51
|
Nukala KM, Lilienthal AJ, Lye SH, Bassuk AG, Chtarbanova S, Manak JR. Downregulation of oxidative stress-mediated glial innate immune response suppresses seizures in a fly epilepsy model. Cell Rep 2023; 42:112004. [PMID: 36641750 PMCID: PMC9942582 DOI: 10.1016/j.celrep.2023.112004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/30/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023] Open
Abstract
Previous work in our laboratory has shown that mutations in prickle (pk) cause myoclonic-like seizures and ataxia in Drosophila, similar to what is observed in humans carrying mutations in orthologous PRICKLE genes. Here, we show that pk mutant brains show elevated, sustained neuronal cell death that correlates with increasing seizure penetrance, as well as an upregulation of mitochondrial oxidative stress and innate immune response (IIR) genes. Moreover, flies exhibiting more robust seizures show increased levels of IIR-associated target gene expression suggesting they may be linked. Genetic knockdown in glia of either arm of the IIR (Immune Deficiency [Imd] or Toll) leads to a reduction in neuronal death, which in turn suppresses seizure activity, with oxidative stress acting upstream of IIR. These data provide direct genetic evidence that oxidative stress in combination with glial-mediated IIR leads to progression of an epilepsy disorder.
Collapse
Affiliation(s)
- Krishna M Nukala
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Shu Hui Lye
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL 35487, USA
| | - Alexander G Bassuk
- Department of Pediatrics, University of Iowa and Carver College of Medicine, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa and Carver College of Medicine, Iowa City, IA 52242, USA; The Iowa Neuroscience Institute, University of Iowa and Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - J Robert Manak
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA; Department of Pediatrics, University of Iowa and Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
52
|
de Almeida GRL, Szczepanik JC, Selhorst I, Cunha MP, Dafre AL. The expanding impact of methylglyoxal on behavior-related disorders. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110635. [PMID: 36103947 DOI: 10.1016/j.pnpbp.2022.110635] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/02/2022] [Accepted: 09/07/2022] [Indexed: 01/17/2023]
Abstract
Methylglyoxal (MGO) is a reactive dicarbonyl compound formed as a byproduct of glycolysis. MGO is a major cell-permeant precursor of advanced glycation end products (AGEs), since it readily reacts with basic phospholipids and nucleotides, as well as amino acid residues of proteins, such as arginine, cysteine, and lysine. The AGEs production induced by MGO are widely associated with several pathologies, including neurodegenerative diseases. However, the impact of MGO metabolism and AGEs formation in the central nervous system (particularly in neurons, astrocytes and oligodendrocytes) on behavior and psychiatric diseases is not fully understood. Here, we briefly present background information on the biological activity of MGO in the central nervous system. It was gathered the available information on the role of MGO metabolism at the physiological processes, as well as at the neurobiology of psychiatry diseases, especially pain-related experiences, anxiety, depression, and cognition impairment-associated diseases. To clarify the role of MGO on behavior and associated diseases, we reviewed primarily the main findings at preclinical studies focusing on genetic and pharmacological approaches. Since monoamine neurotransmitter systems are implicated as pivotal targets on the pathophysiology and treatment of psychiatry and cognitive-related diseases, we also reviewed how MGO affects these neurotransmission systems and the implications of this phenomenon for nociception and pain; learning and cognition; and mood. In summary, this review highlights the pivotal role of glyoxalase 1 (Glo1) and MGO levels in modulating behavioral phenotypes, as well as related cellular and molecular signaling. Conclusively, this review signals dopamine as a new neurochemical MGO target, as well as highlights how MGO metabolism can modulate the pathophysiology and treatment of pain, psychiatric and cognitive-related diseases.
Collapse
Affiliation(s)
- Gudrian R L de Almeida
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | - Jozimar C Szczepanik
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | - Ingrid Selhorst
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| | - Mauricio P Cunha
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, SC, Brazil; Department of Basic Sciences of Life, Federal University of Juiz de Fora, 35010-177 Governador Valadares, MG, Brazil.
| | - Alcir L Dafre
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900 Florianópolis, SC, Brazil
| |
Collapse
|
53
|
Shutevska K, Zhivikj Z, Dimkovski A, Geshkovski N, Petreska Ivanovska T, Kadifkova Panovska T, Kapedanovska Nestorovska A. The importance of AKR1D1 enzyme in drug metabolism. MAKEDONSKO FARMACEVTSKI BILTEN 2022. [DOI: 10.33320/maced.pharm.bull.2022.68.03.155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Kristina Shutevska
- University Ss Cyril and Methodius, Faculty of Pharmacy, Institute of Applied Biochemistry, Mother Theresa 47, 1000 Skopje, Republic of North Macedonia
| | - Zoran Zhivikj
- University Ss Cyril and Methodius, Faculty of Pharmacy, Institute of Applied Biochemistry, Mother Theresa 47, 1000 Skopje, Republic of North Macedonia
| | - Aleksandar Dimkovski
- University Ss Cyril and Methodius, Faculty of Pharmacy, Institute of Pharmaceutical Chemistry, Mother Theresa 47, 1000 Skopje, Republic of North Macedonia
| | - Nikola Geshkovski
- University Ss Cyril and Methodius, Faculty of Pharmacy, Institute of Pharmaceutical Technology, Mother Theresa 47, 1000 Skopje, Republic of North Macedonia
| | - Tanja Petreska Ivanovska
- University Ss Cyril and Methodius, Faculty of Pharmacy, Institute of Applied Biochemistry, Mother Theresa 47, 1000 Skopje, Republic of North Macedonia
| | - Tatjana Kadifkova Panovska
- University Ss Cyril and Methodius, Faculty of Pharmacy, Institute of Applied Biochemistry, Mother Theresa 47, 1000 Skopje, Republic of North Macedonia
| | - Aleksandra Kapedanovska Nestorovska
- University Ss Cyril and Methodius, Faculty of Pharmacy, Institute of Pharmaceutical Chemistry, Mother Theresa 47, 1000 Skopje, Republic of North Macedonia
| |
Collapse
|
54
|
Liu YY, Liu YW, Huang GK, Hung KC, Lin YH, Yeh CH, Yin SM, Tsai CH, Chen YH. Overexpression of AKR1B10 Predicts Poor Prognosis in Gastric Cancer Patients Undergoing Surgical Resection. Curr Oncol 2022; 30:85-99. [PMID: 36661656 PMCID: PMC9857867 DOI: 10.3390/curroncol30010007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Aldo-keto reductase family 1 member B10 (AKR1B10) is associated with several cancers, but the prognostic role in gastric cancer (GC) remains unclear. We enrolled 359 GC patients who underwent a gastrectomy with D2 lymph node dissection. AKR1B10 expression was scored using an immunoreactive scoring system based on immunohistochemistry. Adjuvant chemotherapy with S-1 or oxaliplatin plus capecitabine was administered to pathological stage II or III disease patients. There were 117 (32.6%) and 242 (67.4%) patients with AKR1B10 overexpression and low expression, respectively. Patients overexpressing AKR1B10 had worse 5-year disease-free survival (DFS) and overall survival (OS) rates than those with low expression of AKR1B10. Pathological T3-T4 stage, pathological stage III, lymph node ratio ≥25%, and AKR1B10 overexpression were independent prognostic factors for worse DFS and OS in univariate and multivariate analyses. For 162 stage II or III patients who received adjuvant chemotherapy after surgical resection and 59 patients with signet ring cell carcinoma histology, AKR1B10 overexpression was also associated with inferior DFS and OS. AKR1B10 was not associated with clinical survival in stage I GC patients. In conclusion, AKR1B10 overexpression may be an independent prognostic factor for worse survival in GC patients who underwent gastrectomy with D2 lymph node dissection.
Collapse
Affiliation(s)
- Yu-Yin Liu
- Division of General Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Yueh-Wei Liu
- Division of General Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Gong-Kai Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Kuo-Chen Hung
- Division of Trauma Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Yu-Hung Lin
- Division of General Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Cheng-Hsi Yeh
- Division of General Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Shih-Min Yin
- Division of General Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Ching-Hua Tsai
- Division of Trauma Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Yen-Hao Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Nursing, School of Nursing, Fooyin University, Kaohsiung 831, Taiwan
| |
Collapse
|
55
|
Chang GRL, Lin WY, Fan HC, Tu MY, Liu YH, Yen CC, Cidem A, Chen W, Chen CM. Kefir peptides ameliorate osteoporosis in AKR1A1 knockout mice with vitamin C deficiency by promoting osteoblastogenesis and inhibiting osteoclastogenesis. Biomed Pharmacother 2022; 156:113859. [DOI: 10.1016/j.biopha.2022.113859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022] Open
|
56
|
da Graça Cabreira M, Wang X, Critsinelis A, Setegne M, Lotfi P, Wan YW, Barrios G, Mei Z, Gee AP, Buja LM, Perin E. Environmental oxygen affects ex vivo growth and proliferation of mesenchymal progenitors by modulating mitogen-activated protein kinase and mammalian target of rapamycin signaling. Cytotherapy 2022; 24:1201-1210. [PMID: 36109320 DOI: 10.1016/j.jcyt.2022.06.005] [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: 12/16/2021] [Revised: 04/16/2022] [Accepted: 06/13/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AIMS Stem and progenitor cells of hematopoietic and mesenchymal lineages reside in the bone marrow under low oxygen (O2) saturation. O2 levels used in ex vivo expansion of multipotent mesenchymal stromal cells (MSCs) affect proliferation, metabolism and differentiation. METHODS Using cell-based assays and transcriptome and proteome data, the authors compared MSC cultures simultaneously grown under a conventional 19.95% O2 atmosphere or at 5% O2. RESULTS In 5% O2, MSCs showed better proliferation and higher self-renewal ability, most probably sustained by enhanced signaling activity of mitogen-activated protein kinase and mammalian target of rapamycin pathways. Non-oxidative glycolysis-based energy metabolism supported growth and proliferation in 5% O2 cultures, whereas MSCs grown under 19.95% O2 also utilized oxidative phosphorylation. Cytoprotection mechanisms used by cells under 5% O2 differed from 19.95% O2 suggesting differences in the triggers of cell stress between these two O2 conditions. CONCLUSIONS Based on the potential benefits for the growth and metabolism of MSCs, the authors propose the use of 5% O2 for MSC culture.
Collapse
Affiliation(s)
| | - Xiaohong Wang
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Mekedlawit Setegne
- Chemistry-Biology Interface Predoctoral Training Program, Stanford University, Stanford, California, USA
| | - Parisa Lotfi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Ying-Wooi Wan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Gabriela Barrios
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, Texas, USA
| | - Zhuyong Mei
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas, USA
| | - Adrian P Gee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas, USA
| | - Louis Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, University of Texas Health Science Center, Houston, Texas, USA
| | - Emerson Perin
- Center for Clinical Research, Texas Heart Institute, Houston, Texas, USA
| |
Collapse
|
57
|
Jean-Pierre M, Michalovicz LT, Kelly KA, O'Callaghan JP, Nathanson L, Klimas N, J. A. Craddock T. A pilot reverse virtual screening study suggests toxic exposures caused long-term epigenetic changes in Gulf War Illness. Comput Struct Biotechnol J 2022; 20:6206-6213. [DOI: 10.1016/j.csbj.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
|
58
|
Stancil SL, Voss M, Nolte W, Tumberger J, Adelman W, Abdel‐Rahman S. Effects of genotype and food on naltrexone exposure in adolescents. Clin Transl Sci 2022; 15:2732-2743. [PMID: 36200172 PMCID: PMC9652442 DOI: 10.1111/cts.13399] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 01/26/2023] Open
Abstract
Naltrexone (NTX), an opioid antagonist metabolized by aldo-keto reductase 1C4 (AKR1C4), is prescribed for psychiatric conditions like eating disorders with variable response. Systemic exposure is highly variable in adults, yet no data exist in children. The purpose of this study was to evaluate NTX exposure in adolescents with eating disorders. Adolescents aged 12-21 years with eating disorders underwent postdose blood sampling in the fasted and/or fed state. NTX and primary active metabolite, 6-β-naltrexol, were determined by ultra-high performance liquid chromatography tandem mass spectrometry. Pharmacokinetic parameters were determined by noncompartmental analysis. DNA was genotyped for AKR1C4 missense mutations associated with decreased activity (rs3829125 and rs17134592). Linear mixed effects modeling was performed. In 21 participants, aged 16.9 ± 1.9 years (15-21 years), 81% female participants, maximum concentration (Cmax ) was 90.4 ± 129 nM/mg/kg, area under the concentration-time curve from zero to infinity (AUC0-∞ ) was 166 ± 154 nM h/mg/kg, and varied 63-fold and 21-fold, respectively. Compared with wildtype, those with AKR1C4 allelic variations (n = 7) displayed 3.2-fold higher AUC0-∞ , four-fold higher Cmax and delayed time to Tmax . Linear mixed effects modeling demonstrated a large effect of genotype on AUC0-∞ (Cohen's d -2.3) and Cmax (Cohen's d -1.4). Food effect was large for AUC0-∞ (Cohen's d 2.6), but highly variable and failed to reach significance for Cmax. The respective model accounted for 82% of the variance in NTX AUC0-∞ and 46% of the variance in Cmax . NTX systemic exposure is highly variable in adolescents with eating disorders and modulated, in part, by AKR1C4 genotype and food intake. These findings may, in part, explain the large degree of interindividual variability observed response to NTX.
