1
|
Lin Y, Xu T, Jiang Q, Chen J, Zhang H, Reinach PS, Yan D, Qu J, Chen S. Fluorometholone inhibits corneal epithelial proliferation, migration via targeting Rho GTPases: RhoA, Rac1, and Cdc42. Exp Eye Res 2025:110397. [PMID: 40268157 DOI: 10.1016/j.exer.2025.110397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
Abnormal corneal epithelial hyperplasia is a common complication following refractive surgery. 0.1% fluorometholone (FML) eye drops are commonly used for treatment. However, their efficacy varies among patients, potentially attributed to differences in the patient's microenvironment. The underlying reason remains incompletely understood. This study aimed to elucidate the molecular mechanisms of FML's action on corneal epithelial cells (CECs). The effects of FML on the cell viability, proliferation, cell cycle, and migration of human corneal epithelial cells (HCECs) were evaluated using MTS assay, EdU staining, flow cytometry, and scratch assay, respectively. Mouse corneal sections were immunofluorescently stained to assess cell proliferation. A corneal wound model, monitored by slit-lamp photography, was utilized to evaluate the impact of FML on wound healing. Gene expression alterations were detected via RNA sequencing. RT-qPCR and Western blot were employed to validate gene and protein expression in HCECs and mouse corneal epithelia. Proteomic analysis was conducted on tear samples from patients. FML treatment significantly inhibited CEC proliferation, migration, and wound healing. At the molecular level, FML treatment led to a remarkable downregulation of RhoA, Rac1, and Cdc42. Correspondingly, reductions in the downstream Erk and NF-κB signaling pathways were observed in both HCECs and mouse corneal epithelia. Moreover, these pathways were similarly downregulated in tear samples from clinical patients. In conclusion, FML inhibits CEC proliferation and migration by modulating the Rho GTPase signaling network, especially through RhoA/Rac1/Cdc42, thereby suppressing the Erk/NF-κB pathway.
Collapse
Affiliation(s)
- Yong Lin
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Tianyi Xu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiuruo Jiang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jialu Chen
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hua Zhang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peter Sol Reinach
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Yan
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jia Qu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Shihao Chen
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
2
|
Carmo A, Rocha M, Pereirinha P, Tomé R, Costa E. Antifungals: From Pharmacokinetics to Clinical Practice. Antibiotics (Basel) 2023; 12:884. [PMID: 37237787 PMCID: PMC10215229 DOI: 10.3390/antibiotics12050884] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
The use of antifungal drugs started in the 1950s with polyenes nystatin, natamycin and amphotericin B-deoxycholate (AmB). Until the present day, AmB has been considered to be a hallmark in the treatment of invasive systemic fungal infections. Nevertheless, the success and the use of AmB were associated with severe adverse effects which stimulated the development of new antifungal drugs such as azoles, pyrimidine antimetabolite, mitotic inhibitors, allylamines and echinochandins. However, all of these drugs presented one or more limitations associated with adverse reactions, administration route and more recently the development of resistance. To worsen this scenario, there has been an increase in fungal infections, especially in invasive systemic fungal infections that are particularly difficult to diagnose and treat. In 2022, the World Health Organization (WHO) published the first fungal priority pathogens list, alerting people to the increased incidence of invasive systemic fungal infections and to the associated risk of mortality/morbidity. The report also emphasized the need to rationally use existing drugs and develop new drugs. In this review, we performed an overview of the history of antifungals and their classification, mechanism of action, pharmacokinetic/pharmacodynamic (PK/PD) characteristics and clinical applications. In parallel, we also addressed the contribution of fungi biology and genetics to the development of resistance to antifungal drugs. Considering that drug effectiveness also depends on the mammalian host, we provide an overview on the roles of therapeutic drug monitoring and pharmacogenomics as means to improve the outcome, prevent/reduce antifungal toxicity and prevent the emergence of antifungal resistance. Finally, we present the new antifungals and their main characteristics.
