Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 14.546
1.
Int J Rheum Dis ; 27(5): e15162, 2024 May.
Article En | MEDLINE | ID: mdl-38720421

BACKGROUND: Recent findings suggest a link between gout and the development of dementia. Early treatment with colchicine is recommended as a first-line therapy for gout flares. Animal studies demonstrate that colchicine could induce cognitive impairment. This cohort study aimed to investigate the association between colchicine use and the risk of developing dementia. METHODS: In this nationwide cohort study, we performed comparative analysis on 6147 patients ≥40 years, with gout and colchicine new users against 6147 controls to assess subsequent dementia risk. The colchicine group and the control group (urate lowering therapy group) were matched on the bases of age, sex, index year, and comorbidities. All participants were followed for up to 14 years for a diagnosis of dementia considering medical records were retrospectively checked over this period. Cox proportional hazards models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). Sensitivity analyses were performed to validate our findings. RESULTS: The adjusted hazard ratio (aHR) of dementia for colchicine users was 1.45 (95% CI = 1.05, 1.99) relative to comparison group after adjusting for sex, age, and comorbidities. Sensitivity analysis aiming to minimize underdiagnosed occult dementia at the time of index year yielded consistent positive association. In higher accumulative dose colchicine group (cumulative defined daily dose [cDDD] >30), the aHR of dementia risk for colchicine users was 1.42 (95% CI = 1.03, 1.97) compared with nonusers. For those duration of colchicine use >30 days, the aHR was 1.53 (95% CI = 1.01-2.32) compared to the nonuser group. CONCLUSIONS: A significant risk of dementia was observed in this study in patients with gout using colchicine at higher cDDD and for a longer period. Further research is needed to elucidate the relationship between colchicine, gout, and dementia.


Colchicine , Dementia , Gout Suppressants , Gout , Humans , Colchicine/adverse effects , Colchicine/therapeutic use , Gout/epidemiology , Gout/drug therapy , Dementia/epidemiology , Dementia/chemically induced , Dementia/diagnosis , Female , Male , Retrospective Studies , Aged , Middle Aged , Gout Suppressants/adverse effects , Risk Factors , Risk Assessment , Time Factors , Taiwan/epidemiology , Adult , Aged, 80 and over , Databases, Factual
2.
BMC Plant Biol ; 24(1): 428, 2024 May 21.
Article En | MEDLINE | ID: mdl-38773358

BACKGROUND: Acacia nilotica Linn. is a widely distributed tree known for its applications in post-harvest and medicinal horticulture. However, its seed-based growth is relatively slow. Seed is a vital component for the propagation of A. nilotica due to its cost-effectiveness, genetic diversity, and ease of handling. Colchicine, commonly used for polyploidy induction in plants, may act as a pollutant at elevated levels. Its optimal concentration for Acacia nilotica's improved growth and development has not yet been determined, and the precise mechanism underlying this phenomenon has not been established. Therefore, this study investigated the impact of optimized colchicine (0.07%) seed treatment on A. nilotica's morphological, anatomical, physiological, fluorescent, and biochemical attributes under controlled conditions, comparing it with a control. RESULTS: Colchicine seed treatment significantly improved various plant attributes compared to control. This included increased shoot length (84.6%), root length (53.5%), shoot fresh weight (59.1%), root fresh weight (42.8%), shoot dry weight (51.5%), root dry weight (40%), fresh biomass (23.6%), stomatal size (35.9%), stomatal density (41.7%), stomatal index (51.2%), leaf thickness (11 times), leaf angle (2.4 times), photosynthetic rate (40%), water use efficiency (2.2 times), substomatal CO2 (36.6%), quantum yield of photosystem II (13.1%), proton flux (3.1 times), proton conductivity (2.3 times), linear electron flow (46.7%), enzymatic activities of catalase (25%), superoxide dismutase (33%), peroxidase (13.5%), and ascorbate peroxidase (28%), 2,2-diphenyl-1-picrylhydrazyl-radical scavenging activities(23%), total antioxidant capacity (59%), total phenolic (23%), and flavonoid content (37%) with less number of days to 80% germination (57.1%), transpiration rate (53.9%), stomatal conductance (67.1%), non-photochemical quenching (82.8%), non-regulatory energy dissipation (24.3%), and H2O2 (25%) and O-2 levels (30%). CONCLUSION: These findings elucidate the intricate mechanism behind the morphological, anatomical, physiological, fluorescent, and biochemical transformative effects of colchicine seed treatment on Acacia nilotica Linn. and offer valuable insights for quick production of A. nilotica's plants with modification and enhancement from seeds through an eco-friendly approach.


