1
|
de Freitas KS, Squarisi IS, de Souza LTM, Ozelin SD, de Souza Oliveira LT, Ribeiro VP, Bastos JK, Tavares DC. Evaluation of safety and efficacy of Brazilian brown propolis from Araucaria sp. in preventing colon cancer. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:196-208. [PMID: 39609034 DOI: 10.1080/15287394.2024.2431921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Brazilian propolis produced by honeybees have been widely studied, but few data exist regarding the safety and pharmacological potential of this natural product. The aim of the present study was to examine the toxicity, genotoxicity, and chemoprevention effects attributed to exposure to the brown propolis hydroalcoholic extract (BPHE) of Araucaria sp. Acute oral toxicity test was conducted using Wistar Hannover rats, demonstrating that the highest dose tested (2,000 mg/kg b.w.) produced no apparent adverse effects or lethality. The micronucleus (MN) genotoxicity test was conducted using peripheral blood from Swiss mice, which also noted that BPHE did not induce significant chromosomal damage. It is of interest that BPHE at a dose of 12 mg/kg b.w. exhibited antigenotoxic effects against the doxorubicin (DXR)-induced damage. However, BPHE did not influence the depletion of reduced glutathione induced by DXR in mice. It is noteworthy that BPHE exerted chemopreventive effects at doses 6, 12, and 24 mg/kg b.w. The determination of this effect of BPHE on colon carcinogenesis was examined using aberrant crypt foci (ACF) as evidenced by histological analysis. The colons of animals treated with BPHE (12 mg/kg b.w.) exhibited a significant reduction in staining for proliferating cell nuclear antigen (PCNA) and cyclooxygenase-2 (COX-2) protein following 1,2-dimethylhydrazine (DMH)-and BPHE combined treatments. Hence, it is conceivable that the anti-inflammatory activity of the chemical constituents of BPHE are involved in its chemopreventive action against colon carcinogenesis as evidenced from ACF assay. Therefore, BHPE was found to be a safe product, without any apparent significant acute adverse risk. Further, the extract exhibited antigenotoxic and anticarcinogenic activities which may be considered for beneficial uses in colon carcinogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Victor Pena Ribeiro
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | |
Collapse
|
2
|
Durr-E-Shahwar, Zubair H, Raza MK, Khan Z, Mansour L, Ali A, Imran M. Investigation of GSTP1 and PTEN gene polymorphisms and their association with susceptibility to colorectal cancer. Radiol Oncol 2025:raon-2025-0001. [PMID: 39754630 DOI: 10.2478/raon-2025-0001] [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: 07/12/2024] [Accepted: 10/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND This study investigates the association of single nucleotide polymorphism in glutathione S transferase P1 (rs1695 and rs1138272) and phosphatase and TENsin homolog (rs701848 and rs2735343) with the risk of colorectal cancer (CRC). PATIENTS AND METHODS In this case-control study, 250 healthy controls and 200 CRC patients were enrolled. All subjects were divided into 3 groups: healthy control, patients, and overall (control + patients). Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The demographic information, including age, gender, location, smoking status, cancer stage, and node involvement, were collected. RESULTS The allele frequencies of PTEN rs701848 in overall subjects were 0.78 for C and 0.22 for T. Similarly, in overall individuals, allele frequencies for PTEN rs2735343 were 0.65 and 0.35 for G and C alleles, respectively. The CC genotype or C allele of rs701848 and CG/GG genotype of rs2735343 were observed to be a risk factor for CRC. In overall individuals, a significant (p ≤ 0.05)) association was observed between rs701848 and rs2735343 polymorphisms CRC. Allele frequencies for GSTP1 rs1695 were 0.68 and 0.32 for the A and G alleles, respectively. Allele frequencies for GSTP1 rs1138272 were 0.68 and 0.32 for C and T alleles, respectively. However, a significant (p < 0.05) association was found in males for rs1695, while a non-significant difference was observed for the distribution of any genotypes or alleles at GSTP1 (rs1138272). CONCLUSIONS Both SNPs of PTEN rs701848 and rs2735343 polymorphisms were significantly associated with CRC. However, in GSTP1, rs1695 was significantly associated with CRC risk in males, and rs1138272 showed a non-significant association with colorectal cancer risk.
Collapse
Affiliation(s)
- Durr-E-Shahwar
- 1Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Hina Zubair
- 1Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Muhammad Kashif Raza
- 1Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
- 2Department of Chemistry, Shaheed Benazir Bhutto University Sheringal Dir upper, Sringeri, Pakistan
| | - Zahid Khan
- 1Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| | - Lamjed Mansour
- 3Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Aktar Ali
- 4Biological Screening Core, Warren Family Center for Drug Discovery, University of Notre Dame, Notre Dame, United States
| | - Muhammad Imran
- 1Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Pakistan
| |
Collapse
|
3
|
Ulusu NN. Revealing the secrets of Blue Zones. Front Pharmacol 2024; 15:1428111. [PMID: 39726786 PMCID: PMC11669513 DOI: 10.3389/fphar.2024.1428111] [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: 05/05/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024] Open
Abstract
Aging is influenced by cellular senescence mechanisms that are associated with oxidative stress. Oxidative stress is the imbalance between antioxidants and free radicals. This imbalance affects enzyme activities and causes mitochondrial dysfunction. It also slows down cellular energy production and disrupts cellular homeostasis. Additionally, oxidative stress stimulates inflammation, increases the number of point mutations, and alters intercellular communication. It can lead to epigenetic alterations, genomic instability, telomere attrition, and loss of proteostasis. Ultimately, these factors contribute to aging and the development of chronic diseases. Glucose-6-phosphate dehydrogenase (G6PD) is an antioxidant enzyme that protects cells from oxidative and nitrosative damage. It helps restore redox balance, preserve macromolecule function, and rescue cells from cellular senescence, autophagy, and stress-induced apoptosis. G6PD is considered an anti-senescence enzyme. The World Health Organization classifies G6PD variants into five groups based on the enzyme's residual activity. The first four classes are categorized according to the degree of G6PD deficiency, while the fifth class includes variants with enzyme activities greater than normal. Increased G6PD activity does not exhibit clinical manifestations. Consequently, the full spectrum of mutations and the prevalence of increased G6PD activity in the population remain unknown. The world's oldest and healthiest people live in Blue Zones. These comprise isolated populations, and there may be a geographic prevalence of high-activity G6PD variants that protect against oxidative stress-induced senescence. To uncover the secret of centenarians' longevity, additional research is needed to determine whether the hidden factor is the increased activity of the G6PD enzyme.
Collapse
Affiliation(s)
- N. Nuray Ulusu
- Department of Medical Biochemistry, School of Medicine, Koc University, Istanbul, Türkiye
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| |
Collapse
|
4
|
Wang Y, Gu J, Zhang F, Zhou D, Yu Y, Chang M, Huo H, Tian K. Multi-omics analysis reveals the mechanisms by which C6-HSL enhances the resistance of typical functional bacteria in activated sludge to low-temperature stress. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176454. [PMID: 39341252 DOI: 10.1016/j.scitotenv.2024.176454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/28/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Signaling molecules, particularly acyl-homoserine lactones (AHLs), can enhance microbial activity under low-temperature stress. However, the specific mechanisms underlying this effect remain unclear. This study identified a typical activated sludge functional bacterium that is sensitive to low temperatures and regulated by hexanoyl-L-homoserine lactone (C6-HSL), a representative of AHLs. It elucidates how C6-HSL modulates the bacterium's resistance to low-temperature stress. Experimental results indicated that C6-HSL significantly increased the levels of adenosine triphosphate (ATP), superoxide dismutase (SOD), peroxidase (POD) and glutathione peroxidase (GSH-Px) in strain LB-001 under low-temperature stress, while also decreasing the levels of reactive oxygen species (ROS). Additionally, C6-HSL markedly repaired the damage to cell membrane structure caused by low-temperature stress. At the genetic level, C6-HSL upregulated the expression of 20 key genes related to energy metabolism, antioxidation, and fatty acid synthesis. At the metabolic level, C6-HSL increased the levels of metabolites related to energy metabolism and antioxidation, boosted the content of unsaturated fatty acids, and reduced the content of saturated fatty acids. This study utilized C6-HSL and low-temperature induction in conjunction with 16S microbial diversity sequencing, genomics, transcriptomics, and metabolomics. These methods were employed to elucidate the molecular mechanisms by which exogenous C6-HSL regulates the resistance of activated sludge microbial communities to low-temperature stress. This research lays the foundation for the application of AHLs and cell communication in wastewater biological treatment, fostering deeper exploration and further innovation in related academic research.
