peripheral sites of the lungs and impaired drug distribution to the diseased sites. The efficacy of antituberculosis drugs, with atorvastatin as an adjunct, on the viability of M. tuberculosis cells was demonstrated. A nontoxic statin dosage established phenotypic and normal granuloma vasculature and showed an additive effect with rifampicin and isoniazid. Our data show that statins help to reduce the tuberculosis bacterial burden. Our findings reveal that the bacterial load is connected with impaired drug permeability resulting from lipid accumulation in the bacterial cell wall. Statin therapy combined with antituberculosis medications have the potential to improve treatment in tuberculosis patients. IMPORTANCE Mycobacterium tuberculosis binds to and enters the macrophage via the cell membrane cholesterol. M. tuberculosis limits phagosomal maturation and activation without engaging in phagocytosis. Aggregation of cholesterol in the cell wall of M. tuberculosis and an increase in the vascularity at the granuloma site reduce the permeability of rifampicin and isoniazid concentrations. However, very few studies have assessed the effect of statins on drug penetration, which can be increased through a reduction in cholesterol and vascularity. Herein, we used atorvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, to observe its effect on bacterial burden through increasing the drug concentration at the infection site. Our main research goal is to diminish mycobacterial dissemination and attenuate bacterial growth by increasing drug permeability.
Collapse
Affiliation(s)
- Kusuma Sai Davuluri
- Department of Microbiology and Molecular Biology, ICMR, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India
| | - Amit Kumar Singh
- Department of Microbiology and Molecular Biology, ICMR, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India
| | - Ajay Vir Singh
- Department of Microbiology and Molecular Biology, ICMR, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India
| | - Pooja Chaudhary
- Department of Microbiology and Molecular Biology, ICMR, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India
| | - Sunil Kumar Raman
- Division of Pharmaceutics and Pharmacokinetics, CSIR, Central Drug Research Institute, Lucknow, India
| | - Shweta Kushwaha
- Department of Microbiology and Molecular Biology, ICMR, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India
| | | | - Devendra Singh Chauhan
- Department of Microbiology and Molecular Biology, ICMR, National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra, India
| |
Collapse
|
3
|
Averbukh LD, Turshudzhyan A, Wu DC, Wu GY. Statin-induced Liver Injury Patterns: A Clinical Review. J Clin Transl Hepatol 2022; 10:543-552. [PMID: 35836753 PMCID: PMC9240239 DOI: 10.14218/jcth.2021.00271] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/09/2021] [Accepted: 11/08/2021] [Indexed: 12/04/2022] Open
Abstract
Since their introduction in 1987, hydroxymethyl glutaryl coenzyme A reductase (HMG-CoA) inhibitors, more commonly known as statins, have become some of the most widely prescribed medications in the world. Though generally considered to be safe and well tolerated, statins have been associated with several side effects including mild liver dysfunction manifested by increases in aminotransferases. Rarely, statins have been noted to induce more serious hepatic injury, including liver injury with autoimmune features. Current literature supports statin induced liver injury presenting in either hepatocellular or cholestatic patterns, though with the former being the prevailing pattern of injury. Fortunately, severe liver injury is uncommon with statin use and is generally reversible without any intervention other than offending statin cessation. When evaluating cases of suspected statin-induced liver injury, a complete medical history, laboratory tests including a complete metabolic panel, autoimmune markers, and viral panel, as well as hepatic imaging, are crucial for a complete causality analysis with validated tools such as Roussel Uclaf Causality Assessment Method. The aim of this review is to review the current evidence for statin-induced liver injury and cholestasis.