Acacia , Colchicine , Seeds , Colchicine/pharmacology , Acacia/drug effects , Acacia/physiology , Acacia/growth & development , Acacia/metabolism , Seeds/drug effects , Seeds/growth & development , Photosynthesis/drug effects , Antioxidants/metabolism
3.
BMC Plant Biol ; 24(1): 439, 2024 May 22.
Article En | MEDLINE | ID: mdl-38778255

BACKGROUND: Glehnia littoralis is a medicinal and edible plant species having commercial value and has several hundred years of cultivation history. Polyploid breeding is one of the most important and fastest ways to generate novel varieties. To obtain tetraploids of G. littoralis in vitro, colchicine treatment was given to the seeds and then were screened based on morphology, flow cytometry, and root tip pressing assays. Furthermore, transcriptome analysis was performed to identity the differentially expressed genes associated with phenotypic changes in tetraploid G. littoralis. RESULTS: The results showed that 0.05% (w/v) colchicine treatment for 48 h was effective in inducing tetraploids in G. littoralis. The tetraploid G. littoralis (2n = 4x = 44) was superior in leaf area, leaf thickness, petiole diameter, SPAD value (Chl SPAD), stomatal size, epidermal tissues thickness, palisade tissues thickness, and spongy tissues thickness to the diploid ones, while the stomatal density of tetraploids was significantly lower. Transcriptome sequencing revealed, a total of 1336 differentially expressed genes (DEGs) between tetraploids and diploids. Chromosome doubling may lead to DNA content change and gene dosage effect, which directly affects changes in quantitative traits, with changes such as increased chlorophyll content, larger stomata and thicker tissue of leaves. Several up-regulated DEGs were found related to growth and development in tetraploid G. littoralis such as CKI, PPDK, hisD and MDP1. KEGG pathway enrichment analyses showed that most of DEGs were enriched in metabolic pathways. CONCLUSIONS: This is the first report of the successful induction of tetraploids in G. littoralis. The information presented in this study facilitate breeding programs and molecular breeding of G. littoralis varieties.


Gene Expression Profiling , Phenotype , Tetraploidy , Transcriptome , Colchicine/pharmacology , Caryophyllales/genetics , Gene Expression Regulation, Plant , Plant Leaves/genetics , Plant Leaves/growth & development , Plant Leaves/anatomy & histology
4.
Ann Intern Med ; 177(5): JC54, 2024 May.
Article En | MEDLINE | ID: mdl-38710085

SOURCE CITATION: Roubille F, Bouabdallaoui N, Kouz S, et al. Low-dose colchicine in patients with type 2 diabetes and recent myocardial infarction in the COLchicine Cardiovascular Outcomes Trial (COLCOT). Diabetes Care. 2024;47:467-470. 38181203.


Colchicine , Diabetes Mellitus, Type 2 , Myocardial Infarction , Colchicine/therapeutic use , Colchicine/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Humans , Myocardial Infarction/prevention & control , Male , Middle Aged , Female , Aged
5.
Immun Inflamm Dis ; 12(5): e1273, 2024 May.
Article En | MEDLINE | ID: mdl-38798123

BACKGROUND: Previous research done in Bulgaria demonstrated a fivefold reduction in mortality from COVID-19 with increased doses of colchicine from two hospitals in the country. We report here a further 333 cases of COVID-19 inpatients, treated with different doses of colchicine and its effect on mortality. MATERIALS AND METHODS: A case-control comparison from two additional hospitals was conducted between increased doses of colchicine and added bromhexine to standard of care (SOC) versus current SOC. Risk and odds ratio, as well as subgroup analysis, was conducted with newly reported data, alongside aggregate data from all hospital centers to determine the extent of mortality reduction in COVID-19 inpatients. RESULTS: There was a clear reduction in the mortality of inpatients with increasing doses of colchicine-between twofold and sevenfold. Colchicine loading doses of 4 mg are more effective than those with 2 mg. Despite these doses being higher than the so-called "standard doses," colchicine inpatients experienced lower mortality than SOC patients (5.7% vs. 19.53%). This mortality benefit was evident in different age subgroups, with a 4-mg loading dose of colchicine proving slightly superior to a 2-mg loading dose. Colchicine led to an overall relative risk reduction of 70.7%, with SOC patients having 3.91 higher odds of death. The safety of the doses was not different than the reported in the summary of product characteristics. CONCLUSION: Inpatients in Bulgaria with added colchicine and bromhexine to SOC achieved better clinical and mortality outcomes than those on SOC alone. These results question the World Health Organization-recommended strategy to inhibit viral replication. We posit that our treatment strategy to inhibit the Severe acute respiratory syndrome coronavirus 2 entry into the cell with inhaled bromhexine and the hyperactivated NLRP3 inflammasome with higher doses of colchicine, prevents the development of cytokine storm. The timing of the initiation of treatment seems critical.