Collapse
Affiliation(s)
- Yibing Wang
- School of Environment, Northeast Normal University, Changchun 130024, China
| | - Jinming Gu
- School of Environment, Northeast Normal University, Changchun 130024, China
| | - Fenglin Zhang
- School of Environment, Northeast Normal University, Changchun 130024, China
| | - Dandan Zhou
- School of Environment, Northeast Normal University, Changchun 130024, China; Engineering Research Center of Low-Carbon Treatment and Green Development of Polluted Water in Northeast China, Ministry of Education, Northeast Normal University, Changchun 130117, China; Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun 130117, China
| | - Yue Yu
- School of Environment, Northeast Normal University, Changchun 130024, China
| | - Menghan Chang
- School of Environment, Northeast Normal University, Changchun 130024, China
| | - Hongliang Huo
- School of Environment, Northeast Normal University, Changchun 130024, China; Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun 130117, China.
| | - Kejian Tian
- Engineering Research Center of Low-Carbon Treatment and Green Development of Polluted Water in Northeast China, Ministry of Education, Northeast Normal University, Changchun 130117, China.
| |
Collapse
|
5
|
Kirsanov RS, Khailova LS, Krasnov VS, Firsov AM, Lyamzaev KG, Panteleeva AA, Popova LB, Nazarov PA, Tashlitsky VN, Korshunova GA, Kotova EA, Antonenko YN. Spontaneous reversal of small molecule-induced mitochondrial uncoupling: the case of anilinothiophenes. FEBS J 2024; 291:5523-5539. [PMID: 39570682 DOI: 10.1111/febs.17329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/08/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024]
Abstract
Tissue specificity can render mitochondrial uncouplers more promising as leading compounds for creating drugs against serious diseases. In search of tissue-specific uncouplers, we address anilinothiophenes as possible glutathione-S-transferase substrates (GST). Earlier, 'cyclic' uncoupling activity was reported for 5-bromo-N-(4-chlorophenyl)-3,4-dinitro-2-thiophenamine (BDCT) in isolated rat liver mitochondria (RLM). The mechanism by which BDCT induced two-phase changes in mitochondrial respiration (stimulation followed by deceleration) was unknown. To clarify this issue, we synthesized BDCT and its two analogues. Among these, 5-bromo-3,4-dinitro-N-(4-nitrophenyl)-2-thiophenamine (BDNT) appeared to be the most effective as a mitochondrial uncoupler, decreasing membrane potential and stimulating respiration at submicromolar concentrations. Importantly, BDNT exerted two-phase changes in both mitochondrial membrane potential and respiration rate of RLM, which were enhanced by the addition of glutathione (GSH) but inhibited by the compounds capable of GSH depleting, such as 1-chloro-2,4-dinitrobenzene (CDNB). By contrast, the phase of recoupling was not observed in rat heart mitochondria (RHM). Remarkably, BDNT elicited mitochondrial depolarization in primary human fibroblasts but not in cultured human liver (HepG2) cells. By detecting proton-selective electrical current through planar bilayer lipid membranes, we demonstrated the ability of BDCT and BDNT to transfer protons across membranes. BDNT proved to be an anionic protonophore with a pKa of 7.38. By using LC-MS and capillary electrophoresis, we directly showed the formation of BDNT conjugates with GSH upon incubation with RLM but not RHM. Therefore, we hypothesize that GST is involved in the disappearance of the anilinothiophene uncoupling activity in RLM, ensuring the tissue-specific behavior of the uncoupler.
Collapse
Affiliation(s)
- Roman S Kirsanov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | - Ljudmila S Khailova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | - Vladimir S Krasnov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
- Department of Chemistry, Lomonosov Moscow State University, Russia
| | - Alexander M Firsov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | - Konstantin G Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
- The "Russian Clinical Research Center for Gerontology" of the Ministry of Healthcare of the Russian Federation, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alisa A Panteleeva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | - Lyudmila B Popova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | - Pavel A Nazarov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | | | - Galina A Korshunova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | - Elena A Kotova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| | - Yuri N Antonenko
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Russia
| |
Collapse
|
6
|
Wang S, Ju C, Chen M, Zhai Q, Cheng C, Zhou W, Xue L, Xu C, Tan X, Dai R. Combining untargeted and targeted metabolomics to reveal the mechanisms of herb pair Anemarrhena asphodeloides Bunge and Phellodendron chinense C. K. Schneid on benign prostatic hyperplasia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118539. [PMID: 38986754 DOI: 10.1016/j.jep.2024.118539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/26/2024] [Accepted: 07/07/2024] [Indexed: 07/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Anemarrhena asphodeloides Bunge (Ane) and Phellodendron chinense C. K. Schneid (Phe) is classical herb pair in traditional Chinese medicine, commonly used to ameliorate the symptoms of Benign Prostatic Hyperplasia (BPH). However, the mechanisms underlying this effect are remained indistinct. AIM OF THE STUDY This study aimed to clarify potential therapeutic mechanisms of herb pair on BPH from a metabolic perspective. MATERIALS AND METHODS Testosterone propionate-induced BPH rat model was established, prostatic parameters, histopathology and the levels of serum dihydrotestosterone (DHT) and testosterone (T) were used to evaluate the pharmacological effect of the herb pair on BPH. Subsequently, untargeted metabolomics of prostate tissues samples was performed by UHPLC-Q-Exactive-Orbitrap-MS, followed by multivariate statistical analysis. Targeted metabolomics by UHPLC-QQQ-MS was further utilized to verify and supplement the results of lipids and amino acids found by untargeted metabolomics, clarifying the relationship between disease, herbal pair and metabolism pathway. RESULTS The study found that Ane-Phe could relieve the progression of BPH and regulate metabolic imbalances. The levels of 13 metabolites decreased and 11 increased in prostatic tissues including glycerolphospholipid, arachidonic acid, citric acid and so on, these altered metabolites were primarily associated with TCA cycle, arachidonic acid metabolism, lipid metabolism and amino acid metabolism. Furthermore, targeted metabolomics was fulfilled to further analyze the lipid metabolism disorders, the levels of 5 lipids in serum and 21 in prostatic tissues were changed in the herb pair group compared to the model group, which closely related to glycerophospholipid, sphingolipid and glycerolipid metabolism. Besides, amino acid metabolism may be regulated by activating arginine metabolism pathway. CONCLUSIONS In this study, the combination of untargeted metabolomics and targeted metabolomics was applied to explore therapeutic mechanisms of Ane-Phe on BPH. In summary, Ane-Phe could improve the levels of endogenous metabolites by regulating multiple metabolic pathways and plays a role in energy supply, anti-inflammation and oxidative stress in BPH treatment.
Collapse
Affiliation(s)
- Shuxuan Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Caier Ju
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Meige Chen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Qirui Zhai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Cheng Cheng
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Wei Zhou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Lijuan Xue
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Chenglong Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Xiaojie Tan
- Yujing Technology Shanghai Co., Ltd, Shanghai, 200131, PR China.
| | - Ronghua Dai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| |
Collapse
|
7
|
Roos J, Manolikakes G, Schlomann U, Klinke A, Schopfer FJ, Neumann CA, Maier TJ. Nitro-fatty acids: promising agents for the development of new cancer therapeutics. Trends Pharmacol Sci 2024; 45:1061-1080. [PMID: 39490362 DOI: 10.1016/j.tips.2024.09.009] [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: 06/14/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024]
Abstract
Nitro-fatty acids (NO2-FAs) are endogenous pleiotropic lipid mediators regarded as promising drug candidates for treating inflammatory and fibrotic diseases. Over the past two decades, the anti-inflammatory and cytoprotective actions of NO2-FAs and several molecular targets have been identified. More recently, preclinical studies have demonstrated their potential as prospective cancer therapeutics with favorable safety and tumor-selective profiles. In this review, we describe the mechanisms of action, with a focus on NO2-FA antineoplastic and chemosensitizing effects. We also address the potential therapeutic applications of endogenous and structurally modified NO2-FAs species in cancer treatment.
