1
|
Soudais C, Schaus R, Bachelet C, Minet N, Mouasni S, Garcin C, Souza CL, David P, Cousu C, Asnagli H, Parker A, Palmquist-Gomes P, Sepulveda FE, Storck S, Meilhac SM, Fischer A, Martin E, Latour S. Inactivation of cytidine triphosphate synthase 1 prevents fatal auto-immunity in mice. Nat Commun 2024; 15:1982. [PMID: 38438357 PMCID: PMC10912214 DOI: 10.1038/s41467-024-45805-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 01/25/2024] [Indexed: 03/06/2024] Open
Abstract
De novo synthesis of the pyrimidine, cytidine triphosphate (CTP), is crucial for DNA/RNA metabolism and depends on the CTP synthetases, CTPS1 and -2. Partial CTPS1 deficiency in humans has previously been shown to lead to immunodeficiency, with impaired expansion of T and B cells. Here, we examine the effects of conditional and inducible inactivation of Ctps1 and/or Ctps2 on mouse embryonic development and immunity. We report that deletion of Ctps1, but not Ctps2, is embryonic-lethal. Tissue and cells with high proliferation and renewal rates, such as intestinal epithelium, erythroid and thymic lineages, activated B and T lymphocytes, and memory T cells strongly rely on CTPS1 for their maintenance and growth. However, both CTPS1 and CTPS2 are required for T cell proliferation following TCR stimulation. Deletion of Ctps1 in T cells or treatment with a CTPS1 inhibitor rescued Foxp3-deficient mice from fatal systemic autoimmunity and reduced the severity of experimental autoimmune encephalomyelitis. These findings support that CTPS1 may represent a target for immune suppression.
Collapse
Affiliation(s)
- Claire Soudais
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France.
- Université de Paris Cité, Paris, France.
| | - Romane Schaus
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France
| | - Camille Bachelet
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France
- Université de Paris Cité, Paris, France
| | - Norbert Minet
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France
- Université de Paris Cité, Paris, France
| | - Sara Mouasni
- Laboratory of Molecular Basis of Altered Immune Homeostasis Inserm UMR 1163, Institut Imagine, Paris, France
| | - Cécile Garcin
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France
- Université de Paris Cité, Paris, France
| | - Caique Lopes Souza
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France
- Université de Paris Cité, Paris, France
| | - Pierre David
- Transgenesis Platform, Laboratoire d'Expérimentation Animale et Transgenèse (LEAT), Institut Imagine-Structure Fédérative de Recherche Necker INSERM US24/CNRS, UMS3633, Paris, France
| | - Clara Cousu
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Hélène Asnagli
- Step-Pharma, Technoparc du Pays-de-Gex, Saint-Genis-Pouilly, France
| | - Andrew Parker
- Step-Pharma, Technoparc du Pays-de-Gex, Saint-Genis-Pouilly, France
| | - Paul Palmquist-Gomes
- Université de Paris Cité, Paris, France
- Imagine - Institut Pasteur, Unit of Heart Morphogenesis, INSERM UMR1163, F-75015, Paris, France
| | - Fernando E Sepulveda
- Laboratory of Molecular Basis of Altered Immune Homeostasis Inserm UMR 1163, Institut Imagine, Paris, France
| | - Sébastien Storck
- Université Paris Cité, CNRS UMR 8253, INSERM U1151, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Sigolène M Meilhac
- Université de Paris Cité, Paris, France
- Imagine - Institut Pasteur, Unit of Heart Morphogenesis, INSERM UMR1163, F-75015, Paris, France
| | - Alain Fischer
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France
- Collège de France, Paris, France
| | - Emmanuel Martin
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France
| | - Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, Inserm UMR 1163, Institut Imagine, Paris, France.