Collapse
Affiliation(s)
- Leon D. Averbukh
- Department of Medicine, Division of Gastroenterology-Hepatology, Allegheny Health Network, Pittsburgh, PA, USA
- Correspondence to: Leon D. Averbukh, Allegheny Health Network, 320 East North Avenue, 7th Floor, South Tower, Pittsburgh, PA 15212, USA. ORCID: https://orcid.org/0000-0002-7739-7689. Tel: +1-412-359-3846, Fax: +1-412-442-2139, E-mail:
| | - Alla Turshudzhyan
- Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, USA
| | - David C. Wu
- Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, USA
| | - George Y. Wu
- Department of Medicine, Division of Gastroenterology-Hepatology, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
4
|
Ibrahim SSA, Kandil LS, Ragab GM, El-Sayyad SM. Micro RNAs 26b, 20a inversely correlate with GSK-3 β/NF-κB/NLRP-3 pathway to highlight the additive promising effects of atorvastatin and quercetin in experimental induced arthritis. Int Immunopharmacol 2021; 99:108042. [PMID: 34426107 DOI: 10.1016/j.intimp.2021.108042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 12/23/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease with challenging therapeutic potential due to the implication of cross-talking intracellular pathways in the pathogenesis of the disease. This study aimed to evaluate the effects of the combination therapy of atorvastatin and quercetin on glycogen synthase kinase-3 beta/ nuclear factor kappa-B/ nucleotide-binding oligomerization domain-like receptor family pyrin domain containing-3 or inflammasome (GSK-3β/NF-KB/NLRP-3) pathway as well as on microRNAs 26b and 20a (miR-26b, miR-20a) and to investigate the possible beneficial outcomes of the combination to offer a better treatment option than methotrexate (MTX) in adjuvant-induced arthritis (AIA). Assessment of arthritis progression, serum inflammatory, and oxidative parameters were done. The tibiotarsal tissue expression of the inflammatory parameters was evaluated. Western blot analysis was done to assess the expression level of the important members in the GSK-3β/NF-κB/NLRP-3 pathway. Furthermore, the expression level of both microRNAs and serum level of transaminases were determined. All treatments, especially the combination regimen, abated arthritis progression, the elevated serum level of inflammatory and oxidative stress parameters in arthritic rats. Moreover, They down-regulated the gene expression of the important members of the aforementioned signaling pathway, amended the tissue levels of inflammatory parameters and elevated the expression level of miR-26b and miR-20a. Finally, we concluded that the combination therapy modulated miR-26b and miR-20a as well as GSK-3β/NF-κB/NLRP-3 pathway, provided additive anti-inflammatory and anti-oxidant effects and offered an additional hepatoprotective effect as compared to untreated arthritic rats and MTX-treated groups, suggesting its promising role to be used as replacement therapy to MTX in RA.
Collapse
Affiliation(s)
| | - Lamia Said Kandil
- Department of Pharmacology & Therapeutics, Pharos University in Alexandria, Alexandria, Egypt; Department of Biochemistry, School of Biological Sciences, University of East Anglia, England
| | - Ghada M Ragab
- Department of Pharmacology and Toxicology, Misr University for Science and Technology, 6(th) of October City, Egypt
| | - Shorouk M El-Sayyad
- Department of Pharmacology & Toxicology, October 6 University, 12585, Giza, Egypt
| |
Collapse
|
5
|
Pongpunna S, Pratipanawatr T, Wongtaweepkij K, Jarernsiripornkul N. Evaluation of Patient's Knowledge of Atorvastatin Information in Patient Information Leaflets: A Pre-Post Intervention Study in Thailand. Patient Prefer Adherence 2021; 15:2377-2387. [PMID: 34737553 PMCID: PMC8560174 DOI: 10.2147/ppa.s334668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/12/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE This study aimed to evaluate the effect of using atorvastatin PIL on patients' medication knowledge, perceptions of the PIL usefulness, their anxiety about the medication, and factors related to these aspects. PATIENTS AND METHODS A pre-post intervention study was conducted in outpatients at a university hospital. Patients prescribed atorvastatin were enrolled using systematic random sampling. Participants were asked to complete Self-Administered Questionnaire to assess atorvastatin knowledge at baseline. An atorvastatin-PIL produced by the manufacturer was introduced to the participants as the intervention. One month after receiving the PIL, the participants were re-assessed. Ten questions were developed to assess atorvastatin knowledge and visual analog scale (VAS) was used to assess perceived benefits of using the PIL and patient anxiety about the medication. Multiple linear regression was used to assess the related factors. RESULTS Of 450 questionnaires distributed, 370 were returned. Atorvastatin knowledge significantly increased with mean score of 5.06±1.92 at baseline to 8.34±1.79 at 1-month after intervention. Reading all sections of the PIL (p=0.017) and working for civil service (p=0.006) were associated with higher knowledge scores at baseline and after intervention. Low educational level was associated with lower knowledge scores at baseline (p=0.002), but experience of allergy (p=0.042) was associated with higher knowledge scores after the intervention. Patients had high level of perceived usefulness from the PIL (average scores=8.87±1.83) and low level of anxiety (average scores=3.69±3.06). Reading all sections of the PIL (p=0.007) and taking more than 5 medications (p=0.012) were related to perceived usefulness of the PIL. Females (p<0.001) and herbal supplement users (p=0.048) were related to anxiety about the medication. CONCLUSION PILs could improve medication knowledge in patients. Patients' perceptions of benefits of PILs were high and anxiety about medication was low. Use of PILs should be encouraged to improve patients' knowledge and appropriate use of medications.
