1
|
Pravdić Z, Suvajdžić-Vuković N, Virijević M, Mitrović M, Pantić N, Sabljić N, Pavlović Đ, Marjanović I, Bukumirić Z, Vidović A, Jaković L, Pavlović S, Gašić V. Can pharmacogenetics impact the therapeutic effect of cytarabine and anthracyclines in adult acute myeloid leukaemia patients?: A Serbian experience. J Med Biochem 2024; 43:545-555. [PMID: 39139169 PMCID: PMC11318899 DOI: 10.5937/jomb0-47459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/21/2024] [Indexed: 08/15/2024] Open
Abstract
Background Cytarabine-anthracycline-based induction chemotherapy remains the standard of care for remission induction among patients with newly diagnosed acute myeloid leukaemia (AML). There are remarkable differences in therapy response among AML patients. This fact could be partly explained by the patients' genetic variability related to the metabolic paths of cytarabine and anthracyclines. This study aims to evaluate the effect of variants in pharmacogenes SLC29A1, DCK, ABCB1, GSTM1, and GSTT1, as well as laboratory and AML-related parameters on clinical outcomes in adult AML patients. Methods A total of 100 AML patients were included in the study. Pharmacogenetic variants SLC29A1 rs9394992, DCK rs12648166, ABCB1 rs2032582, and GSTM1 and GSTT1 gene deletions were detected by methodology based on PCR, fragment analysis and direct sequencing. The methods of descriptive and analytic statistics were used. Survival analysis was done using the Kaplan-Meier method using the Log-Rank test. Results This is the first study of adult AML pharmacogenetics in the Serbian population. Clinical outcomes in our cohort of AML patients were not impacted by analysed variants in SLC29A1, DCK, ABCB1 and GSTT1, and GSTM1 genes, independently or in combinations. Achievement of complete remission was identified as an independent prognostic indicator of clinical outcome. Conclusions The population-specific genomic profile has to be considered in pharmacogenetics. Since the data on AML pharmacogenetics in European populations is limited, our results contribute to knowledge in this field and strongly indicate that a high-throughput approach must be applied to find particular pharmacogenetic markers of AML in the European population.
Collapse
Affiliation(s)
- Zlatko Pravdić
- University Clinical Centre of Serbia, Clinic of Haematology, Belgrade
| | | | | | - Mirjana Mitrović
- University Clinical Centre of Serbia, Clinic of Haematology, Belgrade
| | - Nikola Pantić
- University Clinical Centre of Serbia, Clinic of Haematology, Belgrade
| | - Nikica Sabljić
- University Clinical Centre of Serbia, Clinic of Haematology, Belgrade
| | - Đorđe Pavlović
- University of Belgrade, Institute of Molecular Genetics and Genetical Engineering, Belgrade
| | - Irena Marjanović
- University of Belgrade, Institute of Molecular Genetics and Genetical Engineering, Belgrade
| | - Zoran Bukumirić
- University of Belgrade, Faculty of Medicine, Institute of Medical Statistics and Informatics, Belgrade
| | - Ana Vidović
- University Clinical Centre of Serbia, Clinic of Haematology, Belgrade
| | - Ljubomir Jaković
- University Clinical Centre of Serbia, Clinic of Haematology, Belgrade
| | - Sonja Pavlović
- University of Belgrade, Institute of Molecular Genetics and Genetical Engineering, Belgrade
| | - Vladimir Gašić
- University of Belgrade, Institute of Molecular Genetics and Genetical Engineering, Belgrade
| |
Collapse
|
2
|
Rodrigues JCG, Fernandes MR, Ribeiro-dos-Santos AM, de Araújo GS, de Souza SJ, Guerreiro JF, Ribeiro-dos-Santos Â, de Assumpção PP, dos Santos NPC, Santos S. Pharmacogenomic Profile of Amazonian Amerindians. J Pers Med 2022; 12:jpm12060952. [PMID: 35743738 PMCID: PMC9224798 DOI: 10.3390/jpm12060952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/05/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
Given the role of pharmacogenomics in the large variability observed in drug efficacy/safety, an assessment about the pharmacogenomic profile of patients prior to drug prescription or dose adjustment is paramount to improve adherence to treatment and prevent adverse drug reaction events. A population commonly underrepresented in pharmacogenomic studies is the Native American populations, which have a unique genetic profile due to a long process of geographic isolation and other genetic and evolutionary processes. Here, we describe the pharmacogenetic variability of Native American populations regarding 160 pharmacogenes involved in absorption, distribution, metabolism, and excretion processes and biological pathways of different therapies. Data were obtained through complete exome sequencing of individuals from 12 different Amerindian groups of the Brazilian Amazon. The study reports a total of 3311 variants; of this, 167 are exclusive to Amerindian populations, and 1183 are located in coding regions. Among these new variants, we found non-synonymous coding variants in the DPYD and the IFNL4 genes and variants with high allelic frequencies in intronic regions of the MTHFR, TYMS, GSTT1, and CYP2D6 genes. Additionally, 332 variants with either high or moderate (disruptive or non-disruptive impact in protein effectiveness, respectively) significance were found with a minimum of 1% frequency in the Amazonian Amerindian population. The data reported here serve as scientific basis for future design of specific treatment protocols for Amazonian Amerindian populations as well as for populations admixed with them, such as the Northern Brazilian population.