- Université de Paris Cité, Paris, France.
| |
Collapse
|
2
|
Apaolaza I, San José-Enériz E, Valcarcel LV, Agirre X, Prosper F, Planes FJ. A network-based approach to integrate nutrient microenvironment in the prediction of synthetic lethality in cancer metabolism. PLoS Comput Biol 2022; 18:e1009395. [PMID: 35286311 PMCID: PMC8947600 DOI: 10.1371/journal.pcbi.1009395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 03/24/2022] [Accepted: 02/18/2022] [Indexed: 11/18/2022] Open
Abstract
Synthetic Lethality (SL) is currently defined as a type of genetic interaction in which the loss of function of either of two genes individually has limited effect in cell viability but inactivation of both genes simultaneously leads to cell death. Given the profound genomic aberrations acquired by tumor cells, which can be systematically identified with -omics data, SL is a promising concept in cancer research. In particular, SL has received much attention in the area of cancer metabolism, due to the fact that relevant functional alterations concentrate on key metabolic pathways that promote cellular proliferation. With the extensive prior knowledge about human metabolic networks, a number of computational methods have been developed to predict SL in cancer metabolism, including the genetic Minimal Cut Sets (gMCSs) approach. A major challenge in the application of SL approaches to cancer metabolism is to systematically integrate tumor microenvironment, given that genetic interactions and nutritional availability are interconnected to support proliferation. Here, we propose a more general definition of SL for cancer metabolism that combines genetic and environmental interactions, namely loss of gene functions and absence of nutrients in the environment. We extend our gMCSs approach to determine this new family of metabolic synthetic lethal interactions. A computational and experimental proof-of-concept is presented for predicting the lethality of dihydrofolate reductase (DHFR) inhibition in different environments. Finally, our approach is applied to identify extracellular nutrient dependences of tumor cells, elucidating cholesterol and myo-inositol depletion as potential vulnerabilities in different malignancies. Metabolic reprogramming is one of the hallmarks of tumor cells. Synthetic lethality (SL) is a promising approach to exploit these metabolic alterations and elucidate cancer-specific genetic dependences. However, current SL approaches do not systematically consider tumor microenvironment, which is particularly important in cancer metabolism in order to generalize identified genetic dependences. In this article, we directly address this issue and propose a more general approach to SL that integrates both genetic and environmental context of tumor cells. Our definition can help to contextualize genetic dependencies in different environmental scenarios, but it could also reveal nutrient dependencies according to the genetic context. We also provide a computational pipeline to identify this new family of synthetic lethals in genome-scale metabolic networks. A computational and experimental proof-of-concept is presented for predicting the lethality of dihydrofolate reductase (DHFR) inhibition in different environments. Finally, our approach is applied to identify extracellular nutrient dependences of cancer cell lines, elucidating cholesterol and myo-Inositol depletion as potential vulnerabilities in different malignancies.
Collapse
Affiliation(s)
- Iñigo Apaolaza
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, Centro de Ingeniería Biomédica, Pamplona, Spain
- Universidad de Navarra, DATAI Instituto de Ciencia de los Datos e Inteligencia Artificial, Pamplona, Spain
| | - Edurne San José-Enériz
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Luis V. Valcarcel
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Xabier Agirre
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Felipe Prosper
- Universidad de Navarra, CIMA Centro de Investigación de Medicina Aplicada, Pamplona, Spain
- IdiSNA Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
- Clínica Universidad de Navarra, Pamplona, Spain
| | - Francisco J. Planes
- Universidad de Navarra, Tecnun Escuela de Ingeniería, San Sebastián, Spain
- Universidad de Navarra, Centro de Ingeniería Biomédica, Pamplona, Spain
- Universidad de Navarra, DATAI Instituto de Ciencia de los Datos e Inteligencia Artificial, Pamplona, Spain
- * E-mail:
| |
Collapse
|
3
|