Collapse
Affiliation(s)
- Supawinee Pongpunna
- Division of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | | | - Kamonphat Wongtaweepkij
- Division of Clinical Pharmacy, Faculty of Pharmacy, Srinakharinwirot University, Nakhon Nayok, Thailand
| | - Narumol Jarernsiripornkul
- Division of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
- Correspondence: Narumol Jarernsiripornkul Division of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, ThailandTel +66-4334-8353Fax +66-4320-2379 Email
| |
Collapse
|
6
|
Eltellawy YA, El-Kayal M, Abdel-Rahman RF, Salah S, Shaker DS. Optimization of transdermal atorvastatin calcium - Loaded proniosomes: Restoring lipid profile and alleviating hepatotoxicity in poloxamer 407-induced hyperlipidemia. Int J Pharm 2020; 593:120163. [PMID: 33309831 DOI: 10.1016/j.ijpharm.2020.120163] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/28/2020] [Accepted: 12/05/2020] [Indexed: 01/22/2023]
Abstract
In an attempt to optimize the anti- hyperlipidemic effect and reduce statins induced hepatotoxicity, Atorvastatin Calcium (ATC) transdermal proniosomal gel (PNG) was developed. Different non-ionic surfactants (NISs) (Spans, Tweens, Cremophor RH 40 and Brij 52) were incorporated in the vesicle's lipid bilayer, in combination with lecithin. PNG formulae were characterized for encapsulation efficiency percent (% EE), vesicle size, polydispersity index (PDI) and zeta potential (ZP). Ex-vivo permeation study was performed using full thickness rat skin measuring drug flux and skin permeability coefficients. The pharmacodynamic performance of optimized transdermal ATC- PNG on both lipid profile and liver biomarkers was assessed and compared to oral ATC administration in poloxamer 407-induced hyperlipidemic rats. The liver tissues were subjected to histological examination as well. The results revealed nano-size range vesicles with relatively high ATC entrapment efficiency. Ex-vivo results demonstrated the permeation superiority of ATC proniosomes over free drug. Pharmacodynamic study revealed that transdermal administration of ATC- PNG succeeded in retaining the anti-hyperlipidemic efficacy of orally administered ATC without elevating liver biomarkers. The histological examination signified the role of optimized ATC-PNG in hindering statin- induced hepatocellular damage. The obtained results suggested a promising, easy-to-manufacture and effective ATC proniosomal gel for safe treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Yasmin A Eltellawy
- Department of Pharmaceutics & Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt (FUE), Cairo, Egypt.
| | - Maha El-Kayal
- Department of Pharmaceutics & Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt (FUE), Cairo, Egypt
| | | | - Salwa Salah
- Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Dalia S Shaker
- Department of Pharmaceutics & Pharmaceutical Technology, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt (FUE), Cairo, Egypt.