Collapse
Affiliation(s)
- Juliana Carla Gomes Rodrigues
- Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belem 66073-000, Brazil; (M.R.F.); (Â.R.-d.-S.); (P.P.d.A.); (N.P.C.d.S.); (S.S.)
- Correspondence: ; Tel.: +55-(91)-983973173
| | - Marianne Rodrigues Fernandes
- Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belem 66073-000, Brazil; (M.R.F.); (Â.R.-d.-S.); (P.P.d.A.); (N.P.C.d.S.); (S.S.)
| | - André Maurício Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belem 66075-110, Brazil; (A.M.R.-d.-S.); (G.S.d.A.); (J.F.G.)
| | - Gilderlanio Santana de Araújo
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belem 66075-110, Brazil; (A.M.R.-d.-S.); (G.S.d.A.); (J.F.G.)
| | | | - João Farias Guerreiro
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belem 66075-110, Brazil; (A.M.R.-d.-S.); (G.S.d.A.); (J.F.G.)
| | - Ândrea Ribeiro-dos-Santos
- Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belem 66073-000, Brazil; (M.R.F.); (Â.R.-d.-S.); (P.P.d.A.); (N.P.C.d.S.); (S.S.)
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belem 66075-110, Brazil; (A.M.R.-d.-S.); (G.S.d.A.); (J.F.G.)
| | - Paulo Pimentel de Assumpção
- Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belem 66073-000, Brazil; (M.R.F.); (Â.R.-d.-S.); (P.P.d.A.); (N.P.C.d.S.); (S.S.)
| | - Ney Pereira Carneiro dos Santos
- Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belem 66073-000, Brazil; (M.R.F.); (Â.R.-d.-S.); (P.P.d.A.); (N.P.C.d.S.); (S.S.)
| | - Sidney Santos
- Núcleo de Pesquisa em Oncologia, Universidade Federal do Pará, Belem 66073-000, Brazil; (M.R.F.); (Â.R.-d.-S.); (P.P.d.A.); (N.P.C.d.S.); (S.S.)
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belem 66075-110, Brazil; (A.M.R.-d.-S.); (G.S.d.A.); (J.F.G.)
| |
Collapse
|
3
|
GSTM1 and GSTT1 double null genotypes determining cell fate and proliferation as potential risk factors of relapse in children with hematological malignancies after hematopoietic stem cell transplantation. J Cancer Res Clin Oncol 2021; 148:71-86. [PMID: 34499222 PMCID: PMC8752561 DOI: 10.1007/s00432-021-03769-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/14/2021] [Indexed: 10/29/2022]
Abstract
PURPOSE This study aimed to retrospectively evaluate the genetic association of null variants of glutathione S-transferases GSTM1 and GSTT1 with relapse incidence in children with hematological malignancies (HMs) undergoing busulfan (BU)- containing allogeneic hematopoietic stem cell transplantation (HSCT) and to assess the impact of these variants on BU-induced cytotoxicity on the immortalized lymphoblastoid cell lines (LCLs) and tumor THP1 GST gene-edited cell models. METHODS GSTM1- and GSTT1-null alleles were genotyped using germline DNA from whole blood prior to a conditioning BU-based regimen. Association of GSTM1- and GSTT1-null variants with relapse incidence was analyzed using multivariable competing risk analysis. BU-induced cell death studies were conducted in GSTs- null and non-null LCLs and CRISPR-Cas9 gene-edited THP1 leukemia cell lines. RESULTS Carrying GSTM1/GSTT1 double null genotype was found to be an independent risk factor for post-HSCT relapse in 86 children (adjusted HR: 6.52 [95% Cl, 2.76-15.42; p = 1.9 × 10-5]). BU-induced cell death preferentially in THP1GSTM1(non-null) and LCLsGSTM1(non-null) as shown by decreased viability, increased necrosis and levels of the oxidized form of glutathione compared to null cells, while GSTT1 non-null cells showed increased baseline proliferation. CONCLUSION The clinical association suggests that GSTM1/GSTT1 double null genotype could serve as genetic stratification biomarker for the high risk of post-HSCT relapse. Functional studies have indicated that GSTM1 status modulates BU-induced cell death. On the other hand, GSTT1 is proposed to be involved in baseline cell proliferation.