Collapse
Affiliation(s)
- Stephani L. Stancil
- Division of Adolescent MedicineChildren's Mercy Hospital – Kansas CityKansas CityMissouriUSA
- Division of Clinical Pharmacology, Toxicology and Therapeutic InnovationChildren's Mercy Hospital – Kansas CityKansas CityMissouriUSA
- Department of PediatricsUniversity of Missouri‐Kansas City School of MedicineKansas CityMissouriUSA
| | - Michaela Voss
- Division of Adolescent MedicineChildren's Mercy Hospital – Kansas CityKansas CityMissouriUSA
- Department of PediatricsUniversity of Missouri‐Kansas City School of MedicineKansas CityMissouriUSA
| | - Whitney Nolte
- Division of Clinical Pharmacology, Toxicology and Therapeutic InnovationChildren's Mercy Hospital – Kansas CityKansas CityMissouriUSA
- Department of PediatricsUniversity of Missouri‐Kansas City School of MedicineKansas CityMissouriUSA
| | - John Tumberger
- Division of Adolescent MedicineChildren's Mercy Hospital – Kansas CityKansas CityMissouriUSA
| | - William Adelman
- Department of Student Health and Counseling, Division of WellnessThe University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Susan Abdel‐Rahman
- Division of Clinical Pharmacology, Toxicology and Therapeutic InnovationChildren's Mercy Hospital – Kansas CityKansas CityMissouriUSA
- Department of PediatricsUniversity of Missouri‐Kansas City School of MedicineKansas CityMissouriUSA
| |
Collapse
|
59
|
Park J, Paudel SB, Jin CH, Lee G, Choi HI, Ryoo GH, Kil YS, Nam JW, Jung CH, Kim BR, Na MK, Han AR. Comparative Analysis of Coumarin Profiles in Different Parts of Peucedanum japonicum and Their Aldo-Keto Reductase Inhibitory Activities. Molecules 2022; 27:7391. [PMID: 36364218 PMCID: PMC9657185 DOI: 10.3390/molecules27217391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 03/13/2024] Open
Abstract
Peucedanum japonicum (Umbelliferae) is widely distributed throughout Southeast Asian countries. The root of this plant is used in traditional medicine to treat colds and pain, whereas the young leaves are considered an edible vegetable. In this study, the differences in coumarin profiles for different parts of P. japonicum including the flowers, roots, leaves, and stems were compared using ultra-performance liquid chromatography time-of-flight mass spectrometry. Twenty-eight compounds were tentatively identified, including three compounds found in the genus Peucedanum for the first time. Principal component analysis using the data set of the measured mass values and intensities of the compounds exhibited distinct clustering of the flower, leaf, stem, and root samples. In addition, their anticancer activities were screened using an Aldo-keto reductase (AKR)1C1 assay on A549 human non-small-cell lung cancer cells and the flower extract inhibited AKR1C1 activity. Based on these results, seven compounds were selected as potential markers to distinguish between the flower part versus the root, stem, and leaf parts using an orthogonal partial least-squares discriminant analysis. This study is the first to provide information on the comparison of coumarin profiles from different parts of P. japonicum as well as their AKR1C1 inhibitory activities. Taken together, the flowers of P. japonicum offer a new use related to the efficacy of overcoming anticancer drug resistance, and may be a promising source for the isolation of active lead compounds.
Collapse
Affiliation(s)
- Jisu Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeollabuk-do, Jeongeup-si 56212, Korea
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea
| | - Sunil Babu Paudel
- College of Pharmacy, Yeungnam University, Gyeongsangbuk-do, Gyeongsan-si 38541, Korea
| | - Chang Hyun Jin
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeollabuk-do, Jeongeup-si 56212, Korea
| | - Gileung Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeollabuk-do, Jeongeup-si 56212, Korea
| | - Hong-Il Choi
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeollabuk-do, Jeongeup-si 56212, Korea
| | - Ga-Hee Ryoo
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeollabuk-do, Jeongeup-si 56212, Korea
| | - Yun-Seo Kil
- College of Pharmacy, Yeungnam University, Gyeongsangbuk-do, Gyeongsan-si 38541, Korea
| | - Joo-Won Nam
- College of Pharmacy, Yeungnam University, Gyeongsangbuk-do, Gyeongsan-si 38541, Korea
| | - Chan-Hun Jung
- Jeonju AgroBio-Materials Institute, Jeollabuk-do, Jeonju-si 54810, Korea
| | - Bo-Ram Kim
- Natural Product Research Division, Honam National Institute of Biological Resources, Jeollanam-do, Mokpo-si 58762, Korea
| | - Min Kyun Na
- College of Pharmacy, Chungnam National University, Daejeon 34134, Korea
| | - Ah-Reum Han
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeollabuk-do, Jeongeup-si 56212, Korea
| |
Collapse
|
60
|
Soranzo T, Ben Tahar A, Chmayssem A, Zelsmann M, Vadgama P, Lenormand JL, Cinquin P, K. Martin D, Zebda A. Electrochemical Biosensing of Glucose Based on the Enzymatic Reduction of Glucose. SENSORS (BASEL, SWITZERLAND) 2022; 22:s22197105. [PMID: 36236202 PMCID: PMC9572614 DOI: 10.3390/s22197105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 06/12/2023]
Abstract
In this work, the enzyme aldehyde reductase, also known as aldose reductase, was synthesized and cloned from a human gene. Spectrophotometric measurements show that in presence of the nicotinamide adenine dinucleotide phosphate cofactor (NADPH), the aldehyde reductase catalyzed the reduction of glucose to sorbitol. Electrochemical measurements performed on an electrodeposited poly(methylene green)-modified gold electrode showed that in the presence of the enzyme aldehyde reductase, the electrocatalytic oxidation current of NADPH decreased drastically after the addition of glucose. These results demonstrate that aldehyde reductase is an enzyme that allows the construction of an efficient electrochemical glucose biosensor based on glucose reduction.
Collapse
Affiliation(s)
- Thomas Soranzo
- Univ. Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France
| | - Awatef Ben Tahar
- Univ. Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France
| | - Ayman Chmayssem
- Univ. Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France
| | - Marc Zelsmann
- Univ. Grenoble Alpes, CNRS, CEA-LETI, Grenoble INP, LTM, F-38054 Grenoble, France
| | - Pankaj Vadgama
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Jean-Luc Lenormand
- Univ. Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France
| | - Phillipe Cinquin
- Univ. Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France
| | - Donald K. Martin
- Univ. Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France
| | - Abdelkader Zebda
- Univ. Grenoble Alpes, TIMC-IMAG/CNRS/INSERM, UMR 5525, F-38000 Grenoble, France
| |
Collapse
|
61
|
Zuo X, Zeng H, Wang B, Yang X, He D, Wang L, Ouyang H, Yuan J. AKR1C1 Protects Corneal Epithelial Cells Against Oxidative Stress-Mediated Ferroptosis in Dry Eye. Invest Ophthalmol Vis Sci 2022; 63:3. [PMID: 36066316 PMCID: PMC9463717 DOI: 10.1167/iovs.63.10.3] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To evaluate the precise mode of cell death and to investigate the molecular mechanism underlying the initiation of inflammation in dry eye disease (DED). Methods C57BL/6 mice were injected with scopolamine subcutaneously and exposed to desiccating stress to establish a DED mouse model. An immortalized human corneal epithelial cell line (HCEC) was cultured under hyperosmolarity (500 mOsM). Protein expressions were measured using western blot assay and immunofluorescence staining. mRNA expression was analyzed by RNA-sequencing and quantitative RT-PCR. Transmission electron microscopy was used to observe the intracellular ultrastructure. Intracellular Fe2+ was detected by a FerroOrange fluorescent probe. Flow cytometry was used to evaluate the cellular reactive oxygen species and lipid peroxidation. Results Marked changes in ferroptosis-related markers expression, intracellular iron accumulation, and lipid peroxidation were observed in corneal epithelial cells of DED models. When excessive oxidative stress was suppressed, ferroptosis induced by hyperosmolarity in HCECs was restrained, as indicated by decreased iron content and lipid peroxidation levels. Moreover, AKR1C1 was upregulated by the activation of NRF2 in HCECs under hyperosmolarity. When AKR1C1 was knocked down, cell viability was decreased, accompanied by increased lipid peroxidation, whereas overexpression of AKR1C1 produced the opposite results. It was observed consistently that corneal defects and the inflammatory response were promoted after inhibition of AKR1C1 in vivo. Conclusions Excessive oxidative stress-induced ferroptosis participates in DED pathogenesis. The expression of AKR1C1 is triggered by NRF2 to decrease ferroptosis-induced cell damage and inflammation in HCECs. These findings may provide potential makers targeting ferroptosis and AKR1C1 for DED therapy.
Collapse
Affiliation(s)
- Xin Zuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Hao Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Bowen Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xue Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Dalian He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Li Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Hong Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
62
|
Vargas JA, Leonardo DA, D’Muniz Pereira H, Lopes AR, Rodriguez HN, Cobos M, Marapara JL, Castro JC, Garratt RC. Structural Characterization of L-Galactose Dehydrogenase: An Essential Enzyme for Vitamin C Biosynthesis. PLANT & CELL PHYSIOLOGY 2022; 63:1140-1155. [PMID: 35765894 PMCID: PMC9381564 DOI: 10.1093/pcp/pcac090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
In plants, it is well-known that ascorbic acid (vitamin C) can be synthesized via multiple metabolic pathways but there is still much to be learned concerning their integration and control mechanisms. Furthermore, the structural biology of the component enzymes has been poorly exploited. Here we describe the first crystal structure for an L-galactose dehydrogenase [Spinacia oleracea GDH (SoGDH) from spinach], from the D-mannose/L-galactose (Smirnoff-Wheeler) pathway which converts L-galactose into L-galactono-1,4-lactone. The kinetic parameters for the enzyme are similar to those from its homolog from camu camu, a super-accumulator of vitamin C found in the Peruvian Amazon. Both enzymes are monomers in solution and have a pH optimum of 7, and their activity is largely unaffected by high concentrations of ascorbic acid, suggesting the absence of a feedback mechanism acting via GDH. Previous reports may have been influenced by changes of the pH of the reaction medium as a function of ascorbic acid concentration. The structure of SoGDH is dominated by a (β/α)8 barrel closely related to aldehyde-keto reductases (AKRs). The structure bound to NAD+ shows that the lack of Arg279 justifies its preference for NAD+ over NADP+, as employed by many AKRs. This favors the oxidation reaction that ultimately leads to ascorbic acid accumulation. When compared with other AKRs, residue substitutions at the C-terminal end of the barrel (Tyr185, Tyr61, Ser59 and Asp128) can be identified to be likely determinants of substrate specificity. The present work contributes toward a more comprehensive understanding of structure-function relationships in the enzymes involved in vitamin C synthesis.
Collapse
Affiliation(s)
| | | | - Humberto D’Muniz Pereira
- São Carlos Institute of Physics, University of São Paulo, Avenida João Dagnone 1100, São Carlos, SP 13563-120, Brazil
| | - Adriana R Lopes
- Laboratory of Biochemistry, Instituto Butantan, Av. Vital Brasil, São Paulo 1500, Brazil
| | - Hicler N Rodriguez
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru
| | - Marianela Cobos
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru
- Laboratorio de Biotecnología y Bioenergética, Universidad Científica del Perú, Av. Abelardo Quiñones km 2.5, Iquitos 16006, Peru
- Departamento Académico de Ciencias Biomédicas y Biotecnología (DACBB), Facultad de Ciencias Biológicas (FCB), Universidad Nacional de la Amazonia Peruana (UNAP), Ciudad Universitaria - Zungarococha, San Juan Bautista 16000, Peru
| | - Jorge L Marapara
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru
- Departamento Académico de Ciencias Biomédicas y Biotecnología (DACBB), Facultad de Ciencias Biológicas (FCB), Universidad Nacional de la Amazonia Peruana (UNAP), Ciudad Universitaria - Zungarococha, San Juan Bautista 16000, Peru
| | - Juan C Castro
- Unidad Especializada del Laboratorio de Investigación en Biotecnología (UELIB), Centro de Investigaciones de Recursos Naturales de la UNAP (CIRNA), Universidad Nacional de la Amazonia Peruana (UNAP), Psje. Los Paujiles S/N, Iquitos 1600, Peru
- Departamento Académico de Ciencias Biomédicas y Biotecnología (DACBB), Facultad de Ciencias Biológicas (FCB), Universidad Nacional de la Amazonia Peruana (UNAP), Ciudad Universitaria - Zungarococha, San Juan Bautista 16000, Peru
| | | |
Collapse
|
63
|
Morell A, Budagaga Y, Vagiannis D, Zhang Y, Laštovičková L, Novotná E, Haddad A, Haddad M, Portillo R, Hofman J, Wsól V. Isocitrate dehydrogenase 2 inhibitor enasidenib synergizes daunorubicin cytotoxicity by targeting aldo-keto reductase 1C3 and ATP-binding cassette transporters. Arch Toxicol 2022; 96:3265-3277. [PMID: 35972551 DOI: 10.1007/s00204-022-03359-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/03/2022] [Indexed: 12/01/2022]
Abstract
Targeting mutations that trigger acute myeloid leukaemia (AML) has emerged as a refined therapeutic approach in recent years. Enasidenib (Idhifa) is the first selective inhibitor of mutated forms of isocitrate dehydrogenase 2 (IDH2) approved against relapsed/refractory AML. In addition to its use as monotherapy, a combination trial of enasidenib with standard intensive induction therapy (daunorubicin + cytarabine) is being evaluated. This study aimed to decipher enasidenib off-target molecular mechanisms involved in anthracycline resistance, such as reduction by carbonyl reducing enzymes (CREs) and drug efflux by ATP-binding cassette (ABC) transporters. We analysed the effect of enasidenib on daunorubicin (Daun) reduction by several recombinant CREs and different human cell lines expressing aldo-keto reductase 1C3 (AKR1C3) exogenously (HCT116) or endogenously (A549 and KG1a). Additionally, A431 cell models overexpressing ABCB1, ABCG2, or ABCC1 were employed to evaluate enasidenib modulation of Daun efflux. Furthermore, the potential synergism of enasidenib over Daun cytotoxicity was quantified amongst all the cell models. Enasidenib selectively inhibited AKR1C3-mediated inactivation of Daun in vitro and in cell lines expressing AKR1C3, as well as its extrusion by ABCB1, ABCG2, and ABCC1 transporters, thus synergizing Daun cytotoxicity to overcome resistance. This work provides in vitro evidence on enasidenib-mediated targeting of the anthracycline resistance actors AKR1C3 and ABC transporters under clinically achievable concentrations. Our findings may encourage its combination with intensive chemotherapy and even suggest that the effectiveness of enasidenib as monotherapy against AML could lie beyond the targeting of mIDH2.