COVID-19 Drug Treatment , COVID-19 , Colchicine , SARS-CoV-2 , Humans , Colchicine/administration & dosage , Colchicine/therapeutic use , Female , Middle Aged , Male , Aged , COVID-19/mortality , COVID-19/epidemiology , Case-Control Studies , Adult , Aged, 80 and over , Bulgaria/epidemiology , Inpatients , Dose-Response Relationship, Drug , Treatment Outcome
7.
J Am Heart Assoc ; 13(9): e033700, 2024 May 07.
Article En | MEDLINE | ID: mdl-38700005

BACKGROUND: The only clinically approved drug that reduces doxorubicin cardiotoxicity is dexrazoxane, but its application is limited due to the risk of secondary malignancies. So, exploring alternative effective molecules to attenuate its cardiotoxicity is crucial. Colchicine is a safe and well-tolerated drug that helps reduce the production of reactive oxygen species. High doses of colchicine have been reported to block the fusion of autophagosomes and lysosomes in cancer cells. However, the impact of colchicine on the autophagy activity within cardiomyocytes remains inadequately elucidated. Recent studies have highlighted the beneficial effects of colchicine on patients with pericarditis, postprocedural atrial fibrillation, and coronary artery disease. It remains ambiguous how colchicine regulates autophagic flux in doxorubicin-induced heart failure. METHODS AND RESULTS: Doxorubicin was administered to establish models of heart failure both in vivo and in vitro. Prior studies have reported that doxorubicin impeded the breakdown of autophagic vacuoles, resulting in damaged mitochondria and the accumulation of reactive oxygen species. Following the administration of a low dose of colchicine (0.1 mg/kg, daily), significant improvements were observed in heart function (left ventricular ejection fraction: doxorubicin group versus treatment group=43.75%±3.614% versus 57.07%±2.968%, P=0.0373). In terms of mechanism, a low dose of colchicine facilitated the degradation of autolysosomes, thereby mitigating doxorubicin-induced cardiotoxicity. CONCLUSIONS: Our research has shown that a low dose of colchicine is pivotal in restoring the autophagy activity, thereby attenuating the cardiotoxicity induced by doxorubicin. Consequently, colchicine emerges as a promising therapeutic candidate to improve doxorubicin cardiotoxicity.


Autophagy , Cardiotoxicity , Colchicine , Doxorubicin , Lysosomes , Myocytes, Cardiac , Colchicine/toxicity , Colchicine/pharmacology , Doxorubicin/toxicity , Cardiotoxicity/prevention & control , Autophagy/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Disease Models, Animal , Male , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/metabolism , Antibiotics, Antineoplastic/toxicity , Reactive Oxygen Species/metabolism , Mice , Mice, Inbred C57BL , Ventricular Function, Left/drug effects
8.
Am Fam Physician ; 109(5): 441-446, 2024 May.
Article En | MEDLINE | ID: mdl-38804758

Acute pericarditis is defined as inflammation of the pericardium and occurs in approximately 4.4% of patients who present to the emergency department for nonischemic chest pain, with a higher prevalence in men. Although there are numerous etiologies of pericarditis, most episodes are idiopathic and the cause is presumed to be viral. Diagnosis of pericarditis requires at least two of the following criteria: new or worsening pericardial effusion, characteristic pleuritic chest pain, pericardial friction rub, or electrocardiographic changes, including new, widespread ST elevations or PR depressions. Pericardial friction rubs are highly specific but transient, and they have been reported in 18% to 84% of patients with acute pericarditis. Classic electrocardiographic findings include PR-segment depressions; diffuse, concave, upward ST-segment elevations without reciprocal changes; and T-wave inversions. Transthoracic echocardiography should be performed in all patients with acute pericarditis to characterize the size of effusions and evaluate for complications. Nonsteroidal anti-inflammatory drugs are the first-line treatment option. Glucocorticoids should be reserved for patients with contraindications to first-line therapy and those who are pregnant beyond 20 weeks' gestation or have other systemic inflammatory conditions. Colchicine should be used in combination with first- or second-line treatments to reduce the risk of recurrence. Patients with a higher risk of complications should be admitted to the hospital for further workup and treatment.