Collapse
|
4
|
Andrade FG, Feliciano SVM, Sardou-Cezar I, Brisson GD, dos Santos-Bueno FV, Vianna DT, Marques LVC, Terra-Granado E, Zalcberg I, Santos MDO, Costa JT, Noronha EP, Thuler LCS, Wiemels JL, Pombo-de-Oliveira MS. Pediatric Acute Promyelocytic Leukemia: Epidemiology, Molecular Features, and Importance of GST-Theta 1 in Chemotherapy Response and Outcome. Front Oncol 2021; 11:642744. [PMID: 33816294 PMCID: PMC8017304 DOI: 10.3389/fonc.2021.642744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/08/2021] [Indexed: 11/23/2022] Open
Abstract
Previous studies have suggested a variation in the incidence of acute promyelocytic leukemia (APL) among the geographic regions with relatively higher percentages in the Latin American population. We aimed to explore the population burden of pediatric APL, gathering information from the population-based cancer registry (PBCR) and the diagnosis of APL obtained through incident cases from a hospital-based cohort. The homozygous deletion in glutathione S-transferases (GSTs) leads to a loss of enzyme detoxification activity, possibly affecting the treatment response. Mutations in the RAS pathway genes are also considered to be a key component of the disease both in the pathogenesis and in the outcomes. We have assessed mutations in a RAS-MAP kinase pathway (FLT3, PTPN11, and K-/NRAS) and GST variant predisposition risk in the outcome. Out of the 805 children and adolescents with acute myeloid leukemia (AML) who are registered in the PBCR, 35 (4.3%) were APL cases. The age-adjusted incidence rate (AAIR) was 0.03 per 100,000 person-years. One-hundred and sixty-three patients with APL were studied out of 931 AML cases (17.5%) from a hospital-based cohort. Mutations in FLT3, KRAS, and NRAS accounted for 52.1% of the cases. Patients with APL presented a 5-year probability of the overall survival (OS) of 67.3 ± 5.8%. A GST-theta 1 (GSTT1) null genotype conferred adverse prognosis, with an estimated hazard ratio of 2.8, 95% confidence interval (CI) 1.2-6.9. We speculate that the GSTT1 polymorphism is associated with therapeutics and would allow better OS of patients with APL with a GSTT1 null genotype.