| |
Collapse
|
7
|
Hepatoprotective Effects of a Novel Trihoney against Nonalcoholic Fatty Liver Disease: A Comparative Study with Atorvastatin. ScientificWorldJournal 2020; 2020:4503253. [PMID: 33132768 PMCID: PMC7568805 DOI: 10.1155/2020/4503253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 09/07/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disorder worldwide with no curative therapy. The aim of this study was to investigate the hepatoprotective effects of a novel Trihoney against biochemical and histological manifestations of NAFLD in hypercholesterolemic rabbits. Methodology. Forty-eight male New Zealand white (NZW) rabbits were grouped into normal diet (C), normal diet with 0.6 g/kg/day of Trihoney (C + H), 1% cholesterol diet (HCD), 1% cholesterol diet with 0.3 g/kg/day of Trihoney (HCD + H1), 1% cholesterol diet with 0.6 g/kg/day of Trihoney (HCD + H2), and 1% cholesterol diet with 2 mg/kg/day of atorvastatin (HCD + At.). Animals were sacrificed after 12 weeks of treatment. Serum lipids and liver function test (LFT) were measured prior to and at the endpoint of the experiment for total cholesterol (TC), low-density lipoprotein (LDL-c), alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), gamma-glutamyl transferase (GGT), and total bilirubin (T. Bil.). Liver was processed for histopathology study. Liver homogenate was analysed for oxidative stress parameters: superoxide dismutase (SOD), glutathione peroxidase (GPx), and malondialdehyde (MDA). Results. Lipid analysis approved the induction of hypercholesterolemia. A significant elevation (p < 0.01) of serum AST and ALT levels showed by the HCD group was compared to C and C + H groups. Trihoney exhibited a significant reduction (p < 0.001) of AST and ALT compared to the HCD group. Likewise, AST and ALT reduced significantly in the HCD + At. group (p < 0.001). Trihoney supplementation induced significant (p < 0.05) enhancement of SOD and GPx activities. Atorvastatin treatment was associated with significant (p < 0.05) reduction of SOD and GPx activities in the liver. Trihoney and atorvastatin showed marked (p < 0.001) reduction of hepatic lipid peroxidation. Trihoney showed histological protection against progression of NAFLD to nonalcoholic steatohepatitis (NASH). Atorvastatin exhibited no beneficial impact on hepatic architecture. Conclusion. Trihoney was able to maintain normal liver function and showed hepatoprotection against progression of NAFLD to NASH probably through hypocholesterolaemic and antioxidant functions.
Collapse
|
8
|
Moustapha ME. Radioiodination of Atorvastatin as a Model Radiopharmaceutical for Targeting Liver. RADIOCHEMISTRY 2020. [DOI: 10.1134/s1066362220040104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
9
|
Hossaini Nasr S, Rashidijahanabad Z, Ramadan S, Kauffman N, Parameswaran N, Zinn KR, Qian C, Arora R, Agnew D, Huang X. Effective atherosclerotic plaque inflammation inhibition with targeted drug delivery by hyaluronan conjugated atorvastatin nanoparticles. NANOSCALE 2020; 12:9541-9556. [PMID: 32314997 PMCID: PMC7234819 DOI: 10.1039/d0nr00308e] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Atherosclerosis is associated with inflammation in the arteries, which is a major cause of heart attacks and strokes. Reducing the extent of local inflammation at atherosclerotic plaques can be an attractive strategy to combat atherosclerosis. While statins can exhibit direct anti-inflammatory activities, the high dose required for such a therapy renders it unrealistic due to their low systemic bioavailabilities and potential side effects. To overcome this, a new hyaluronan (HA)-atorvastatin (ATV) conjugate was designed with the hydrophobic statin ATV forming the core of the nanoparticle (HA-ATV-NP). The HA on the NPs can selectively bind with CD44, a cell surface receptor overexpressed on cells residing in atherosclerotic plaques and known to play important roles in plaque development. HA-ATV-NPs exhibited significantly higher anti-inflammatory effects on macrophages compared to ATV alone in vitro. Furthermore, when administered in an apolipoprotein E (ApoE)-knockout mouse model of atherosclerosis following a 1-week treatment regimen, HA-ATV-NPs markedly decreased inflammation in advanced atherosclerotic plaques, which were monitored through contrast agent aided magnetic resonance imaging. These results suggest CD44 targeting with HA-ATV-NPs is an attractive strategy to reduce harmful inflammation in atherosclerotic plaques.