Collapse
Affiliation(s)
- Jessica Roos
- Division of Immunology, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, 63225, Hesse, Germany.
| | - Georg Manolikakes
- Department of Chemistry, RPTU Kaiserslautern-Landau, Kaiserslautern, 67663, Rhineland-Palatinate, Germany
| | - Uwe Schlomann
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Anna Klinke
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Liver Research Center (PLRC), University of Pittsburgh, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine (C3M) University of Pittsburgh, Pittsburgh, PA, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Thorsten J Maier
- Division of Immunology, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, 63225, Hesse, Germany
| |
Collapse
|
8
|
Ma X, Jiang P, Geng J, Li X, Jin Y, Li B, Liu W. Magnetic nanoparticle-decorated metal organic frameworks for chemiluminescence detection of glutathione. Mikrochim Acta 2024; 191:618. [PMID: 39316193 DOI: 10.1007/s00604-024-06686-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024]
Abstract
A chemiluminescence (CL) method for determination of glutathione (GSH) was developed with magnetic nanoparticle-decorated metal organic frameworks (Fe3O4 NPs@Cu-TATB). The composite material was synthesized via a hydrothermal method and glutathione (GSH) can be detected by both visual and chemiluminescence (CL) methods. The synthesized Fe3O4 NPs@Cu-TATB exhibited excellent catalytic activity in the luminol-H2O2 CL system. The mechanism revealed that three types of oxygen-containing radicals (ROS) were generated in this system. As GSH can reduce the catalytic effect of generated ROS radicals, the inhibiting CL signal was produced in the Fe3O4 NPs@Cu-TATB-luminol-H2O2 system. Based on the established CL system, the detection limits for GSH using CL and visual methods were found to be 0.3 µM and 0.7 µM, respectively. This low-cost and convenient detection method can be applied to the determination of GSH content in human blood.
Collapse
Affiliation(s)
- Xiaohu Ma
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Street, Xi'an, 710119, Shaanxi, P. R. China
| | - Peiyu Jiang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Street, Xi'an, 710119, Shaanxi, P. R. China
| | - Jingbo Geng
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Street, Xi'an, 710119, Shaanxi, P. R. China
| | - Xinyi Li
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Street, Xi'an, 710119, Shaanxi, P. R. China
| | - Yan Jin
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Street, Xi'an, 710119, Shaanxi, P. R. China
| | - Baoxin Li
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Street, Xi'an, 710119, Shaanxi, P. R. China
| | - Wei Liu
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, 620 West Chang'an Street, Xi'an, 710119, Shaanxi, P. R. China.
| |
Collapse
|
9
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
10
|
Ong HW, Yang X, Smith JL, Taft-Benz S, Howell S, Dickmander RJ, Havener TM, Sanders MK, Brown JW, Couñago RM, Chang E, Krämer A, Moorman NJ, Heise M, Axtman AD, Drewry DH, Willson TM. Strategic Fluorination to Achieve a Potent, Selective, Metabolically Stable, and Orally Bioavailable Inhibitor of CSNK2. Molecules 2024; 29:4158. [PMID: 39275006 PMCID: PMC11397024 DOI: 10.3390/molecules29174158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
The host kinase casein kinase 2 (CSNK2) has been proposed to be an antiviral target against β-coronaviral infection. To pharmacologically validate CSNK2 as a drug target in vivo, potent and selective CSNK2 inhibitors with good pharmacokinetic properties are required. Inhibitors based on the pyrazolo[1,5-a]pyrimidine scaffold possess outstanding potency and selectivity for CSNK2, but bioavailability and metabolic stability are often challenging. By strategically installing a fluorine atom on an electron-rich phenyl ring of a previously characterized inhibitor 1, we discovered compound 2 as a promising lead compound with improved in vivo metabolic stability. Compound 2 maintained excellent cellular potency against CSNK2, submicromolar antiviral potency, and favorable solubility, and was remarkably selective for CSNK2 when screened against 192 kinases across the human kinome. We additionally present a co-crystal structure to support its on-target binding mode. In vivo, compound 2 was orally bioavailable, and demonstrated modest and transient inhibition of CSNK2, although antiviral activity was not observed, possibly attributed to its lack of prolonged CSNK2 inhibition.
Collapse
Affiliation(s)
- Han Wee Ong
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xuan Yang
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jeffery L. Smith
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sharon Taft-Benz
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Stefanie Howell
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rebekah J. Dickmander
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tammy M. Havener
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marcia K. Sanders
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason W. Brown
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Rafael M. Couñago
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), University of Campinas, Campinas 13083-886, SP, Brazil
| | - Edcon Chang
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Andreas Krämer
- Structural Genomics Consortium (SGC), Institute of Pharmaceutical Chemistry, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Nathaniel J. Moorman
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark Heise
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alison D. Axtman
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David H. Drewry
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Timothy M. Willson
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, USA (T.M.W.)
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
11
|
Fan T, Shen L, Huang Y, Wang X, Zhao L, Zhong R, Wang P, Sun G. Lonidamine Increases the Cytotoxic Effect of 1-[(4-Amino-2-methyl-5-pyrimidinyl)methyl]-3-(2-chloroethyl)-3-nitrosourea via Energy Inhibition, Disrupting Redox Homeostasis, and Downregulating MGMT Expression in Human Lung Cancer Cell Line. ACS OMEGA 2024; 9:36134-36147. [PMID: 39220482 PMCID: PMC11360010 DOI: 10.1021/acsomega.4c00641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/30/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Lung cancer ranks as the second most diagnosed cancer and the leading cause of cancer-related deaths worldwide. Novel chemotherapeutic strategies are crucial to efficiently target tumor cells while minimizing toxicity to normal cells. In this study, we proposed a combination strategy using energy blocker lonidamine (LND) and cytotoxic drug nimustine (ACNU, 1-[(4-amino-2-methyl-5-pyrimidinyl)methyl]-3-(2-chloroethyl)-3-nitrosourea) to enhance the killing of a human lung cancer cell line and investigated the potential chemo-sensitizing mechanism of LND. LND was found to remarkably increase the cytotoxicity of ACNU to A549 and H1299 cells without significantly affecting normal lung BEAS2B cells. The combination of LND and ACNU also produced significant effects on cell apoptosis, colony formation, cell migration, and invasion assays compared to single drug treatment. Mechanistically, LND decreased intracellular ATP levels by inhibiting glycolysis and inducing mitochondrial dysfunction. Furthermore, the combination of LND and ACNU could intensify cellular oxidative stress, decrease cellular GSH contents, and increase reactive oxygen species (ROS) production. Notably, LND alone dramatically downregulated the expression of DNA repair protein MGMT (O6-methylguanine-DNA methyltransferase), enhancing DNA interstrand cross-link formation induced by ACNU. Overall, LND represents a potential chemo-sensitizer to enhance ACNU therapy through energy inhibition, disrupting redox homeostasis and downregulating MGMT expression in human lung cancer cell line under preclinical and clinical background.
Collapse
Affiliation(s)
- Tengjiao Fan
- Department
of Medical Technology, Beijing Pharmaceutical
University of Staff and Workers, Beijing 100079, P. R. China
- Beijing
Key Laboratory of Environment & Viral Oncology, College of Chemistry
and Life Science, Beijing University of
Technology, Beijing 100124, P. R. China
| | - Lin Shen
- Department
of Dermatology, the First Medical Center of PLA General Hospital, Beijing 100853, P. R. China
| | - Yaxin Huang
- Beijing
Key Laboratory of Environment & Viral Oncology, College of Chemistry
and Life Science, Beijing University of
Technology, Beijing 100124, P. R. China
| | - Xin Wang
- Department
of Clinical Trials Center, National Cancer Center/National Clinical
Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100029, P. R. China
| | - Lijiao Zhao
- Beijing
Key Laboratory of Environment & Viral Oncology, College of Chemistry
and Life Science, Beijing University of
Technology, Beijing 100124, P. R. China
| | - Rugang Zhong
- Beijing
Key Laboratory of Environment & Viral Oncology, College of Chemistry
and Life Science, Beijing University of
Technology, Beijing 100124, P. R. China
| | - Peng Wang
- Department
of Neurosurgery, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, P. R. China
| | - Guohui Sun
- Beijing
Key Laboratory of Environment & Viral Oncology, College of Chemistry
and Life Science, Beijing University of
Technology, Beijing 100124, P. R. China
| |
Collapse
|
12
|
Nakken CL, Berntssen MHG, Meier S, Bijlsma L, Mjøs SA, Sørhus E, Donald CE. Exposure of Polycyclic Aromatic Hydrocarbons (PAHs) and Crude Oil to Atlantic Haddock ( Melanogrammus aeglefinus): A Unique Snapshot of the Mercapturic Acid Pathway. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:14855-14863. [PMID: 39101928 PMCID: PMC11340023 DOI: 10.1021/acs.est.4c05112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/06/2024]
Abstract
Fish exposed to xenobiotics like petroleum-derived polycyclic aromatic hydrocarbons (PAHs) will immediately initiate detoxification systems through effective biotransformation reactions. Yet, there is a discrepancy between recognized metabolic pathways and the actual metabolites detected in fish following PAH exposure like oil pollution. To deepen our understanding of PAH detoxification, we conducted experiments exposing Atlantic haddock (Melanogrammus aeglefinus) to individual PAHs or complex oil mixtures. Bile extracts, analyzed by using an ion mobility quadrupole time-of-flight mass spectrometer, revealed novel metabolites associated with the mercapturic acid pathway. A dominant spectral feature recognized as PAH thiols set the basis for a screening strategy targeting (i) glutathione-, (ii) cysteinylglycine-, (iii) cysteine-, and (iv) mercapturic acid S-conjugates. Based on controlled single-exposure experiments, we constructed an interactive library of 33 metabolites originating from 8 PAHs (anthracene, phenanthrene, 1-methylphenanthrene, 1,4-dimethylphenanthrene, chrysene, benz[a]anthracene, benzo[a]pyrene, and dibenz[a,h]anthracene). By incorporation of the library in the analysis of samples from crude oil exposed fish, PAHs conjugated with glutathione and cysteinylglycine were uncovered. This qualitative study offers an exclusive glimpse into the rarely acknowledged mercapturic acid detoxification pathway in fish. Furthermore, this furnishes evidence that this metabolic pathway also succeeds for PAHs in complex pollution sources, a notable discovery not previously reported.