Collapse
Affiliation(s)
- Anália Carmo
- Advanced Unit for Pharmacokinetics and Personalized Therapeutics, Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
| | - Marilia Rocha
- Advanced Unit for Pharmacokinetics and Personalized Therapeutics, Pharmacy Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal (P.P.)
| | - Patricia Pereirinha
- Advanced Unit for Pharmacokinetics and Personalized Therapeutics, Pharmacy Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal (P.P.)
| | - Rui Tomé
- Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal;
| | - Eulália Costa
- Advanced Unit for Pharmacokinetics and Personalized Therapeutics, Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal
| |
Collapse
|
3
|
New reactions of 2,3-diaminonaphthaline with carbonyl electrophiles. MENDELEEV COMMUNICATIONS 2023. [DOI: 10.1016/j.mencom.2023.01.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
4
|
Guin D, Yadav S, Singh P, Singh P, Thakran S, Kukal S, Kanojia N, Paul PR, Pattnaik B, Sardana V, Grover S, Hasija Y, Saso L, Agrawal A, Kukreti R. Human genetic factors associated with pneumonia risk, a cue for COVID-19 susceptibility. INFECTION, GENETICS AND EVOLUTION 2022; 102:105299. [PMID: 35545162 PMCID: PMC9080029 DOI: 10.1016/j.meegid.2022.105299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/30/2022] [Accepted: 05/02/2022] [Indexed: 01/08/2023]
Abstract
Pneumonia, an acute respiratory tract infection, is one of the major causes of mortality worldwide. Depending on the site of acquisition, pneumonia can be community acquired pneumonia (CAP) or nosocomial pneumonia (NP). The risk of pneumonia, is partially driven by host genetics. CYP1A1 is a widely studied pulmonary CYP family gene primarily expressed in peripheral airway epithelium. The CYP1A1 genetic variants, included in this study, alter the gene activity and are known to contribute in lung inflammation, which may cause pneumonia pathogenesis. In this study, we performed a meta-analysis to establish the possible contribution of CYP1A1 gene, and its three variants (rs2606345, rs1048943 and rs4646903) towards the genetic etiology of pneumonia risk. Using PRISMA guidelines, we systematically reviewed and meta-analysed case-control studies, evaluating risk of pneumonia in patients carrying the risk alleles of CYP1A1 variants. Heterogeneity across the studies was evaluated using I2 statistics. Based on heterogeneity, a random-effect (using maximum likelihood) or fixed-effect (using inverse variance) model was applied to estimate the effect size. Pooled odds ratio (OR) was calculated to estimate the overall effect of the risk allele association with pneumonia susceptibility. Egger's regression test and funnel plot were used to assess publication bias. Subgroup analysis was performed based on pneumonia type (CAP and NP), population, as well as age group. A total of ten articles were identified as eligible studies, which included 3049 cases and 2249 healthy controls. The meta-analysis findings revealed CYP1A1 variants, rs2606345 [T vs G; OR = 1.12 (0.75–1.50); p = 0.02; I2 = 84.89%], and rs1048943 [G vs T; OR = 1.19 (0.76–1.61); p = 0.02; I2 = 0.00%] as risk markers whereas rs4646903 showed no statistical significance for susceptibility to pneumonia. On subgroup analysis, both the genetic variants showed significant association with CAP but not with NP. We additionally performed a spatial analysis to identify the key factors possibly explaining the variability across countries in the prevalence of the coronavirus disease 2019 (COVID-19), a viral pneumonia. We observed a significant association between the risk allele of rs2606345 and rs1048943, with a higher COVID-19 prevalence worldwide, providing us important links in understanding the variability in COVID-19 prevalence.