Anti-Inflammatory Agents, Non-Steroidal , Electrocardiography , Pericarditis , Humans , Pericarditis/diagnosis , Pericarditis/physiopathology , Pericarditis/therapy , Acute Disease , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colchicine/therapeutic use , Echocardiography , Female , Pericardial Effusion/diagnosis , Pericardial Effusion/therapy , Pericardial Effusion/etiology , Chest Pain/etiology , Chest Pain/diagnosis , Male , Glucocorticoids/therapeutic use
9.
Int Immunopharmacol ; 132: 111967, 2024 May 10.
Article En | MEDLINE | ID: mdl-38569431

OBJECTIVES: Canakinumab, a human monoclonal antibody targeted at interleukin-1 beta, has demonstrated safety and efficacy in preventing familial Mediterranean fever (FMF) attacks among individuals with colchicine-resistant (crFMF). The manufacturer orders prescribe monthly subcutaneous injections. However, a subset of our patients is treated with an "canakinumab on demand " (COD) strategy, with wider intervals between drug administrations. Therefore, we aimed to compare disease activity and drug safety between COD and "canakinumab fixed frequency" (CFF) policies. METHODS: This retrospective study collected data from three Israeli paediatric rheumatology centres, of children with crFMF who were treated with canakinumab. Epidemiological and clinical parameters, cumulative drug dosages, and adverse events were compared between children treated by both policies. RESULTS: Twenty-five (49 %) children were treated according to COD policy and 26 according to CFF policy. Demographic parameters and most of the disease features did not differ significantly between the groups. Both groups showed significant reduction in attacks after canakinumab introduction. The median number (interquartile range) of attacks per month did not differ significantly between the COD and CFF groups (0.33 (0.08, 0.58) and 0.13 (0, 0.5), respectively, p = 0.485 (even though, per definition, COD patients presumably had an attack before receiving the second canakinumab dose). The mean monthly dose was lower for the COD than the CFF group (1.13 ± 1.13 vs. 3.16 ± 1.46 mg/kg, p < 0.001). Adverse events were similar between the groups. CONCLUSION: For individuals with crFMF, COD compared to CFF policy can achieve similar efficacy and safety, with a lower accumulated canakinumab dose, rendering it less immunosuppressive and less expensive.


Antibodies, Monoclonal, Humanized , Colchicine , Drug Resistance , Familial Mediterranean Fever , Humans , Familial Mediterranean Fever/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Child , Male , Female , Retrospective Studies , Colchicine/therapeutic use , Colchicine/administration & dosage , Colchicine/adverse effects , Adolescent , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/immunology , Treatment Outcome , Child, Preschool , Israel , Drug Administration Schedule
10.
Int J Pharm ; 657: 124170, 2024 May 25.
Article En | MEDLINE | ID: mdl-38679244

Improving the anticancer efficacy of chemotherapeutic drugs and photosensitizers requires innovative multifunctional nanoplatforms. This study introduces a chemo- and phototherapeutic drug delivery system (DDS) based on poly(lactide-co-glycolide) (PLGA) nanoparticles (NPs), both PEGylated and non-PEGylated, with a mean size of 200 ± 75 nm. Colchicine (Colch) and purpurin18 (P18) were co-encapsulated into these NPs, and their in vitro drug release profiles were investigated. The anticancer potential of these systems was evaluated across various cell lines (i.e., CaCo-2, PC-3, MCF-7, and MRC-5 cells), demonstrating enhanced NP uptake by cancer cells compared to free drugs. Co-administration of Colch and P18 in 2D and 3D cell line models exhibited a synergistic effect, harnessing both chemotherapeutic and photodynamic effects, leading to higher cancer cell elimination efficacy. This newly developed multifunctional DDS presents a promising platform for combined chemo- and photodynamic therapy in cancer treatment.


Colchicine , Drug Carriers , Drug Liberation , Polylactic Acid-Polyglycolic Acid Copolymer , Humans , Colchicine/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Drug Carriers/chemistry , Cell Line, Tumor , Spheroids, Cellular/drug effects , Nanoparticles/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Photochemotherapy/methods , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Neoplasms/drug therapy , Cell Survival/drug effects , Drug Delivery Systems/methods
11.
Int J Cardiovasc Imaging ; 40(4): 945-948, 2024 Apr.
Article En | MEDLINE | ID: mdl-38558330

In the last century, there has been more than enough research that proved the association of high lipid and glucose levels with cardiovascular disease, thus establishing the current well-known traditional cardiovascular risk factors such as dyslipidemia, diabetes, and metabolic syndrome. Hence, these cardiovascular risk factors are target therapy for glucose and lipid-lowering agents to prevent adverse cardiovascular events. However, despite controlling the lipid and glucose levels, some studies demonstrated the subclinical atherosclerosis suggesting that these cardiovascular risk factors alone cannot account for the entire atherosclerosis burden. In the last years, large-scale clinical trials demonstrated the operation of the inflammatory pathway in atherosclerotic cardiovascular disease (ASCVD) by the immune system, both the innate (neutrophils, macrophages) and adaptive (T cell and other lymphocytes) limbs, contribute to atherosclerosis and atherothrombosis. In this regard, some studies that use antiinflammatory therapy targeting the immune system by modulating or blocking interleukins, also known as anti-cytokine therapy, have been shown to reduce the risk of adverse cardiovascular events in patients with previous coronary artery disease. In this regard, the U.S. Food and Drug Administration (FDA) approved the use of colchicine 0.5 mg once daily for reducing cardiovascular events in patients who have established ASCVD and high residual systemic inflammation. Therefore, measuring the systemic inflammation can improve the cardiovascular risk assessment and identify the subsets of patients that will benefit from anti-cytokine therapy after diagnosis of ASCVD or after myocardial revascularization.