Collapse
Affiliation(s)
- Charlotte L. Nakken
- Department
of Chemistry, University of Bergen, Bergen 5007, Norway
- Marine
Toxicology, Institute of Marine Research, Bergen 5817, Norway
| | | | - Sonnich Meier
- Marine
Toxicology, Institute of Marine Research, Bergen 5817, Norway
| | - Lubertus Bijlsma
- Environmental
and Public Health Analytical Chemistry, Research Institute for Pesticides and Water, University Jaume I, Castellón 12071, Spain
| | - Svein A. Mjøs
- Department
of Chemistry, University of Bergen, Bergen 5007, Norway
| | - Elin Sørhus
- Marine
Toxicology, Institute of Marine Research, Bergen 5817, Norway
| | - Carey E. Donald
- Marine
Toxicology, Institute of Marine Research, Bergen 5817, Norway
| |
Collapse
|
13
|
Federici L, Masulli M, De Laurenzi V, Allocati N. The Role of S-Glutathionylation in Health and Disease: A Bird's Eye View. Nutrients 2024; 16:2753. [PMID: 39203889 PMCID: PMC11357436 DOI: 10.3390/nu16162753] [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: 07/29/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Protein glutathionylation is a reversible post-translational modification that involves the attachment of glutathione to cysteine residues. It plays a role in the regulation of several cellular processes and protection against oxidative damage. Glutathionylation (GS-ylation) modulates protein function, inhibits or enhances enzymatic activity, maintains redox homeostasis, and shields several proteins from irreversible oxidative stress. Aberrant GS-ylation patterns are thus implicated in various diseases, particularly those associated with oxidative stress and inflammation, such as cardiovascular diseases, neurodegenerative disorders, cancer, and many others. Research in the recent years has highlighted the potential to manipulate protein GS-ylation for therapeutic purposes with strategies that imply both its enhancement and inhibition according to different cases. Moreover, it has become increasingly evident that monitoring the GS-ylation status of selected proteins offers diagnostic potential in different diseases. In this review, we try to summarize recent research in the field with a focus on our current understanding of the molecular mechanisms related to aberrant protein GS-ylation.
Collapse
Affiliation(s)
- Luca Federici
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’ Annunzio”, 66100 Chieti, Italy; (L.F.); (M.M.); (V.D.L.)
- CAST (Center for Advanced Studies and Technology), University “G. d’ Annunzio”, 66100 Chieti, Italy
| | - Michele Masulli
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’ Annunzio”, 66100 Chieti, Italy; (L.F.); (M.M.); (V.D.L.)
| | - Vincenzo De Laurenzi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’ Annunzio”, 66100 Chieti, Italy; (L.F.); (M.M.); (V.D.L.)
- CAST (Center for Advanced Studies and Technology), University “G. d’ Annunzio”, 66100 Chieti, Italy
| | - Nerino Allocati
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’ Annunzio”, 66100 Chieti, Italy; (L.F.); (M.M.); (V.D.L.)
| |
Collapse
|
14
|
Koirala M, DiPaola M. Overcoming Cancer Resistance: Strategies and Modalities for Effective Treatment. Biomedicines 2024; 12:1801. [PMID: 39200265 PMCID: PMC11351918 DOI: 10.3390/biomedicines12081801] [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: 07/11/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Resistance to cancer drugs is a complex phenomenon that poses a significant challenge in the treatment of various malignancies. This review comprehensively explores cancer resistance mechanisms and discusses emerging strategies and modalities to overcome this obstacle. Many factors contribute to cancer resistance, including genetic mutations, activation of alternative signaling pathways, and alterations in the tumor microenvironment. Innovative approaches, such as targeted protein degradation, immunotherapy combinations, precision medicine, and novel drug delivery systems, hold promise for improving treatment outcomes. Understanding the intricacies of cancer resistance and leveraging innovative modalities are essential for advancing cancer therapy.
Collapse
|
15
|
An X, Yu W, Liu J, Tang D, Yang L, Chen X. Oxidative cell death in cancer: mechanisms and therapeutic opportunities. Cell Death Dis 2024; 15:556. [PMID: 39090114 PMCID: PMC11294602 DOI: 10.1038/s41419-024-06939-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Reactive oxygen species (ROS) are highly reactive oxygen-containing molecules generated as natural byproducts during cellular processes, including metabolism. Under normal conditions, ROS play crucial roles in diverse cellular functions, including cell signaling and immune responses. However, a disturbance in the balance between ROS production and cellular antioxidant defenses can lead to an excessive ROS buildup, causing oxidative stress. This stress damages essential cellular components, including lipids, proteins, and DNA, potentially culminating in oxidative cell death. This form of cell death can take various forms, such as ferroptosis, apoptosis, necroptosis, pyroptosis, paraptosis, parthanatos, and oxeiptosis, each displaying distinct genetic, biochemical, and signaling characteristics. The investigation of oxidative cell death holds promise for the development of pharmacological agents that are used to prevent tumorigenesis or treat established cancer. Specifically, targeting key antioxidant proteins, such as SLC7A11, GCLC, GPX4, TXN, and TXNRD, represents an emerging approach for inducing oxidative cell death in cancer cells. This review provides a comprehensive summary of recent progress, opportunities, and challenges in targeting oxidative cell death for cancer therapy.
Collapse
Affiliation(s)
- Xiaoqin An
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Wenfeng Yu
- Provincial Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, PR China
| | - Jinbao Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Li Yang
- Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, PR China.
| | - Xin Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
16
|
Ong HW, Yang X, Smith JL, Dickmander RJ, Brown JW, Havener TM, Taft-Benz S, Howell S, Sanders MK, Capener JL, Couñago RM, Chang E, Krämer A, Moorman NJ, Heise M, Axtman AD, Drewry DH, Willson TM. More than an Amide Bioisostere: Discovery of 1,2,4-Triazole-containing Pyrazolo[1,5- a]pyrimidine Host CSNK2 Inhibitors for Combatting β-Coronavirus Replication. J Med Chem 2024; 67:12261-12313. [PMID: 38959455 DOI: 10.1021/acs.jmedchem.4c00962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The pyrazolo[1,5-a]pyrimidine scaffold is a promising scaffold to develop potent and selective CSNK2 inhibitors with antiviral activity against β-coronaviruses. Herein, we describe the discovery of a 1,2,4-triazole group to substitute a key amide group for CSNK2 binding present in many potent pyrazolo[1,5-a]pyrimidine inhibitors. Crystallographic evidence demonstrates that the 1,2,4-triazole replaces the amide in forming key hydrogen bonds with Lys68 and a water molecule buried in the ATP-binding pocket. This isosteric replacement improves potency and metabolic stability at a cost of solubility. Optimization for potency, solubility, and metabolic stability led to the discovery of the potent and selective CSNK2 inhibitor 53. Despite excellent in vitro metabolic stability, rapid decline in plasma concentration of 53 in vivo was observed and may be attributed to lung accumulation, although in vivo pharmacological effect was not observed. Further optimization of this novel chemotype may validate CSNK2 as an antiviral target in vivo.