Musdalita E, Hidayat R, Sumariyono S, Kusumo Wibowo SA, Ariane A, Shatri H, Rengganis I, Antono D. Correlation between serum methotrexate-polyglutamate 3 (MTX-PG3) level and disease activity in rheumatoid arthritis patients: A prospective cohort study. F1000Res 2022; 11:187. [PMID: 35284067 PMCID: PMC8891717 DOI: 10.12688/f1000research.108714.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Rheumatoid arthritis (RA) is one of the most common autoimmune diseases, characterized by systemic inflammation, joint destruction and disability. Methotrexate (MTX) is used as the primary treatment for RA patients. However, the response to MTX therapy is highly varied and difficult to predict. This study sought to determine the role of MTX by measuring the MTX polyglutamate 3 (MTX-PG3) levels and the disease activity score 28 based on C-reactive protein (DAS28-CRP) of RA patients. Method: A prospective cohort study was conducted at the Rheumatology Polyclinic of Dr. Cipto Mangunkusumo General Hospital. Thirty-four patients with RA were included and followed up to 12 weeks. The RA patients were treated with MTX 10 mg per week and an increased dose of 5 mg per week every month. DAS28-CRP and MTX-PG3 level were assessed at week 8 and 12. Multivariate logistic regression analysis was used to determine the correlation between MTX-PG3 and DAS28-CRP. Result: A total of 34 RA patients were followed and the MTX was well tolerated in which no increase of serum glutamic oxaloacetic transaminase (SGOT), serum glutamic pyruvic transaminase (SGPT) and glomerular filtration rate (GFR) were observed. The mean scores of DAS28-CRP decreased following the MTX-treatment: 3.93, 3.22 and 2.82 at week 0, 8 and 12, respectively. In contrast, the median concentration of MTX-PG3 increased from week 8 to week 12 followed by increasing the dose of MTX. Our analysis suggested there was a moderate positive correlation between MTX-PG3 levels and DAS28-CRP score at week 8 and week 12 post-MTX treatment. Conclusion: The level of MTX-PG3 is correlated with DAS28-CRP score suggesting that MTX-PG3 could be used as an indicator to assess the disease activity in RA patients. Nevertheless, a prospective study with a higher number of patients is needed to confirm this finding.
Collapse
Affiliation(s)
- Eva Musdalita
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,, Universitas Indonesia/ Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
- Department of Internal Medicine, Faculty of Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Rudy Hidayat
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,, Universitas Indonesia/ Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Sumariyono Sumariyono
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,, Universitas Indonesia/ Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Suryo Anggoro Kusumo Wibowo
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,, Universitas Indonesia/ Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Anna Ariane
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,, Universitas Indonesia/ Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Hamzah Shatri
- Division of Psychosomatic and Palliative Care, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia/Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Iris Rengganis
- Division of Allergy and Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia/Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Dono Antono
- Division of Cardiovascular, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia/Dr Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| |
Collapse
|
4
|
Kaye AD, Jeha GM, Pham AD, Fuller MC, Lerner ZI, Sibley GT, Cornett EM, Urits I, Viswanath O, Kevil CG. Folic Acid Supplementation in Patients with Elevated Homocysteine Levels. Adv Ther 2020; 37:4149-4164. [PMID: 32845472 PMCID: PMC7497502 DOI: 10.1007/s12325-020-01474-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Indexed: 12/12/2022]
Abstract
Introduction Folic acid is the most important dietary determinant of homocysteine (Hcy). Hcy serves as a critical intermediate in methylation reactions. It is created from methionine and either converted back to methionine or transformed into cysteine. This process is aided through several enzymes and three vitamins, folic acid, B12, and B6. Daily supplementation with 0.5–5.0 mg of folic acid typically lowers plasma Hcy levels by approximately 25%. Hyperhomocysteinemia is a known risk factor for coronary artery disease. In this regard, elevated levels of Hcy have been found in a majority of patients with vascular disease. Methods A literature review of folic acid supplementation for various disease states including cardiovascular disease was conducted. This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors. Results In this review, we discuss the biochemistry of folic acid, Hcy biosynthesis, Hcy and hydrogen sulfide bioavailability, pathogenesis of hyperhomocysteinemia and its role as a risk factor for disease, and treatment studies with folic acid supplementation in disease states. Conclusion Folic acid supplementation should be recommended to any patient who has an elevated Hcy level, and this level should be measured and treated at an early age, since folic acid is easily obtained and may likely reduce vascular disease and other deleterious pathologic processes in high-risk populations.
