1
|
Psaltis PJ, Nguyen MT, Singh K, Sinhal A, Wong DTL, Alcock R, Rajendran S, Dautov R, Barlis P, Patel S, Salagaras T, Marathe JA, Bursill CA, Montarello NJ, Nidorf SM, Thompson PL, Butters J, Cuthbert AR, Yelland LN, Ottaway JL, Kataoka Y, Di Giovanni G, Nicholls SJ. Optical coherence tomography assessment of the impact of colchicine on non-culprit coronary plaque composition after myocardial infarction. Cardiovasc Res 2025; 121:468-478. [PMID: 39189611 DOI: 10.1093/cvr/cvae191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 08/28/2024] Open
Abstract
AIMS Low-dose colchicine reduces the risk of cardiovascular events after myocardial infarction (MI). The purpose of this study was to assess the effect of colchicine post-MI on coronary plaque morphology in non-culprit segments by optical coherence tomography (OCT). METHODS AND RESULTS COCOMO-ACS was a double-blind, placebo-controlled trial that randomized 64 patients (median age 61.5 years; 9.4% female) with acute non-ST-segment elevation MI to colchicine 0.5 mg daily or placebo for a median of 17.8 months in addition to guideline-recommended therapy. Participants underwent serial OCT imaging within a matched segment of non-culprit coronary artery that contained at least one lipid-rich plaque causing ≥20% stenosis. The primary outcome was the change in minimum fibrous cap thickness (FCT) in non-culprit segments from baseline to final visit. Of those randomized, 57 (29 placebo, 28 colchicine) had evaluable imaging at baseline and follow-up. Overall, colchicine had no effect on relative (placebo +48.0 ± 35.1% vs. colchicine +62.4 ± 38.1%, P = 0.18) or absolute changes in minimum FCT (+29.2 ± 20.9 µm vs. + 37.2 ± 21.3 µm, P = 0.18), or change in maximum lipid arc (-38.8 ± 32.2° vs. -54.8 ± 46.9°, P = 0.18) throughout the imaged non-culprit segment. However, in patients assigned colchicine, cap rupture was less frequent (placebo 27.6% vs. colchicine 3.6%, P = 0.03). In post hoc analysis of 43 participants who had been followed for at least 16 months, minimum FCT increased to a greater extent in the colchicine group (placebo +38.7 ± 25.4% vs. colchicine +64.7 ± 34.1%, P = 0.005). CONCLUSION In this study, OCT failed to detect an effect of colchicine on the minimum FCT or maximum lipid arc of plaques in non-culprit segments post-MI. The post hoc observation that minimum FCT increased to a greater extent with colchicine after more prolonged treatment suggests that longer-term studies may be required to detect the effect of anti-inflammatory therapies on plaque morphology by OCT. CLINICAL TRIAL NUMBER Australian New Zealand Clinical Trials Registry Identifier, ACTRN12618000809235, registered on the 11 May 2018.
Collapse
Affiliation(s)
- Peter J Psaltis
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Department of Cardiology, Central Adelaide Local Health Network, Port Road, Adelaide 5000, Australia
- Adelaide Medical School, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Mau T Nguyen
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Department of Cardiology, Central Adelaide Local Health Network, Port Road, Adelaide 5000, Australia
- Adelaide Medical School, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Kuljit Singh
- Department of Cardiology, Gold Coast University Hospital, Gold Coast, Australia
| | - Ajay Sinhal
- Department of Cardiology, Flinders Medical Center, Adelaide, Australia
| | - Dennis T L Wong
- Victorian Heart Institute, Monash University, Clayton, Australia
| | - Richard Alcock
- Department of Cardiology, Royal Perth Hospital, Perth, Australia
| | | | - Rustem Dautov
- Department of Cardiology, The Prince Charles Hospital, Brisbane, Australia
| | - Peter Barlis
- Department of Cardiology, The Northern Hospital, Melbourne, Australia
| | - Sanjay Patel
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
- Heart Research Institute, The University of Sydney, Sydney, Australia
| | - Thalia Salagaras
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
| | - Jessica A Marathe
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Department of Cardiology, Central Adelaide Local Health Network, Port Road, Adelaide 5000, Australia
- Adelaide Medical School, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Christina A Bursill
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Adelaide Medical School, University of Adelaide, North Terrace, Adelaide 5000, Australia
| | - Nicholas J Montarello
- Department of Cardiology, Central Adelaide Local Health Network, Port Road, Adelaide 5000, Australia
| | - Stefan M Nidorf
- Advara HeartCare, Western Australia, Perth, Australia
- Cardiovascular Science and Diabetes Program, Harry Perkins Institute of Medical Research, Perth, Australia
| | - Peter L Thompson
- Advara HeartCare, Western Australia, Perth, Australia
- Cardiovascular Science and Diabetes Program, Harry Perkins Institute of Medical Research, Perth, Australia
- Heart Research Institute, Sir Charles Gairdner Hospital, Perth, Australia
| | - Julie Butters
- Victorian Heart Institute, Monash University, Clayton, Australia
| | - Alana R Cuthbert
- SAHMRI Women & Kids, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Lisa N Yelland
- SAHMRI Women & Kids, South Australian Health and Medical Research Institute, Adelaide, Australia
- School of Public Health, The University of Adelaide, Adelaide, Australia
| | - Juanita L Ottaway
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
| | - Yu Kataoka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Giuseppe Di Giovanni
- Vascular Research Center, Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide 5000, Australia
- Victorian Heart Institute, Monash University, Clayton, Australia
| | | |
Collapse
|
2
|