Anti-Inflammatory Agents , Biomarkers , Blood Glucose , Cytokines , Heart Disease Risk Factors , Inflammation Mediators , Inflammation , Triglycerides , Humans , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/adverse effects , Atherosclerosis/immunology , Atherosclerosis/drug therapy , Atherosclerosis/blood , Biomarkers/blood , Blood Glucose/drug effects , Blood Glucose/metabolism , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/immunology , Colchicine/therapeutic use , Colchicine/adverse effects , Cytokines/blood , Cytokines/metabolism , Inflammation/immunology , Inflammation/drug therapy , Inflammation/blood , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Risk Assessment , Risk Factors , Treatment Outcome , Triglycerides/blood
12.
Sci Rep ; 14(1): 9270, 2024 04 23.
Article En | MEDLINE | ID: mdl-38649421

The present study explored the anticancer activity of a Chitosan-based nanogel incorporating thiocolchicoside and lauric acid (CTL) against oral cancer cell lines (KB-1). Cell viability, AO/EtBr dual staining and Cell cycle analysis were done to evaluate the impact of CTL nanogel on oral cancer cells. Real-time PCR was performed to analyze proapoptotic and antiapoptotic gene expression in CTL-treated KB-1 cells. Further, molecular docking analysis was conducted to explore the interaction of our key ingredient, thiocolchicoside and its binding affinities. The CTL nanogel demonstrated potent anticancer activity by inhibiting oral cancer cell proliferation and inducing cell cycle arrest in cancer cells. Gene expression analysis indicated alterations in Bax and Bcl-2 genes; CTL nanogel treatment increased Bax mRNA expression and inhibited the Bcl-2 mRNA expression, which showed potential mechanisms of the CTL nanogel's anticancer action. It was found that thiocolchicoside can stabilize the protein's function or restore it as a tumour suppressor. The CTL nanogel exhibited excellent cytotoxicity and potent anticancer effects, making it a potential candidate for non-toxic chemotherapy in cancer nanomedicine. Furthermore, the nanogel's ability to modulate proapoptotic gene expression highlights its potential for targeted cancer therapy. This research contributes to the growing interest in Chitosan-based nanogels and their potential applications in cancer treatment.


Antineoplastic Agents , Apoptosis , Chitosan , Colchicine , Colchicine/analogs & derivatives , Lauric Acids , Mouth Neoplasms , Nanogels , Polyethyleneimine , Humans , Chitosan/analogs & derivatives , Chitosan/chemistry , Chitosan/pharmacology , Lauric Acids/chemistry , Lauric Acids/pharmacology , Cell Line, Tumor , Nanogels/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Colchicine/pharmacology , Apoptosis/drug effects , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Molecular Docking Simulation , Cell Proliferation/drug effects , Cell Survival/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology
13.
Bioorg Med Chem Lett ; 105: 129745, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38614151

A series of 8 novel pyridinyl 4-(2-oxoimidazolidin-1-yl)benzenesulfonates (PYRIB-SOs) were designed, prepared and evaluated for their mechanism of action. PYRIB-SOs were found to have antiproliferative activity in the nanomolar to submicromolar range on several breast cancer cell lines. Moreover, subsequent biofunctional assays indicated that the most potent PYRIB-SOs 1-3 act as antimitotics binding to the colchicine-binding site (C-BS) of α, ß-tubulin and that they arrest the cell cycle progression in the G2/M phase. Microtubule immunofluorescence and tubulin polymerisation assay confirm that they disrupt the cytoskeleton through inhibition of tubulin polymerisation as observed with microtubule-destabilising agents. They also show good overall theoretical physicochemical, pharmacokinetic and druglike properties. Overall, these results show that PYRIB-SOs is a new family of promising antimitotics to be further studied in vivo for biopharmaceutical and pharmacodynamic evaluations.