Collapse
Affiliation(s)
- Anselm Morell
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Youssif Budagaga
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Dimitrios Vagiannis
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Yu Zhang
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Lenka Laštovičková
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Eva Novotná
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Andrew Haddad
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Melodie Haddad
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Ramon Portillo
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Jakub Hofman
- Department of Pharmacology, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic
| | - Vladimír Wsól
- Department of Biochemical Sciences, Charles University, Faculty of Pharmacy, Akademika Heyrovskeho 1203, 50005, Hradec Kralove, Czech Republic.
| |
Collapse
|
64
|
Li H, Yang Y, Hu Y, Chen CC, Huang JW, Min J, Dai L, Guo RT. Structural analysis and engineering of aldo-keto reductase from glyphosate-resistant Echinochloa colona. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129191. [PMID: 35739721 DOI: 10.1016/j.jhazmat.2022.129191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
Glyphosate is a dominant organophosphate herbicide that inhibits 5-enolpyruvylshikimate-3-phosphate synthase (EPSPS) of the shikimate pathway. Glyphosate is extensively applied since manufactured, which has led to the emergence of various glyphosate-resistant crops and weeds. However, the molecular mechanism of many glyphosate-resistance machineries remains unclear. Recently, the upregulated expression of two homologous aldo-keto reductases (AKRs), designated as AKR4C16 and AKR4C17, were found to contribute to the glyphosate resistance in Echinochloa colona. This represents the first naturally evolved glyphosate-degrading machinery reported in plants. Here, we report the three-dimensional structure of these two AKR enzymes in complex with cofactor by performing X-ray crystallography. Furthermore, the binding-mode of glyphosate were elucidated in a ternary complex of AKR4C17. Based on the structural information and the previous study, we proposed a possible mechanism of action of AKR-mediated glyphosate degradation. In addition, a variant F291D of AKR4C17 that was constructed based on structure-based engineering showed a 70% increase in glyphosate degradation. In conclusion, these results demonstrate the structural features and glyphosate-binding mode of AKR4C17, which increases our understanding of the enzymatic mechanism of glyphosate bio-degradation and provides an important basis for the designation of AKR-based glyphosate-resistance for further applications.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Yu Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Yumei Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Chun-Chi Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Jian-Wen Huang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Jian Min
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China
| | - Longhai Dai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China.
| | - Rey-Ting Guo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan 430062, PR China.
| |
Collapse
|
65
|
Liu Z, Zhang R, Zhang W, Xu Y. Structure-based rational design of hydroxysteroid dehydrogenases for improving and diversifying steroid synthesis. Crit Rev Biotechnol 2022:1-17. [PMID: 35834355 DOI: 10.1080/07388551.2022.2054770] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
A group of steroidogenic enzymes, hydroxysteroid dehydrogenases are involved in steroid metabolism which is very important in the cell: signaling, growth, reproduction, and energy homeostasis. The enzymes show an inherent function in the interconversion of ketosteroids and hydroxysteroids in a position- and stereospecific manner on the steroid nucleus and side-chains. However, the biocatalysis of steroids reaction is a vital and demanding, yet challenging, task to produce the desired enantiopure products with non-natural substrates or non-natural cofactors, and/or in non-physiological conditions. This has driven the use of protein design strategies to improve their inherent biosynthetic efficiency or activate their silent catalytic ability. In this review, the innate features and catalytic characteristics of enzymes based on sequence-structure-function relationships of steroidogenic enzymes are reviewed. Combining structure information and catalytic mechanisms, progress in protein redesign to stimulate potential function, for example, substrate specificity, cofactor dependence, and catalytic stability are discussed.
Collapse
Affiliation(s)
- Zhiyong Liu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
| | - Rongzhen Zhang
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
| | - Wenchi Zhang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yan Xu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key Laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi, China
| |
Collapse
|
66
|
Cinnamamide derivatives with 4-hydroxypiperidine moiety enhance effect of doxorubicin to cancer cells and protect cardiomyocytes against drug-induced toxicity through CBR1 inhibition mechanism. Life Sci 2022; 305:120777. [DOI: 10.1016/j.lfs.2022.120777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/21/2022] [Accepted: 06/29/2022] [Indexed: 12/06/2022]
|
67
|
Endo S, Morikawa Y, Matsunaga T, Hara A, Nishinaka T. Porcine aldo-keto reductase 1C subfamily members AKR1C1 and AKR1C4: Substrate specificity, inhibitor sensitivity and activators. J Steroid Biochem Mol Biol 2022; 221:106113. [PMID: 35398259 DOI: 10.1016/j.jsbmb.2022.106113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 01/13/2023]
Abstract
Most members of the aldo-keto reductase (AKR) 1 C subfamily are hydroxysteroid dehydrogenases (HSDs). Similarly to humans, four genes for AKR1C proteins (AKR1C1-AKR1C4) have been identified in the pig, which is a suitable species for biomedical research model of human diseases and optimal organ donor for xenotransplantation. Previous study suggested that, among the porcine AKR1Cs, AKR1C1 and AKR1C4 play important roles in steroid hormone metabolism in the reproductive tissues; however, their biological functions are still unknown. Herein, we report the biochemical properties of the two recombinant enzymes. Kinetic and product analyses of steroid specificity indicated that AKR1C1 is a multi-specific reductase, which acts as 3α-HSD for 3-keto-5β-dihydro-C19/C21-steroids, 3β-HSD for 3-keto-5α-dihydro-C19-steroids including androstenone, 17β-HSD for 17-keto-C19-steroids including estrone, and 20α-HSD for progesterone, showing Km values of 0.5-11 µM. By contrast, AKR1C4 exhibited only 3α-HSD activity for 3-keto groups of 5α/β-dihydro-C19-steroids, 5β-dihydro-C21-steroids and bile acids (Km: 1.0-1.9 µM). AKR1C1 and AKR1C4 also showed broad substrate specificity for nonsteroidal carbonyl compounds including endogenous 4-oxo-2-nonenal, 4-hydroxy-nonenal, acrolein, isocaproaldehyde, farnesal, isatin and methylglyoxal, of which 4-oxo-2-nonenal was reduced with the lowest Km value of 0.9 µM. Moreover, AKR1C1 had the characteristic of reducing aliphatic ketones and all-trans-retinal. The enzymes were inhibited by flavonoids, synthetic estrogens, nonsteroidal anti-inflammatory drugs, triterpenoids and phenolphthalein, whereas only AKR1C4 was activated by bromosulfophthalein. These results suggest that AKR1C1 and AKR1C4 function as 3α/3β/17β/20α-HSD and 3α-HSD, respectively, in metabolism of steroid hormones and a sex pheromone androstenone, both of which also play roles in metabolism of nonsteroidal carbonyl compounds.
Collapse
Affiliation(s)
- Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan.
| | - Yoshifumi Morikawa
- Forensic Science Laboratory, Gifu Prefectural Police Headquarters, Gifu 500-8501, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| | - Akira Hara
- Faculty of Engineering, Gifu University, Gifu 501-1193, Japan
| | - Toru Nishinaka
- Faculty of Pharmacy, Osaka-Ohtani University, Osaka 584-8540, Japan
| |
Collapse
|
68
|
Yasar Yildiz S, Finore I, Leone L, Romano I, Lama L, Kasavi C, Nicolaus B, Toksoy Oner E, Poli A. Genomic Analysis Provides New Insights Into Biotechnological and Industrial Potential of Parageobacillus thermantarcticus M1. Front Microbiol 2022; 13:923038. [PMID: 35756030 PMCID: PMC9218356 DOI: 10.3389/fmicb.2022.923038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/18/2022] [Indexed: 02/02/2023] Open
Abstract
Parageobacillus thermantarcticus strain M1 is a Gram-positive, motile, facultative anaerobic, spore forming, and thermophilic bacterium, isolated from geothermal soil of the crater of Mount Melbourne (74°22′ S, 164°40′ E) during the Italian Antarctic Expedition occurred in Austral summer 1986–1987. Strain M1 demonstrated great biotechnological and industrial potential owing to its ability to produce exopolysaccharides (EPSs), ethanol and thermostable extracellular enzymes, such as an xylanase and a β-xylosidase, and intracellular ones, such as xylose/glucose isomerase and protease. Furthermore, recent studies revealed its high potential in green chemistry due to its use in residual biomass transformation/valorization and as an appropriate model for microbial astrobiology studies. In the present study, using a systems-based approach, genomic analysis of P. thermantarcticus M1 was carried out to enlighten its functional characteristics. The elucidation of whole-genome organization of this thermophilic cell factory increased our understanding of biological mechanisms and pathways, by providing valuable information on the essential genes related to the biosynthesis of nucleotide sugar precursors, monosaccharide unit assembly, as well as the production of EPSs and ethanol. In addition, gene prediction and genome annotation studies identified genes encoding xylanolytic enzymes that are required for the conversion of lignocellulosic materials to high-value added molecules. Our findings pointed out the significant potential of strain M1 in various biotechnological and industrial applications considering its capacity to produce EPSs, ethanol and thermostable enzymes via the utilization of lignocellulosic waste materials.
Collapse
Affiliation(s)
- Songul Yasar Yildiz
- Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ilaria Finore
- Institute of Biomolecular Chemistry (ICB), National Research Council, Naples, Italy
| | - Luigi Leone
- Institute of Biomolecular Chemistry (ICB), National Research Council, Naples, Italy
| | - Ida Romano
- Institute of Biomolecular Chemistry (ICB), National Research Council, Naples, Italy
| | - Licia Lama
- Institute of Biomolecular Chemistry (ICB), National Research Council, Naples, Italy
| | - Ceyda Kasavi
- Department of Bioengineering, Industrial Biotechnology and Systems Biology (IBSB), Marmara University, Istanbul, Turkey
| | - Barbara Nicolaus
- Institute of Biomolecular Chemistry (ICB), National Research Council, Naples, Italy
| | - Ebru Toksoy Oner
- Department of Bioengineering, Industrial Biotechnology and Systems Biology (IBSB), Marmara University, Istanbul, Turkey
| | - Annarita Poli
- Institute of Biomolecular Chemistry (ICB), National Research Council, Naples, Italy
| |
Collapse
|
69
|
Bolleddula J, Chen H, Cohen L, Zhou X, Pusalkar S, Berger A, Sedarati F, Venkatakrishnan K, Chowdhury SK. Metabolism and Disposition of [14C]Pevonedistat, a First-In-Class NEDD8 ‑Activating Enzyme Inhibitor, After Intravenous Infusion to Patients With Advanced Solid Tumors. Drug Metab Dispos 2022; 50:989-997. [PMID: 35504658 DOI: 10.1124/dmd.122.000842] [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: 01/27/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022] Open
Abstract
Metabolism and disposition of pevonedistat, an investigational, first-in-class inhibitor of the NEDD8-activating enzyme (NAE), were characterized in patients with advanced solid tumors after intravenous infusion of [14C]pevonedistat at 25 mg/m2 (~60-85mCi radioactive dose). More than 94% of the administered dose was recovered, with ~41% and ~53% of drug-related material eliminated in urine and feces, respectively. The metabolite profiles of [14C]pevonedistat were established in plasma using an accelerator mass spectrometer (AMS) and excreta with traditional radiometric analysis. In plasma, unchanged parent drug accounted for approximately 49% of the total drug-related material. Metabolites M1 and M2 were major (>10% of the total drug-related material) circulating metabolites and accounted for approximately 15% and 22% of the drug related material, respectively. Unchanged [14C]pevonedistat accounted for approximately 4% and 17% of the dose in urine and feces, respectively. Oxidative metabolites M1, M2, and M3 appeared as the most abundant drug-related components in the excreta and represented approximately 27%, 26%, and 15% of the administered dose, respectively. Based on the unbound plasma exposure in cancer patients and in vitro NAE inhibition, the contribution of metabolites M1 and M2 to overall in vivo pharmacological activity is anticipated to be minimal. The exposure to these metabolites was higher at safe and well tolerated doses in rat and dog (the two preclinical species used in toxicology evaluation) plasma than that observed in human plasma. Reaction phenotyping studies revealed that CYP3A4/5 are primary enzymes responsible for the metabolic clearance of pevonedistat. Significance Statement This study details the metabolism and clearance mechanisms of pevonedistat, a first-in-class NEDD8‑activating enzyme inhibitor, after intravenous administration to patients with cancer. Pevonedistat is biotransformed to 2 major circulating metabolites with higher exposure in non-clinical toxicological species than in humans. The pharmacological activity contribution of these metabolites is minimal compared to the overall target pharmacological effect of pevonedistat. Renal clearance was not an important route of excretion of unchanged pevonedistat (~4% of the dose).