Collapse
|
5
|
Beck KR, Odermatt A. Antifungal therapy with azoles and the syndrome of acquired mineralocorticoid excess. Mol Cell Endocrinol 2021; 524:111168. [PMID: 33484741 DOI: 10.1016/j.mce.2021.111168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/31/2020] [Accepted: 01/06/2021] [Indexed: 10/22/2022]
Abstract
The syndromes of mineralocorticoid excess describe a heterogeneous group of clinical manifestations leading to endocrine hypertension, typically either through direct activation of mineralocorticoid receptors or indirectly by impaired pre-receptor enzymatic regulation or through disturbed renal sodium homeostasis. The phenotypes of these disorders can be caused by inherited gene variants and somatic mutations or may be acquired upon exposures to exogenous substances. Regarding the latter, the symptoms of an acquired mineralocorticoid excess have been reported during treatment with azole antifungal drugs. The current review describes the occurrence of mineralocorticoid excess particularly during the therapy with posaconazole and itraconazole, addresses the underlying mechanisms as well as inter- and intra-individual differences, and proposes a therapeutic drug monitoring strategy for these two azole antifungals. Moreover, other therapeutically used azole antifungals and ongoing efforts to avoid adverse mineralocorticoid effects of azole compounds are shortly discussed.
Collapse
Affiliation(s)
- Katharina R Beck
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology and Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
6
|
Costa-de-Oliveira S, Rodrigues AG. Candida albicans Antifungal Resistance and Tolerance in Bloodstream Infections: The Triad Yeast-Host-Antifungal. Microorganisms 2020; 8:E154. [PMID: 31979032 PMCID: PMC7074842 DOI: 10.3390/microorganisms8020154] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/10/2020] [Accepted: 01/16/2020] [Indexed: 01/08/2023] Open
Abstract
Candida albicans represents the most frequent isolated yeast from bloodstream infections. Despite the remarkable progress in diagnostic and therapeutic approaches, these infections continue to be a critical challenge in intensive care units worldwide. The economic cost of bloodstream fungal infections and its associated mortality, especially in debilitated patients, remains unacceptably high. Candida albicans is a highly adaptable microorganism, being able to develop resistance following prolonged exposure to antifungals. Formation of biofilms, which diminish the accessibility of the antifungal, selection of spontaneous mutations that increase expression or decreased susceptibility of the target, altered chromosome abnormalities, overexpression of multidrug efflux pumps and the ability to escape host immune defenses are some of the factors that can contribute to antifungal tolerance and resistance. The knowledge of the antifungal resistance mechanisms can allow the design of alternative therapeutically options in order to modulate or revert the resistance. We have focused this review on the main factors that are involved in antifungal resistance and tolerance in patients with C. albicans bloodstream infections.
Collapse
Affiliation(s)
- Sofia Costa-de-Oliveira
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, Al. Hernâni Monteiro, 4200-319 Porto, Portugal;
- Center for Research in Health Technologies and Information Systems (CINTESIS), R. Dr. Plácido da Costa, 4200-450 Porto, Portugal
| | - Acácio G. Rodrigues
- Division of Microbiology, Department of Pathology, Faculty of Medicine, University of Porto, Al. Hernâni Monteiro, 4200-319 Porto, Portugal;
- Center for Research in Health Technologies and Information Systems (CINTESIS), R. Dr. Plácido da Costa, 4200-450 Porto, Portugal
- Burn Unit, São João Hospital Center, Al. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
7
|
John J, Loo A, Mazur S, Walsh TJ. Therapeutic drug monitoring of systemic antifungal agents: a pragmatic approach for adult and pediatric patients. Expert Opin Drug Metab Toxicol 2019; 15:881-895. [PMID: 31550939 DOI: 10.1080/17425255.2019.1671971] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Introduction: Therapeutic drug monitoring (TDM) has been shown to optimize the management of invasive fungal infections (IFIs), particularly for select antifungal agents with a well-defined exposure-response relationship and an unpredictable pharmacokinetic profile or a narrow therapeutic index. Select triazoles (itraconazole, voriconazole, and posaconazole) and flucytosine fulfill these criteria, while the echinocandins, fluconazole, isavuconazole, and amphotericin B generally do not do so. Given the morbidity and mortality associated with IFIs and the challenges surrounding the use of currently available antifungal agents, TDM plays an important role in therapy.Areas covered: This review seeks to describe the rationale for TDM of antifungal agents, summarize their pharmacokinetic and pharmacodynamic properties, identify treatment goals for efficacy and safety, and provide recommendations for optimal dosing and therapeutic monitoring strategies.Expert opinion: Several new antifungal agents are currently in development, including compounds from existing antifungal classes with enhanced pharmacokinetic or safety profiles as well as agents with novel targets for the treatment of IFIs. Given the predictable pharmacokinetics of these newly developed agents, use of routine TDM is not anticipated. However, expanded knowledge of exposure-response relationships of these compounds may yield a role for TDM to improve outcomes for adult and pediatric patients.