Al-Kuraishy HM, Sulaiman GM, Mohammed HA, Dawood RA, Albuhadily AK, Al-Gareeb AI, Klionsky DJ, Abomughaid MM. Insight into the Mechanistic role of Colchicine in Atherosclerosis. Curr Atheroscler Rep 2025; 27:40. [PMID: 40111634 DOI: 10.1007/s11883-025-01291-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE OF REVIEW Globally, the prevalence of atherosclerosis (AS) is rising. Currently, there is no specific drug for AS. Therefore, this review aims to discuss the protective mechanisms of colchicine against the development and progression of atherosclerosis (AS). RECENT FINDINGS Many studies highlighted that the anti-inflammatory drug colchicine reduces the severity of AS, although the underlying mechanism for the beneficial effect of colchicine was not fully clarified. AS is a chronic progressive vascular disorder characterized by the formation of atherosclerotic plaques. Endothelial dysfunction is an initial stage in the pathogenesis of AS that is induced by oxidized low-density lipoprotein (oxLDL). Engulfment of oxLDL by macrophages triggers the development of inflammation due to the release of pro-inflammatory cytokines and growth factors. Inflammatory and adhesion molecules are involved in the pathogenesis of AS. Infiltration and accumulation of leukocytes provoke erosion, rupture, and thrombosis of the atherosclerotic plaque. Therefore, targeting inflammation and leukocyte infiltration by anti-inflammatory agents may reduce AS progression and complications. The anti-inflammatory drug colchicine reduces the severity of AS, although the underlying mechanism for the beneficial effect of colchicine was not fully elucidated. IN CONCLUSION colchicine through inhibition of vascular inflammation, oxidative stress, platelet aggregation and the modulation of autophagy reduces the development and progression of AS.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ghassan M Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq.
| | - Hamdoon A Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, 51452, Qassim, Saudi Arabia
| | - Retaj A Dawood
- Department of Biology, College of Science, Al-Mustaqbal University, Hilla, 51001, Iraq
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Jabir Ibn Hayyan Medical University, Al-Ameer Qu, PO.Box13 Kufa, Najaf, Iraq
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Mosleh M Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 255, 67714, Bisha, Saudi Arabia
| |
Collapse
|
3
|
Chuang YT, Yen CY, Tang JY, Chang FR, Tsai YH, Wu KC, Chien TM, Chang HW. The modulation of immune cell death in connection to microRNAs and natural products. Front Immunol 2024; 15:1425602. [PMID: 39759512 PMCID: PMC11695430 DOI: 10.3389/fimmu.2024.1425602] [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: 04/30/2024] [Accepted: 11/27/2024] [Indexed: 01/07/2025] Open
Abstract
Immunogenic cell death (ICD) spatiotemporally regulates damage-associated molecular patterns (DAMPs) derived from dying cancer cells to signal the immune response. Intriguingly, these DAMPs and cytokines also induce cellular responses in non-immune cells, particularly cancer cells. Several ICD-modulating natural products and miRNAs have been reported to regulate the DAMP, cytokine, and cell death responses, but they lack systemic organization and connection. This review summarizes the impacts of natural products and miRNAs on the DAMP and cytokine responses and cancer cell death responses (apoptosis, autophagy, ferroptosis, necroptosis, and pyroptosis). We establish the rationale that ICD inducers of natural products have modulating effects on miRNAs, targeting DAMPs and cytokines for immune and cancer cell death responses. In conclusion, DAMP, cytokine, and cell death responses are intricately linked in cancer cells, and they are influenced by ICD-modulating natural products and miRNAs.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei, Taiwan
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung, Taiwan
| | - Kuo-Chuan Wu
- Department of Computer Science and Information Engineering, National Pingtung University, Pingtung, Taiwan
| | - Tsu-Ming Chien
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Gangshan Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
4
|
Tucker B, Goonetilleke N, Patel S, Keech A. Colchicine in atherosclerotic cardiovascular disease. Heart 2024; 110:618-625. [PMID: 38331560 DOI: 10.1136/heartjnl-2023-323177] [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: 10/03/2023] [Accepted: 12/07/2023] [Indexed: 02/10/2024] Open
Abstract
Inflammation has a direct role in the development of atherosclerotic vascular disease, and oral colchicine displays broad anti-inflammatory properties. Several large, randomised controlled trials (RCTs) have evaluated colchicine's impact on cardiovascular outcomes. Results from a meta-analysis of these trials demonstrate that colchicine reduces the risk of recurrent major adverse cardiovascular events (MACEs) by 25%, leading to its recent approval by the Food and Drug Administration for the treatment and prevention of cardiovascular disease. Despite this, colchicine has not been shown to confer any survival benefit in these trials. The non-significant reduction in cardiovascular death of 18% (95% CI: 45% decrease to 23% increase) is outweighed by a more prominent, borderline non-significant increase in the risk of non-cardiovascular death by 38% (95% CI: 1% decrease to 92% increase). Key populations including those with heart failure, those undergoing surgical revascularisation, women, elderly individuals and non-Caucasians are under-represented in completed trials, which limits generalisability. C reactive protein has been proposed as a biomarker for colchicine response and shows promise for identifying a high-risk population where the benefit on MACE reduction and specifically reduced cardiovascular death might outweigh any real increased risk of non-cardiovascular death; however, this approach is still to be validated in ongoing RCTs. In conclusion, while colchicine shows promise in reducing MACE, its net risk-benefit profile requires further elucidation before its widespread adoption into clinical practice for the secondary prevention of atherosclerotic cardiovascular disease. Much more large-scale, long-term trial data are still needed in this space.