Collapse
Affiliation(s)
- Han Wee Ong
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Xuan Yang
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jeffery L Smith
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rebekah J Dickmander
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jason W Brown
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Tammy M Havener
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Sharon Taft-Benz
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Stefanie Howell
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Marcia K Sanders
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jacob L Capener
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rafael M Couñago
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Centro de Química Medicinal (CQMED), Centro de Biologia Molecular e Engenharia Genética (CBMEG), University of Campinas, Campinas, São Paulo 13083-886, Brazil
| | - Edcon Chang
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Andreas Krämer
- SGC, Institute of Pharmaceutical Chemistry, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Nathaniel J Moorman
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mark Heise
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Alison D Axtman
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - David H Drewry
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Timothy M Willson
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Structural Genomics Consortium (SGC) and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
17
|
Naughton KJ, Song X, Childress AR, Skaggs EM, Byrd AL, Gosser CM, Esoe DP, DuCote TJ, Plaugher DR, Lukyanchuk A, Goettl RA, Liu J, Brainson CF. Methionine Restriction Reduces Lung Cancer Progression and Increases Chemotherapy Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.599795. [PMID: 38979225 PMCID: PMC11230185 DOI: 10.1101/2024.06.25.599795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Targeting tumor metabolism through dietary interventions is an area of growing interest, and may help to improve the significant mortality of aggressive cancers, including non-small cell lung cancer (NSCLC). Here we show that the restriction of methionine in the aggressive KRAS/Lkb1-mutant NSCLC autochthonous mouse model drives decreased tumor progression and increased carboplatin treatment efficacy. Importantly, methionine restriction during early stages of tumorigenesis prevents the lineage switching known to occur in the model, and alters the tumor immune microenvironment (TIME) to have fewer tumor-infiltrating neutrophils. Mechanistically, mutations in LKB1 are linked to anti-oxidant production through changes to cystathionine-β-synthase (CBS) expression. Human cell lines with rescued LKB1 show increased CBS levels and resistance to carboplatin, which can be partially rescued by methionine restriction. Furthermore, LKB1 rescued cells, but not mutant cells, show less G2-M arrest and apoptosis in high methionine conditions. Knock-down of CBS sensitized both LKB1 mutant and non-mutated lines to carboplatin, again rescuing the carboplatin resistance of the LKB1 rescued lines. Given that immunotherapy is commonly combined with chemotherapy for NSCLC, we next wanted to understand if T cells are impaired by MR. Therefore, we examined the ability of T cells from MR and control tumor bearing mice to proliferate in culture and found that T cells from MR treated mice had no defects in proliferation, even though we continued the MR conditions ex vivo. We also identified that CBS is most highly correlated with smoking, adenocarcinomas with alveolar and bronchiolar features, and adenosquamous cell carcinomas, implicating its roles in oxidative stress response and lineage fate in human tumors. Taken together, we have shown the importance of MR as a dietary intervention to slow tumor growth and improve treatment outcomes for NSCLC.
Collapse
Affiliation(s)
- Kassandra J Naughton
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Xiulong Song
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Avery R Childress
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Erika M Skaggs
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Aria L Byrd
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Christian M Gosser
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Dave-Preston Esoe
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Tanner J DuCote
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Daniel R Plaugher
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Alexsandr Lukyanchuk
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
| | - Ryan A Goettl
- Markey Cancer Center, University of Kentucky, Lexington KY 40536
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington KY 40536
- Department of Internal Medicine, University of Kentucky, Lexington KY 40536
| | - Christine F Brainson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington KY 40536
- Markey Cancer Center, University of Kentucky, Lexington KY 40536
- Corresponding author
| |
Collapse
|
18
|
Wong G, Wu SY, Chen WM, Hsu PJ, Chou TC, Chiang MF, Wu MS, Lee MC, Soong RS. Effects of N-acetylcysteine on hepatocellular carcinoma in chronic hepatitis C. Am J Cancer Res 2024; 14:3533-3544. [PMID: 39113878 PMCID: PMC11301300 DOI: 10.62347/mtlw1449] [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: 05/07/2024] [Accepted: 06/19/2024] [Indexed: 08/10/2024] Open
Abstract
Hepatitis C virus (HCV) infection significantly contributes to global hepatocellular carcinoma (HCC) incidence. N-Acetylcysteine (NAC), known for its antioxidant properties, is a potential therapeutic agent. However, evidence on its efficacy in reducing HCC risk among HCV patients is limited. A retrospective cohort analysis using Taiwan's National Health Insurance Research Database (2008-2018) included ≥18-year-old HCV patients. NAC usage (≥28 cumulative defined daily doses [cDDDs]) was assessed for its association with HCC risk using Cox regression models and propensity score matching. The study comprised 269,647 HCV patients, with detailed NAC dosage characterization and hazard ratios (HRs) for HCC risk. Post-matching, NAC usage emerged as the significant predictor of reduced HCC risk (adjusted HR: 0.39, 95% CI: 0.37-0.41, P<0.0001). Dose-response analysis showed reduced HCC risk with increasing cDDDs of NAC (P<0.0001). Higher daily NAC dosage (≥1 DDD) was associated with significantly lower HCC risk (adjusted HR: 0.33, 95% CI: 0.31-0.36, P<0.0001). The study provides compelling evidence for NAC's potential in reducing HCC risk among HCV patients. Insights into dose-dependent effects and optimal daily intensity thresholds offer valuable directions for future therapeutic strategies and clinical trials targeting HCC burden in HCV-infected individuals.
Collapse
Affiliation(s)
- Gary Wong
- Division of General Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical UniversityNo. 111, Sec. 3, Xinglong Road, Wenshan District, Taipei 116, Taiwan
| | - Szu-Yuan Wu
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic UniversityNew Taipei 242, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic UniversityNew Taipei 242, Taiwan
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia UniversityTaichung 413, Taiwan
- Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
- Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia UniversityTaichung 413, Taiwan
- Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical UniversityTaipei 110, Taiwan
| | - Wan-Ming Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic UniversityNew Taipei 242, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic UniversityNew Taipei 242, Taiwan
| | - Po-Jung Hsu
- Division of General Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical UniversityNo. 111, Sec. 3, Xinglong Road, Wenshan District, Taipei 116, Taiwan
- College of Medicine, Taipei Medical UniversityNo. 250 Wu-Hsing Street, Taipei 110, Taiwan
- Division of Transplantation Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical UniversityTaipei 116, Taiwan
- TMU Research Center for Organ Transplantation, College of Medicine, Taipei Medical UniversityTaipei 110, Taiwan
- Taipei Cancer Center, Taipei Medical UniversityTaipei 110, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
| | - Ta-Chun Chou
- Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical UniversityTaipei 110, Taiwan
- Division of Transplantation Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical UniversityTaipei 116, Taiwan
- TMU Research Center for Organ Transplantation, College of Medicine, Taipei Medical UniversityTaipei 110, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
| | - Ming-Feng Chiang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
| | - Ming-Shun Wu
- College of Medicine, Taipei Medical UniversityNo. 250 Wu-Hsing Street, Taipei 110, Taiwan
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical UniversityTaipei 116, Taiwan
| | - Ming-Che Lee
- College of Medicine, Taipei Medical UniversityNo. 250 Wu-Hsing Street, Taipei 110, Taiwan
- Division of Transplantation Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical UniversityTaipei 116, Taiwan
- TMU Research Center for Organ Transplantation, College of Medicine, Taipei Medical UniversityTaipei 110, Taiwan
- Taipei Cancer Center, Taipei Medical UniversityTaipei 110, Taiwan
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical UniversityNo. 291, Jhongjheng Road, Jhonghe, New Taipei 23561, Taiwan
| | - Ruey-Shyang Soong
- Division of General Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical UniversityNo. 111, Sec. 3, Xinglong Road, Wenshan District, Taipei 116, Taiwan
- College of Medicine, Taipei Medical UniversityNo. 250 Wu-Hsing Street, Taipei 110, Taiwan
- Division of Transplantation Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical UniversityTaipei 116, Taiwan
- TMU Research Center for Organ Transplantation, College of Medicine, Taipei Medical UniversityTaipei 110, Taiwan
- Taipei Cancer Center, Taipei Medical UniversityTaipei 110, Taiwan
| |
Collapse
|
19
|
Himmelbauer M, Bajrami B, Basile R, Capacci A, Chen T, Choi CK, Gilfillan R, Gonzalez-Lopez de Turiso F, Gu C, Hoemberger M, Johnson DS, Jones JH, Kadakia E, Kirkland M, Lin EY, Liu Y, Ma B, Magee T, Mantena S, Marx IE, Metrick CM, Mingueneau M, Murugan P, Muste CA, Nadella P, Nevalainen M, Parker Harp CR, Pattaropong V, Pietrasiewicz A, Prince RJ, Purgett TJ, Santoro JC, Schulz J, Sciabola S, Tang H, Vandeveer HG, Wang T, Yousaf Z, Helal CJ, Hopkins BT. Discovery and Preclinical Characterization of BIIB129, a Covalent, Selective, and Brain-Penetrant BTK Inhibitor for the Treatment of Multiple Sclerosis. J Med Chem 2024; 67:8122-8140. [PMID: 38712838 PMCID: PMC11129193 DOI: 10.1021/acs.jmedchem.4c00220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 05/08/2024]
Abstract
Multiple sclerosis (MS) is a chronic disease with an underlying pathology characterized by inflammation-driven neuronal loss, axonal injury, and demyelination. Bruton's tyrosine kinase (BTK), a nonreceptor tyrosine kinase and member of the TEC family of kinases, is involved in the regulation, migration, and functional activation of B cells and myeloid cells in the periphery and the central nervous system (CNS), cell types which are deemed central to the pathology contributing to disease progression in MS patients. Herein, we describe the discovery of BIIB129 (25), a structurally distinct and brain-penetrant targeted covalent inhibitor (TCI) of BTK with an unprecedented binding mode responsible for its high kinome selectivity. BIIB129 (25) demonstrated efficacy in disease-relevant preclinical in vivo models of B cell proliferation in the CNS, exhibits a favorable safety profile suitable for clinical development as an immunomodulating therapy for MS, and has a low projected total human daily dose.