Collapse
Affiliation(s)
- Jamie John
- Department of Pharmacy, New York-Presbyterian Hospital, New York, NY, USA
| | - Angela Loo
- Department of Pharmacy, New York-Presbyterian Hospital, New York, NY, USA
| | - Shawn Mazur
- Department of Pharmacy, New York-Presbyterian Hospital, New York, NY, USA
| | - Thomas J Walsh
- Transplantation-Oncology Infectious Diseases Program, New York-Presbyterian Hospital, Weill Cornell Medical Center, New York, NY, USA
| |
Collapse
|
8
|
Zahari Z, Lee CS, Ibrahim MA, Musa N, Mohd Yasin MA, Lee YY, Tan SC, Mohamad N, Ismail R. Relationship Between ABCB1
Polymorphisms and Cold Pain Sensitivity Among Healthy Opioid-naive Malay Males. Pain Pract 2017; 17:930-940. [DOI: 10.1111/papr.12546] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/21/2016] [Accepted: 10/26/2016] [Indexed: 02/03/2023]
Affiliation(s)
- Zalina Zahari
- Department of Pharmacy; Hospital Universiti Sains Malaysia; Kelantan Malaysia
- Pharmacogenetics and Novel Therapeutics Cluster; Institute for Research in Molecular Medicine; Universiti Sains Malaysia; Kelantan Malaysia
| | - Chee Siong Lee
- Department of Emergency Medicine; School of Medical Sciences; Universiti Sains Malaysia; Kelantan Malaysia
| | - Muslih Abdulkarim Ibrahim
- Pharmacogenetics and Novel Therapeutics Cluster; Institute for Research in Molecular Medicine; Universiti Sains Malaysia; Kelantan Malaysia
- Department of Pharmacology and Toxicology; College of Pharmacy; Hawler Medical University; Hawler Iraq
| | - Nurfadhlina Musa
- Pharmacogenetics and Novel Therapeutics Cluster; Institute for Research in Molecular Medicine; Universiti Sains Malaysia; Kelantan Malaysia
| | - Mohd Azhar Mohd Yasin
- Pharmacogenetics and Novel Therapeutics Cluster; Institute for Research in Molecular Medicine; Universiti Sains Malaysia; Kelantan Malaysia
- Department of Psychiatry; School of Medical Sciences; Universiti Sains Malaysia; Kelantan Malaysia
| | - Yeong Yeh Lee
- School of Medical Sciences; Universiti Sains Malaysia; Kelantan Malaysia
| | - Soo Choon Tan
- Pharmacogenetics and Novel Therapeutics Cluster; Institute for Research in Molecular Medicine; Universiti Sains Malaysia; Kelantan Malaysia
| | - Nasir Mohamad
- Pharmacogenetics and Novel Therapeutics Cluster; Institute for Research in Molecular Medicine; Universiti Sains Malaysia; Kelantan Malaysia
- Faculty of Medicine & Health Sciences; Universiti Sultan Zainal Abidin; Terengganu Malaysia
| | - Rusli Ismail
- Pharmacogenetics and Novel Therapeutics Cluster; Institute for Research in Molecular Medicine; Universiti Sains Malaysia; Kelantan Malaysia
- Centre of Excellence for Research in AIDS; University of Malaya; Kuala Lumpur Malaysia
| |
Collapse
|
9
|
Patterson TF, Thompson GR, Denning DW, Fishman JA, Hadley S, Herbrecht R, Kontoyiannis DP, Marr KA, Morrison VA, Nguyen MH, Segal BH, Steinbach WJ, Stevens DA, Walsh TJ, Wingard JR, Young JAH, Bennett JE. Practice Guidelines for the Diagnosis and Management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin Infect Dis 2016; 63:e1-e60. [PMID: 27365388 DOI: 10.1093/cid/ciw326] [Citation(s) in RCA: 1789] [Impact Index Per Article: 198.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 05/11/2016] [Indexed: 12/12/2022] Open
Abstract
It is important to realize that guidelines cannot always account for individual variation among patients. They are not intended to supplant physician judgment with respect to particular patients or special clinical situations. IDSA considers adherence to these guidelines to be voluntary, with the ultimate determination regarding their application to be made by the physician in the light of each patient's individual circumstances.