Antimitotic Agents , Cell Proliferation , Colchicine , Drug Screening Assays, Antitumor , Humans , Colchicine/chemistry , Colchicine/metabolism , Colchicine/pharmacology , Binding Sites , Antimitotic Agents/pharmacology , Antimitotic Agents/chemistry , Antimitotic Agents/chemical synthesis , Structure-Activity Relationship , Cell Proliferation/drug effects , Cell Line, Tumor , Benzenesulfonates/chemistry , Benzenesulfonates/pharmacology , Benzenesulfonates/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Tubulin/metabolism , Molecular Structure , Tubulin Modulators/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Pyridines/chemical synthesis , Dose-Response Relationship, Drug
14.
Int J Cardiol ; 406: 132068, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38648916

BACKGROUND: Atrial fibrillation (AF), the most common arrhythmia, is closely related to inflammation. Colchicine has the potent anti-inflammatory effects. Several randomized clinical trials (RCTs) have evaluated the efficacy and safety of colchicine in the prevention of AF but the results are inconsistent. OBJECTIVE: The purpose of our study was to evaluate the impact of colchicine on AF. METHOD AND RESULTS: PubMed, EMBASE, Web of Science, Cochrane Library and ClinicalTrials.gov were searched for related studies until Jan 8, 2024. A total of 17 studies including 16,238 participants were included. Compared to the placebo group, there were fewer incidences of AF in the colchicine group (RR: 0.75, 95%CI: 0.68-0.83, P < 0.001). The incidence of overall adverse events and overall gastrointestinal intolerance did not differ significantly between the two groups. However, diarrhea, nausea, and discontinuation occurred more frequently in patients treated with colchicine. CONCLUSION: Colchicine can prevent patients from the incidence of AF, regardless of the mean age of patients, type of atrial fibrillation, maintenance dose, duration of colchicine use, cumulative daily dose, and follow-up time with more diarrhea, nausea and discontinuation. These adverse events can be avoided by low doses (0.5 mg once daily) and long period time of colchicine use.


Atrial Fibrillation , Colchicine , Randomized Controlled Trials as Topic , Colchicine/therapeutic use , Colchicine/adverse effects , Atrial Fibrillation/prevention & control , Atrial Fibrillation/drug therapy , Humans , Randomized Controlled Trials as Topic/methods , Treatment Outcome
15.
Spectrochim Acta A Mol Biomol Spectrosc ; 315: 124272, 2024 Jul 05.
Article En | MEDLINE | ID: mdl-38603960

Visualization of cell apoptosis is a critical task playing central roles in the fundamental studies in biology, pathology, and biomedicine. Dual-emissive fluorescent probes are desired molecular tools for study on apoptosis, which however were rarely reported. Herein, utilizing the polarity differences between lysosomes and nucleus, a translocation type of fluorescent probe (NA-S) was developed for the dual-color visualization of cell apoptosis. NA-S was designed to be polarity sensitive, bearing alkalescence group, and with DNA affinity. In living cells, NA-S targeted the lysosomes to give blue fluorescence, which translocated into the nucleus during cell apoptosis to give green emission. Thereby, the cell apoptosis could be visualized with NA-S in dual-emissive manner. With the unique probe, the cell apoptosis induced by oxidative stress, UV irradiation, rotenone, colchicine, and paclitaxel have been successfully visualized.


Apoptosis , Cell Nucleus , Fluorescent Dyes , Lysosomes , Apoptosis/drug effects , Lysosomes/metabolism , Humans , Fluorescent Dyes/chemistry , Cell Nucleus/metabolism , Spectrometry, Fluorescence , HeLa Cells , Oxidative Stress , Colchicine/pharmacology , Rotenone/pharmacology , Paclitaxel/pharmacology
16.
Curr Cardiol Rep ; 26(5): 423-434, 2024 May.
Article En | MEDLINE | ID: mdl-38573553

PURPOSE OF REVIEW: Targeting traditional cardiovascular risk factors is effective in reducing recurrent cardiovascular events, yet the presence of residual cardiovascular risk due to underlying systemic inflammation is a largely unaddressed opportunity. This review aims to comprehensively assess the evolving role of colchicine as a therapeutic approach targeting residual inflammatory risk in the context of those with coronary artery disease (CAD). RECENT FINDINGS: Inflammation plays a significant role in promoting atherosclerosis, and targeting anti-inflammatory pathways has the potential to decrease cardiovascular events. Low-dose colchicine (0.5 mg/day orally), when added to guideline-directed medical care for CAD, safely decreases major adverse cardiovascular events (MACE) by 31% in stable atherosclerosis patients and 23% in those after recent myocardial infarctions. Meta-analyses of recent randomized control trials further support both the efficacy and safety of colchicine, particularly when added to other standard cardiovascular therapies, including statin therapy. The European Society of Cardiology and other national guidelines endorse the use of low-dose colchicine in patients across the spectrum of CAD. Recently, colchicine was FDA-approved in the United States as the first anti-inflammatory therapy for the reduction of cardiovascular events. In a period of a rising incidence of CAD across the globe, colchicine represents a unique opportunity to decrease MACE due to its large magnitude of benefits and general affordability. However, challenges with drug interactions must be addressed, especially in those regions where HIV, hepatitis, and tuberculosis are prevalent. Colchicine is safe and effective at reducing cardiovascular events across a broad spectrum of coronary syndromes. The ability to simultaneously target traditional risk factors and mitigate residual inflammatory risk marks a substantial advancement in cardiovascular prevention strategies, heralding a new era in the global battle against CAD.