Collapse
|
5
|
Funk RS, Singh RK, Becker ML. Metabolomic Profiling to Identify Molecular Biomarkers of Cellular Response to Methotrexate In Vitro. Clin Transl Sci 2020; 13:137-146. [PMID: 31651077 PMCID: PMC6951846 DOI: 10.1111/cts.12694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/06/2019] [Indexed: 01/21/2023] Open
Abstract
Variation in methotrexate (MTX) efficacy represents a significant barrier to early and effective disease control in the treatment of autoimmune arthritis. We hypothesize that the utilization of metabolomic techniques will allow for an improved understanding of the biochemical basis for the pharmacological activity of MTX, and can promote the identification and evaluation of novel molecular biomarkers of MTX response. In this work, erythroblastoid cells were exposed to MTX at the physiologic concentration of 1,000 nM and analyzed using three metabolomic platforms to give a broad spectrum of cellular metabolites. MTX pharmacological activity, defined as cellular growth inhibition, was associated with an altered cellular metabolomic profile based on the analysis of 724 identified metabolites. By discriminant analysis, MTX treatment was associated with increases in ketoisovaleric acid, fructose, galactose, and 2-deoxycytidine, and corresponding reductions in 2-deoxyuridine, phosphatidylinositol 32:0, orotic acid, and inosine monophosphate. Inclusion of data from analysis of folate metabolism in combination with chemometric and metabolic network analysis demonstrated that MTX treatment is associated with dysregulated folate metabolism and nucleotide biosynthesis, which is in line with its known mechanism of action. However, MTX treatment was also associated with alterations in a diversity of metabolites, including intermediates of amino acid, carbohydrate, and lipid metabolism. Collectively, these findings support a robust metabolic response following exposure to physiologic concentrations of MTX. They also identify various metabolic intermediates that are associated with the pharmacological activity of MTX, and are, therefore, potential molecular biomarker candidates in future preclinical and clinical studies of MTX efficacy in autoimmune arthritis.
Collapse
Affiliation(s)
- Ryan S. Funk
- Department of Pharmacy PracticeMedical CenterUniversity of KansasKansas CityKansasUSA
| | - Rakesh K. Singh
- Department of Pharmacy PracticeMedical CenterUniversity of KansasKansas CityKansasUSA
| | - Mara L. Becker
- Division of RheumatologyDepartment of PediatricsDuke Children's HospitalDurhamNorth CarolinaUSA
| |
Collapse
|
6
|
The Protective Effects of Syzygium aromaticum Essential Oil Extract against Methotrexate Induced Hepatic and Renal Toxicity in Rats. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2019. [DOI: 10.22207/jpam.13.1.57] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
7
|
Singh RK, van Haandel L, Heruth DP, Ye SQ, Leeder JS, Becker ML, Funk RS. Nicotinamide Phosphoribosyltransferase Deficiency Potentiates the Antiproliferative Activity of Methotrexate through Enhanced Depletion of Intracellular ATP. J Pharmacol Exp Ther 2018; 365:96-106. [PMID: 29420256 PMCID: PMC5830637 DOI: 10.1124/jpet.117.246199] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/26/2018] [Indexed: 12/11/2022] Open
Abstract