Collapse
Affiliation(s)
- Martin
K. Himmelbauer
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Bekim Bajrami
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Rebecca Basile
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Andrew Capacci
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - TeYu Chen
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Colin K. Choi
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Rab Gilfillan
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | | | - Chungang Gu
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Marc Hoemberger
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Douglas S. Johnson
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - J. Howard Jones
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ekta Kadakia
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Melissa Kirkland
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Edward Y. Lin
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ying Liu
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Bin Ma
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Tom Magee
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Srinivasa Mantena
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Isaac E. Marx
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Claire M. Metrick
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Michael Mingueneau
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Paramasivam Murugan
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Cathy A. Muste
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Prasad Nadella
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Marta Nevalainen
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Chelsea R. Parker Harp
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Vatee Pattaropong
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Alicia Pietrasiewicz
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Robin J. Prince
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Thomas J. Purgett
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Joseph C. Santoro
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Jurgen Schulz
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Simone Sciabola
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Hao Tang
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - H. George Vandeveer
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ti Wang
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Zain Yousaf
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Christopher J. Helal
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Brian T. Hopkins
- Biogen Research and Development, 225 Binney Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
20
|
Song S, Zhang X, Cui L, Wang Y, Tian X, Wang K, Ji K. Mechanisms of lipopolysaccharide protection in tumor drug-induced macrophage damage. Int J Biol Macromol 2024; 266:131006. [PMID: 38522696 DOI: 10.1016/j.ijbiomac.2024.131006] [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: 12/26/2023] [Revised: 03/04/2024] [Accepted: 03/17/2024] [Indexed: 03/26/2024]
Abstract
Malignant tumors contribute significantly to human mortality. Chemotherapy is a commonly used treatment for tumors. However, due to the low selectivity of chemotherapeutic drugs, immune cells can be damaged during antitumor treatment, resulting in toxicity. Lipopolysaccharide (LPS) can stimulate immune cells to respond to foreign substances. Here, we found that 10 ng/mL LPS could induce tolerance to antitumor drugs in macrophages without altering the effect of the drugs on tumor cells. Differentially expressed genes (DEGs) were identified between cells before and after LPS administration using transcriptome sequencing and found to be mainly associated with ATP-binding cassette (ABC)-resistant transporters and glutathione S-transferase (GST). LPS was shown by qRT-PCR and western blotting to promote the expression of ABCC1, GSTT1, and GSTP1 by 38.3 %, 194.8 %, and 27.0 %. Furthermore, three inhibitors (inhibitors of GST, glutathione synthesis, and ABCC1) were used for further investigation, showing that these inhibitors reduced macrophage survival rates by 44.0 %, 52.3 %, and 43.3 %, while the intracellular adriamycin content increased by 28.9 %, 42.9 %, and 51.3 %, respectively. These findings suggest that the protective mechanism of LPS on macrophages is associated with increased GST activity, the consumption of glutathione, and increased expression of ABCC1 protein. Therefore, LPS has a potential role in enhancing immunity.
Collapse
Affiliation(s)
- Shuliang Song
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Xiao Zhang
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Lei Cui
- Pharmacy Department, Yellow Sea Road Street Community Health Service Center, YanTai, Shandong, 264000, China
| | - Yan Wang
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Xiao Tian
- Marine College, Shandong University, Weihai, Shandong 264209, China.
| | - Ke Wang
- Pharmacy Department, Heping Hospital Affiliated to Changzhi Medical College, Changzhi 046500, China.
| | - Kai Ji
- Department of Plastic Surgery, China-Japan Friendship Hospital, Beijing 100029, China.
| |
Collapse
|
21
|
Li Y, Han D, Luo Z, Lv X, Liu B. The Chan-Lam-type synthesis of thioimidazolium salts for thiol-(hetero)arene conjugation. Chem Commun (Camb) 2024; 60:4675-4678. [PMID: 38591667 DOI: 10.1039/d4cc00704b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
The design of stable and variable aryl linkers for conjugating drug moieties to the metabolism-related thiols is of importance in drug discovery. We disclosed that thioimidazolium groups are unique scaffolds for the thiol-(hetero)arene conjugation under mild conditions. The drug bound thioimidazolium salts, which are easily accessible via a copper-mediated Chan-Lam process in gram-scale, could be successfully applied to the late-stage coupling of bioactive thiols to construct a broad array of drug-like molecules.
Collapse
Affiliation(s)
- Yue Li
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China.
| | - Dongchang Han
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China.
| | - Zhibin Luo
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China.
| | - Xiaomeng Lv
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China.
| | - Bin Liu
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
22
|
Iori S, D'Onofrio C, Laham-Karam N, Mushimiyimana I, Lucatello L, Lopparelli RM, Gelain ME, Capolongo F, Pauletto M, Dacasto M, Giantin M. Establishment and characterization of cytochrome P450 1A1 CRISPR/Cas9 Knockout Bovine Foetal Hepatocyte Cell Line (BFH12). Cell Biol Toxicol 2024; 40:18. [PMID: 38528259 PMCID: PMC10963470 DOI: 10.1007/s10565-024-09856-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
The cytochrome P450 1A (CYP1A) subfamily of xenobiotic metabolizing enzymes (XMEs) consists of two different isoforms, namely CYP1A1 and CYP1A2, which are highly conserved among species. These two isoenzymes are involved in the biotransformation of many endogenous compounds as well as in the bioactivation of several xenobiotics into carcinogenic derivatives, thereby increasing the risk of tumour development. Cattle (Bos taurus) are one of the most important food-producing animal species, being a significant source of nutrition worldwide. Despite daily exposure to xenobiotics, data on the contribution of CYP1A to bovine hepatic metabolism are still scarce. The CRISPR/Cas9-mediated knockout (KO) is a useful method for generating in vivo and in vitro models for studying xenobiotic biotransformations. In this study, we applied the ribonucleoprotein (RNP)-complex approach to successfully obtain the KO of CYP1A1 in a bovine foetal hepatocyte cell line (BFH12). After clonal expansion and selection, CYP1A1 excision was confirmed at the DNA, mRNA and protein level. Therefore, RNA-seq analysis revealed significant transcriptomic changes associated with cell cycle regulation, proliferation, and detoxification processes as well as on iron, lipid and mitochondrial homeostasis. Altogether, this study successfully generates a new bovine CYP1A1 KO in vitro model, representing a valuable resource for xenobiotic metabolism studies in this important farm animal species.