Cardiovascular Diseases , Colchicine , Colchicine/therapeutic use , Humans , Cardiovascular Diseases/prevention & control , Coronary Artery Disease/prevention & control , Coronary Artery Disease/drug therapy , Anti-Inflammatory Agents/therapeutic use , Inflammation/drug therapy , Inflammation/prevention & control , Heart Disease Risk Factors , Randomized Controlled Trials as Topic , Global Health , Risk Factors
17.
Int J Biol Sci ; 20(6): 2092-2110, 2024.
Article En | MEDLINE | ID: mdl-38617538

Development of non-surgical treatment of human abdominal aortic aneurysm (AAA) has clinical significance. Colchicine emerges as an effective therapeutic regimen in cardiovascular diseases. Yet, whether colchicine slows AAA growth remain controversy. Here, we demonstrated that daily intragastric administration of low-dose colchicine blocked AAA formation, prevented vascular smooth muscle cell (SMC) phenotype switching and apoptosis, and vascular inflammation in both peri-aortic CaPO4 injury and subcutaneous angiotensin-II infusion induced experimental AAA mice models. Mechanistically, colchicine increased global mRNA stability by inhibiting the METTL14/YTHDC1-mediated m6A modification, resulting in increased sclerostin (SOST) expression and consequent inactivation of the WNT/ß-catenin signaling pathway in vascular SMCs from mouse AAA lesions and in cultured human aortic SMCs. Moreover, human and mouse AAA lesions all showed increased m6A methylation, decreased SOST expression, and skewed synthetic SMC de-differentiation phenotype, compared to those without AAA. This study uncovers a novel mechanism of colchicine in slowing AAA development by using the METTL14/SOST/WNT/ß-catenin axis to control vascular SMC homeostasis in mouse aortic vessels and in human aortic SMCs. Therefore, use of colchicine may benefit AAA patients in clinical practice.


Aortic Aneurysm, Abdominal , Muscle, Smooth, Vascular , Humans , Animals , Mice , Aortic Aneurysm, Abdominal/drug therapy , Homeostasis , Aorta , Colchicine/therapeutic use
18.
Eur Heart J ; 45(18): 1596-1601, 2024 May 13.
Article En | MEDLINE | ID: mdl-38596868

Low-dose colchicine (0.5 mg daily) is now FDA-approved for secondary prevention in patients with coronary disease and will be increasingly prescribed in clinical practice. In this State-of-the-Art Review, data were collated from contemporary systemic reviews of case reports, drug registries, and placebo-controlled trials that assessed specific issues of safety related to the continuous use of colchicine in a range of clinical settings to inform physicians, pharmacists, and patients of the absolute risks of continuous use of low-dose colchicine, including among individuals taking statin therapy. Based upon these collective data, it is concluded that aside mild diarrhoea on initiation of colchicine that typically subsides in the vast majority of patients within a week of therapy, continuous use of low-dose colchicine is well tolerated and very safe. It does not affect renal, liver, or cognitive function, has no adverse effects on bleeding, wound healing, fertility, or pregnancy, and does not increase risks of cancer, serious infection, or cause-specific mortality. When appropriately prescribed to patients without significant renal or hepatic impairment, reports of myelosuppression, myotoxicity, and serious drug-drug interactions are rare and no more frequent than placebo, including in patients taking statin therapy. Physicians, pharmacists, and patients can be reassured that in the absence of significant renal or hepatic impairment continuous use of low-dose colchicine can be used safely in patients with atherosclerosis for the purpose of reducing cardiovascular risk.


Colchicine , Colchicine/administration & dosage , Colchicine/adverse effects , Humans , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Drug Interactions , Female , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use
19.
BMC Pulm Med ; 24(1): 190, 2024 Apr 20.
Article En | MEDLINE | ID: mdl-38641775