Collapse
Affiliation(s)
| | - Hao Chen
- Takeda Development Center Americas Inc., United States
| | | | - Xiaofei Zhou
- Clinical Pharmacology, Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, United States
| | | | | | | | | | | |
Collapse
|
70
|
Zeng W, Li W, Chen H, Zhou L. Relay Photocatalytic Reaction of N-Aryl Amino Acids and 2-Bromo-3,3,3-trifluoropropene: Synthesis of 4-(Difluoromethylidene)-tetrahydroquinolines. Org Lett 2022; 24:3265-3269. [PMID: 35467357 DOI: 10.1021/acs.orglett.2c01117] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The bulk industrial chemical 2-bromo-3,3,3-trifluoropropene (BTP) was first employed as a coupling partner in photocatalytic defluorinative reactions with N-aryl amino acids. Photoredox activation of the C(sp2)-Br bond of the resultant 2-bromo-1,1-difluoroalkenes generates gem-difluoro vinyl radicals for further radical cyclization. Various 4-(difluoromethylidene)-tetrahydroquinolines were assembled in good yields by combining two photoredox cycles with a single photocatalyst.
Collapse
Affiliation(s)
- Weidi Zeng
- School of Chemistry, Sun Yat-Sen University, Panyu District, Guangzhou 510006, China
| | - Weiyu Li
- School of Chemistry, Sun Yat-Sen University, Panyu District, Guangzhou 510006, China
| | - Haoguo Chen
- School of Chemistry, Sun Yat-Sen University, Panyu District, Guangzhou 510006, China
| | - Lei Zhou
- School of Chemistry, Sun Yat-Sen University, Panyu District, Guangzhou 510006, China
| |
Collapse
|
71
|
Ahvenainen T, Kaukomaa J, Kämpjärvi K, Uimari O, Ahtikoski A, Mäkinen N, Heikinheimo O, Aaltonen LA, Karhu A, Bützow R, Vahteristo P. Comparison of 2SC, AKR1B10, and FH Antibodies as Potential Biomarkers for FH-deficient Uterine Leiomyomas. Am J Surg Pathol 2022; 46:537-546. [PMID: 34678832 DOI: 10.1097/pas.0000000000001826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a tumor predisposition syndrome caused by germline fumarate hydratase (FH) mutations and characterized by uterine and cutaneous leiomyomas and renal cell cancer. Currently, there is no generally approved method to differentiate FH-deficient uterine leiomyomas from other leiomyomas. Here, we analyzed 3 antibodies (S-(2-succino)-cysteine [2SC], aldo-keto reductase family 1, member B10 [AKR1B10], and FH) as potential biomarkers. The study consisted of 2 sample series. The first series included 155 formalin-fixed paraffin-embedded uterine leiomyomas, of which 90 were from HLRCC patients and 65 were sporadic. The second series included 1590 unselected fresh frozen leiomyomas. Twenty-seven tumors were from known HLRCC patients, while the FH status for the remaining 1563 tumors has been determined by copy number analysis and Sanger sequencing revealing 45 tumors with monoallelic (n=33) or biallelic (n=12) FH loss. Altogether 197 samples were included in immunohistochemical analyses: all 155 samples from series 1 and 42 available corresponding formalin-fixed paraffin-embedded samples from series 2 (15 tumors with monoallelic and 7 with biallelic FH loss, 20 with no FH deletion). Results show that 2SC performed best with 100% sensitivity and specificity. Scoring was straightforward with unambiguously positive or negative results. AKR1B10 identified most tumors accurately with 100% sensitivity and 99% specificity. FH was 100% specific but showed slightly reduced 91% sensitivity. Both FH and AKR1B10 displayed also intermediate staining intensities. We suggest that when patient's medical history and/or histopathologic tumor characteristics indicate potential FH-deficiency, the tumor's FH status is determined by 2SC staining. When aberrant staining is observed, the patient can be directed to genetic counseling and mutation screening.
Collapse
Affiliation(s)
- Terhi Ahvenainen
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
- iCAN Digital Precision Cancer Medicine Flagship
| | - Jaana Kaukomaa
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
| | - Kati Kämpjärvi
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
| | - Outi Uimari
- Department of Obstetrics and Gynecology, Oulu University Hospital
- PEDEGO Research Unit, University of Oulu and Oulu University Hospital
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu
| | - Anne Ahtikoski
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu
- Department of Pathology, Oulu University Hospital
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland
| | - Netta Mäkinen
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
| | - Oskari Heikinheimo
- Applied Tumor Genomics Research Program
- Department of Obstetrics and Gynecology, Helsinki University Hospital
| | - Lauri A Aaltonen
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
- iCAN Digital Precision Cancer Medicine Flagship
| | - Auli Karhu
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
- iCAN Digital Precision Cancer Medicine Flagship
| | - Ralf Bützow
- Applied Tumor Genomics Research Program
- Department of Pathology, Laboratory of Helsinki University Hospital (HUSLAB), Helsinki University Hospital and University of Helsinki, Helsinki
| | - Pia Vahteristo
- Applied Tumor Genomics Research Program
- Department of Medical and Clinical Genetics, University of Helsinki
- iCAN Digital Precision Cancer Medicine Flagship
| |
Collapse
|
72
|
Balestri F, Moschini R, Mura U, Cappiello M, Del Corso A. In Search of Differential Inhibitors of Aldose Reductase. Biomolecules 2022; 12:biom12040485. [PMID: 35454074 PMCID: PMC9024650 DOI: 10.3390/biom12040485] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 11/22/2022] Open
Abstract
Aldose reductase, classified within the aldo-keto reductase family as AKR1B1, is an NADPH dependent enzyme that catalyzes the reduction of hydrophilic as well as hydrophobic aldehydes. AKR1B1 is the first enzyme of the so-called polyol pathway that allows the conversion of glucose into sorbitol, which in turn is oxidized to fructose by sorbitol dehydrogenase. The activation of the polyol pathway in hyperglycemic conditions is generally accepted as the event that is responsible for a series of long-term complications of diabetes such as retinopathy, cataract, nephropathy and neuropathy. The role of AKR1B1 in the onset of diabetic complications has made this enzyme the target for the development of molecules capable of inhibiting its activity. Virtually all synthesized compounds have so far failed as drugs for the treatment of diabetic complications. This failure may be partly due to the ability of AKR1B1 to reduce alkenals and alkanals, produced in oxidative stress conditions, thus acting as a detoxifying agent. In recent years we have proposed an alternative approach to the inhibition of AKR1B1, suggesting the possibility of a differential inhibition of the enzyme through molecules able to preferentially inhibit the reduction of either hydrophilic or hydrophobic substrates. The rationale and examples of this new generation of aldose reductase differential inhibitors (ARDIs) are presented.
Collapse
Affiliation(s)
- Francesco Balestri
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56127 Pisa, Italy; (F.B.); (R.M.); (U.M.); (A.D.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
| | - Roberta Moschini
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56127 Pisa, Italy; (F.B.); (R.M.); (U.M.); (A.D.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
| | - Umberto Mura
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56127 Pisa, Italy; (F.B.); (R.M.); (U.M.); (A.D.C.)
| | - Mario Cappiello
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56127 Pisa, Italy; (F.B.); (R.M.); (U.M.); (A.D.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
- Correspondence:
| | - Antonella Del Corso
- Biochemistry Unit, Department of Biology, University of Pisa, Via S. Zeno 51, 56127 Pisa, Italy; (F.B.); (R.M.); (U.M.); (A.D.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
| |
Collapse
|
73
|
The metabolism and biotransformation of AFB 1: Key enzymes and pathways. Biochem Pharmacol 2022; 199:115005. [PMID: 35318037 DOI: 10.1016/j.bcp.2022.115005] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 02/05/2023]
Abstract
Aflatoxins B1 (AFB1) is a hepatoxic compound produced by Aspergillus flavus and Aspergillus parasiticus, seriously threatening food safety and the health of humans and animals. Understanding the metabolism of AFB1 is important for developing detoxification and intervention strategies. In this review, we summarize the AFB1 metabolic fates in humans and animals and the key enzymes that metabolize AFB1, including cytochrome P450s (CYP450s) for AFB1 bioactivation, glutathione-S-transferases (GSTs) and aflatoxin-aldehyde reductases (AFARs) in detoxification. Furthermore, AFB1 metabolism in microbes is also summarized. Microorganisms specifically and efficiently transform AFB1 into less or non-toxic products in an environmental-friendly approach which could be the most desirable detoxification strategy in the future. This review provides a wholistic insight into the metabolism and biotransformation of AFB1 in various organisms, which also benefits the development of protective strategies in humans and animals.
Collapse
|
74
|
Chabert C, Vitte AL, Iuso D, Chuffart F, Trocme C, Buisson M, Poignard P, Lardinois B, Debois R, Rousseaux S, Pepin JL, Martinot JB, Khochbin S. AKR1B10, One of the Triggers of Cytokine Storm in SARS-CoV2 Severe Acute Respiratory Syndrome. Int J Mol Sci 2022; 23:ijms23031911. [PMID: 35163833 PMCID: PMC8836815 DOI: 10.3390/ijms23031911] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 02/07/2023] Open
Abstract
Preventing the cytokine storm observed in COVID-19 is a crucial goal for reducing the occurrence of severe acute respiratory failure and improving outcomes. Here, we identify Aldo-Keto Reductase 1B10 (AKR1B10) as a key enzyme involved in the expression of pro-inflammatory cytokines. The analysis of transcriptomic data from lung samples of patients who died from COVID-19 demonstrates an increased expression of the gene encoding AKR1B10. Measurements of the AKR1B10 protein in sera from hospitalised COVID-19 patients suggests a significant link between AKR1B10 levels and the severity of the disease. In macrophages and lung cells, the over-expression of AKR1B10 induces the expression of the pro-inflammatory cytokines Interleukin-6 (IL-6), Interleukin-1β (IL-1β) and Tumor Necrosis Factor a (TNFα), supporting the biological plausibility of an AKR1B10 involvement in the COVID-19-related cytokine storm. When macrophages were stressed by lipopolysaccharides (LPS) exposure and treated by Zopolrestat, an AKR1B10 inhibitor, the LPS-induced production of IL-6, IL-1β, and TNFα is significantly reduced, reinforcing the hypothesis that the pro-inflammatory expression of cytokines is AKR1B10-dependant. Finally, we also show that AKR1B10 can be secreted and transferred via extracellular vesicles between different cell types, suggesting that this protein may also contribute to the multi-organ systemic impact of COVID-19. These experiments highlight a relationship between AKR1B10 production and severe forms of COVID-19. Our data indicate that AKR1B10 participates in the activation of cytokines production and suggest that modulation of AKR1B10 activity might be an actionable pharmacological target in COVID-19 management.
Collapse
Affiliation(s)
- Clovis Chabert
- Institute for Advanced Biosciences—UGA—INSERM U1209—CNRS UMR 5309, 38700 La Tronche, France; (A.-L.V.); (D.I.); (F.C.); (S.R.); (S.K.)
- Correspondence: ; Tel.: +33-6-8898-4506
| | - Anne-Laure Vitte
- Institute for Advanced Biosciences—UGA—INSERM U1209—CNRS UMR 5309, 38700 La Tronche, France; (A.-L.V.); (D.I.); (F.C.); (S.R.); (S.K.)
| | - Domenico Iuso
- Institute for Advanced Biosciences—UGA—INSERM U1209—CNRS UMR 5309, 38700 La Tronche, France; (A.-L.V.); (D.I.); (F.C.); (S.R.); (S.K.)
| | - Florent Chuffart
- Institute for Advanced Biosciences—UGA—INSERM U1209—CNRS UMR 5309, 38700 La Tronche, France; (A.-L.V.); (D.I.); (F.C.); (S.R.); (S.K.)
| | - Candice Trocme
- Laboratoire BEP (Biochimie des Enzymes et les Protéines), Institut de Biologie et de Pathologie, CHU Grenoble Alpes, 38700 La Tronche, France;
| | - Marlyse Buisson
- Institut de Biologie Structurale, CEA, CNRS and Centre Hospitalier Universitaire Grenoble Alpes, Université Grenoble Alpes, 38000 Grenoble, France; (M.B.); (P.P.)
| | - Pascal Poignard
- Institut de Biologie Structurale, CEA, CNRS and Centre Hospitalier Universitaire Grenoble Alpes, Université Grenoble Alpes, 38000 Grenoble, France; (M.B.); (P.P.)
| | - Benjamin Lardinois
- Laboratory Department, CHU UCL Namur Site de Ste Elisabeth, 5000 Namur, Belgium; (B.L.); (R.D.)
| | - Régis Debois
- Laboratory Department, CHU UCL Namur Site de Ste Elisabeth, 5000 Namur, Belgium; (B.L.); (R.D.)
| | - Sophie Rousseaux
- Institute for Advanced Biosciences—UGA—INSERM U1209—CNRS UMR 5309, 38700 La Tronche, France; (A.-L.V.); (D.I.); (F.C.); (S.R.); (S.K.)
| | - Jean-Louis Pepin
- HP2 Laboratory, INSERM U1300, Grenoble Alpes University, 38000 Grenoble, France;
- Sleep Laboratory, Pole Thorax et Vaisseaux, Grenoble Alpes University Hospital, 38000 Grenoble, France
| | - Jean-Benoit Martinot
- Sleep Laboratory and Pulmonology and Allergy Department—CHU UCL Namur, St. Elisabeth Site, 5000 Namur, Belgium;
- Institute of Experimental and Clinical Research, UCL Bruxelles Woluwe, 1200 Brussels, Belgium
| | - Saadi Khochbin
- Institute for Advanced Biosciences—UGA—INSERM U1209—CNRS UMR 5309, 38700 La Tronche, France; (A.-L.V.); (D.I.); (F.C.); (S.R.); (S.K.)