Collapse
Affiliation(s)
- Bradley Tucker
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | | | - Sanjay Patel
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Anthony Keech
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- NHMRC Clinical Trials Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
5
|
Michalak-Stoma A, Walczak K, Adamczyk M, Kowal M, Krasowska D. Selected miRNA and Psoriasis-Cardiovascular Disease (CVD)-Overweight/Obesity Network-A Pilot Study. Int J Mol Sci 2023; 24:13916. [PMID: 37762217 PMCID: PMC10530775 DOI: 10.3390/ijms241813916] [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: 08/03/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Psoriasis is nowadays recognized as a multifactorial systemic disease with complex and not fully understood pathogenesis. In psoriatic patients, the increased cardiovascular disease (CVD) risk and frequent comorbidities like obesity are observed. The aim of this study was to investigate differences in miRNA (miR-22-3p, miR-133a-3p, miR-146a-5p, miR-369-3p, and Let-7b-5p) involved in CVD risk among psoriatic patients with overweight/obesity and with normal weight. The study comprised 28 male psoriatic patients and 16 male healthy controls. miRNA isolated from peripheral blood mononuclear cells was reverse-transcribed and RT-qPCR was performed. We have found decreased levels of miR-22, miR-133a, miR-146a, and miR-369 among the psoriatic patients. There was a statistically significant difference in miR-22 and miR-146a levels between psoriatic patients with overweight/obesity and with normal weight. There were positive correlations between miR-22 and miR-146a levels and psoriatic arthritis (PsA) in psoriatic patients with normal weight and between the miR-133a level and PsA in the overweight/obese patients. The decreased levels of selected miRNA are consistent with the levels observed in CVD indicating their impact on the CVD risk in psoriatic patients. miR-22 and miR-146 may be recognized as one of the contributing factors in the obesity-CVD-psoriasis network.
Collapse
Affiliation(s)
- Anna Michalak-Stoma
- Chair and Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, ul. Staszica 16, 20-081 Lublin, Poland; (K.W.); (M.A.); (M.K.); (D.K.)
| | | | | | | | | |
Collapse
|
6
|
Nardin M, Verdoia M, Laera N, Cao D, De Luca G. New Insights into Pathophysiology and New Risk Factors for ACS. J Clin Med 2023; 12:jcm12082883. [PMID: 37109221 PMCID: PMC10146393 DOI: 10.3390/jcm12082883] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Cardiovascular disease still represents the main cause of mortality worldwide. Despite huge improvements, atherosclerosis persists as the principal pathological condition, both in stable and acute presentation. Specifically, acute coronary syndromes have received substantial research and clinical attention in recent years, contributing to improve overall patients' outcome. The identification of different evolution patterns of the atherosclerotic plaque and coronary artery disease has suggested the potential need of different treatment approaches, according to the mechanisms and molecular elements involved. In addition to traditional risk factors, the finer portrayal of other metabolic and lipid-related mediators has led to higher and deep knowledge of atherosclerosis, providing potential new targets for clinical management of the patients. Finally, the impressive advances in genetics and non-coding RNAs have opened a wide field of research both on pathophysiology and the therapeutic side that are extensively under investigation.
Collapse
Affiliation(s)
- Matteo Nardin
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
- Third Medicine Division, Department of Medicine, ASST Spedali Civili, 25123 Brescia, Italy
| | - Monica Verdoia
- Division of Cardiology, Ospedale degli Infermi, ASL Biella, 13900 Biella, Italy
- Department of Translational Medicine, Eastern Piedmont University, 13100 Novara, Italy
| | - Nicola Laera
- Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
| | - Davide Cao
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy
| | - Giuseppe De Luca
- Division of Cardiology, AOU "Policlinico G. Martino", Department of Clinical and Experimental Medicine, University of Messina, 98166 Messina, Italy
- Division of Cardiology, IRCCS Hospital Galeazzi-Sant'Ambrogio, 20161 Milan, Italy
| |
Collapse
|
7
|
Da'as SI, Ahmed I, Hasan WH, Abdelrahman DA, Aliyev E, Nisar S, Bhat AA, Joglekar MV, Hardikar AA, Fakhro KA, Akil ASAS. The link between glycemic control measures and eye microvascular complications in a clinical cohort of type 2 diabetes with microRNA-223-3p signature. J Transl Med 2023; 21:171. [PMID: 36869348 PMCID: PMC9985290 DOI: 10.1186/s12967-023-03893-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/16/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a critical healthcare challenge and priority in Qatar which is listed amongst the top 10 countries in the world, with its prevalence presently at 17% double the global average. MicroRNAs (miRNAs) are implicated in the pathogenesis of (T2D) and long-term microvascular complications including diabetic retinopathy (DR). METHODS In this study, a T2D cohort that accurately matches the characteristics of the general population was employed to find microRNA (miRNA) signatures that are correlated with glycemic and β cell function measurements. Targeted miRNA profiling was performed in (471) T2D individuals with or without DR and (491) (non-diabetic) healthy controls from the Qatar Biobank. Discovery analysis identified 20 differentially expressed miRNAs in T2D compared to controls, of which miR-223-3p was significantly upregulated (fold change:5.16, p = 3.6e-02) and positively correlated with glucose and hemoglobin A1c (HbA1c) levels (p-value = 9.88e-04 and 1.64e-05, respectively), but did not show any significant associations with insulin or C-peptide. Accordingly, we performed functional validation using a miR-223-3p mimic (overexpression) under control and hyperglycemia-induced conditions in a zebrafish model. RESULTS Over-expression of miR-223-3p alone was associated with significantly higher glucose (42.7 mg/dL, n = 75 vs 38.7 mg/dL, n = 75, p = 0.02) and degenerated retinal vasculature, and altered retinal morphology involving changes in the ganglion cell layer and inner and outer nuclear layers. Assessment of retinal angiogenesis revealed significant upregulation in the expression of vascular endothelial growth factor and its receptors, including kinase insert domain receptor. Further, the pancreatic markers, pancreatic and duodenal homeobox 1, and the insulin gene expressions were upregulated in the miR-223-3p group. CONCLUSION Our zebrafish model validates a novel correlation between miR-223-3p and DR development. Targeting miR-223-3p in T2D patients may serve as a promising therapeutic strategy to control DR in at-risk individuals.