BACKGROUND: The Coronavirus disease 2019 (COVID-19) pandemic has robustly affected the global healthcare and economic systems and it was caused by coronavirus-2 (SARS-CoV-2). The clinical presentation of the disease ranges from a flu-like illness to severe pneumonia and death. Till September 2022, the cumulative number of cases exceeded 600 million worldwide and deaths were more than 6 million. Colchicine is an alkaloid drug that is used in many autoinflammatory conditions e.g., gout, familial Mediterranean fever, and Behçet's syndrome. Colchicine inhibits the production of superoxide and the release of interleukins that stimulate the inflammatory cascade. Colchicine decreases the differentiation of myofibroblast and the release of fibrotic mediators including transforming growth factor (TGF-ß1) that are related to the fibrosis. Moreover, colchicine has been used to traet viral myocarditis caused by CMV or EBV, interstitial pneumonia, and pericarditis resulting from influenza B infection. Additionally, colchicine is considered safe and affordable with wide availability. OBJECTIVE: The aim of the current study was to assess the evidence of colchicine effectiveness in COVID-19 treatment. METHODS: A comprehensive review of the literature was done till May 2022 and yielded 814 articles after ranking the articles according to authors and year of publication. Only 8 clinical trials and cohort studies fulfilling the inclusion criteria were included for further steps of data collection, analysis, and reporting. RESULTS: This meta-analysis involved 16,488 patients; 8146 patients in the treatment group and 8342 patients in the control group. The results showed that colchicine resulted in a significant reduction in the mortality rate among patients received colchicine in comparison with placebo or standard care (RR 0.35, 95%CI: 0.15-0.79). Colchicine resulted in a significant decrease in the need for O2 therapy in patients with COVID-19 (RR 0.07, 95%CI 0.02-0.27, P = 0.000024). However, colchicine had no significant effect on the following outcomes among COVID-19 patients: the need for hospitalization, ICU admission, artificial ventilation, and hospital discharge rate. Among the PCR confirmed COVID-19 patients, colchicine decreased the hospitalization rate (RR 0.75, 95%CI 0.57-0.99, P = 0.042). However, colchicine had no effect on mortality and the need for mechanical ventilation among this subgroup. CONCLUSION: Colchicine caused a significant clinical improvement among COVID-19 patients as compared with the standard care or placebo, in terms of the need for O2, and mortality. This beneficial effect could play a role in the management of COVID-19 especially severe cases to decrease need for oxygen and to decrease mortality among these patients.


COVID-19 , Virus Diseases , Humans , SARS-CoV-2 , Colchicine/therapeutic use , COVID-19 Drug Treatment
20.
Eur J Med Res ; 29(1): 245, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38649928

BACKGROUND: To determine the effect of colchicine on cancer risk in patients with the immune-mediated inflammatory diseases (IMIDs)-related to colchicine use. METHODS: This is a time-dependent propensity-matched general population study based on the National Health Insurance Research Database (NHIRD) of Taiwan. We identified the IMIDs patients (n = 111,644) newly diagnosed between 2000 and 2012 based on the International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM)-274,712, 135, 136.1, 279.49, 518.3, 287.0, 696.0, 696.1, 696.8, 420, 429.4, 710.0, 710.1, 710.3, 710.4, 714.0, 720, 55.0, 55.1, 55.9, 556. INCLUSION CRITERIA: aged ≧ 20 years, if a patient had at least these disease diagnosis requirements within 1 year of follow-up, and, these patients had at least two outpatient visits or an inpatient visit. After propensity-matched according to age, sex, comorbidities, medications and index date, the IMIDs patients enter into colchicine users (N = 16,026) and colchicine nonusers (N = 16,026). Furthermore, time-dependent Cox models were used to analyze cancer risk in propensity-matched colchicine users compared with the nonusers. The cumulative cancer incidence was analyzed using Cox proportional regression analysis. We calculated adjusted hazard ratios (aHRs) and their 95% confidence intervals (95% CIs) for cancer after adjusting for sex, age, comorbidities, and use of medicine including acetylcysteine, medication for smoking cessation such as nicotine replacement medicines (the nicotine patch) and pill medicines (varenicline), anti-inflammatory drugs and immunosuppressant drugs. RESULTS: Comparing the colchicine nonusers, all cancer risk were mildly attenuated, the (aHR (95% CI)) of all cancer is (0.84 (0.55, 0.99)). Meanwhile, the colchicine users were associated with the lower incidence of the colorectal cancer, the (aHRs (95% CI)) is (0.22 (0.19, 0.89)). Those aged < 65 years and male/female having the colchicine users were associated with lower risk the colorectal cancer also. Moreover, the colchicine > 20 days use with the lower aHR for colorectal cancer. CONCLUSION: Colchicine was associated with the lower aHR of the all cancer and colorectal cancer formation in patients with the IMIDs.


Colchicine , Databases, Factual , National Health Programs , Neoplasms , Humans , Colchicine/therapeutic use , Female , Male , Taiwan/epidemiology , Middle Aged , Neoplasms/epidemiology , Aged , National Health Programs/statistics & numerical data , Adult , Risk Factors , Inflammation/drug therapy , Incidence
...