Collapse
Affiliation(s)
- Thomas F Patterson
- University of Texas Health Science Center at San Antonio and South Texas Veterans Health Care System
| | | | - David W Denning
- National Aspergillosis Centre, University Hospital of South Manchester, University of Manchester, United Kingdom
| | - Jay A Fishman
- Massachusetts General Hospital and Harvard Medical School
| | | | | | | | - Kieren A Marr
- Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Vicki A Morrison
- Hennepin County Medical Center and University of Minnesota, Minneapolis
| | | | - Brahm H Segal
- University at Buffalo Jacobs School of Medicine and Biomedical Sciences, and Roswell Park Cancer Institute, New York
| | | | | | - Thomas J Walsh
- New York-Presbyterian Hospital/Weill Cornell Medical Center, New York
| | | | | | - John E Bennett
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
10
|
Therapeutic drug monitoring for triazoles: A needs assessment review and recommendations from a Canadian perspective. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2015; 25:327-43. [PMID: 25587296 PMCID: PMC4277162 DOI: 10.1155/2014/340586] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Invasive fungal infections cause significant morbidity and mortality in patients with concomitant underlying immunosuppressive diseases. The recent addition of new triazoles to the antifungal armamentarium has allowed for extended-spectrum activity and flexibility of administration. Over the years, clinical use has raised concerns about the degree of drug exposure following standard approved drug dosing, questioning the need for therapeutic drug monitoring (TDM). Accordingly, the present guidelines focus on TDM of triazole antifungal agents. A review of the rationale for triazole TDM, the targeted patient populations and available laboratory methods, as well as practical recommendations based on current evidence from an extended literature review are provided in the present document.
Collapse
|
11
|
Castelli MV, Butassi E, Monteiro MC, Svetaz LA, Vicente F, Zacchino SA. Novel antifungal agents: a patent review (2011 – present). Expert Opin Ther Pat 2014; 24:323-38. [DOI: 10.1517/13543776.2014.876993] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
12
|
Salnikova LE, Smelaya TV, Golubev AM, Rubanovich AV, Moroz VV. CYP1A1, GCLC, AGT, AGTR1 gene-gene interactions in community-acquired pneumonia pulmonary complications. Mol Biol Rep 2013; 40:6163-76. [PMID: 24068433 DOI: 10.1007/s11033-013-2727-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 09/14/2013] [Indexed: 11/26/2022]
Abstract
This study was conducted to establish the possible contribution of functional gene polymorphisms in detoxification/oxidative stress and vascular remodeling pathways to community-acquired pneumonia (CAP) susceptibility in the case-control study (350 CAP patients, 432 control subjects) and to predisposition to the development of CAP complications in the prospective study. All subjects were genotyped for 16 polymorphic variants in the 14 genes of xenobiotics detoxification CYP1A1, AhR, GSTM1, GSTT1, ABCB1, redox-status SOD2, CAT, GCLC, and vascular homeostasis ACE, AGT, AGTR1, NOS3, MTHFR, VEGFα. Risk of pulmonary complications (PC) in the single locus analysis was associated with CYP1A1, GCLC and AGTR1 genes. Extra PC (toxic shock syndrome and myocarditis) were not associated with these genes. We evaluated gene-gene interactions using multi-factor dimensionality reduction, and cumulative gene risk score approaches. The final model which included >5 risk alleles in the CYP1A1 (rs2606345, rs4646903, rs1048943), GCLC, AGT, and AGTR1 genes was associated with pleuritis, empyema, acute respiratory distress syndrome, all PC and acute respiratory failure (ARF). We considered CYP1A1, GCLC, AGT, AGTR1 gene set using Set Distiller mode implemented in GeneDecks for discovering gene-set relations via the degree of sharing descriptors within a given gene set. N-acetylcysteine and oxygen were defined by Set Distiller as the best descriptors for the gene set associated in the present study with PC and ARF. Results of the study are in line with literature data and suggest that genetically determined oxidative stress exacerbation may contribute to the progression of lung inflammation.