| |
Collapse
|
75
|
Aldehyde Dehydrogenase 2 Family Member (ALDH2) Is a Therapeutic Index for Oxaliplatin Response on Colorectal Cancer Therapy with Dysfunction p53. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1322788. [PMID: 35178443 PMCID: PMC8844434 DOI: 10.1155/2022/1322788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
Oxaliplatin resistance is a major issue in the treatment of p53 mutant colorectal cancer (CRC). Finding the specific biomarkers would improve therapeutic efficacy of patients with CRC. In order to figure out the biomarker for CRC patients with mutant p53 access oxaliplatin, a Gene Expression Omnibus dataset (GSE42387) was used to determine differentially expressed genes (DEGs). The Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape software were used to predict protein-protein interactions. The Database for Annotation, Visualization, and Integrated Discovery online tool was used to group the DEGs into their common pathways. 138 DEGs were identified with 46 upregulated and 92 downregulated. In the PPI networks, 7 of the upregulated genes and 13 of the downregulated genes were identified as hub genes (high degrees). Four hub genes, aldehyde dehydrogenase 2 family member (ALDH2), aldo-keto reductase family 1 member B1 (AKR1B1), aldo-keto reductase family 1 member B10 (AKR1B10), and monoglyceride lipase (MGLL) were enriched in the most significant pathway, glycerolipid metabolism. Further, we found that low expression of ALDH2 is correlated with poor overall survival and oxaliplatin resistance. Finally, we found that combined treatment with ALDH2 inhibitor and oxaliplatin will reduce the sensitivity to oxaliplatin in p53 mutant HT29 cells. In conclusion, we demonstrate that ALDH2 may be a biomarker for oxaliplatin resistance status in CRC patients and bring new insight into treatment strategy for p53 mutant CRC patients.
Collapse
|
76
|
Stancil SL, Nolte W, Pearce RE, Staggs VS, Leeder JS. The Impact of Age and Genetics on Naltrexone Biotransformation. Drug Metab Dispos 2022; 50:168-173. [PMID: 34728519 PMCID: PMC9621334 DOI: 10.1124/dmd.121.000646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/01/2021] [Indexed: 02/03/2023] Open
Abstract
Naltrexone, an opioid antagonist primarily metabolized by aldo-keto reductase 1C4 (AKR1C4), treats pediatric conditions involving compulsiveness (e.g., autism spectrum, Prader-Willi, eating disorders, non-suicidal self-injury). Pharmacokinetic variability is apparent in adults, yet no data are available for children. This study aimed to examine the impact of age and genetic variation on naltrexone biotransformation. Human liver cytosol (HLC) samples (n = 158) isolated from children and adult organ donors were incubated with therapeutically relevant concentrations of naltrexone (0.1, 1 µM). Naltrexone biotransformation was determined by ultraperformance mass spectrometry quantification of the primary metabolite, 6-beta-naltrexol (6βN), and 6βN formation rates (pmol/mg protein/min) were calculated. HLCs from organ donors, age range 0-79 y (mean 16.0 ± 18.2 y), 37% (n = 60) female, 20% (n = 33) heterozygous and 1.2% (n = 2) homozygous for co-occurring AKR1C4 variants (S145C/L311V) showed >200-fold range in 6βN formation (0.37-76.5 pmol/mg protein/min). Source of donor samples was found to be a substantial contributor to variability. Model estimates for a trimmed data set of source-adjusted pediatric samples (aged 0-18 y) suggested that AKR1C4 genetic variation, age, and sex explained 36% of the variability in 6βN formation. Although activity increased steadily from birth and peaked in middle childhood (2-5 years), genetic variation (S145C/L311V) demonstrated a greater effect on activity than did age. Naltrexone biotransformation is highly variable in pediatric and adult livers and can be partly accounted for by individual factors feasible to obtain (e.g., genetic variability, age, sex). These data may inform a precision therapeutics approach (e.g., exposure optimization) to further study Naltrexone responsiveness in children and adults. SIGNIFICANCE STATEMENT: Biotransformation of the commonly used opioid antagonist naltrexone is highly variable and may contribute to reduced therapeutic response. Age, sex, and genetic variation in the drug-metabolizing enzyme, AKR1C4, are potential factors contributing to this variability. In pediatric samples, genetic variation (S145C/L311V) demonstrates a greater impact on activity than age. Additionally, the source of donor samples was identified as an important contributor and must be accounted for to confidently elucidate the biological variables most impactful to drug biotransformation.
Collapse
Affiliation(s)
- Stephani L Stancil
- Division of Adolescent Medicine (S.L.S.), Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation (S.L.S., W.N., R.E.P., J.S.L.), and Biostatistics and Epidemiology Core (V.S.S.) Children's Mercy Kansas City, Kansas City, Missouri; Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri (S.L.S., W.N., R.E.P., V.S.S., J.S.L.)
| | - Whitney Nolte
- Division of Adolescent Medicine (S.L.S.), Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation (S.L.S., W.N., R.E.P., J.S.L.), and Biostatistics and Epidemiology Core (V.S.S.) Children's Mercy Kansas City, Kansas City, Missouri; Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri (S.L.S., W.N., R.E.P., V.S.S., J.S.L.)
| | - Robin E Pearce
- Division of Adolescent Medicine (S.L.S.), Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation (S.L.S., W.N., R.E.P., J.S.L.), and Biostatistics and Epidemiology Core (V.S.S.) Children's Mercy Kansas City, Kansas City, Missouri; Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri (S.L.S., W.N., R.E.P., V.S.S., J.S.L.)
| | - Vincent S Staggs
- Division of Adolescent Medicine (S.L.S.), Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation (S.L.S., W.N., R.E.P., J.S.L.), and Biostatistics and Epidemiology Core (V.S.S.) Children's Mercy Kansas City, Kansas City, Missouri; Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri (S.L.S., W.N., R.E.P., V.S.S., J.S.L.)
| | - J Steven Leeder
- Division of Adolescent Medicine (S.L.S.), Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation (S.L.S., W.N., R.E.P., J.S.L.), and Biostatistics and Epidemiology Core (V.S.S.) Children's Mercy Kansas City, Kansas City, Missouri; Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri (S.L.S., W.N., R.E.P., V.S.S., J.S.L.)
| |
Collapse
|
77
|
Sinha J, Karatza E, Gonzalez D. Physiologically-based pharmacokinetic modeling of oxcarbazepine and levetiracetam during adjunctive antiepileptic therapy in children and adolescents. CPT Pharmacometrics Syst Pharmacol 2022; 11:225-239. [PMID: 34816634 PMCID: PMC8846633 DOI: 10.1002/psp4.12750] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
Oxcarbazepine (OXZ) and levetiracetam (LEV) are two new generation anti‐epileptic drugs, often co‐administered in children with enzyme‐inducing antiepileptic drugs (EIAEDs). The anti‐epileptic effect of OXZ and LEV are linked to the exposure of OXZ’s active metabolite 10‐monohydroxy derivative (MHD) and (the parent) LEV, respectively. However, little is known about the confounding effect of age and EIAEDs on the pharmacokinetics (PKs) of MHD and LEV. To address this knowledge gap, physiologically‐based pharmacokinetic (PBPK) modeling was performed in the PK‐Sim software using literature data from children greater than or equal to 2 years of age. Age‐related changes in clearance (CL) of MHD and LEV were characterized, both in the presence (group 1) and absence (group 2) of concomitant EIAEDs. The drug‐drug interaction effect of EIAEDs was estimated as the difference in CL estimates between groups 1 and 2. PBPK modeling suggests that bodyweight normalized CL (ml/min/kg) is higher in younger children than their older counterparts (i.e., due to an influence of age). Concomitant EIAEDs further increase MHD’s CL to a fixed extent of 25% at any age, but EIAEDs’ effect on LEV’s CL increases with age from 20% (at 2 years) to 30% (at adolescence). Simulations with the maximum recommended doses (MRDs) revealed that children between 2 and 4 years and greater than 4 years, who are not on EIAEDs, are at risk of exceeding the reference exposure range for OXZ and LEV, respectively. This analysis demonstrates the use of PBPK modeling in understanding the confounding effect of age and comedications on PKs in children and adolescents.
Collapse
Affiliation(s)
- Jaydeep Sinha
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Department of Pediatrics, UNC School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eleni Karatza
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
78
|
Ito Y, Takasawa A, Takasawa K, Murakami T, Akimoto T, Kyuno D, Kawata Y, Shano K, Kirisawa K, Ota M, Aoyama T, Murata M, Sugimoto K, Chiba H, Saito T, Osanai M. Aberrant expression of claudin-6 contributes to malignant potentials and drug resistance of cervical adenocarcinoma. Cancer Sci 2022; 113:1519-1530. [PMID: 35100472 PMCID: PMC8990859 DOI: 10.1111/cas.15284] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/09/2022] [Accepted: 01/24/2022] [Indexed: 11/30/2022] Open
Abstract
Recent studies have revealed that aberrant expression of tight junction (TJ) proteins is a hallmark of various solid tumors and it is recognized as a useful therapeutic target. Claudin‐6 (CLDN6), a member of the family of TJ transmembrane proteins, is an ideal therapeutic target because it is not expressed in human adult normal tissues. In this study, we found that CLDN6 is highly expressed in uterine cervical adenocarcinoma (ADC) and that high CLDN6 expression was correlated with lymph node metastasis and lymphovascular infiltration and was an independent prognostic factor. Shotgun proteome analysis revealed that cell‐cell adhesion‐related proteins and drug metabolism‐associated proteins (aldo‐keto reductase [AKR] family proteins) were significantly increased in CLDN6‐overexpressing cells. Furthermore, overexpression of CLDN6 enhanced cell‐cell adhesion properties and attenuated sensitivity to anticancer drugs including doxorubicin, daunorubicin, and cisplatin. Taken together, the results indicate that aberrant expression of CLDN6 enhances malignant potentials and drug resistance of cervical ADC, possibly due to increased cell‐cell adhesion properties and drug metabolism. Our findings provide an insight into a new therapeutic strategy, a CLDN6‐targeting therapy, against cervical ADC.
Collapse
Affiliation(s)
- Yui Ito
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Akira Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Kumi Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Taro Murakami
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Taishi Akimoto
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Daisuke Kyuno
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Yuka Kawata
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Kodai Shano
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Kurara Kirisawa
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Misaki Ota
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Tomoyuki Aoyama
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Masaki Murata
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Kotaro Sugimoto
- Department of Basic Pathology, Graduate School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Hideki Chiba
- Department of Basic Pathology, Graduate School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Makoto Osanai
- Department of Pathology, Sapporo Medical University School of Medicine, S1 W17, Chuo-ku, Sapporo, 060-8556, Japan
| |
Collapse
|
79
|
Mateo-Otero Y, Ribas-Maynou J, Delgado-Bermúdez A, Llavanera M, Recuero S, Barranco I, Yeste M. Aldose Reductase B1 in Pig Sperm Is Related to Their Function and Fertilizing Ability. Front Endocrinol (Lausanne) 2022; 13:773249. [PMID: 35173684 PMCID: PMC8842650 DOI: 10.3389/fendo.2022.773249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/03/2022] [Indexed: 11/30/2022] Open
Abstract
Aldose reductase B1 (AKR1B1) has been reported to participate in the modulation of male and female reproductive physiology in several mammalian species. In spite of this, whether or not AKR1B1 could be related to sperm quality, functionality and fertilizing ability is yet to be elucidated. The present study, therefore, aimed to investigate: i) the presence of AKR1B1 in epididymal and ejaculated sperm; ii) the relationship between the AKR1B1 present in sperm and the physiology of the male gamete; iii) the liaison between the relative content of AKR1B1 in sperm and their ability to withstand preservation for 72 h; and iv) the potential link between sperm AKR1B1 and in vitro fertility outcomes. Immunoblotting revealed that AKR1B1 is present in both epididymal and ejaculated sperm with a similar relative content. Moreover, the relative levels of AKR1B1 in sperm (36 kDa band) were found to be negatively related to several kinematic parameters and intracellular calcium levels, and positively to the percentage of sperm with distal cytoplasmic droplets after storage. Finally, AKR1B1 amounts in sperm (36 kDa band) were negatively associated to fertilization rate at two days post-fertilization and embryo development at six days post-fertilization. The results of the present work suggest that AKR1B1 in sperm is probably acquired during maturation rather than at ejaculation and could play a role in that process. Moreover, AKR1B1 seems to be related to the sperm resilience to preservation and to their fertilizing capacity, as lower levels of the 36 kDa band (putative inactive form of this protein) result in better reproductive outcomes.