Collapse
Affiliation(s)
- Silvia Iori
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Caterina D'Onofrio
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Nihay Laham-Karam
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Neulaniementie 2, 70211, Kuopio, Finland
| | - Isidore Mushimiyimana
- University of Eastern Finland, A.I. Virtanen Institute for Molecular Sciences, Neulaniementie 2, 70211, Kuopio, Finland
| | - Lorena Lucatello
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Rosa Maria Lopparelli
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Maria Elena Gelain
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Francesca Capolongo
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Mauro Dacasto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy
| | - Mery Giantin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale Dell'Università 16, Legnaro, 35020, Padua, Italy.
| |
Collapse
|
23
|
Soong TH, Hotze C, Khandelwal NK, Tomasiak TM. Structural Basis for Oxidized Glutathione Recognition by the Yeast Cadmium Factor 1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578287. [PMID: 38352558 PMCID: PMC10862839 DOI: 10.1101/2024.01.31.578287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Transporters from the ABCC family have an essential role in detoxifying electrophilic compounds including metals, drugs, and lipids, often through conjugation with glutathione complexes. The Yeast Cadmium Factor 1 (Ycf1) transports glutathione alone as well as glutathione conjugated to toxic heavy metals including Cd2+, Hg2+, and As3+. To understand the complicated selectivity and promiscuity of heavy metal substrate binding, we determined the cryo-EM structure of Ycf1 bound to the substrate, oxidized glutathione. We systematically tested binding determinants with cellular survival assays against cadmium to determine how the substrate site accommodates different-sized metal complexes. We identify a "flex-pocket" for substrate binding that binds glutathione complexes asymmetrically and flexes to accommodate different size complexes.
Collapse
Affiliation(s)
- Tik Hang Soong
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Clare Hotze
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Nitesh Kumar Khandelwal
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
- Department of Biochemistry and Physics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Thomas M Tomasiak
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
24
|
Tahir R, Samra, Afzal F, Liang J, Yang S. Novel protective aspects of dietary polyphenols against pesticidal toxicity and its prospective application in rice-fish mode: A Review. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109418. [PMID: 38301811 DOI: 10.1016/j.fsi.2024.109418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
The rice fish system represents an innovative and sustainable approach to integrated farming, combining rice cultivation with fish rearing in the same ecosystem. However, one of the major challenges in this system is the pesticidal pollution resulting from various sources, which poses risks to fish health and overall ecosystem balance. In recent years, dietary polyphenols have emerged as promising bioactive compounds with potential chemo-preventive and therapeutic properties. These polyphenols, derived from various plant sources, have shown great potential in reducing the toxicity of pesticides and improving the health of fish within the rice fish system. This review aims to explore the novel aspects of using dietary polyphenols to mitigate pesticidal toxicity and enhance fish health in the rice fish system. It provides comprehensive insights into the mechanisms of action of dietary polyphenols and their beneficial effects on fish health, including antioxidant, anti-inflammatory, and detoxification properties. Furthermore, the review discusses the potential application methods of dietary polyphenols, such as direct supplementation in fish diets or through incorporation into the rice fields. By understanding the interplay between dietary polyphenols and pesticides in the rice fish system, researchers can develop innovative and sustainable strategies to promote fish health, minimize pesticide impacts, and ensure the long-term viability of this integrated farming approach. The information presented in this review will be valuable for scientists, aqua-culturists, and policymakers aiming to implement eco-friendly and health-enhancing practices in the rice fish system.
Collapse
Affiliation(s)
- Rabia Tahir
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, Punjab, 63100, Pakistan
| | - Samra
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Fozia Afzal
- Department of Zoology, The Islamia University of Bahawalpur, Bahawalpur, Punjab, 63100, Pakistan
| | - Ji Liang
- School of Humanities, Universiti Sains Malaysia, Minden, Penang, 11800, Malaysia
| | - Song Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
25
|
Sharma NK, Bahot A, Sekar G, Bansode M, Khunteta K, Sonar PV, Hebale A, Salokhe V, Sinha BK. Understanding Cancer's Defense against Topoisomerase-Active Drugs: A Comprehensive Review. Cancers (Basel) 2024; 16:680. [PMID: 38398072 PMCID: PMC10886629 DOI: 10.3390/cancers16040680] [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: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, the emergence of cancer drug resistance has been one of the crucial tumor hallmarks that are supported by the level of genetic heterogeneity and complexities at cellular levels. Oxidative stress, immune evasion, metabolic reprogramming, overexpression of ABC transporters, and stemness are among the several key contributing molecular and cellular response mechanisms. Topo-active drugs, e.g., doxorubicin and topotecan, are clinically active and are utilized extensively against a wide variety of human tumors and often result in the development of resistance and failure to therapy. Thus, there is an urgent need for an incremental and comprehensive understanding of mechanisms of cancer drug resistance specifically in the context of topo-active drugs. This review delves into the intricate mechanistic aspects of these intracellular and extracellular topo-active drug resistance mechanisms and explores the use of potential combinatorial approaches by utilizing various topo-active drugs and inhibitors of pathways involved in drug resistance. We believe that this review will help guide basic scientists, pre-clinicians, clinicians, and policymakers toward holistic and interdisciplinary strategies that transcend resistance, renewing optimism in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Nilesh Kumar Sharma
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Anjali Bahot
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Gopinath Sekar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Mahima Bansode
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Kratika Khunteta
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Priyanka Vijay Sonar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Ameya Hebale
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Vaishnavi Salokhe
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Birandra Kumar Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
26
|
Bhattacharya K, Mahato S, Deka S, Chanu NR, Shrivastava AK, Khanal P. Netting into the Sophoretin pool: An approach to trace GSTP1 inhibitors for reversing chemoresistance. Comput Biol Chem 2024; 108:107981. [PMID: 37976621 DOI: 10.1016/j.compbiolchem.2023.107981] [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: 09/27/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Chemoresistance, a significant challenge in cancer treatment, is often associated with the cellular glutathione-related detoxification system. The GSTP1 isoenzyme (glutathione S-transferases) plays a critical role in the cytoplasmic inactivation of anticancer drugs. This suggests the identification of GSTP1 inhibitors to combat chemoresistance. We screened Sophoretin (also called quercetin) derivatives for molecular properties, pharmacokinetics, and toxicity profiles. Following that, we conducted molecular docking and simulations between selected derivatives and GSTP1. The best-docked complex, GSTP1-quercetin 7-O-β-D-glucoside, exhibited a binding affinity of -8.1 kcal/mol, with no predicted toxicity and good pharmacokinetic properties. Molecular dynamics simulations confirmed the stability of this complex. Quercetin 7-O-β-D-glucoside shows promise as a lead candidate for addressing chemoresistance in cancer patients, although further experimental studies are needed to validate its efficacy and therapeutic potential.
Collapse
Affiliation(s)
- Kunal Bhattacharya
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam 781026, India; Royal School of Pharmacy, The Assam Royal Global University, Assam 781035, India.
| | - Shikha Mahato
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam 781026, India
| | - Satyendra Deka
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam 781026, India
| | - Nongmaithem Randhoni Chanu
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam 781026, India; Faculty of Pharmaceutical Science, Assam Downtown University, Assam, India
| | - Amit Kumar Shrivastava
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicine Research Institute, Wonkwang University, Iksan 570-749, Republic of Korea
| | - Pukar Khanal
- Department of Pharmacology and Toxicology, KLE College of Pharmacy, Belagavi, KLE Academy of Higher Education and Research (KAHER), Belagavi 590010, India.
| |
Collapse
|
27
|
Meshanni JA, Lee JM, Vayas KN, Sun R, Jiang C, Guo GL, Gow AJ, Laskin JD, Laskin DL. Suppression of Lung Oxidative Stress, Inflammation, and Fibrosis following Nitrogen Mustard Exposure by the Selective Farnesoid X Receptor Agonist Obeticholic Acid. J Pharmacol Exp Ther 2024; 388:586-595. [PMID: 37188530 PMCID: PMC10801770 DOI: 10.1124/jpet.123.001557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/26/2023] [Accepted: 04/22/2023] [Indexed: 05/17/2023] Open
Abstract
Nitrogen mustard (NM) is a cytotoxic vesicant known to cause pulmonary injury that can progress to fibrosis. NM toxicity is associated with an influx of inflammatory macrophages in the lung. Farnesoid X receptor (FXR) is a nuclear receptor involved in bile acid and lipid homeostasis that has anti-inflammatory activity. In these studies, we analyzed the effects of FXR activation on lung injury, oxidative stress, and fibrosis induced by NM. Male Wistar rats were exposed to phosphate-buffered saline (vehicle control) or NM (0.125 mg/kg) by intratracheal Penncentury-MicroSprayer aerosolization; this was followed by treatment with the FXR synthetic agonist, obeticholic acid (OCA, 15 mg/kg), or vehicle control (0.13-0.18 g peanut butter) 2 hours later and then once per day, 5 days per week thereafter for 28 days. NM caused histopathological changes in the lung, including epithelial thickening, alveolar circularization, and pulmonary edema. Picrosirius red staining and lung hydroxyproline content were increased, indicative of fibrosis; foamy lipid-laden macrophages were also identified in the lung. This was associated with aberrations in pulmonary function, including increases in resistance and hysteresis. Following NM exposure, lung expression of HO-1 and iNOS, and the ratio of nitrates/nitrites in bronchoalveolar lavage fluid (BAL), markers of oxidative stress increased, along with BAL levels of inflammatory proteins, fibrinogen, and sRAGE. Administration of OCA attenuated NM-induced histopathology, oxidative stress, inflammation, and altered lung function. These findings demonstrate that FXR plays a role in limiting NM-induced lung injury and chronic disease, suggesting that activating FXR may represent an effective approach to limiting NM-induced toxicity. SIGNIFICANCE STATEMENT: In this study, the role of farnesoid-X-receptor (FXR) in mustard vesicant-induced pulmonary toxicity was analyzed using nitrogen mustard (NM) as a model. This study's findings that administration of obeticholic acid, an FXR agonist, to rats reduces NM-induced pulmonary injury, oxidative stress, and fibrosis provide novel mechanistic insights into vesicant toxicity, which may be useful in the development of efficacious therapeutics.