Collapse
Affiliation(s)
- Lyubov E Salnikova
- N.I. Vavilov Institute of General Genetics, Russian Academy of Sciences, 3 Gubkin Street, Moscow, 117971, Russia,
| | | | | | | | | |
Collapse
|
13
|
The potential effect of gender in CYP1A1 and GSTM1 genotype-specific associations with pediatric brain tumor. Tumour Biol 2013; 34:2709-19. [DOI: 10.1007/s13277-013-0823-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 04/24/2013] [Indexed: 12/21/2022] Open
|
14
|
Zhivkova Z, Doytchinova I. Quantitative structure—plasma protein binding relationships of acidic drugs. J Pharm Sci 2012; 101:4627-41. [DOI: 10.1002/jps.23303] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 07/24/2012] [Accepted: 08/02/2012] [Indexed: 11/08/2022]
|
15
|
Salnikova LE, Smelaya TV, Moroz VV, Golubev AM, Rubanovich AV. Host genetic risk factors for community-acquired pneumonia. Gene 2012; 518:449-56. [PMID: 23107763 DOI: 10.1016/j.gene.2012.10.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 10/04/2012] [Accepted: 10/21/2012] [Indexed: 11/27/2022]
Abstract
This study was conducted to establish the contribution of genetic host factors in the susceptibility to community acquired pneumonia (CAP) in the Russian population. Patients with CAP (n=334), volunteers without a previous history of CAP, constantly exposed to infectious agents, control A group (n=141) and a second control group B consisted of healthy persons (n=314) were included in the study. All subjects were genotyped for 13 polymorphic variants in the genes of xenobiotics detoxification CYP1A1 (rs2606345, rs4646903, and rs1048943), GSTM1 (Ins/del), GSTT1 (Ins/del), ABCB1 rs1045642); immune and inflammation response IL-6 (rs1800795), TNF-a (rs1800629), MBL2 (rs7096206), CCR5 (rs333), NOS3 (rs1799983), angiotensin-converting enzyme ACE (rs4340), and occlusive vascular disease/hyperhomocysteinemia MTHFR (rs1801133). Seven polymorphic variants in genes CYP1A1, GSTM1, ABCB1, NOS3, IL6, CCR5 and ACE were associated with CAP. For two genes CYP1A1 and GSTM1 associations remained significant after correction for multiple comparisons. Multiple analysis by the number of all risk genotypes showed a highly significant association with CAP (P=2.4×10(-7), OR=3.03, 95% CI 1.98-4.64) with the threshold for three risk genotypes. Using the ROC-analysis, the AUC value for multi-locus model was estimated as 68.38.