Collapse
Affiliation(s)
- Seyedmehdi Hossaini Nasr
- Department of Chemistry Michigan State University, East Lansing, Michigan 48824, USA. and Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA
| | - Zahra Rashidijahanabad
- Department of Chemistry Michigan State University, East Lansing, Michigan 48824, USA. and Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA
| | - Sherif Ramadan
- Department of Chemistry Michigan State University, East Lansing, Michigan 48824, USA. and Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Chemistry Department, Faculty of Science, Benha University, Benha, Qaliobiya 13518, Egypt
| | - Nate Kauffman
- Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Biomedical Engineering Michigan State University, East Lansing, Michigan 48824, USA
| | | | - Kurt R Zinn
- Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Biomedical Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Radiology Michigan State University, East Lansing, Michigan 48824, USA
| | - Chunqi Qian
- Department of Radiology Michigan State University, East Lansing, Michigan 48824, USA
| | - Ripla Arora
- Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Obstetrics, Gynecology and Reproductive Biology Michigan State University, East Lansing, Michigan 48824, USA
| | - Dalen Agnew
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824, USA
| | - Xuefei Huang
- Department of Chemistry Michigan State University, East Lansing, Michigan 48824, USA. and Institute for Quantitative Health Science and Engineering Michigan State University, East Lansing, Michigan 48824, USA and Department of Biomedical Engineering Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
10
|
Khan AA, Ahmed S, Mohammed A, Elzouki ANY. Autoimmune-like Drug-induced Liver Injury Caused by Atorvastatin and Demonstration of the Safety Profile of Pravastatin: A Case Report and Literature Review. Cureus 2020; 12:e7299. [PMID: 32313740 PMCID: PMC7163344 DOI: 10.7759/cureus.7299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/17/2020] [Indexed: 01/01/2023] Open
Abstract
Statin-induced liver injury is a well-recognized but rare phenomenon with hepatocellular, cholestatic, and mixed phenotypes. Most studies do not recommend regular monitoring of liver function tests (LFTs) after starting statins unless clinically indicated. We report a case of autoimmune-like atorvastatin-induced liver injury (aminotransferases > 5 times the upper limit of normal) that was detected on routine follow-up after three months in an asymptomatic patient. In addition to elevation in transaminases, the patient had weakly positive ANAs. Anti-smooth muscle antibody (ASMA) was positive in titers of 1:680. Screening for viral hepatitis A-E was negative. Other diagnostic investigations showed complete blood examination, including eosinophils, renal function tests, electrolytes, total protein, albumin, prothrombin time, activated partial thromboplastin time (aPTT), international normalized ratio (INR), serum ferritin, and iron saturation to be in the normal range. Ultrasound and computed tomography (CT) abdomen showed normal liver, gall bladder, biliary tree, and pancreas. The patient was managed as a case of autoimmune-like drug-induced liver injury (DILI) caused by atorvastatin and the medication was discontinued. LFTs returned to completely normal 30 days after the discontinuation of atorvastatin. Furthermore, switching to pravastatin for dyslipidemia management four months after stopping atorvastatin did not lead to hepatotoxicity, illustrating the safety profile of pravastatin in patients who are unable to tolerate atorvastatin.
Collapse
Affiliation(s)
- Adeel A Khan
- Internal Medicine, Hamad Medical Corporation, Doha, QAT
| | - Salma Ahmed
- Community Medicine, Hamad Medical Corporation, Doha, QAT
| | | | - Abdel-Naser Y Elzouki
- Internal Medicine, Hamad Medical Corporation, Doha, QAT
- Medicine, Weill Cornell Medical College, Doha, QAT
| |
Collapse
|
11
|
Cholangiolytic Changes in Statin-Induced Liver Injury. Case Rep Pathol 2020; 2020:9650619. [PMID: 32099709 PMCID: PMC7040411 DOI: 10.1155/2020/9650619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/31/2020] [Indexed: 01/17/2023] Open
Abstract
Atorvastatin is a commonly used oral cholesterol-lowering agent. Side effects associated with statin therapy include arthralgia, myalgia, dyspepsia, weakness, and headache. Prospective and retrospective studies of drug-induced liver injury have identified statin-induced hepatotoxicity, with atorvastatin being the most commonly cited. Associated liver function test elevations have varied from hepatocellular to cholestatic/mixed pattern. We report a case of a 58-year-old woman that illustrates unusual histologic findings associated with a mixed pattern of statin-induced liver injury. While being treated with atorvastatin, the patient exhibited repeated bouts of abdominal pain over a year associated with biliary tree dilation, variably attributed to postcholecystectomy dilation and stenosis of the ampulla of Vater. Following sphincterotomy, the patient's bilirubin normalized but the other liver function tests remained elevated. Liver biopsy revealed portal and lobular inflammation with cholangiolysis. The patient's liver function tests normalized following cessation of atorvastatin therapy.