Lower plasma nicotinamide phosphoribosyltransferase (NAMPT) levels are associated with improved response to methotrexate (MTX) in patients with juvenile idiopathic arthritis. Cell-based studies confirmed that reduced cellular NAMPT activity potentiates the pharmacologic activity of MTX; however, the mechanism of this interaction has yet to be defined. Therefore, in this study, we investigate the mechanism of enhanced pharmacologic activity of MTX in NAMPT-deficient A549 cells. Small interfering RNA-based silencing of NAMPT expression resulted in a greater than 3-fold increase in sensitivity to MTX (P < 0.005) that was completely reversed by supplementation with folinic acid. Despite a 68% reduction in cellular NAD levels in NAMPT-deficient cells, no change in expression or activity of dihydrofolate reductase was observed and uptake of MTX was not significantly altered. MTX did not potentiate the depletion of cellular NAD levels, but NAMPT-deficient cells had significant elevations in levels of intermediates of de novo purine biosynthesis and were 4-fold more sensitive to depletion of ATP by MTX (P < 0.005). Supplementation with hypoxanthine and thymidine completely reversed the antiproliferative activity of MTX in NAMPT-deficient cells and corresponded to repletion of the cellular ATP pool without any effect on NAD levels. Together, these findings demonstrate that increased MTX activity with decreased NAMPT expression is dependent on the antifolate activity of MTX and is driven by enhanced sensitivity to the ATP-depleting effects of MTX. For the first time, these findings provide mechanistic details to explain the increase in pharmacological activity of MTX under conditions of reduced NAMPT activity.
Collapse
Affiliation(s)
- Rakesh K Singh
- Departments of Pharmacy Practice (R.K.S., R.S.F.) and Pharmacology, Toxicology, and Therapeutics (J.S.L., R.S.F.), University of Kansas Medical Center, Kansas City, Kansas; Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (L.v.H., J.S.L., M.L.B.), Rheumatology (M.L.B.), and Experimental and Translational Genetics (D.P.H., S.Q.Y.), Children's Mercy Kansas City, Kansas City, Missouri; and Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri (S.Q.Y.)
| | - Leon van Haandel
- Departments of Pharmacy Practice (R.K.S., R.S.F.) and Pharmacology, Toxicology, and Therapeutics (J.S.L., R.S.F.), University of Kansas Medical Center, Kansas City, Kansas; Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (L.v.H., J.S.L., M.L.B.), Rheumatology (M.L.B.), and Experimental and Translational Genetics (D.P.H., S.Q.Y.), Children's Mercy Kansas City, Kansas City, Missouri; and Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri (S.Q.Y.)
| | - Daniel P Heruth
- Departments of Pharmacy Practice (R.K.S., R.S.F.) and Pharmacology, Toxicology, and Therapeutics (J.S.L., R.S.F.), University of Kansas Medical Center, Kansas City, Kansas; Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (L.v.H., J.S.L., M.L.B.), Rheumatology (M.L.B.), and Experimental and Translational Genetics (D.P.H., S.Q.Y.), Children's Mercy Kansas City, Kansas City, Missouri; and Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri (S.Q.Y.)
| | - Shui Q Ye
- Departments of Pharmacy Practice (R.K.S., R.S.F.) and Pharmacology, Toxicology, and Therapeutics (J.S.L., R.S.F.), University of Kansas Medical Center, Kansas City, Kansas; Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (L.v.H., J.S.L., M.L.B.), Rheumatology (M.L.B.), and Experimental and Translational Genetics (D.P.H., S.Q.Y.), Children's Mercy Kansas City, Kansas City, Missouri; and Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri (S.Q.Y.)
| | - J Steven Leeder
- Departments of Pharmacy Practice (R.K.S., R.S.F.) and Pharmacology, Toxicology, and Therapeutics (J.S.L., R.S.F.), University of Kansas Medical Center, Kansas City, Kansas; Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (L.v.H., J.S.L., M.L.B.), Rheumatology (M.L.B.), and Experimental and Translational Genetics (D.P.H., S.Q.Y.), Children's Mercy Kansas City, Kansas City, Missouri; and Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri (S.Q.Y.)