Collapse
Affiliation(s)
- Francianne G. Andrade
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Suellen V. M. Feliciano
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Ingrid Sardou-Cezar
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Gisele D. Brisson
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Filipe V. dos Santos-Bueno
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Danielle T. Vianna
- Laboratory of Molecular Biology, Bone Marrow Transplantation Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Luísa V. C. Marques
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Eugênia Terra-Granado
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Ilana Zalcberg
- Laboratory of Molecular Biology, Bone Marrow Transplantation Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Marceli de O. Santos
- Surveillance and Prevention, Instituto Nacional de Cancer, Rio de Janeiro, Brazil
| | - Juliana T. Costa
- Department of Pediatric Hematology-Oncology, Hospital Martagão Gesteira, Salvador, Brazil
| | - Elda P. Noronha
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Luiz C. S. Thuler
- Clinical Research Department, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Joseph L. Wiemels
- Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Maria S. Pombo-de-Oliveira
- Pediatric Hematology-Oncology Program, Research Center, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Lee N, Park SM, Yee J, Yoon HY, Han JM, Gwak HS. Association Between Glutathione-S-Transferase Gene Polymorphisms and Responses to Tyrosine Kinase Inhibitor Treatment in Patients with Chronic Myeloid Leukemia: A Meta-analysis. Target Oncol 2021; 15:47-54. [PMID: 31974831 DOI: 10.1007/s11523-020-00696-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Although many earlier studies revealed an effect of glutathione-S-transferase (GST) gene polymorphisms on tyrosine kinase inhibitor (TKI) treatment responses in chronic myeloid leukemia (CML) patients, the significance of this relationship remains controversial. OBJECTIVE This study aimed to review and meta-analyze treatment responses to TKIs in patients with CML and GST gene polymorphisms, including GSTT1, GSTM1, and GSTP1. PATIENTS AND METHODS We searched four medical databases, PubMed, Web of Science, the Cochrane Library, and Embase, by using keywords related to GST gene polymorphisms and clinical responses in CML patients receiving TKI treatment. The meta-analysis was performed using RevMan version 5.3 and Comprehensive Meta-Analysis software version 3.0. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to examine the association between GSTT1, GSTM1, and GSTP1 polymorphisms and TKI treatment responses in patients with CML. RESULTS The null polymorphisms of GSTT1 and GSTM1 did not affect TKI treatment responses, while the GSTP1 Ile105Val polymorphism had a significant impact on responses to TKI. Patients who were GSTP1 variant allele carriers (AG + GG) had poor responses to TKI treatment compared to patients who were wild-type homozygote carriers (AA) (OR 1.85, 95% CI 1.31-2.62; p < 0.001). CONCLUSIONS This meta-analysis of patients with CML showed that G allele carriers with GSTP1 Ile105Val polymorphism had significantly worse responses to TKI treatment than wild-type homozygote carriers.
Collapse
Affiliation(s)
- Nari Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Su Min Park
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Jeong Yee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Ha Young Yoon
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Ji Min Han
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
| | - Hye Sun Gwak
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
- College of Pharmacy, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Republic of Korea.
| |
Collapse
|
6
|
Lamba JK, Cao X, Raimondi S, Downing J, Ribeiro R, Gruber TA, Rubnitz J, Pounds S. DNA Methylation Clusters and Their Relation to Cytogenetic Features in Pediatric AML. Cancers (Basel) 2020; 12:cancers12103024. [PMID: 33080932 PMCID: PMC7603219 DOI: 10.3390/cancers12103024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022] Open
Abstract
Acute Myeloid Leukemia (AML) is characterized by recurrent genetic and cytogenetic lesions that are utilized for risk stratification and for making treatment decisions. In recent years, methylation dysregulation has been extensively studied and associated with risk groups and prognosis in adult AML, however, such studies in pediatric AML are limited. Moreover, the mutations in epigenetic genes such as DNMT3A, IDH1 or IDH2 are almost absent or rare in pediatric patients as compared to their abundance in adult AML. In the current study, we evaluated methylation patterns that occur with or independent of the well-defined cytogenetic features in pediatric AML patients enrolled on multi-site AML02 clinical trial (NCT00136084). Our results demonstrate that unlike adult AML, cytosine DNA methylation does not result in significant unique clusters in pediatric AML, however, DNA methylation signatures correlated significantly with the most common and recurrent cytogenetic features. Paired evaluation of DNA methylation and expression identified genes and pathways of biological relevance that hold promise for novel therapeutic strategies. Our results further demonstrate that epigenetic signatures occur complimentary to the well-established chromosomal/mutational landscape, implying that dysregulation of oncogenes or tumor suppressors might be leveraging both genetic and epigenetic mechanisms to impact biological pathways critical for leukemogenesis.