Collapse
Affiliation(s)
- Lyubov E Salnikova
- N.I. Vavilov Institute of General Genetics, Russian Academy of Sciences, 3 Gubkin Street, Moscow 117971, Russia.
| | | | | | | | | |
Collapse
|
16
|
|
17
|
Sal’nikova LE, Chumachenko AG, Akayeva EA, Kuznetsova GI, Vesnina IN, Lapteva NS, Abilev SK, Rubanovich AV. Somatic mutagenesis in human lymphocytes depending on genotypes for detoxification and oxidative response loci. RUSS J GENET+ 2010. [DOI: 10.1134/s1022795410120112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
18
|
Li Y, Theuretzbacher U, Clancy CJ, Nguyen MH, Derendorf H. Pharmacokinetic/Pharmacodynamic Profile of Posaconazole. Clin Pharmacokinet 2010; 49:379-96. [DOI: 10.2165/11319340-000000000-00000] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
19
|
|
20
|
|
21
|
Hallucinations during voriconazole therapy: who is at higher risk and could benefit from therapeutic drug monitoring? Ther Drug Monit 2009; 31:135-6. [PMID: 19077926 DOI: 10.1097/ftd.0b013e3181947901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Meletiadis J, Chanock S, Walsh TJ. Defining targets for investigating the pharmacogenomics of adverse drug reactions to antifungal agents. Pharmacogenomics 2008; 9:561-84. [DOI: 10.2217/14622416.9.5.561] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Adverse drug reactions (ADRs) associated with antifungal therapy are major problems in patients with invasive fungal infections. Whether by clinical history or patterns of genetic variation, the identification of patients at risk for ADRs should result in improved outcomes while minimizing deleterious side effects. A major contributing factor to ADRs with antifungal agents relates to drug distribution, metabolism and excretion. Genetic variation in key genes can alter the structure and expression of genes and gene products (e.g., proteins). Thus far, the effort has focused on identifying polymorphisms with either empirical or predicted in silico functional consequences; the best candidate genes encode phase I and II drug-metabolizing enzymes (e.g., CYP2C19 and N-acetyltransferase), plasma proteins (albumin and lipoproteins) and drug transporters (P-glycoprotein and multidrug resistance proteins), which can affect the disposition of antifungal agents, eventually leading to dose-dependent (type A) toxicity. Less is known regarding the key genes that interact with antifungal agents, resulting in idiosyncratic (type B) ADRs. The possible role of certain gene products and genetic polymorphisms in the toxicities of antifungal agents are discussed in this review. The preliminary data address the following: low-density lipoproteins and cholesteryl ester transfer protein in amphotericin B renal toxicity; toll-like receptor 1 and 2 in amphotericin B infusion-related ADRs; phosphodiesterase 6 in voriconazole visual adverse events; flavin-containing monooxygenase, glutathione transferases and multidrug resistance proteins 1 and 2 in ketoconazole and terbinafine hepatotoxicity; CYP enzymes and P-glycoprotein in drug interactions between azoles and coadministered medications; multidrug resistance proteins 8 and 9 on 5-flucytosine bone marrow toxicity; and mast cell activation in caspofungin histamine release. This will focus on high-priority candidate genes, which could provide a starting point for molecular studies to elucidate the potential mechanisms for understanding toxicity associated with antifungal drugs as well as identifying candidate genes for large population prospective genetic association studies.
Collapse
Affiliation(s)
- Joseph Meletiadis
- National Cancer Institute, National Institutes of Health, Pediatric Oncology Branch, Bethesda, MD 20814, USA
- Attikon University General Hospital, Laboratoty for Clinical Microbiology, 1 Rimini Street, Athens 124 62, Greece
| | - Stephen Chanock
- National Cancer Institute, National Institutes of Health, Pediatric Oncology Branch, Bethesda, MD 20814, USA
| | - Thomas J Walsh
- National Cancer Institute, National Institutes of Health, Pediatric Oncology Branch, Bethesda, MD 20814, USA
| |
Collapse
|
23
|
Pharmacogenetics and pharmacogenomics for the infectious diseases practitioner. Pediatr Infect Dis J 2008; 27:263-4. [PMID: 18418196 DOI: 10.1097/inf.0b013e318168d5d0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Nagappan V, Deresinski S. Posaconazole: A Broad-Spectrum Triazole Antifungal Agent. Clin Infect Dis 2007; 45:1610-7. [DOI: 10.1086/523576] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|