Collapse
|
12
|
Drug-Drug Interactions in Acute Coronary Syndrome Patients: Systematic Review. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2019-0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Drug-drug interaction (DDI) is defined as a clinically significant change in the exposure and/or response to a drug caused by co-administration of another drug which may result in a precipitation of an adverse event or alteration of its therapeutic effects. The aim of this systematic review was to provide an overview of DDIs that were actually observed or evaluated in acute coronary syndrome (ACS) patients with particular focus on DDIs with clinical relevance. Electronic searches of the literature were conducted in the following databases: MEDLINE, EBSCO, Scopus, Google Scholar and SCIndeks. A total of 117 articles were included in the review. This review showed that ACS patients can be exposed to a variety of DDIs with diverse outcomes which include decreased efficacy of antiplatelet drugs, thrombolytics or anticoagulants, increased risk of bleeding, rhabdomyolysis, hepatotoxicity, adverse effects on cardiovascular system (e.g. QT interval prolongation, arrhythmias, excessive bradycardia, severe hypotension), serotonin syndrome and drug-induced fever. Majority of the DDIs involved antiplatelet drugs (e.g. aspirin, clopidogrel and ticagrelor). Evidence of some of the reported DDIs is inconclusive as some of the studies have shown conflicting results. There is a need for additional post-marketing and population-based studies to evaluate the true effects of disease states and other factors on the clinical outcomes of DDIs. Clinicians should be attentive to the potential for DDIs and their associated harm in order to minimize or, if possible, avoid medication-related adverse events in ACS patients.
Collapse
|
13
|
Fukunaga K, Nakagawa H, Ishikawa T, Kubo M, Mushiroda T. ABCB1 polymorphism is associated with atorvastatin-induced liver injury in Japanese population. BMC Genet 2016; 17:79. [PMID: 27296832 PMCID: PMC4906899 DOI: 10.1186/s12863-016-0390-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 06/07/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To investigate the associations between atorvastatin-induced liver injury (AILI) and polymorphisms in eight genes possibly involved in the hepatic metabolism (CYP2C9, CYP2C19, CYP3A4, CYP3A5 and UGT1A1) and membrane transport (ABCB1, ABCG2 and SLCO1B1) of atorvastatin, we genotyped 30 AILI and 414 non-AILI patients recruited at BioBank Japan for 15 single nucleotide polymorphisms (SNPs). RESULTS An SNP in ABCB1 (rs2032582: 2677G > T/A) was significantly associated with AILI (P = 0.00068, odds ratio (OR) = 2.59 with 95 % confidence interval (CI) of 1.49-4.50, G allele versus T and A alleles), indicating that the G allele might be a risk factor for AILI. The cytotoxicity test demonstrated that IC50 value of atorvastatin to inhibit the growth and/or viability of Flp-In-293/ABCB1 (2677G) cells was 5.44 ± 0.10 mM, which was significantly lower than those in Flp-In-293/ABCB1 (2677 T) (6.02 ± 0.07 mM) and Flp-In-293/ABCB1 (2677A) cells (5.95 ± 0.08 mM). CONCLUSIONS These results indicate that ABCB1 rs2032582 may predict the risk of AILI in Japanese population.
Collapse
Affiliation(s)
- Koya Fukunaga
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hiroshi Nakagawa
- Department of Biological Chemistry, College of Bioscience and Biotechnology, Chubu University, Aichi, Japan
| | | | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Taisei Mushiroda
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| |
Collapse
|
14
|
Oleaga C, Bernabini C, Smith AS, Srinivasan B, Jackson M, McLamb W, Platt V, Bridges R, Cai Y, Santhanam N, Berry B, Najjar S, Akanda N, Guo X, Martin C, Ekman G, Esch MB, Langer J, Ouedraogo G, Cotovio J, Breton L, Shuler ML, Hickman JJ. Multi-Organ toxicity demonstration in a functional human in vitro system composed of four organs. Sci Rep 2016; 6:20030. [PMID: 26837601 PMCID: PMC4738272 DOI: 10.1038/srep20030] [Citation(s) in RCA: 288] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/18/2015] [Indexed: 12/26/2022] Open
Abstract
We report on a functional human model to evaluate multi-organ toxicity in a 4-organ system under continuous flow conditions in a serum-free defined medium utilizing a pumpless platform for 14 days. Computer simulations of the platform established flow rates and resultant shear stress within accepted ranges. Viability of the system was demonstrated for 14 days as well as functional activity of cardiac, muscle, neuronal and liver modules. The pharmacological relevance of the integrated modules were evaluated for their response at 7 days to 5 drugs with known side effects after a 48 hour drug treatment regime. The results of all drug treatments were in general agreement with published toxicity results from human and animal data. The presented phenotypic culture model exhibits a multi-organ toxicity response, representing the next generation of in vitro systems, and constitutes a step towards an in vitro "human-on-a-chip" assay for systemic toxicity screening.