| | - Mara L Becker
- Departments of Pharmacy Practice (R.K.S., R.S.F.) and Pharmacology, Toxicology, and Therapeutics (J.S.L., R.S.F.), University of Kansas Medical Center, Kansas City, Kansas; Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (L.v.H., J.S.L., M.L.B.), Rheumatology (M.L.B.), and Experimental and Translational Genetics (D.P.H., S.Q.Y.), Children's Mercy Kansas City, Kansas City, Missouri; and Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri (S.Q.Y.)
| | - Ryan S Funk
- Departments of Pharmacy Practice (R.K.S., R.S.F.) and Pharmacology, Toxicology, and Therapeutics (J.S.L., R.S.F.), University of Kansas Medical Center, Kansas City, Kansas; Divisions of Clinical Pharmacology, Toxicology and Therapeutic Innovation (L.v.H., J.S.L., M.L.B.), Rheumatology (M.L.B.), and Experimental and Translational Genetics (D.P.H., S.Q.Y.), Children's Mercy Kansas City, Kansas City, Missouri; and Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri (S.Q.Y.)
| |
Collapse
|
8
|
Sramek M, Neradil J, Veselska R. Much more than you expected: The non-DHFR-mediated effects of methotrexate. Biochim Biophys Acta Gen Subj 2016; 1861:499-503. [PMID: 27993660 DOI: 10.1016/j.bbagen.2016.12.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/10/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND For decades, methotrexate (MTX; amethopterin) has been known as an antifolate inhibitor of dihydrofolate reductase (DHFR), and it is widely used for the treatment of various malignancies and autoimmune diseases. Although the inclusion of MTX in various therapeutic regimens is based on its ability to inhibit DHFR and consequently to suppress the synthesis of pyrimidine and purine precursors, recent studies have shown that MTX is also able to target other intracellular pathways that are independent of folate metabolism. SCOPE OF REVIEW The main aim of this review is to summarize the most important, up-to-date findings of studies regarding the non-DHFR-mediated mechanisms of MTX action. MAJOR CONCLUSIONS The effectiveness of MTX is undoubtedly caused by its capability to affect various intracellular pathways at many levels. Although the most important therapeutic mechanism of MTX is strongly based on the inhibition of DHFR, many other effects of this compound have been described and new studies bring new insights into the pharmacology of MTX every year. GENERAL SIGNIFICANCE Identification of these new targets for MTX is especially important for a better understanding of MTX action in new protocols of combination therapy.
Collapse
Affiliation(s)
- Martin Sramek
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, Brno 656 91, Czech Republic
| | - Jakub Neradil
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, Brno 656 91, Czech Republic
| | - Renata Veselska
- International Clinical Research Center, St. Anne's University Hospital Brno, Pekarska 53, Brno 656 91, Czech Republic.
| |
Collapse
|
9
|
Funk RS, Singh R, Pramann L, Gigliotti N, Islam S, Heruth DP, Ye SQ, Chan MA, Leeder JS, Becker ML. Nicotinamide Phosphoribosyltransferase Attenuates Methotrexate Response in Juvenile Idiopathic Arthritis and In Vitro. Clin Transl Sci 2016; 9:149-57. [PMID: 27166432 PMCID: PMC4902726 DOI: 10.1111/cts.12399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/07/2016] [Accepted: 04/13/2016] [Indexed: 12/22/2022] Open
Abstract
Variability in response to methotrexate (MTX) in the treatment of juvenile idiopathic arthritis (JIA) remains unpredictable and poorly understood. Based on previous studies implicating an interaction between nicotinamide phosphoribosyltransferase (NAMPT) expression and MTX therapy in inflammatory arthritis, we hypothesized that increased NAMPT expression would be associated with reduced therapeutic response to MTX in patients with JIA. A significant association was found between increased plasma concentrations of NAMPT and reduced therapeutic response in patients with JIA treated with MTX. Inhibition of NAMPT in cell culture by either siRNA‐based gene silencing or pharmacological inhibition with FK‐866 was found to result in a fourfold increase in the pharmacological activity of MTX. Collectively, these findings provide evidence that NAMPT inhibits the pharmacological activity of MTX and may represent a predictive biomarker of response, as well as a therapeutic target, in the treatment of JIA with MTX.