Collapse
Affiliation(s)
- Yentel Mateo-Otero
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
| | - Jordi Ribas-Maynou
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
- *Correspondence: Marc Yeste, ; Jordi Ribas-Maynou,
| | - Ariadna Delgado-Bermúdez
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
| | - Marc Llavanera
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
| | - Sandra Recuero
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
| | - Isabel Barranco
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia, Bologna, Italy
| | - Marc Yeste
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
- *Correspondence: Marc Yeste, ; Jordi Ribas-Maynou,
| |
Collapse
|
80
|
Perspective on the Structural Basis for Human Aldo-Keto Reductase 1B10 Inhibition. Metabolites 2021; 11:metabo11120865. [PMID: 34940623 PMCID: PMC8708191 DOI: 10.3390/metabo11120865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022] Open
Abstract
Human aldo-keto reductase 1B10 (AKR1B10) is overexpressed in many cancer types and is involved in chemoresistance. This makes AKR1B10 to be an interesting drug target and thus many enzyme inhibitors have been investigated. High-resolution crystallographic structures of AKR1B10 with various reversible inhibitors were deeply analyzed and compared to those of analogous complexes with aldose reductase (AR). In both enzymes, the active site included an anion-binding pocket and, in some cases, inhibitor binding caused the opening of a transient specificity pocket. Different structural conformers were revealed upon inhibitor binding, emphasizing the importance of the highly variable loops, which participate in the transient opening of additional binding subpockets. Two key differences between AKR1B10 and AR were observed regarding the role of external loops in inhibitor binding. The first corresponded to the alternative conformation of Trp112 (Trp111 in AR). The second difference dealt with loop A mobility, which defined a larger and more loosely packed subpocket in AKR1B10. From this analysis, the general features that a selective AKR1B10 inhibitor should comply with are the following: an anchoring moiety to the anion-binding pocket, keeping Trp112 in its native conformation (AKR1B10-like), and not opening the specificity pocket in AR.
Collapse
|
81
|
Concepcion JCT, Kaundun SS, Morris JA, Hutchings S, Strom SA, Lygin AV, Riechers DE. Resistance to a nonselective 4-hydroxyphenylpyruvate dioxygenase-inhibiting herbicide via novel reduction-dehydration-glutathione conjugation in Amaranthus tuberculatus. THE NEW PHYTOLOGIST 2021; 232:2089-2105. [PMID: 34480751 PMCID: PMC9292532 DOI: 10.1111/nph.17708] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/25/2021] [Indexed: 05/06/2023]
Abstract
Metabolic resistance to 4-hydroxyphenylpyruvate dioxygenase (HPPD)-inhibiting herbicides is a threat in controlling waterhemp (Amaranthus tuberculatus) in the USA. We investigated resistance mechanisms to syncarpic acid-3 (SA3), a nonselective, noncommercial HPPD-inhibiting herbicide metabolically robust to Phase I oxidation, in multiple-herbicide-resistant (MHR) waterhemp populations (SIR and NEB) and HPPD inhibitor-sensitive populations (ACR and SEN). Dose-response experiments with SA3 provided ED50 -based resistant : sensitive ratios of at least 18-fold. Metabolism experiments quantifying parent SA3 remaining in excised leaves during a time course indicated MHR populations displayed faster rates of SA3 metabolism compared to HPPD inhibitor-sensitive populations. SA3 metabolites were identified via LC-MS-based untargeted metabolomics in whole plants. A Phase I metabolite, likely generated by cytochrome P450-mediated alkyl hydroxylation, was detected but was not associated with resistance. A Phase I metabolite consistent with ketone reduction followed by water elimination was detected, creating a putative α,β-unsaturated carbonyl resembling a Michael acceptor site. A Phase II glutathione-SA3 conjugate was associated with resistance. Our results revealed a novel reduction-dehydration-GSH conjugation detoxification mechanism. SA3 metabolism in MHR waterhemp is thus atypical compared to commercial HPPD-inhibiting herbicides. This previously uncharacterized detoxification mechanism presents a unique opportunity for future biorational design by blocking known sites of herbicide metabolism in weeds.
Collapse
Affiliation(s)
| | - Shiv S. Kaundun
- Herbicide BioscienceSyngentaJealott’s Hill International Research CentreBracknell,RG42 6EYUK
| | - James A. Morris
- Herbicide BioscienceSyngentaJealott’s Hill International Research CentreBracknell,RG42 6EYUK
| | - Sarah‐Jane Hutchings
- Herbicide BioscienceSyngentaJealott’s Hill International Research CentreBracknell,RG42 6EYUK
| | - Seth A. Strom
- Department of Crop SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Anatoli V. Lygin
- Department of Crop SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Dean E. Riechers
- Department of Crop SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| |
Collapse
|
82
|
Singh RP, Saini N, Sharma G, Rahisuddin R, Patel M, Kaushik A, Kumaran S. Moonlighting Biochemistry of Cysteine Synthase: A Species-specific Global Regulator. J Mol Biol 2021; 433:167255. [PMID: 34547327 DOI: 10.1016/j.jmb.2021.167255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 11/18/2022]
Abstract
Cysteine Synthase (CS), the enzyme that synthesizes cysteine, performs non-canonical regulatory roles by binding and modulating functions of disparate proteins. Beyond its role in catalysis and regulation in the cysteine biosynthesis pathway, it exerts its moonlighting effect by binding to few other proteins which possess a C-terminal "CS-binding motif", ending with a terminal ILE. Therefore, we hypothesized that CS might regulate many other disparate proteins with the "CS-binding motif". In this study, we developed an iterative sequence matching method for mapping moonlighting biochemistry of CS and validated our prediction by analytical and structural approaches. Using a minimal protein-peptide interaction system, we show that five previously unknown CS-binder proteins that participate in diverse metabolic processes interact with CS in a species-specific manner. Furthermore, results show that signatures of protein-protein interactions, including thermodynamic, competitive-inhibition, and structural features, highly match the known CS-Binder, serine acetyltransferase (SAT). Together, the results presented in this study allow us to map the extreme multifunctional space (EMS) of CS and reveal the biochemistry of moonlighting space, a subset of EMS. We believe that the integrated computational and experimental workflow developed here could be further modified and extended to study protein-specific moonlighting properties of multifunctional proteins.
Collapse
Affiliation(s)
- Ravi Pratap Singh
- G. N. Ramachandran Protein Centre, Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology (IMTECH), Sector 39-A, Chandigarh 160036, India
| | - Neha Saini
- G. N. Ramachandran Protein Centre, Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology (IMTECH), Sector 39-A, Chandigarh 160036, India
| | - Gaurav Sharma
- Institute of Bioinformatics and Applied Biotechnology (IBAB), Electronic city, Bengaluru, Karnataka 560100, India
| | - R Rahisuddin
- G. N. Ramachandran Protein Centre, Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology (IMTECH), Sector 39-A, Chandigarh 160036, India. https://twitter.com/RahisuddinAlig
| | - Madhuri Patel
- G. N. Ramachandran Protein Centre, Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology (IMTECH), Sector 39-A, Chandigarh 160036, India
| | - Abhishek Kaushik
- G. N. Ramachandran Protein Centre, Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology (IMTECH), Sector 39-A, Chandigarh 160036, India
| | - S Kumaran
- G. N. Ramachandran Protein Centre, Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology (IMTECH), Sector 39-A, Chandigarh 160036, India.
| |
Collapse
|
83
|
Cheng T, Lam AK, Gopalan V. Diet derived polycyclic aromatic hydrocarbons and its pathogenic roles in colorectal carcinogenesis. Crit Rev Oncol Hematol 2021; 168:103522. [PMID: 34748942 DOI: 10.1016/j.critrevonc.2021.103522] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022] Open
Abstract
Polycyclic aromatic hydrocarbon (PAHs) are molecules that contaminate meat products during the high-temperature cooking of meat. This study reviewed the pathogenic roles of meat derived polycyclic aromatic hydrocarbons in the carcinogenesis of colorectal cancer (CRC). Ingested PAHs undergo xenobiotic metabolism resulting in the activation of genotoxic metabolites that can induce DNA damage in the colorectum. Genetic polymorphisms in PAH xenobiotic enzymes are linked to the risk of CRC and suggest a role for PAH-meat ingestion in carcinogenesis of colorectal malignancies. Furthermore, PAH specific DNA adducts have been identified in colorectal cancer tissue and linked to high meat intake. DNA adduct resolution is mediated by the nucleotide excision repair, and polymorphisms within genes of this repair pathway and high meat intake are associated with increased CRC risk. In the literature, there is evidence from metabolic enzyme gene variants, DNA repair genes, PAH metabolites, and epidemiological studies suggesting PAH involvement in CRC.
Collapse
Affiliation(s)
- Tracie Cheng
- Cancer Molecular Pathology, School of Medicine & Dentistry, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Alfred K Lam
- Cancer Molecular Pathology, School of Medicine & Dentistry, Griffith University, Gold Coast, Queensland 4222, Australia
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine & Dentistry, Griffith University, Gold Coast, Queensland 4222, Australia.
| |
Collapse
|
84
|
Rai R, Singh S, Rai KK, Raj A, Sriwastaw S, Rai LC. Regulation of antioxidant defense and glyoxalase systems in cyanobacteria. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2021; 168:353-372. [PMID: 34700048 DOI: 10.1016/j.plaphy.2021.09.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/09/2021] [Accepted: 09/28/2021] [Indexed: 05/19/2023]
Abstract
Oxidative stress is common consequence of abiotic stress in plants as well as cyanobacteria caused by generation of reactive oxygen species (ROS), an inevitable product of respiration and photosynthetic electron transport. ROS act as signalling molecule at low concentration however, when its production exceeds the endurance capacity of antioxidative defence system, the organisms suffer oxidative stress. A highly toxic metabolite, methylglyoxal (MG) is also produced in cyanobacteria in response to various abiotic stresses which consequently augment the ensuing oxidative damage. Taking recourse to the common lineage of eukaryotic plants and cyanobacteria, it would be worthwhile to explore the regulatory role of glyoxalase system and antioxidative defense mechanism in combating abiotic stress in cyanobacteria. This review provides comprehensive information on the complete glyoxalase system (GlyI, GlyII and GlyIII) in cyanobacteria. Furthermore, it elucidates the recent understanding regarding the production of ROS and MG, noteworthy link between intracellular MG and ROS and its detoxification via synchronization of antioxidants (enzymatic and non-enzymatic) and glyoxalase systems using glutathione (GSH) as common co-factor.
Collapse
Affiliation(s)
- Ruchi Rai
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Shilpi Singh
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Krishna Kumar Rai
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Alka Raj
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Sonam Sriwastaw
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - L C Rai
- Molecular Biology Section, Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
85
|
Kobayashi M, Yonezawa A, Takasawa H, Nagao Y, Iguchi K, Endo S, Ikari A, Matsunaga T. Development of cisplatin resistance in breast cancer MCF7 cells by up-regulating aldo-keto reductase 1C3 expression, glutathione synthesis, and proteasomal proteolysis. J Biochem 2021; 171:97-108. [PMID: 34676395 DOI: 10.1093/jb/mvab117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/18/2021] [Indexed: 02/04/2023] Open
Abstract
Cisplatin (CDDP) is widely prescribed for the treatment of various cancers including bladder cancers, whereas its clinical use for breast cancer chemotherapy is restricted owing to easy acquisition of the chemoresistance. Here, we established a highly CDDP-resistant variant of human breast cancer MCF7 cells and found that procuring the resistance aberrantly elevates the expression of aldo-keto reductase (AKR) 1C3. Additionally, MCF7 cell sensitivity to CDDP was decreased and increased by overexpression and knockdown, respectively, of AKR1C3, clearly inferring that the enzyme plays a crucial role in acquiring the CDDP resistance. The CDDP-resistant cells suppressed the formation of cytotoxic reactive aldehydes by CDDP treatment, and the suppressive effects were almost completely abolished by pretreating with AKR1C3 inhibitor. The resistant cells also exhibited the elevated glutathione amount and 26S proteasomal proteolytic activities, and their CDDP sensitivity was significantly augmented by pretreatment with an inhibitor of glutathione synthesis or proteasomal proteolysis. Moreover, the combined treatment with inhibitors of AKR1C3, glutathione synthesis, and/or proteasomal proteolysis potently overcame the CDDP resistance and docetaxel cross-resistance. Taken together, our results suggest that the combination of inhibitors of AKR1C3, glutathione synthesis, and/or proteasomal proteolysis is effective as an adjuvant therapy to enhance CDDP sensitivity of breast cancer cells.
Collapse
Affiliation(s)
- Mio Kobayashi
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Ayano Yonezawa
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Hiroaki Takasawa
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Yukino Nagao
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, 5-6-1 Mitahora-higashi, Gifu 502-8585, Japan
| | - Kazuhiro Iguchi
- Laboratory of Community Pharmacy, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, 5-6-1 Mitahora-higashi, Gifu 502-8585, Japan
| |
Collapse
|
86
|
Schwartz M, Neiers F, Charles JP, Heydel JM, Muñoz-González C, Feron G, Canon F. Oral enzymatic detoxification system: Insights obtained from proteome analysis to understand its potential impact on aroma metabolization. Compr Rev Food Sci Food Saf 2021; 20:5516-5547. [PMID: 34653315 DOI: 10.1111/1541-4337.12857] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/02/2021] [Accepted: 09/10/2021] [Indexed: 12/17/2022]
Abstract
The oral cavity is an entry path into the body, enabling the intake of nutrients but also leading to the ingestion of harmful substances. Thus, saliva and oral tissues contain enzyme systems that enable the early neutralization of xenobiotics as soon as they enter the body. Based on recently published oral proteomic data from several research groups, this review identifies and compiles the primary detoxification enzymes (also known as xenobiotic-metabolizing enzymes) present in saliva and the oral epithelium. The functions and the metabolic activity of these enzymes are presented. Then, the activity of these enzymes in saliva, which is an extracellular fluid, is discussed with regard to the salivary parameters. The next part of the review presents research evidencing oral metabolization of aroma compounds and the putative involved enzymes. The last part discusses the potential role of these enzymatic reactions on the perception of aroma compounds in light of recent pieces of evidence of in vivo oral metabolization of aroma compounds affecting their release in mouth and their perception. Thus, this review highlights different enzymes appearing as relevant to explain aroma metabolism in the oral cavity. It also points out that further works are needed to unravel the effect of the oral enzymatic detoxification system on the perception of food flavor in the context of the consumption of complex food matrices, while considering the impact of food oral processing. Thus, it constitutes a basis to explore these biochemical mechanisms and their impact on flavor perception.