Collapse
Affiliation(s)
- Sahar I Da'as
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.,Zebrafish Functional Genomics, Integrated Genomic Services Core Facility, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar.,College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar
| | - Ikhlak Ahmed
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Waseem H Hasan
- Zebrafish Functional Genomics, Integrated Genomic Services Core Facility, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Doua A Abdelrahman
- Zebrafish Functional Genomics, Integrated Genomic Services Core Facility, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Elbay Aliyev
- Laboratory of Genomic Medicine-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sabah Nisar
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Ajaz Ahmad Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Mugdha V Joglekar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW, 2560, Australia
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Narellan Road & Gilchrist Drive, Campbelltown, NSW, 2560, Australia.,Department of Science and Environment, Roskilde University, Universitetsvej 1, 4000, Roskilde, Denmark
| | - Khalid A Fakhro
- Laboratory of Genomic Medicine-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.,College of Health and Life Sciences, Hamad Bin Khalifa University, P.O. Box 34110, Doha, Qatar.,Department of Genetic Medicine, Weill Cornell Medical College, P.O. Box 24144, Doha, Qatar
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar. .,Laboratory of Genomic Medicine-Precision Medicine Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
| |
Collapse
|
8
|
Brown C, Mantzaris M, Nicolaou E, Karanasiou G, Papageorgiou E, Curigliano G, Cardinale D, Filippatos G, Memos N, Naka KK, Papakostantinou A, Vogazianos P, Ioulianou E, Shammas C, Constantinidou A, Tozzi F, Fotiadis DI, Antoniades A. A systematic review of miRNAs as biomarkers for chemotherapy-induced cardiotoxicity in breast cancer patients reveals potentially clinically informative panels as well as key challenges in miRNA research. CARDIO-ONCOLOGY 2022; 8:16. [PMID: 36071532 PMCID: PMC9450324 DOI: 10.1186/s40959-022-00142-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/02/2022] [Indexed: 12/01/2022]
Abstract
Breast cancer patients are at a particularly high risk of cardiotoxicity from chemotherapy having a detrimental effect on quality-of-life parameters and increasing the risk of mortality. Prognostic biomarkers would allow the management of therapies to mitigate the risks of cardiotoxicity in vulnerable patients and a key potential candidate for such biomarkers are microRNAs (miRNA). miRNAs are post-transcriptional regulators of gene expression which can also be released into the circulatory system and have been associated with the progression of many chronic diseases including many types of cancer. In this review, the evidence for the potential application of miRNAs as biomarkers for chemotherapy-induced cardiotoxicity (CIC) in breast cancer patientsis evaluated and a simple meta-analysis is performed to confirm the replication status of each reported miRNA. Further selection of miRNAs is performed by reviewing the reported associations of each miRNA with other cardiovascular conditions. Based on this research, the most representative panels targeting specific chemotherapy agents and treatment regimens are suggested, that contain several informative miRNAs, including both general markers of cardiac damage as well as those for the specific cancer treatments.
Collapse
|
9
|
Mortazavi-Jahromi SS, Aslani M. Dysregulated miRNAs network in the critical COVID-19: An important clue for uncontrolled immunothrombosis/thromboinflammation. Int Immunopharmacol 2022; 110:109040. [PMID: 35839566 PMCID: PMC9271492 DOI: 10.1016/j.intimp.2022.109040] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022]
Abstract
Known as a pivotal immunohemostatic response, immunothrombosis is activated to restrict the diffusion of pathogens. This beneficial intravascular defensive mechanism represents the close interaction between the immune and coagulation systems. However, its uncontrolled form can be life-threatening to patients with the critical coronavirus disease 2019 (COVID-19). Hyperinflammation and ensuing cytokine storm underlie the activation of the coagulation system, something which results in the provocation of more immune-inflammatory responses by the thrombotic mediators. This vicious cycle causes grave clinical complications and higher risks of mortality. Classified as an evolutionarily conserved family of the small non-coding RNAs, microRNAs (miRNAs) serve as the fine-tuners of genes expression and play a key role in balancing the pro/anticoagulant and pro-/anti-inflammatory factors maintaining homeostasis. Therefore, any deviation from their optimal expression levels or efficient functions can lead to severe complications. Despite their extensive effects on the molecules and processes involved in uncontrolled immunothrombosis, some genetic agents and uncontrolled immunothrombosis-induced interfering factors (e.g., miRNA-single nucleotide polymorphysms (miR-SNPs), the complement system components, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, and reactive oxygen species (ROS)) have apparently disrupted their expressions/functions. This review study aims to give an overview of the role of miRNAs in the context of uncontrolled immunothrombosis/thromboinflammation accompanied by some presumptive interfering factors affecting their expressions/functions in the critical COVID-19. Detecting, monitoring, and resolving these interfering agents mafy facilitate the design and development of the novel miRNAs-based therapeutic approaches to the reduction of complications incidence and mortality in patients with the critical COVID-19.