Collapse
Affiliation(s)
- Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Catia Bernabini
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Alec S.T. Smith
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Balaji Srinivasan
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Max Jackson
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - William McLamb
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Vivien Platt
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Richard Bridges
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Yunqing Cai
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Navaneetha Santhanam
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Bonnie Berry
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Sarah Najjar
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Nesar Akanda
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Candace Martin
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Gail Ekman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| | - Mandy B. Esch
- Department of Biomedical Engineering, Cornell University, 115 and 305 Weill Hall, Ithaca, NY 14853
| | - Jessica Langer
- L’Oreal Research and Innovation, Clark, NJ, 07666/ Aulnay sous Bois, France, 93600
| | | | - Jose Cotovio
- L’Oreal Research and Innovation, Aulnay sous Bois, France
| | - Lionel Breton
- L’Oreal Research and Innovation, Aulnay sous Bois, France
| | - Michael L. Shuler
- Department of Biomedical Engineering, Cornell University, 115 and 305 Weill Hall, Ithaca, NY 14853
| | - James J. Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway Suite 400, Orlando, FL 32828
| |
Collapse
|
15
|
Abstract
Drug-induced hepatotoxicity is a common cause of acute hepatitis, and the recognition of the responsible drug may be difficult. We describe a case of clopidogrel-related acute hepatitis. The diagnosis is strongly suggested by an accurate medical history and liver biopsy. Reports about cases of hepatotoxicity due to clopidogrel are increasing in the last few years, after the increased use of this drug. In conclusion, we believe that physicians should carefully consider the risk of drug-induced hepatic injury when clopidogrel is prescribed.
Collapse
|
16
|
El Shazly RI, Mohammed WHS, Mohamed SF, Elnasr MIS. Prevalence and risk factors of liver biochemical abnormalities in patients with systemic lupus erythematosus. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2014. [DOI: 10.4103/1110-161x.147352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
17
|
Chang JH, Ly J, Plise E, Zhang X, Messick K, Wright M, Cheong J. Differential Effects of Rifampin and Ketoconazole on the Blood and Liver Concentration of Atorvastatin in Wild-Type and Cyp3a and Oatp1a/b Knockout Mice. Drug Metab Dispos 2014; 42:1067-73. [DOI: 10.1124/dmd.114.057968] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
18
|
Shakib MC, Gabrial S, Gabrial G. Rice Bran Oil Compared to Atorvastatin for Treatment of Dyslipidemia in Patients with Type 2 Diabetes. Open Access Maced J Med Sci 2014. [DOI: 10.3889/oamjms.2014.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Objective: To compare the effect of rice bran oil versus statins (atorvastatin drug) on blood glucose, glycosylated hemoglobin (HbA1C) and serum lipid profiles in patients with type 2 diabetes. The safety of the tested rice bran oil and atorvastatin were investigated. Fatty acids contents of RBO, olive and sesame oil were also assessed.Materials and Methods: Forty four eligible patients with type 2 diabetes and moderately hyperlipidemic were randomly and equally allocated into two groups, rice bran oil (RBO) group and atorvastatin group. The RBO group received a low-calorie diet and consumed 30Â g / day RBO oil as salad dressing and for use as main cooking oil for 6 months. The Atorvastatin group received a low-calorie diet and 40 mg/day of atorvastatin drug for 6 months. At baseline and after 6 months of study intervention, blood glucose, glycosylated hemoglobin (HbA1c), serum lipid profiles; hepatic, renal and inflammatory biomarkers were estimated.Results: Results showed significant increase in fasting and postprandial blood glucose, HbA1C and liver transaminases (alanine transaminase ALT and aspartate transaminase AST) in the atorvastatin group while a significant reduction was shown in RBO group. Moreover, significant reductions in lipid profile levels, blood urea, serum uric acid and erythrocyte sedimentation rate (ESR) were observed in both RBO and atorvastatin groups after 6 months of the study intervention.Conclusion: The use of rice bran oil together with dietary modifications may have implications in lowering fasting and postprandial blood glucose, suppressing serum lipid levels, reduce the TC/HDL-C ratio and therefore reducing the risk of cardiovascular disease. Moreover, RBO exerts a hypouricemic action and anti-inflammatory effects. The findings obtained from the current study reinforce the use of RBO as an alternative natural potent hypolipidemic agent safer than atorvastatin drug that may induce side effects in some cases in patients intolerant to statins.