Collapse
Affiliation(s)
- R S Funk
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, Kansas, USA.,Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA.,Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - R Singh
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - L Pramann
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - N Gigliotti
- Division of Immunology Research, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - S Islam
- Division of Experimental and Translational Genetics, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - D P Heruth
- Division of Experimental and Translational Genetics, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - S Q Ye
- Division of Experimental and Translational Genetics, Children's Mercy Kansas City, Kansas City, Missouri, USA.,Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - M A Chan
- Division of Immunology Research, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - J S Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA.,Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - M L Becker
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA.,Division of Rheumatology, Children's Mercy Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
10
|
Funk RS, Becker ML. Disease modifying anti-rheumatic drugs in juvenile idiopathic arthritis: striving for individualized therapy. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1133234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
11
|
Funk RS, van Haandel L, Leeder JS, Becker ML. Folate depletion and increased glutamation in juvenile idiopathic arthritis patients treated with methotrexate. Arthritis Rheumatol 2015; 66:3476-85. [PMID: 25186097 DOI: 10.1002/art.38865] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 08/26/2014] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Folates exist as a fluctuating pool of polyglutamated metabolites that may serve as a clinical marker of methotrexate (MTX) activity. This study was undertaken to evaluate circulating folate content and folate polyglutamate distribution in juvenile idiopathic arthritis (JIA) patients and in a cell culture model based on MTX exposure and folate supply. METHODS Blood, plasma, and red blood cell (RBC) measurements of MTX and folates were obtained from previously published data sets and an additional analysis of JIA patients receiving MTX (n = 98) and those not receiving MTX (n = 78). Erythroblastoid cells maintained in culture were exposed to MTX and grown under varying levels of folic acid supplementation. Samples were analyzed for cellular folate and MTX content. RESULTS Circulating folate levels were lower in JIA patients receiving MTX, with reduced levels of blood, plasma, and RBC 5-methyl-tetrahydrofolate (5mTHF) (P < 0.0001). Average polyglutamate chain length (Gluavg ) of RBC 5mTHF was elevated in JIA patients receiving MTX (median ± interquartile range 5.63 ± 0.15 versus 5.54 ± 0.11 in those not receiving MTX; P < 0.001) and correlated with both RBC MTX accumulation (P = 0.02) and reduced plasma 5mTHF levels (P = 0.008). MTX exposure and folate deprivation in erythroblastoid cells resulted in a depletion of bioactive folate species that was associated with a shift to higher Gluavg values for several species, most notably tetrahydrofolate (THF) and 5,10-methylene-tetrahydrofolate (CH2 THF). Increased Gluavg resulted from the depletion of short-chain and the accumulation of long-chain glutamate species. CONCLUSION Our findings indicate that folate content and polyglutamate distribution are responsive markers of MTX activity and folate supply in vivo and in vitro, and may provide novel clinical markers of pharmacologic activity of MTX.