Collapse
Affiliation(s)
- Seyed Shahabeddin Mortazavi-Jahromi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Department of Cellular and Molecular Biology, Kish International Campus, University of Tehran, Kish, Iran.
| | - Mona Aslani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
González L, Bulnes JF, Orellana MP, Muñoz Venturelli P, Martínez Rodriguez G. The Role of Colchicine in Atherosclerosis: From Bench to Bedside. Pharmaceutics 2022; 14:pharmaceutics14071395. [PMID: 35890291 PMCID: PMC9323936 DOI: 10.3390/pharmaceutics14071395] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a key feature of atherosclerosis. The inflammatory process is involved in all stages of disease progression, from the early formation of plaque to its instability and disruption, leading to clinical events. This strongly suggests that the use of anti-inflammatory agents might improve both atherosclerosis progression and cardiovascular outcomes. Colchicine, an alkaloid derived from the flower Colchicum autumnale, has been used for years in the treatment of inflammatory pathologies, including Gout, Mediterranean Fever, and Pericarditis. Colchicine is known to act over microtubules, inducing depolymerization, and over the NLRP3 inflammasome, which might explain its known anti-inflammatory properties. Recent evidence has shown the therapeutic potential of colchicine in the management of atherosclerosis and its complications, with limited adverse effects. In this review, we summarize the current knowledge regarding colchicine mechanisms of action and pharmacokinetics, as well as the available evidence on the use of colchicine for the treatment of coronary artery disease, covering basic, translational, and clinical studies.
Collapse
Affiliation(s)
- Leticia González
- Centro de Imágenes Biomédicas, Departamento de Radiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Instituto Milenio de Ingeniería e Inteligencia Artificial para la Salud, iHEALTH, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Juan Francisco Bulnes
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (J.F.B.); (M.P.O.)
| | - María Paz Orellana
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (J.F.B.); (M.P.O.)
| | - Paula Muñoz Venturelli
- Centro de Estudios Clínicos, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana, Universidad de Desarrollo, Santiago 7610658, Chile;
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, NSW 2042, Australia
| | - Gonzalo Martínez Rodriguez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile; (J.F.B.); (M.P.O.)
- Correspondence:
| |
Collapse
|
11
|
Chen T, Liu G, Yu B. Colchicine for Coronary Artery Disease: A Review. Front Cardiovasc Med 2022; 9:892588. [PMID: 35783861 PMCID: PMC9246049 DOI: 10.3389/fcvm.2022.892588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Coronary artery disease is a serious threat to human health. More and more evidences indicate chronic inflammatory plays a key role in the development of this disease. Inflammation markers are gradually used in the diagnosis and treatment. Although the treatment of coronary heart disease with colchicine is still controversial, more and more studies showed that patients can benefit from this medicine. In this review, we discuss and summarize colchicine on essential pharmacology, anti-inflammatory mechanism of action, and the most important and recent clinical studies. According to these literatures, colchicine possibly will possibly become a new valuable and cheap medicine for the treatment of coronary artery disease.
Collapse
Affiliation(s)
- Tao Chen
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guihong Liu
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Bo Yu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Bo Yu,
| |
Collapse
|
12
|
Zhou J, Liu L, Wu P, Zhao L, Wu Y. Fusobacterium nucleatum Accelerates Atherosclerosis via Macrophage-Driven Aberrant Proinflammatory Response and Lipid Metabolism. Front Microbiol 2022; 13:798685. [PMID: 35359716 PMCID: PMC8963492 DOI: 10.3389/fmicb.2022.798685] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/03/2022] [Indexed: 12/18/2022] Open
Abstract
Periodontitis, an oral chronic inflammatory disease, is reported to show an association with atherosclerotic vascular disease. Fusobacterium nucleatum is an oral commensal bacterium that is abundantly implicated in various forms of periodontal diseases; however, its role in the pathogenesis of atherosclerosis is unclear. This study aimed to elucidate the underlying pathogenic mechanisms of atherosclerosis induced by F. nucleatum to provide new insight on the prevention and treatment of atherosclerosis. We used an animal model, that is, ApoE–/– mice were infected with F. nucleatum by oral gavage, and in vitro co-culture models to assess the pathogenicity of F. nucleatum. The results indicate that F. nucleatum ATCC 25586 invaded aortic tissues and substantially increased the progression of atherosclerotic lesions. In addition, F. nucleatum changed plaque composition into a less-stable phenotype, characterized with increased subcutaneous macrophage infiltration, M1 polarization, lipid deposition, cell apoptosis, and reduced extracellular matrix and collagen content. The serum levels of pro-atherosclerotic factors, such as interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, monocyte chemoattractant protein-1 (MCP-1), c-reactive protein, and oxidized low-density lipoprotein (ox-LDL) and microRNAs (miR-146a, miR-155, and miR-23b) were considerably increased after F. nucleatum stimulation, whereas HDL-c level was reduced. F. nucleatum induced in vitro macrophage apoptosis in a time- and dose-dependent manner. F. nucleatum facilitated ox-LDL–induced cholesterol phagocytosis and accumulation by regulating the expression of lipid metabolism-related genes (AR-A1, ACAT1, ABCA1, and ABCG1). F. nucleatum further worsened the atherosclerotic plaque microenvironment by considerably increasing the levels of IL-6; IL-1β; TNF-α; MCP-1; and MMP-2, 8, and 9 and by suppressing fibronectin (FN) 1 levels during foam cell formation. This study shows that F. nucleatum ATCC 25586 is implicated in atherosclerosis by causing aberrant activation and lipid metabolism in macrophage.