Collapse
Affiliation(s)
- Mathieu Schwartz
- Centre des Sciences du Goût et de l'Alimentation (CSGA), AgroSup Dijon, CNRS, INRAE, Université de Bourgogne Franche Comté, Dijon, France
| | - Fabrice Neiers
- Centre des Sciences du Goût et de l'Alimentation (CSGA), AgroSup Dijon, CNRS, INRAE, Université de Bourgogne Franche Comté, Dijon, France
| | - Jean-Philippe Charles
- Centre des Sciences du Goût et de l'Alimentation (CSGA), AgroSup Dijon, CNRS, INRAE, Université de Bourgogne Franche Comté, Dijon, France
| | - Jean-Marie Heydel
- Centre des Sciences du Goût et de l'Alimentation (CSGA), AgroSup Dijon, CNRS, INRAE, Université de Bourgogne Franche Comté, Dijon, France
| | - Carolina Muñoz-González
- Instituto de investigación en Ciencias de la Alimentación (CIAL), (CSIC-UAM), C/ Nicolás Cabrera, Madrid, Spain
| | - Gilles Feron
- Centre des Sciences du Goût et de l'Alimentation (CSGA), AgroSup Dijon, CNRS, INRAE, Université de Bourgogne Franche Comté, Dijon, France
| | - Francis Canon
- Centre des Sciences du Goût et de l'Alimentation (CSGA), AgroSup Dijon, CNRS, INRAE, Université de Bourgogne Franche Comté, Dijon, France
| |
Collapse
|
87
|
Zhou T, Liu H, Huang Y, Wang Z, Shan Y, Yue Y, Xia Z, Liang Y, An M, Wu Y. ε-poly- L-lysine Affects the Vegetative Growth, Pathogenicity and Expression Regulation of Necrotrophic Pathogen Sclerotinia sclerotiorum and Botrytis cinerea. J Fungi (Basel) 2021; 7:jof7100821. [PMID: 34682242 PMCID: PMC8540936 DOI: 10.3390/jof7100821] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 02/04/2023] Open
Abstract
Microbial secondary metabolites produced by Streptomyces are applied to control plant diseases. The metabolite, ε-poly-l-lysine (ε-PL), is a non-toxic food preservative, but the potential application of this compound as a microbial fungicide in agriculture is rarely reported. In this study, the effect and mode of action of ε-PL on two necrotrophic pathogenic fungi, Sclerotinia sclerotiorum and Botrytis cinerea, were investigated. The results showed that ε-PL effectively inhibited the mycelial growth of S. sclerotiorum and B. cinerea with EC50 values of 283 μg/mL and 281 μg/mL, respectively. In addition, ε-PL at the dose of 150 and 300 μg/mL reduced S. sclerotiorum sclerotia formation. The results of the RNA-seq and RT-qPCR validation indicated that ε-PL significantly regulated the gene expression of critical differential expressed genes (DEGs) involved in fungal growth, metabolism, pathogenicity, and induced an increase in the expression of the fungal stress responses and the detoxification genes. These results provided new insights for understanding the modes of action of ε-PL on S. sclerotiorum and B. cinerea and improved the sustainable management of these plant diseases.
Collapse
|
88
|
Alfarouk KO, Alqahtani SS, Alshahrani S, Morgenstern J, Supuran CT, Reshkin SJ. The possible role of methylglyoxal metabolism in cancer. J Enzyme Inhib Med Chem 2021; 36:2010-2015. [PMID: 34517737 PMCID: PMC8451662 DOI: 10.1080/14756366.2021.1972994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Tumours reprogram their metabolism to acquire an evolutionary advantage over normal cells. However, not all such metabolic pathways support energy production. An example of these metabolic pathways is the Methylglyoxal (MG) one. This pathway helps maintain the redox state, and it might act as a phosphate sensor that monitors the intracellular phosphate levels. In this work, we discuss the biochemical step of the MG pathway and interrelate it with cancer.
Collapse
Affiliation(s)
- Khalid O Alfarouk
- Department of Evolutionary Pharmacology, and Tumor Metabolism, Hala Alfarouk Cancer Center, Khartoum, Sudan
| | - Saad S Alqahtani
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan University, Jazan, KSA
| | - Saeed Alshahrani
- Pharmacology and Toxicology Department, College of Pharmacy, Jazan University, Jazan, KSA
| | - Jakob Morgenstern
- Department of Internal Medicine I, Endocrinology and Metabolism, Heidelberg University, Germany
| | - Claudiu T Supuran
- Neurofarba Department, Universita Degli Studi di Firenze, Florence, Italy
| | - Stephan J Reshkin
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, Bari, Italy
| |
Collapse
|
89
|
Little RF, Hertweck C. Chain release mechanisms in polyketide and non-ribosomal peptide biosynthesis. Nat Prod Rep 2021; 39:163-205. [PMID: 34622896 DOI: 10.1039/d1np00035g] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Review covering up to mid-2021The structure of polyketide and non-ribosomal peptide natural products is strongly influenced by how they are released from their biosynthetic enzymes. As such, Nature has evolved a diverse range of release mechanisms, leading to the formation of bioactive chemical scaffolds such as lactones, lactams, diketopiperazines, and tetronates. Here, we review the enzymes and mechanisms used for chain release in polyketide and non-ribosomal peptide biosynthesis, how these mechanisms affect natural product structure, and how they could be utilised to introduce structural diversity into the products of engineered biosynthetic pathways.
Collapse
Affiliation(s)
- Rory F Little
- Leibniz Institute for Natural Product Research and Infection Biology, HKI, Germany.
| | - Christian Hertweck
- Leibniz Institute for Natural Product Research and Infection Biology, HKI, Germany.
| |
Collapse
|
90
|
Idda ML, Campesi I, Fiorito G, Vecchietti A, Urru SAM, Solinas MG, Franconi F, Floris M. Sex-Biased Expression of Pharmacogenes across Human Tissues. Biomolecules 2021; 11:1206. [PMID: 34439872 PMCID: PMC8393247 DOI: 10.3390/biom11081206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/05/2021] [Accepted: 08/11/2021] [Indexed: 12/18/2022] Open
Abstract
Individual response to drugs is highly variable and largely influenced by genetic variants and gene-expression profiles. In addition, it has been shown that response to drugs is strongly sex-dependent, both in terms of efficacy and toxicity. To expand current knowledge on sex differences in the expression of genes relevant for drug response, we generated a catalogue of differentially expressed human transcripts encoded by 289 genes in 41 human tissues from 838 adult individuals of the Genotype-Tissue Expression project (GTEx, v8 release) and focused our analysis on relevant transcripts implicated in drug response. We detected significant sex-differentiated expression of 99 transcripts encoded by 59 genes in the tissues most relevant for human pharmacology (liver, lung, kidney, small intestine terminal ileum, skin not sun-exposed, and whole blood). Among them, as expected, we confirmed significant differences in the expression of transcripts encoded by the cytochromes in the liver, CYP2B6, CYP3A7, CYP3A5, and CYP1A1. Our systematic investigation on differences between male and female in the expression of drug response-related genes, reinforce the need to overcome the sex bias of clinical trials.
Collapse
Affiliation(s)
- Maria Laura Idda
- Institute of Genetics and Biomedical research, 07100 Sassari, Italy;
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (I.C.); (G.F.); (A.V.); (M.G.S.)
| | - Giovanni Fiorito
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (I.C.); (G.F.); (A.V.); (M.G.S.)
- Unit of Environmental Epidemiology, School of Public Health, Imperial College, London SW7 2AZ, UK
| | - Andrea Vecchietti
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (I.C.); (G.F.); (A.V.); (M.G.S.)
| | - Silvana Anna Maria Urru
- Hospital Pharmacy Unit, Trento General Hospital, Autonomous Province of Trento, 38122 Trento, Italy;
- Department of Chemistry and Pharmacy, School of Hospital Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Maria Giuliana Solinas
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (I.C.); (G.F.); (A.V.); (M.G.S.)
| | - Flavia Franconi
- National Laboratory of Pharmacology and Gender medicine, National Institute of Biostructure and Biosystems, 00136 Rome, Italy;
| | - Matteo Floris
- Institute of Genetics and Biomedical research, 07100 Sassari, Italy;
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (I.C.); (G.F.); (A.V.); (M.G.S.)
| |
Collapse
|
91
|
Dong H, Chang MCY. Structural Basis for Branched Substrate Selectivity in a Ketoreductase from Ascaris suum. ACS Catal 2021. [DOI: 10.1021/acscatal.1c01642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Hongjun Dong
- Department of Chemistry, University of California, Berkeley, Berkeley, California 94720-1460, United States
| | - Michelle C. Y. Chang
- Department of Chemistry, University of California, Berkeley, Berkeley, California 94720-1460, United States
- Department of Molecular & Cell Biology, University of California, Berkeley, California 94720-3200, United States
- Department of Chemical & Biomolecular Engineering, University of California, Berkeley, California 94720-1462, United States
| |
Collapse
|
92
|
Sonowal H, Ramana KV. Development of Aldose Reductase Inhibitors for the Treatment of Inflammatory Disorders and Cancer: Current Drug Design Strategies and Future Directions. Curr Med Chem 2021; 28:3683-3712. [PMID: 33109031 DOI: 10.2174/0929867327666201027152737] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 11/22/2022]
Abstract
Aldose Reductase (AR) is an enzyme that converts glucose to sorbitol during the polyol pathway of glucose metabolism. AR has been shown to be involved in the development of secondary diabetic complications due to its involvement in causing osmotic as well as oxidative stress. Various AR inhibitors have been tested for their use to treat secondary diabetic complications, such as retinopathy, neuropathy, and nephropathy in clinical studies. Recent studies also suggest the potential role of AR in mediating various inflammatory complications. Therefore, the studies on the development and potential use of AR inhibitors to treat inflammatory complications and cancer besides diabetes are currently on the rise. Further, genetic mutagenesis studies, computer modeling, and molecular dynamics studies have helped design novel and potent AR inhibitors. This review discussed the potential new therapeutic use of AR inhibitors in targeting inflammatory disorders and cancer besides diabetic complications. Further, we summarized studies on how AR inhibitors have been designed and developed for therapeutic purposes in the last few decades.
Collapse
Affiliation(s)
- Himangshu Sonowal
- Moores Cancer Center, University of California San Diego, La Jolla, California 92037, United States
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, United States
| |
Collapse
|
93
|
Protective Effect of Aldo-keto Reductase 1B1 Against Neuronal Cell Damage Elicited by 4'-Fluoro-α-pyrrolidinononanophenone. Neurotox Res 2021; 39:1360-1371. [PMID: 34043181 DOI: 10.1007/s12640-021-00380-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/17/2021] [Accepted: 05/21/2021] [Indexed: 10/21/2022]
Abstract
Chronic exposure to cathinone derivatives increases the risk of severe health hazards, whereas little is known about the detailed pathogenic mechanisms triggered by the derivatives. We have recently shown that treatment with α-pyrrolidinononanophenone (α-PNP, a highly lipophilic cathinone derivative possessing a long hydrocarbon main chain) provokes neuronal cell apoptosis and its 4'-fluorinated analog (F-α-PNP) potently augments the apoptotic effect. In this study, we found that neuronal SK-N-SH cell damage elicited by F-α-PNP treatment is augmented most potently by pre-incubation with an AKR1B1 inhibitor tolrestat, among specific inhibitors of four aldo-keto reductase (AKR) family members (1B1, 1C1, 1C2, and 1C3) expressed in the neuronal cells. In addition, forced overexpression of AKR1B1 remarkably lowered the cell sensitivity to F-α-PNP toxicity, clearly indicating that AKR1B1 protects from neurotoxicity of the derivative. Treatment of SK-N-SH cells with F-α-PNP resulted in a dose-dependent up-regulation of AKR1B1 expression and activation of its transcription factor NF-E2-related factor 2. Metabolic analyses using liquid chromatography/mass spectrometry/mass spectrometry revealed that AKR1B1 is hardly involved in the F-α-PNP metabolism. The F-α-PNP treatment resulted in production of reactive oxygen species and lipid peroxidation byproduct 4-hydroxy-2-nonenal (HNE) in the cells. The enhanced HNE level was reduced by overexpression of AKR1B1, which also lessened the cell damage elicited by HNE. These results suggest that the AKR1B1-mediated neuronal cell protection is due to detoxification of HNE formed by F-α-PNP treatment, but not to metabolism of the derivative.