Collapse
Affiliation(s)
- Jatinder K. Lamba
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32608, USA
- Correspondence:
| | - Xueyuan Cao
- Department of Acute and Tertiary Care, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Susana Raimondi
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (S.R.); (J.D.)
| | - James Downing
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (S.R.); (J.D.)
| | - Raul Ribeiro
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.); (J.R.)
| | - Tanja A. Gruber
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Jeffrey Rubnitz
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.); (J.R.)
| | - Stanley Pounds
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| |
Collapse
|
7
|
Cardona-Echeverry A, Prada-Arismendy J. Deciphering the role of Wnt signaling in acute myeloid leukemia prognosis: how alterations in DNA methylation come into play in patients' prognosis. J Cancer Res Clin Oncol 2020; 146:3097-3109. [PMID: 32980885 DOI: 10.1007/s00432-020-03407-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 09/21/2020] [Indexed: 10/23/2022]
Abstract
Acute myeloid leukemia (AML) is a malignant clonal disorder affecting myeloid differentiation through mechanisms that include epigenetic dysregulation. Abnormal changes in DNA methylation and gene expression profiles of pathways involved in hematopoietic development, such as Wnt/β-catenin, contribute to the transformation, development, and maintenance of leukemic cells. This review summarizes the alterations of Wnt signaling-related genes at the epigenetic and transcriptional level and their implications for AML prognosis. Among the implications of epigenetic alterations in AML, methylation of Wnt antagonists is related to poor prognosis, whereas their upregulation has been associated with a better clinical outcome. Furthermore, Wnt target genes c-Myc and LEF-1 present distinct implications. LEF-1 expression positively influences the patient overall survival. c-Myc upregulation has been associated with treatment resistance in AML, although c-Myc expression is not exclusively dependent of Wnt signaling. Understanding the signaling abnormalities could help us to further understand leukemogenesis, improve the current risk stratification for AML patients, and even serve to propose novel therapeutic targets.
Collapse
Affiliation(s)
- Andrés Cardona-Echeverry
- Grupo de Investigación e innovación Biomédica-GI2B, Facultad de Ciencias Exactas y Aplicadas, Instituto Tecnológico Metropolitano-ITM, 050034, Medellín, Colombia
| | - Jeanette Prada-Arismendy
- Grupo de Investigación e innovación Biomédica-GI2B, Facultad de Ciencias Exactas y Aplicadas, Instituto Tecnológico Metropolitano-ITM, 050034, Medellín, Colombia.
| |
Collapse
|
8
|
Abstract
The mercapturic acid pathway is a major route for the biotransformation of xenobiotic and endobiotic electrophilic compounds and their metabolites. Mercapturic acids (N-acetyl-l-cysteine S-conjugates) are formed by the sequential action of the glutathione transferases, γ-glutamyltransferases, dipeptidases, and cysteine S-conjugate N-acetyltransferase to yield glutathione S-conjugates, l-cysteinylglycine S-conjugates, l-cysteine S-conjugates, and mercapturic acids; these metabolites constitute a "mercapturomic" profile. Aminoacylases catalyze the hydrolysis of mercapturic acids to form cysteine S-conjugates. Several renal transport systems facilitate the urinary elimination of mercapturic acids; urinary mercapturic acids may serve as biomarkers for exposure to chemicals. Although mercapturic acid formation and elimination is a detoxication reaction, l-cysteine S-conjugates may undergo bioactivation by cysteine S-conjugate β-lyase. Moreover, some l-cysteine S-conjugates, particularly l-cysteinyl-leukotrienes, exert significant pathophysiological effects. Finally, some enzymes of the mercapturic acid pathway are described as the so-called "moonlighting proteins," catalytic proteins that exert multiple biochemical or biophysical functions apart from catalysis.
Collapse
Affiliation(s)
- Patrick E Hanna
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
9
|
Pravdić Z, Suvajdžić-Vuković N. Pharmacogenomics and regulation of apoptosis in acute myeloid leukaemia. MEDICINSKI PODMLADAK 2020. [DOI: 10.5937/mp71-28140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a heterogenous clonal hematopoietic malignancy primarily treated with combination of cytarabine (ara-C) and anthracyclines. Despite high remission rates, especially in younger patients, a vast majority of patients die due to relapse or chemotherapy/stem cell transplantation-related toxicity. The partial explanation for this grim clinical outcome lies in the patients' genetic variability. In this review, we will summarize how genetic polymorphisms of proteins, in metabolic paths of cytarabine and anthracyclines and proteins involved in regulation of apoptosis, influence efficacy and toxicity in the AML treatment.