Collapse
Affiliation(s)
- Jieyu Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiyao Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Zheng Z, Wu B, Chen Q, Luo Y, Tang X, Wang J, Xie X, Dong R, Li W, Zhu J, Li S. The feasibility and safety of a simple method for coronary sinus blood sampling during catheter ablation of arrhythmias. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:170. [PMID: 35280379 PMCID: PMC8908160 DOI: 10.21037/atm-21-6973] [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: 11/30/2021] [Accepted: 02/11/2022] [Indexed: 11/20/2022]
Abstract
Background Coronary sinus (CS) blood sampling gives an insight into the localized pathophysiology of heart diseases. However, additional specifically-designed or modified catheters were needed in most previous studies, making the convenience of clinical application unsatisfactory. This study aimed to introduce a simple method for CS blood sampling without using additional catheters during catheter ablation (CA) of arrhythmias, and to investigate its feasibility and safety. Methods A total of 119 patients undergoing CA were prospectively enrolled, including 25 with paroxysmal supraventricular tachycardia (PSVT), 30 with premature ventricular complexes (PVC), and 64 with atrial fibrillation (AF). Cannulation and sampling of CS was performed via the femoral vein using a conventional 8F SR0TM or 8.5F SL1TM introducer sheath (St. Jude Medical) guided by a 6F steerable diagnostic catheter (St. Jude Medical). The success rate and any suspicious complications were recorded. Untargeted liquid chromatograph-mass spectrometer (LC-MS)-based metabonomics of CS samples versus peripheral venous samples were also performed. Results CS blood samples were successfully collected from 114 patients, with an overall success rate of 95.8%. Among patients with different arrhythmias, the success rates were similar, with 96.0% in PSVT, 96.7% in PVC, and 95.3% in AF (P=0.223). Adverse events occurred in four (3.4%) patients, including two patients with occasional atrial ectopic beats without causing any discomfort, and two patients with new-onset paroxysmal AF lasting for about 2 min. No serious complications were noted. Metabonomics analysis showed that CS samples provided a number of different metabolites (93 in PVST, 217 in PVC, and 109 in AF) versus peripheral samples. Conclusions Our method for CS blood sampling during CA is feasible and safe, and can provide useful cardiometabolic information that is significantly different from a peripheral sample.
Collapse
Affiliation(s)
- Zhenda Zheng
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bingyuan Wu
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qian Chen
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanting Luo
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xixiang Tang
- VIP Medical Service Center, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiafu Wang
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xujing Xie
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruiming Dong
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jieming Zhu
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Suhua Li
- Department of Cardiovascular Medicine, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Manipulating cellular microRNAs and analyzing high-dimensional gene expression data using machine learning workflows. STAR Protoc 2021; 2:100910. [PMID: 34746868 PMCID: PMC8554629 DOI: 10.1016/j.xpro.2021.100910] [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] [Indexed: 11/29/2022] Open
Abstract
MicroRNAs (miRNAs) are elements of the gene regulatory network and manipulating their abundance is essential toward elucidating their role in patho-physiological conditions. We present a detailed workflow that identifies important miRNAs using a machine learning algorithm. We then provide optimized techniques to validate the identified miRNAs through over-expression/loss-of-function studies. Overall, these protocols apply to any field in biology where high-dimensional data are produced. For complete details on the use and execution of this protocol, please refer to Wong et al. (2021a). LASSO and bootstrapping identify important miRNAs associated with gene of interest Generating puromycin resistant, doxycycline inducible, miRNA overexpressing cells Transient miRNA knockdown using LNA inhibitors in human primary cells Optimized reagent concentrations and cell densities for miRNA manipulation
Collapse
|
15
|
Yu J, Li Y, Leng D, Cao C, Yu Y, Wang Y. microRNA-3646 serves as a diagnostic marker and mediates the inflammatory response induced by acute coronary syndrome. Bioengineered 2021; 12:5632-5640. [PMID: 34519257 PMCID: PMC8806520 DOI: 10.1080/21655979.2021.1967066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Acute coronary syndrome (ACS) is one of the main syndromes of coronary artery disease with high mortality. The identification of biomarkers associated with disease occurrence and progression could improve early detection and risk prediction. This study was aimed to reveal the clinical significance and function of miR-3646 in ACS. The expression of miR-3646 was evaluated in ACS patients, healthy volunteers, and non-ACS patients and estimated the clinical significance of miR-3646. The ACS modeling rats were also established in this study to explore the potential mechanism underlying the function of miR-3646. miR-3646 was upregulated in ACS patients compared with healthy volunteers and non-ACS patients. The expression of miR-3646 was positively correlated with the severity and progression of ACS patients and could discriminate ACS patients from healthy volunteers and non-ACS patients. The knockdown of miR-3646 could reverse the inflammatory response induced by ACS.miR-3646 serves as a diagnostic biomarker for ACS. The knockdown of miR-3646 could alleviate ACS by reversing inflammatory response. These results provide a potential therapeutic target of ACS.