Collapse
Affiliation(s)
- Ryan S Funk
- Children's Mercy Kansas City, Kansas City, Missouri and the University of Kansas Medical Center, Kansas City, Kansas
| | | | | | | |
Collapse
|
12
|
Zebala JA, Mundell A, Messinger L, Griffin CE, Schuler AD, Kahn SJ. LD-aminopterin in the canine homologue of human atopic dermatitis: a randomized, controlled trial reveals dosing factors affecting optimal therapy. PLoS One 2014; 9:e108303. [PMID: 25255447 PMCID: PMC4177985 DOI: 10.1371/journal.pone.0108303] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 08/28/2014] [Indexed: 01/09/2023] Open
Abstract
Background Options are limited for patients with atopic dermatitis (AD) who do not respond to topical treatments. Antifolate therapy with systemic methotrexate improves the disease, but is associated with adverse effects. The investigational antifolate LD-aminopterin may offer improved safety. It is not known how antifolate dose and dosing frequency affect efficacy in AD, but a primary mechanism is thought to involve the antifolate-mediated accumulation of 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR). However, recent in vitro studies indicate that AICAR increases then decreases as a function of antifolate concentration. To address this issue and understand how dosing affects antifolate efficacy in AD, we examined the efficacy and safety of different oral doses and schedules of LD-aminopterin in the canine model of AD. Methods and Findings This was a multi-center, double-blind trial involving 75 subjects with canine AD randomized to receive up to 12 weeks of placebo, once-weekly (0.007, 0.014, 0.021 mg/kg) or twice-weekly (0.007 mg/kg) LD-aminopterin. The primary efficacy outcome was the Global Score (GS), a composite of validated measures of disease severity and itch. GS improved in all once-weekly cohorts, with 0.014 mg/kg being optimal and significant (43%, P<0.01). The majority of improvement was seen by 8 weeks. In contrast, GS in the twice-weekly cohort was similar to placebo and worse than all once-weekly cohorts. Adverse events were similar across all treated cohorts and placebo. Conclusions Once-weekly LD-aminopterin was safe and efficacious in canine AD. Twice-weekly dosing negated efficacy despite having the same daily and weekly dose as effective once-weekly regimens. Optimal dosing in this homologue of human AD correlated with the concentration-selective accumulation of AICAR in vitro, consistent with AICAR mediating LD-aminopterin efficacy in AD.
Collapse
Affiliation(s)
- John A. Zebala
- Syntrix Biosystems, Inc., Auburn, Washington, United States of America
- * E-mail:
| | - Alan Mundell
- Animal Dermatology Service, Edmonds, Washington, United States of America
| | - Linda Messinger
- Veterinary Referral Center of Colorado, Englewood, Colorado, United States of America
| | - Craig E. Griffin
- Animal Dermatology Clinic, San Diego, California, United States of America
| | - Aaron D. Schuler
- Syntrix Biosystems, Inc., Auburn, Washington, United States of America
| | - Stuart J. Kahn
- Syntrix Biosystems, Inc., Auburn, Washington, United States of America
| |
Collapse
|
13
|
The methotrexate polyglutamate assay supports the efficacy of methotrexate for severe inflammatory skin disease in children. J Am Acad Dermatol 2014; 70:252-6. [DOI: 10.1016/j.jaad.2013.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 09/30/2013] [Accepted: 10/01/2013] [Indexed: 11/24/2022]
|
14
|
Inoue K, Yuasa H. Molecular basis for pharmacokinetics and pharmacodynamics of methotrexate in rheumatoid arthritis therapy. Drug Metab Pharmacokinet 2013; 29:12-9. [PMID: 24284432 DOI: 10.2133/dmpk.dmpk-13-rv-119] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Methotrexate (MTX) is a derivative of folic acid (folate) and commonly used as an anchor drug for the treatment of rheumatoid arthritis (RA). The pharmacokinetics (PK) and pharmacodynamics (PD) of MTX entirely depends on the function of specific transporters that belong to the two major superfamilies, solute carrier transporters and ATP-binding cassette transporters. Several transporters have been identified as being able to mediate the transport of MTX, and suggested to be involved in the disposition in the body and in the regulation of intracellular metabolism in target cells, together with several enzymes involved in folate metabolism. Thus, drug-drug interactions through the transporters and their genetic polymorphisms may alter the PK and PD of MTX, resulting in an interpatient variability of efficacy. This review summarizes the PK and PD of MTX, particularly in relation to RA therapy and focuses on the roles of transporters involved in PK and PD with the aim of facilitating an understanding of the molecular basis of the mechanism of MTX action to achieve its effective use in RA therapy.
Collapse
Affiliation(s)
- Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | | |
Collapse
|