Collapse
|
10
|
Muddathir ARM, Abdallah EI, Khabour OF, Abdelgader RE, Elgari MM. Age- and gender-independent association of glutathione S-transferase null polymorphisms with chronic myeloid leukemia. Bosn J Basic Med Sci 2019; 19:350-354. [PMID: 30995900 DOI: 10.17305/bjbms.2019.4176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 03/11/2019] [Indexed: 02/06/2023] Open
Abstract
The glutathione S-transferase (GST) genes encode enzymes that mediate the detoxification of xenobiotics by catalyzing the conjugation of glutathione (GSH) to xenobiotic substrates. The aim of the current study is to investigate the association between GSTT1 and GSTM1 polymorphisms and chronic myeloid leukemia (CML) among Sudanese patients. Patients with CML (n = 115) were recruited to the study from the Radiation and Isotope Centre Khartoum (RICK)-Sudan. Healthy individuals (n = 104) were included as controls. Genotyping of GSTT1 and GSTM1 polymorphisms was performed using multiplex PCR. Null deletions in the GSTT1 and GSTM1 genes are common in the Sudanese population (control group), with frequencies of 33.9% and 38.2%, respectively. The frequencies of GSTT1 (OR: 3.25, 95% CI: 1.87-5.65, p < 0.001) and GSTM1 (OR: 2.14, 95% CI: 1.25-3.67, p < 0.005) null genotypes were significantly higher in CML patients vs. controls. The distribution of GSTT1 and GSTM1 null polymorphisms was not different between male and female (p > 0.01) and young and old CML patients (p > 0.05). Hematological parameters were not affected by null polymorphisms in the patient group (p > 0.05). In addition, the frequency of GSTM1 null polymorphism was lower in advanced-phase CML patients compared to chronic-phase patients (p < 0.05). The GSTT1 and GSTM1 null polymorphisms are associated with CML among Sudanese patients, independently of their age and gender.
Collapse
Affiliation(s)
- Abdel Rahim Mahmoud Muddathir
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Taibah University, Medina, Saudi Arabia; Department of Hematology and Blood Transfusion, Faculty of Medical Laboratory Sciences, Alzaeim Alazhari University, Khartoum, Sudan.
| | | | | | | | | |
Collapse
|
11
|
Morales-Prieto N, Ruiz-Laguna J, Sheehan D, Abril N. Transcriptome signatures of p,p´-DDE-induced liver damage in Mus spretus mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 238:150-167. [PMID: 29554563 DOI: 10.1016/j.envpol.2018.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/19/2018] [Accepted: 03/04/2018] [Indexed: 06/08/2023]
Abstract
The use of DDT (1,1,1-trichloro-2,2-bis(p-chlorophenyl) ethane) in some countries, although regulated, is contributing to an increased worldwide risk of exposure to this organochlorine pesticide or its derivative p,p'-DDE [1,1-dichloro-2,2-bis(p-chlorophenyl) ethylene]. Many studies have associated p,p'-DDE exposure to type 2 diabetes, obesity and alterations of the reproductive system, but their molecular mechanisms of toxicity remain poorly understood. We have addressed this issue by using commercial microarrays based on probes for the entire Mus musculus genome to determine the hepatic transcriptional signatures of p,p'-DDE in the phylogenetically close mouse species Mus spretus. High-stringency hybridization conditions and analysis assured reliable results, which were also verified, in part, by qRT-PCR, immunoblotting and/or enzymatic activity. Our data linked 198 deregulated genes to mitochondrial dysfunction and perturbations of central signaling pathways (kinases, lipids, and retinoic acid) leading to enhanced lipogenesis and aerobic glycolysis, inflammation, cell proliferation and testosterone catabolism and excretion. Alterations of transcript levels of genes encoding enzymes involved in testosterone catabolism and excretion would explain the relationships established between p,p´-DDE exposure and reproductive disorders, obesity and diabetes. Further studies will help to fully understand the molecular basis of p,p´-DDE molecular toxicity in liver and reproductive organs, to identify effective exposure biomarkers and perhaps to design efficient p,p'-DDE exposure counteractive strategies.