Collapse
Affiliation(s)
- Jinming Yu
- Department of Clinical Laboratory, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Yongmei Li
- Department of Clinical Laboratory, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Deguo Leng
- Department of Clinical Laboratory, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Cheng Cao
- Department of Clinical Laboratory, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Yongzhi Yu
- Department of Cardiology, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Yijuan Wang
- Department of Clinical Laboratory, Zibo Municipal Hospital, Zibo, Shandong, China
| |
Collapse
|
16
|
Joglekar MV, Wong WKM, Ema FK, Georgiou HM, Shub A, Hardikar AA, Lappas M. Postpartum circulating microRNA enhances prediction of future type 2 diabetes in women with previous gestational diabetes. Diabetologia 2021; 64:1516-1526. [PMID: 33755745 DOI: 10.1007/s00125-021-05429-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/14/2021] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes mellitus is a major cause of morbidity and death worldwide. Women with gestational diabetes mellitus (GDM) have greater than a sevenfold higher risk of developing type 2 diabetes in later life. Accurate methods for postpartum type 2 diabetes risk stratification are lacking. Circulating microRNAs (miRNAs) are well recognised as biomarkers/mediators of metabolic disease. We aimed to determine whether postpartum circulating miRNAs can predict the development of type 2 diabetes in women with previous GDM. METHODS In an observational study, plasma samples were collected at 12 weeks postpartum from 103 women following GDM pregnancy. Utilising a discovery approach, we measured 754 miRNAs in plasma from type 2 diabetes non-progressors (n = 11) and type 2 diabetes progressors (n = 10) using TaqMan-based real-time PCR on an OpenArray platform. Machine learning algorithms involving penalised logistic regression followed by bootstrapping were implemented. RESULTS Fifteen miRNAs were selected based on their importance in discriminating type 2 diabetes progressors from non-progressors in our discovery cohort. The levels of miRNA miR-369-3p remained significantly different (p < 0.05) between progressors and non-progressors in the validation sample set (n = 82; 71 non-progressors, 11 progressors) after adjusting for age and correcting for multiple comparisons. In a clinical model of prediction of type 2 diabetes that included six traditional risk factors (age, BMI, pregnancy fasting glucose, postpartum fasting glucose, cholesterol and triacylglycerols), the addition of the circulating miR-369-3p measured at 12 weeks postpartum improved the prediction of future type 2 diabetes from traditional AUC 0.83 (95% CI 0.68, 0.97) to an AUC 0.92 (95% CI 0.84, 1.00). CONCLUSIONS This is the first demonstration of miRNA-based type 2 diabetes prediction in women with previous GDM. Improved prediction will facilitate early lifestyle/drug intervention for type 2 diabetes prevention.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- Diabetes and Islet Biology Group, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Wilson K M Wong
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
- Diabetes and Islet Biology Group, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Fahmida K Ema
- Diabetes and Islet Biology Group, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| | - Harry M Georgiou
- Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Alexis Shub
- Department of Obstetrics and Gynaecology, University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Anandwardhan A Hardikar
- Diabetes and Islet Biology Group, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.
- Diabetes and Islet Biology Group, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia.
- Department of Science and Environment, Roskilde University, Roskilde, Denmark.
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, VIC, Australia.
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, VIC, Australia.
| |
Collapse
|
17
|
Elgebaly SA, Christenson RH, Kandil H, El-Khazragy N, Rashed L, Yacoub B, Eldeeb H, Ali M, Sharafieh R, Klueh U, Kreutzer DL. Nourin-Dependent miR-137 and miR-106b: Novel Early Inflammatory Diagnostic Biomarkers for Unstable Angina Patients. Biomolecules 2021; 11:368. [PMID: 33670982 PMCID: PMC7997347 DOI: 10.3390/biom11030368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/19/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Currently, no blood biomarkers exist that can diagnose unstable angina (UA) patients. Nourin is an early inflammatory mediator rapidly released within 5 min by reversible ischemic myocardium, and if ischemia persists, it is also released by necrosis. Nourin is elevated in acute coronary syndrome (ACS) patients but not in symptomatic noncardiac and healthy subjects. Recently, circulating microRNAs (miRNAs) have been established as markers of disease, including cardiac injury and inflammation. OBJECTIVES To profile and validate the potential diagnostic value of Nourin-dependent miR-137 (marker of cell damage) and miR-106b-5p (marker of inflammation) as early biomarkers in suspected UA patients and to investigate the association of their target and regulating genes. METHODS Using Nourin amino acid sequence, an integrated bioinformatics analysis was conducted. Analysis indicated that Nourin is a direct target for miR-137 and miR-106b-5p in myocardial ischemic injury. Two linked molecular networks of lncRNA/miRNAs/mRNAs were also retrieved, including CTB89H12.4/miR-137/FTHL-17 and CTB89H12.4/miR-106b-5p/ANAPC11. Gene expression profiling was assessed in serum samples collected at presentation to an emergency department (ED) from: (1) UA patients (n = 30) (confirmed by invasive coronary angiography with stenosis greater than 50% and troponin level below the clinical decision limit); (2) patients with acute ST elevation myocardial infarction (STEMI) (n = 16) (confirmed by persistent ST-segment changes and elevated troponin level); and 3) healthy subjects (n = 16). RESULTS Gene expression profiles showed that miR-137 and miR-106b-5p were significantly upregulated by 1382-fold and 192-fold in UA compared to healthy, and by 2.5-fold and 4.6-fold in STEMI compared to UA, respectively. Healthy subjects showed minimal expression profile. Receiver operator characteristics (ROC) analysis revealed that the two miRNAs were sensitive and specific biomarkers for assessment of UA and STEMI patients. Additionally, Spearman's correlation analysis revealed a significant association of miRNAs with the associated mRNA targets and the regulating lncRNA. CONCLUSIONS Nourin-dependent gene expression of miR-137 and miR-106b-5p are novel blood-based biomarkers that can diagnose UA and STEMI patients at presentation and stratify severity of myocardial ischemia, with higher expression in STEMI compared to UA. Early diagnosis of suspected UA patients using the novel Nourin biomarkers is key for initiating guideline-based therapy that improves patients' health outcomes.