Collapse
|
19
|
Alfazari AS, Al-Dabbagh B, Almarzooqi S, Albawardi A, Souid AK. Bioenergetic study of murine hepatic tissue treated in vitro with atorvastatin. BMC Pharmacol Toxicol 2013; 14:15. [PMID: 23448291 PMCID: PMC3602108 DOI: 10.1186/2050-6511-14-15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 02/22/2013] [Indexed: 12/15/2022] Open
Abstract
Atorvastatin (a 3-hydroxy-3-methylglutaryl coenzyme-A reductase inhibitor) is a widely used cholesterol-lowering drug, which is recognized for its potential hepatotoxicity. This study investigated in vitro effects of this agent on hepatic tissue respiration, ATP content, caspase activity, urea synthesis and histology. Liver fragments from Taylor Outbred and C57Bl/6 mice were incubated at 37°C in Krebs-Henseleit buffer continuously gassed with 95% O2: 5% CO2 in the presence and absence of atorvastatin. Phosphorescence O2 analyzer that measured dissolved [O2] as a function of time was used to monitor cellular mitochondrial O2 consumption. The caspase-3 substrate N-acetyl-asp-glu-val-asp-7-amino-4-methylcoumarin was used to monitor caspase activity. The rates of hepatocyte respiration (μM O2 min-1 mg-1) in untreated samples were 0.15 ± 0.07 (n = 31). The corresponding rates for samples treated with 50 nM (therapeutic concentration), 150 nM or 1.0 μM atorvastatin for ≤13 h were 0.13 ± 0.05 (n = 19), p = 0.521. The contents of hepatocyte ATP (pmol-1 mg-1) in untreated samples were 40.3 ± 14.0 and in samples treated with 1.0 μM atorvastatin for ≤4.5 h were 48.7 ± 23.9 (p = 0.7754). The concentrations of urea (mg/dL mg-1, produced over 50 min) for untreated samples were 0.061 ± 0.020 (n = 6) and for samples treated with 1.0 μM atorvastatin for ≤6 h were 0.072 ± 0.022 (n = 6), p = 0.3866. Steadily, hepatocyte caspase activity and histology were unaffected by treatments with up to 1.0 μM atorvastatin for ≤6 h. Thus, the studied murine model showed preserved hepatocyte function and structure in the presence of high concentrations of atorvastatin.
Collapse
Affiliation(s)
- Ali S Alfazari
- Department of Internal Medicine, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates.
| | | | | | | | | |
Collapse
|
20
|
Gazzerro P, Proto MC, Gangemi G, Malfitano AM, Ciaglia E, Pisanti S, Santoro A, Laezza C, Bifulco M. Pharmacological actions of statins: a critical appraisal in the management of cancer. Pharmacol Rev 2011; 64:102-46. [PMID: 22106090 DOI: 10.1124/pr.111.004994] [Citation(s) in RCA: 316] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Statins, among the most commonly prescribed drugs worldwide, are cholesterol-lowering agents used to manage and prevent cardiovascular and coronary heart diseases. Recently, a multifaceted action in different physiological and pathological conditions has been also proposed for statins, beyond anti-inflammation and neuroprotection. Statins have been shown to act through cholesterol-dependent and -independent mechanisms and are able to affect several tissue functions and modulate specific signal transduction pathways that could account for statin pleiotropic effects. Typically, statins are prescribed in middle-aged or elderly patients in a therapeutic regimen covering a long life span during which metabolic processes, aging, and concomitant novel diseases, including cancer, could occur. In this context, safety, toxicity, interaction with other drugs, and the state of health have to be taken into account in subjects treated with statins. Some evidence has shown a dichotomous effect of statins with either cancer-inhibiting or -promoting effects. To date, clinical trials failed to demonstrate a reduced cancer occurrence in statin users and no sufficient data are available to define the long-term effects of statin use over a period of 10 years. Moreover, results from clinical trials performed to evaluate the therapeutic efficacy of statins in cancer did not suggest statin use as chemotherapeutic or adjuvant agents. Here, we reviewed the pharmacology of the statins, providing a comprehensive update of the current knowledge of their effects on tissues, biological processes, and pathological conditions, and we dissected the disappointing evidence on the possible future use of statin-based drugs in cancer therapy.
Collapse
Affiliation(s)
- Patrizia Gazzerro
- Department of Pharmaceutical and Biomedical Sciences, University of Salerno, Via Ponte Don Melillo, 84084 Fisciano (Salerno), Italy
| | | | | | | | | | | | | | | | | |
Collapse
|