Collapse
Affiliation(s)
- Noelia Morales-Prieto
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - Julia Ruiz-Laguna
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - David Sheehan
- College of Arts and Science, Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain.
| |
Collapse
|
12
|
Perperopoulou F, Pouliou F, Labrou NE. Recent advances in protein engineering and biotechnological applications of glutathione transferases. Crit Rev Biotechnol 2017; 38:511-528. [PMID: 28936894 DOI: 10.1080/07388551.2017.1375890] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Glutathione transferases (GSTs, EC 2.5.1.18) are a widespread family of enzymes that play a central role in the detoxification, metabolism, and transport or sequestration of endogenous or xenobiotic compounds. During the last two decades, delineation of the important structural and catalytic features of GSTs has laid the groundwork for engineering GSTs, involving both rational and random approaches, aiming to create new variants with new or altered properties. These approaches have expanded the usefulness of native GSTs, not only for understanding the fundamentals of molecular detoxification mechanisms, but also for the development medical, analytical, environmental, and agricultural applications. This review article attempts to summarize successful examples and current developments on GST engineering, highlighting in parallel the recent knowledge gained on their phylogenetic relationships, structural/catalytic features, and biotechnological applications.
Collapse
Affiliation(s)
- Fereniki Perperopoulou
- a Department of Biotechnology, Laboratory of Enzyme Technology , School of Food, Biotechnology and Development, Agricultural University of Athens , Athens , Greece
| | - Fotini Pouliou
- a Department of Biotechnology, Laboratory of Enzyme Technology , School of Food, Biotechnology and Development, Agricultural University of Athens , Athens , Greece
| | - Nikolaos E Labrou
- a Department of Biotechnology, Laboratory of Enzyme Technology , School of Food, Biotechnology and Development, Agricultural University of Athens , Athens , Greece
| |
Collapse
|
13
|
Ang HX, Chan SL, Sani LL, Quah CB, Brunham LR, Tan BOP, Winther MD. Pharmacogenomics in Asia: a systematic review on current trends and novel discoveries. Pharmacogenomics 2017; 18:891-910. [DOI: 10.2217/pgs-2017-0009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
While early pharmacogenomic studies have primarily been carried out in Western populations, there has been a notable increase in the number of Asian studies over the past decade. We systematically reviewed all pharmacogenomic studies conducted in Asia published before 2016 to highlight trends and identify research gaps in Asia. We observed that pharmacogenomic research in Asia was dominated by larger developed countries, notably Japan and Korea, and mainly driven by local researchers. Studies were focused on drugs acting on the CNS, chemotherapeutics and anticoagulants. Significantly, several novel pharmacogenomic associations have emerged from Asian studies. These developments are highly encouraging for the strength of regional scientific and clinical community and propound the importance of discovery studies in different populations.
Collapse
Affiliation(s)
- Hazel Xiaohui Ang
- Genome Institute of Singapore, Agency for Science, Technology & Research, Singapore
| | - Sze Ling Chan
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology & Research, Singapore
| | - Levana L Sani
- Genome Institute of Singapore, Agency for Science, Technology & Research, Singapore
| | | | - Liam R Brunham
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology & Research, Singapore
- Department of Medicine, Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Boon Ooi Patrick Tan
- Genome Institute of Singapore, Agency for Science, Technology & Research, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Cancer & Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore
- Division of Cellular & Molecular Research, National Cancer Centre Singapore, Singapore
| | - Michael D Winther
- Genome Institute of Singapore, Agency for Science, Technology & Research, Singapore
| |
Collapse
|
14
|
Megías-Vericat JE, Montesinos P, Herrero MJ, Bosó V, Martínez-Cuadrón D, Poveda JL, Sanz MÁ, Aliño SF. Pharmacogenomics and the treatment of acute myeloid leukemia. Pharmacogenomics 2016; 17:1245-1272. [DOI: 10.2217/pgs-2016-0055] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clinically and biologically heterogeneous malignancy that is primarily treated with combinations of cytarabine and anthracyclines. Although this scheme remains effective in most of the patients, variability of outcomes in patients has been partly related with their genetic variability. Several pharmacogenetic studies have analyzed the impact of polymorphisms in genes encoding transporters, metabolizers or molecular targets of chemotherapy agents. A systematic review on all eligible studies was carried out in order to estimate the effect of polymorphisms of anthracyclines and cytarabine pathways on efficacy and toxicity of AML treatment. Other emerging genes recently studied in AML, such as DNA repair genes, genes potentially related to chemotherapy response or AML prognosis, have also been included.