Collapse
Affiliation(s)
- Salwa A. Elgebaly
- Research & Development, Nour Heart, Inc., Vienna, VA 22180, USA
- Department of Surgery, School of Medicine, UConn Health, Farmington, CT 06032, USA; (R.S.); (D.L.K.)
| | - Robert H. Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Hossam Kandil
- Department of Cardiology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo 11562, Egypt; (H.K.); (B.Y.); (H.E.); (M.A.)
| | - Nashwa El-Khazragy
- Department of Clinical Pathology-Hematology, Ain Shams Medical Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11566, Egyp;
| | - Laila Rashed
- Department of Biochemistry and Molecular Biology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo 11562, Egypt;
| | - Beshoy Yacoub
- Department of Cardiology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo 11562, Egypt; (H.K.); (B.Y.); (H.E.); (M.A.)
| | - Heba Eldeeb
- Department of Cardiology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo 11562, Egypt; (H.K.); (B.Y.); (H.E.); (M.A.)
| | - Mahmoud Ali
- Department of Cardiology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo 11562, Egypt; (H.K.); (B.Y.); (H.E.); (M.A.)
| | - Roshanak Sharafieh
- Department of Surgery, School of Medicine, UConn Health, Farmington, CT 06032, USA; (R.S.); (D.L.K.)
- Cell & Molecular Tissue Engineering, LLC Farmington, CT 06032, USA;
| | - Ulrike Klueh
- Cell & Molecular Tissue Engineering, LLC Farmington, CT 06032, USA;
- Integrative Biosciences Center (IBio), Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Donald L. Kreutzer
- Department of Surgery, School of Medicine, UConn Health, Farmington, CT 06032, USA; (R.S.); (D.L.K.)
- Cell & Molecular Tissue Engineering, LLC Farmington, CT 06032, USA;
| |
Collapse
|
18
|
Kurup R, Galougahi KK, Figtree G, Misra A, Patel S. The Role of Colchicine in Atherosclerotic Cardiovascular Disease. Heart Lung Circ 2021; 30:795-806. [PMID: 33461916 DOI: 10.1016/j.hlc.2020.11.010] [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: 08/09/2020] [Revised: 10/10/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023]
Abstract
Colchicine, an inexpensive immunomodulatory drug used traditionally to treat gout and familial Mediterranean fever, is rapidly accumulating basic and clinical evidence for a therapeutic role in atherosclerotic cardiovascular disease. Its athero-protective properties are thought to be mainly related to its effect on tubulin polymerisation, enabling a broad range of effect on multiple atherosclerotic plaque cell types and cellular processes, including cell division, cell migration as well as pro-inflammatory cytokine and chemokine secretion. These properties indicate the potential to favourably affect all stages of atherosclerotic plaque development including formation, progression, destabilisation, and plaque rupture. This review focusses on the pharmacology of colchicine, the mechanisms by which it modulates atherosclerosis pathobiology, and summarises the current clinical evidence for its use along with the upcoming clinical trial landscape. Given the current lack of primary immunomodulatory drugs in the treatment of atherosclerosis, colchicine is a promising candidate to fill this therapeutic gap.
Collapse
Affiliation(s)
- Rahul Kurup
- The Heart Research Institute, Sydney, NSW, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia. https://twitter.com/drrahulkurup
| | - Keyvan Karimi Galougahi
- The Heart Research Institute, Sydney, NSW, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Gemma Figtree
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Ashish Misra
- The Heart Research Institute, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Sanjay Patel
- The Heart Research Institute, Sydney, NSW, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
19
|
Mortazavi-Jahromi SS, Aslani M, Mirshafiey A. A comprehensive review on miR-146a molecular mechanisms in a wide spectrum of immune and non-immune inflammatory diseases. Immunol Lett 2020; 227:8-27. [PMID: 32810557 DOI: 10.1016/j.imlet.2020.07.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/10/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are single-strand endogenous and non-coding RNA molecules with a length of about 22 nucleotides, which regulate genes expression, through modulating the translation and stability of their target mRNAs. miR-146a is one of the most studied miRNAs, due to its central role in immune system homeostasis and control of the innate and acquired immune responses. Accordingly, abnormal expression or function of miR-146a results in the incidence and progression of immune and non-immune inflammatory diseases. Its deregulated expression pattern and inefficient function have been reported in a wide spectrum of these illnesses. Based on the existing evidence, this miRNA qualifies as an ideal biomarker for diagnosis, prognosis, and activity evaluation of immune and non-immune inflammatory disorders. Moreover, much attention has recently been paid to therapeutic potential of miR-146a and several researchers have assessed the effects of different drugs on expression and function of this miRNA at diverse experimental, animal, besides human levels, reporting motivating results in the treatment of the diseases. Here, in this comprehensive review, we provide an overview of miR-146a role in the pathogenesis and progression of several immune and non-immune inflammatory diseases such as Rheumatoid arthritis, Systemic lupus erythematosus, Inflammatory bowel disease, Multiple sclerosis, Psoriasis, Graves' disease, Atherosclerosis, Hepatitis, Chronic obstructive pulmonary disease, etc., discuss about its eligibility for being a desirable biomarker for these disorders, and also highlight its therapeutic potential. Understanding these mechanisms underlies the selecting and designing the proper therapeutic targets and medications, which eventually facilitate the treatment process.
Collapse
Affiliation(s)
| | - Mona Aslani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|