Collapse
|
94
|
Shi Z, Kong X, Li C, Liu H, Aliagan AI, Liu L, Shi Y, Shi X, Ma B, Jin R, Wang S, Pan D, Tang J. Bioinformatic analysis of differentially expressed genes as prognostic markers in pheochromocytoma and paraganglioma tumors. Genes Genet Syst 2021; 96:55-69. [PMID: 34039789 DOI: 10.1266/ggs.20-00057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The pathogenesis of pheochromocytoma and paraganglioma (PCPG) catecholamine-producing tumors is exceedingly complicated. Here, we sought to identify important genes affecting the prognosis and survival rate of patients suffering from PCPG. We analyzed 95 samples obtained from two microarray data series, GSE19422 and GSE60459, from the Gene Expression Omnibus (GEO) repository. First, differentially expressed genes (DEGs) were identified by comparing 87 PCPG tumor samples and eight normal adrenal tissue samples using R language. The GEO2R tool and Venn diagram software were applied to the Database for Annotation, Visualization and Integrated Discovery (DAVID) to analyze Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO). We further employed Cytoscape with the Molecular Complex Detection (MCODE) tool to make protein-protein interactions visible for the Search Tool for Retrieval of Interacting Genes (STRING). These procedures resulted in 30 candidate DEGs, which were subjected to Kaplan-Meier analysis and validated by Gene Expression Profiling Interactive Analysis (GEPIA) to determine their influence on overall survival rate. Finally, we identified ALDH3A2 and AKR1B1, two genes in the glycerolipid metabolism pathway, as being particularly enriched in PCPG tumors and correlated with T and B tumor-infiltrating immune cells. Our results suggest that these two DEGs are closely associated with the prognosis of malignant PCPG tumors.
Collapse
Affiliation(s)
- Zhen Shi
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University.,Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio
| | - Xiaodi Kong
- Department of Urology, Xiangya Hospital, Central South University
| | - Cheng Li
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| | - Hui Liu
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| | - Abdulhafiz Imam Aliagan
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio
| | - Li Liu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio
| | - Yue Shi
- School of Mechanical Engineering, Northwestern Polytechnical University
| | - Xiao Shi
- The Third Xiangya Hospital, Central South University
| | - Binbin Ma
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| | - Ruiqi Jin
- The Third Xiangya Hospital, Central South University
| | - Shizhuo Wang
- College of Life Science and Technology, Beijing University of Chemical Technology
| | - Ding Pan
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| | - Juyu Tang
- Department of Hand and Microsurgery, Xiangya Hospital, Central South University
| |
Collapse
|
95
|
Fujii J, Homma T, Miyata S, Takahashi M. Pleiotropic Actions of Aldehyde Reductase (AKR1A). Metabolites 2021; 11:343. [PMID: 34073440 PMCID: PMC8227408 DOI: 10.3390/metabo11060343] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
We provide an overview of the physiological roles of aldehyde reductase (AKR1A) and also discuss the functions of aldose reductase (AKR1B) and other family members when necessary. Many types of aldehyde compounds are cytotoxic and some are even carcinogenic. Such toxic aldehydes are detoxified via the action of AKR in an NADPH-dependent manner and the resulting products may exert anti-diabetic and anti-tumorigenic activity. AKR1A is capable of reducing 3-deoxyglucosone and methylglyoxal, which are reactive intermediates that are involved in glycation, a non-enzymatic glycosylation reaction. Accordingly, AKR1A is thought to suppress the formation of advanced glycation end products (AGEs) and prevent diabetic complications. AKR1A and, in part, AKR1B are responsible for the conversion of d-glucuronate to l-gulonate which constitutes a process for ascorbate (vitamin C) synthesis in competent animals. AKR1A is also involved in the reduction of S-nitrosylated glutathione and coenzyme A and thereby suppresses the protein S-nitrosylation that occurs under conditions in which the production of nitric oxide is stimulated. As the physiological functions of AKR1A are currently not completely understood, the genetic modification of Akr1a could reveal the latent functions of AKR1A and differentiate it from other family members.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan;
| | - Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan;
| | - Satoshi Miyata
- Miyata Diabetes and Metabolism Clinic, 5-17-21 Fukushima, Fukushima-ku, Osaka 553-0003, Japan;
| | - Motoko Takahashi
- Department of Biochemistry, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan;
| |
Collapse
|
96
|
Aldo-keto reductase inhibitors increase the anticancer effects of tyrosine kinase inhibitors in chronic myelogenous leukemia. J Pharmacol Sci 2021; 147:1-8. [PMID: 34294359 DOI: 10.1016/j.jphs.2021.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 01/06/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are widely utilized in clinical practice to treat carcinomas, but secondary tumor resistance during chronic treatment can be problematic. AKR1B1 and AKR1B10 of the aldo-keto reductase (AKR) superfamily are highly expressed in cancer cells and are believed to be involved in drug resistance. The aim of this study was to understand how TKI treatment of chronic myelogenous leukemia (CML) cells changes their glucose metabolism and if inhibition of AKRs can sensitize CML cells to TKIs. K562 cells were treated with the TKIs imatinib, nilotinib, or bosutinib, and the effects on glucose metabolism, cell death, glutathione levels, and AKR levels were assessed. To assess glucose dependence, cells were cultured in normal and low-glucose media. Pretreatment with AKR inhibitors, including epalrestat, were used to determine AKR-dependence. Treatment with TKIs increased intracellular glucose, AKR1B1/10 levels, glutathione oxidation, and nuclear translocation of nuclear factor erythroid 2-related factor 2, but with minimal cell death. These effects were dependent on intracellular glucose accumulation. Pretreatment with epalrestat, or a selective inhibitor of AKR1B10, exacerbated TKI-induced cell death, suggesting that especially AKR1B10 was involved in protection against TKIs. Thus, by disrupting cell protective mechanisms, AKR inhibitors may render CML more susceptible to TKI treatments.
Collapse
|
97
|
Xiong R, Wu Y, Wu Q, Muskhelishvili L, Davis K, Tripathi P, Chen Y, Chen T, Bryant M, Rosenfeldt H, Healy SM, Cao X. Integration of transcriptome analysis with pathophysiological endpoints to evaluate cigarette smoke toxicity in an in vitro human airway tissue model. Arch Toxicol 2021; 95:1739-1761. [PMID: 33660061 PMCID: PMC8113308 DOI: 10.1007/s00204-021-03008-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/16/2021] [Indexed: 01/04/2023]
Abstract
Exposure to cigarette smoke (CS) is a known risk factor in the pathogenesis of smoking-caused diseases, such as chronic obstructive pulmonary diseases (COPD) and lung cancer. To assess the effects of CS on the function and phenotype of airway epithelial cells, we developed a novel repeated treatment protocol and comprehensively evaluated the progression of key molecular, functional, and structural abnormalities induced by CS in a human in vitro air-liquid-interface (ALI) airway tissue model. Cultures were exposed to CS (diluted with 0.5 L/min, 1.0 L/min, and 4.0 L/min clean air) generated from smoking five 3R4F University of Kentucky reference cigarettes under the International Organization for Standardization (ISO) machine smoking regimen, every other day for 4 weeks (3 days per week, 40 min/day). By integrating the transcriptomics-based approach with the in vitro pathophysiological measurements, we demonstrated CS-mediated effects on oxidative stress, pro-inflammatory cytokines and matrix metalloproteinases (MMPs), ciliary function, expression and secretion of mucins, and squamous cell differentiation that are highly consistent with abnormalities observed in airways of smokers. Enrichment analysis on the transcriptomic profiles of the ALI cultures revealed key molecular pathways, such as xenobiotic metabolism, oxidative stress, and inflammatory responses that were perturbed in response to CS exposure. These responses, in turn, may trigger aberrant tissue remodeling, eventually leading to the onset of respiratory diseases. Furthermore, changes of a panel of genes known to be disturbed in smokers with COPD were successfully reproduced in the ALI cultures exposed to CS. In summary, findings from this study suggest that such an integrative approach may be a useful tool for identifying genes and adverse cellular events caused by inhaled toxicants, like CS.
Collapse
Affiliation(s)
- Rui Xiong
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Yue Wu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | | | - Kelly Davis
- Toxicologic Pathology Associates, Jefferson, AR, 72079, USA
| | - Priya Tripathi
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Ying Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tao Chen
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Matthew Bryant
- Office of Scientific Coordination, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Hans Rosenfeldt
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Sheila M Healy
- Division of Nonclinical Science, Center for Tobacco Products, US Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
98
|
Zhang X, Wang Y, Zhang S, Kong X, Liu F, Zhang Z. RNAi-Mediated Silencing of the Chitinase 5 Gene for Fall Webworm ( Hyphantria cunea) Can Inhibit Larval Molting Depending on the Timing of dsRNA Injection. INSECTS 2021; 12:insects12050406. [PMID: 33946562 PMCID: PMC8147239 DOI: 10.3390/insects12050406] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022]
Abstract
Chitinases, which are crucial enzymes required for chitin degradation and reconstruction, are often selectively considered to be effective molecular targets for pest control due to their critical roles in insect development. Although the Hyphantria cunea chitinase gene has been reported previously, its sequence characteristics, gene function, and feasibility as a potential target for pest management were absent. In the present study, we characterized the H. cunea chitinase gene and designated it HcCht5. Phylogenic and domain structure analysis suggested that HcCht5 contained the typical chitinase features and was clustered into chitinase group I. Tissue-specific and developmental expression pattern analysis with Real-Time Quantitative PCR (RT-qPCR) showed that HcCht5 was mainly expressed in the integument tissues and that the transcript levels peaked during molting. RNA interference (RNAi)-mediated silencing of HcCht5 caused 33.3% (2 ug) and 66.7% (4 ug) mortality rates after double-stranded RNA (dsRNA) injection. Importantly, the interference efficiency of HcCht5 depended on the injection time of double-stranded RNA (dsRNA), as the pre-molting treatment achieved molt arrest more effectively. In addition, transcriptome sequencing (RNA-seq) analysis of RNAi samples demonstrated silencing of the down-regulated HcCht5 genes related to chitin metabolism and molting hormone signaling, as well as genes related to detoxification metabolism. Our results indicate the essential role of HcCht5 in H. cunea development and detail the involvement of its gene function in the larval molting process.
Collapse
|
99
|
Hadtstein F, Vrolijk M. Vitamin B-6-Induced Neuropathy: Exploring the Mechanisms of Pyridoxine Toxicity. Adv Nutr 2021; 12:1911-1929. [PMID: 33912895 PMCID: PMC8483950 DOI: 10.1093/advances/nmab033] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/26/2022] Open
Abstract
Vitamin B-6 in the form of pyridoxine (PN) is commonly used by the general population. The use of PN-containing supplements has gained lots of attention over the past years as they have been related to the development of peripheral neuropathy. In light of this, the number of reported cases of adverse health effects due to the use of vitamin B-6 have increased. Despite a long history of study, the pathogenic mechanisms associated with PN toxicity remain elusive. Therefore, the present review is focused on investigating the mechanistic link between PN supplementation and sensory peripheral neuropathy. Excessive PN intake induces neuropathy through the preferential injury of sensory neurons. Recent reports on hereditary neuropathy due to pyridoxal kinase (PDXK) mutations may provide some insight into the mechanism, as genetic deficiencies in PDXK lead to the development of axonal sensory neuropathy. High circulating concentrations of PN may lead to a similar condition via the inhibition of PDXK. The mechanism behind PDXK-induced neuropathy is unknown; however, there is reason to believe that it may be related to γ-aminobutyric acid (GABA) neurotransmission. Compounds that inhibit PDXK lead to convulsions and reductions in GABA biosynthesis. The absence of central nervous system-related symptoms in PDXK deficiency could be due to differences in the regulation of PDXK, where PDXK activity is preserved in the brain but not in peripheral tissues. As PN is relatively impermeable to the blood-brain barrier, PDXK inhibition would similarly be confined to the peripheries and, as a result, GABA signaling may be perturbed within peripheral tissues, such as sensory neurons. Perturbed GABA signaling within sensory neurons may lead to excitotoxicity, neurodegeneration, and ultimately, the development of peripheral neuropathy. For several reasons, we conclude that PDXK inhibition and consequently disrupted GABA neurotransmission is the most plausible mechanism of toxicity.
Collapse
Affiliation(s)
- Felix Hadtstein
- University College Venlo, Campus Venlo, Maastricht University, Maastricht, The Netherlands
| | | |
Collapse
|
100
|
Nishihara M, Ramsden D, Balani SK. Evaluation of the drug-drug interaction potential for trazpiroben (TAK-906), a D 2/D 3 receptor antagonist for gastroparesis, towards cytochrome P450s and transporters. Xenobiotica 2021; 51:668-679. [PMID: 33879032 DOI: 10.1080/00498254.2021.1912438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Trazpiroben (TAK-906), a peripherally selective dopamine D2/D3 receptor antagonist, is being developed for the treatment of patients with gastroparesis. The potential of trazpiroben to act as a perpetrator or a victim for cytochrome P450 (CYP)- or transporter- mediated drug-drug interactions (DDIs) was evaluated following the latest regulatory guidelines.In vitro studies revealed that trazpiroben is metabolised mainly through a non-CYP pathway (56.7%) by multiple cytosolic, NADPH-dependent reductase, such as aldo-keto reductase and short-chain dehydrogenase/reductase including carbonyl reductases. Remaining metabolism occurs through CYP3A4 and CYP2C8 (43.3%). Trazpiroben is neither an inhibitor nor an inducer of major CYP enzymes at a clinically relevant dose. It is a substrate of P-glycoprotein (P-gp) and organic anion transporting polypeptide (OATP) 1B1/1B3, but is not an inhibitor of transporters listed in the DDI guidelines at a clinically relevant dose. This is consistent with findings from CYP3A and P-gp-based clinical assessment showing no substantial change (≤2-fold) in trazpiroben exposure when co-administered with itraconazole.Collectively, trazpiroben has low potential of enzyme-mediated DDIs and is unlikely to act as a perpetrator of transporter-mediated DDIs but there may be a potential to act as a victim of OATP1B1/1B3 DDI that will be evaluated clinically.
Collapse
Affiliation(s)
- Mitsuhiro Nishihara
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa, Japan
| | - Diane Ramsden
- Global Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co, Cambridge, MA, USA
| | - Suresh K Balani
- Global Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co, Cambridge, MA, USA
| |
Collapse
|