Collapse
Affiliation(s)
- Juan Eduardo Megías-Vericat
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Pau Montesinos
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - María José Herrero
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Departamento Farmacología, Facultad de Medicina, Universidad de Valencia, Avda, Blasco Ibáñez 15, 46010 – Valencia, Spain
| | - Virginia Bosó
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - David Martínez-Cuadrón
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - José Luis Poveda
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Miguel Ángel Sanz
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Salvador F Aliño
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Departamento Farmacología, Facultad de Medicina, Universidad de Valencia, Avda, Blasco Ibáñez 15, 46010 – Valencia, Spain
- Unidad de Farmacología Clínica, Área del Medicamento, Hospital Universitario y Politécnico La Fe. Avda. Fernando Abril Martorell 106, 46026 – Valencia, Spain
| |
Collapse
|
15
|
Lu L, Pandey AK, Houseal MT, Mulligan MK. The Genetic Architecture of Murine Glutathione Transferases. PLoS One 2016; 11:e0148230. [PMID: 26829228 PMCID: PMC4734686 DOI: 10.1371/journal.pone.0148230] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 01/14/2016] [Indexed: 12/17/2022] Open
Abstract
Glutathione S-transferase (GST) genes play a protective role against oxidative stress and may influence disease risk and drug pharmacokinetics. In this study, massive multiscalar trait profiling across a large population of mice derived from a cross between C57BL/6J (B6) and DBA2/J (D2)—the BXD family—was combined with linkage and bioinformatic analyses to characterize mechanisms controlling GST expression and to identify downstream consequences of this variation. Similar to humans, mice show a wide range in expression of GST family members. Variation in the expression of Gsta4, Gstt2, Gstz1, Gsto1, and Mgst3 is modulated by local expression QTLs (eQTLs) in several tissues. Higher expression of Gsto1 in brain and liver of BXD strains is strongly associated (P < 0.01) with inheritance of the B6 parental allele whereas higher expression of Gsta4 and Mgst3 in brain and liver, and Gstt2 and Gstz1 in brain is strongly associated with inheritance of the D2 parental allele. Allele-specific assays confirmed that expression of Gsto1, Gsta4, and Mgst3 are modulated by sequence variants within or near each gene locus. We exploited this endogenous variation to identify coexpression networks and downstream targets in mouse and human. Through a combined systems genetics approach, we provide new insight into the biological role of naturally occurring variants in GST genes.
Collapse
Affiliation(s)
- Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38106, United States of America
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Ashutosh K. Pandey
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38106, United States of America
| | - M. Trevor Houseal
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38106, United States of America
| | - Megan K. Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38106, United States of America
- * E-mail:
| |
Collapse
|
16
|
GSTT1 and GSTM1 polymorphisms predict treatment outcome for breast cancer: a systematic review and meta-analysis. Tumour Biol 2015; 37:151-62. [PMID: 26577857 DOI: 10.1007/s13277-015-4401-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/05/2015] [Indexed: 02/07/2023] Open
Abstract
Observational studies have reported controversial results on the association between GSTT1 and GSTM1 genotypes and treatment outcome of breast cancer. The purpose of this study is to evaluate the association between GSTT1 and GSTM1 and treatment outcome in breast cancer patients. Eligible studies were searched in PubMed, EMBASE, Cochrane Library, and China National Knowledge Infrastructure databases. A random-effect model or fixed-effect model was used to calculate the overall combined risk estimates. Twenty-one studies with a total of 4990 patients were included in this meta-analysis. The GSTM1 null genotype (odds ratio (OR) = 1.33, 95 % confidence interval (CI) 1.01-1.75, P = 0.046) and GSTT1/GSTM1 double null genotype (OR = 2.22, 95 % CI 1.02-4.84, P = 0.045) were significantly associated with an increased tumor response. A reduced overall survival (hazard ratio (HR) = 0.84, 95 % CI 0.72-0.98, P = 0.024) was observed in GSTM1 null genotype, especially in mixed descent (HR = 0.77, 95 % CI 0.61-0.96, P = 0.018) and large sample size (HR = 0.85, 95 % CI 0.72-0.99, P = 0.033). Evidence of publication bias was observed in GSTM1 genotype rather than in GSTT1 genotype. This meta-analysis suggests that GSTM1 null and GSTT1/GSTM1 double null polymorphisms might be significantly associated with an increased tumor response. However, the GSTM1 null genotype might be significantly associated with a reduced overall survival. Future studies are warranted to confirm these findings.
Collapse
|