Collapse
Affiliation(s)
- Jaclynn A Meshanni
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Jordan M Lee
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Kinal N Vayas
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Rachel Sun
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Chenghui Jiang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Andrew J Gow
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Jeffrey D Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| | - Debra L Laskin
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (J.A.M., J.M.L., K.N.V., R.S., C.J., G.L.G., A.J.G., D.L.L.) and Department of Environmental and Occupational Health and Justice, School of Public Health (J.D.L.), Rutgers University, Piscataway, New Jersey
| |
Collapse
|
28
|
Zhao S, Zhang J, Chen Y, Cui X, Liu H, Yan Y, Sun Y, Qi Y, Liu Y. The comprehensive mechanism underlying Schisandra polysaccharide in AD-like symptoms of Aβ25-35-induced rats based on hippocampal metabolomics and serum lipidomics techniques. J Pharm Biomed Anal 2023; 236:115717. [PMID: 37716276 DOI: 10.1016/j.jpba.2023.115717] [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: 05/28/2023] [Revised: 08/30/2023] [Accepted: 09/10/2023] [Indexed: 09/18/2023]
Abstract
As is well documented, Alzheimer's disease (AD) is the most prevalent neurodegenerative disease. Meanwhile, Schisandra polysaccharide (SCP) has been reported to exert a protective effect on the nervous system and can regulate metabolic disorders in AD-like symptoms of amyloid β-peptide (Aβ) 25-35-induced rats. Nevertheless, the underlying mechanisms and metabolic markers for the diagnosis of AD are yet to be determined. This study aimed to explore the neuroprotective effect and potential mechanism of action of SCP in AD-like symptoms of Aβ25-35-induced rats by combining pharmacodynamics, metabolomics, and lipidomics. The pharmacodynamic results revealed that SCP significantly improved the spatial learning and long-term memory function and the morphology of neurons in the hippocampal CA1 region, alleviated inflammatory damage and oxidative stress, inhibited the activation of microglia and astrocytes, and increased the proportion of mature neurons of AD-like symptoms of Aβ25-35-induced rats. The results of hippocampal metabolomics and serum lipidomics showed 46 and 48 potential biomarkers were identified for the SCP treatment of AD, respectively. The involved pathways principally comprised lipid metabolism, amino acid metabolism, and energy metabolism. This study elucidates the neuroprotective effect of SCP in AD and its mechanism from the perspective of metabolomics and lipidomics and provides a theoretical basis for the therapeutic effect of SCP in AD.
Collapse
Affiliation(s)
- Shuo Zhao
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China
| | - Jinpeng Zhang
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China
| | - Yufeng Chen
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China
| | - Xinyuan Cui
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China
| | - Haiqing Liu
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China
| | - Ying Yan
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China
| | - Yuexiang Sun
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China
| | - Yongxiu Qi
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China
| | - Yuanyuan Liu
- Pharmaceutical Analysis Teaching Experimental Center, School of Pharmacy, Shandong First Medical University, Taian 271000, China.
| |
Collapse
|
29
|
Georgiou-Siafis SK, Tsiftsoglou AS. The Key Role of GSH in Keeping the Redox Balance in Mammalian Cells: Mechanisms and Significance of GSH in Detoxification via Formation of Conjugates. Antioxidants (Basel) 2023; 12:1953. [PMID: 38001806 PMCID: PMC10669396 DOI: 10.3390/antiox12111953] [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: 09/30/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Glutathione (GSH) is a ubiquitous tripeptide that is biosynthesized in situ at high concentrations (1-5 mM) and involved in the regulation of cellular homeostasis via multiple mechanisms. The main known action of GSH is its antioxidant capacity, which aids in maintaining the redox cycle of cells. To this end, GSH peroxidases contribute to the scavenging of various forms of ROS and RNS. A generally underestimated mechanism of action of GSH is its direct nucleophilic interaction with electrophilic compounds yielding thioether GSH S-conjugates. Many compounds, including xenobiotics (such as NAPQI, simvastatin, cisplatin, and barbital) and intrinsic compounds (such as menadione, leukotrienes, prostaglandins, and dopamine), form covalent adducts with GSH leading mainly to their detoxification. In the present article, we wish to present the key role and significance of GSH in cellular redox biology. This includes an update on the formation of GSH-S conjugates or GSH adducts with emphasis given to the mechanism of reaction, the dependence on GST (GSH S-transferase), where this conjugation occurs in tissues, and its significance. The uncovering of the GSH adducts' formation enhances our knowledge of the human metabolome. GSH-hematin adducts were recently shown to have been formed spontaneously in multiples isomers at hemolysates, leading to structural destabilization of the endogenous toxin, hematin (free heme), which is derived from the released hemoglobin. Moreover, hemin (the form of oxidized heme) has been found to act through the Kelch-like ECH associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor-2 (Nrf2) signaling pathway as an epigenetic modulator of GSH metabolism. Last but not least, the implications of the genetic defects in GSH metabolism, recorded in hemolytic syndromes, cancer and other pathologies, are presented and discussed under the framework of conceptualizing that GSH S-conjugates could be regarded as signatures of the cellular metabolism in the diseased state.
Collapse
Affiliation(s)
| | - Asterios S. Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health Sciences, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece;
| |
Collapse
|
30
|
Yang X, Ong HW, Dickmander RJ, Smith JL, Brown JW, Tao W, Chang E, Moorman NJ, Axtman AD, Willson TM. Optimization of 3-Cyano-7-cyclopropylamino-pyrazolo[1,5- a]pyrimidines toward the Development of an In Vivo Chemical Probe for CSNK2A. ACS OMEGA 2023; 8:39546-39561. [PMID: 37901516 PMCID: PMC10600890 DOI: 10.1021/acsomega.3c05377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/21/2023] [Indexed: 10/31/2023]
Abstract
3-Cyano-7-cyclopropylamino-pyrazolo[1,5-a]pyrimidines, including the chemical probe SGC-CK2-1, are potent and selective inhibitors of CSNK2A in cells but have limited utility in animal models due to their poor pharmacokinetic properties. While developing analogues with reduced intrinsic clearance and the potential for sustained exposure in mice, we discovered that phase II conjugation by GST enzymes was a major metabolic transformation in hepatocytes. A protocol for codosing with ethacrynic acid, a covalent reversible GST inhibitor, was developed to improve the exposure of analogue 2h in mice. A double codosing protocol, using a combination of ethacrynic acid and irreversible P450 inhibitor 1-aminobenzotriazole, increased the blood level of 2h by 40-fold at a 5 h time point.
Collapse
Affiliation(s)
- Xuan Yang
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
| | - Han Wee Ong
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
| | - Rebekah J. Dickmander
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department
of Microbiology & Immunology, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Lineberger
Comprehensive Cancer Center, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department
of Chemistry, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Jeffery L. Smith
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jason W. Brown
- Takeda Development
Center Americas, Inc., San Diego, California 92121, United States
| | - William Tao
- Takeda Development
Center Americas, Inc., San Diego, California 92121, United States
| | - Edcon Chang
- Takeda Development
Center Americas, Inc., San Diego, California 92121, United States
| | - Nathaniel J. Moorman
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
- Department
of Microbiology & Immunology, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Lineberger
Comprehensive Cancer Center, University
of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Alison D. Axtman
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
| | - Timothy M. Willson
- Structural
Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Rapidly
Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
31
|
Perjési P. Preface to the Special Issue "Glutathione: Chemistry and Biochemistry". Molecules 2023; 28:5993. [PMID: 37630245 PMCID: PMC10460063 DOI: 10.3390/molecules28165993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
This year we celebrate the 135th anniversary of the discovery of glutathione (L-γ-glutamyl-L-cysteinyl-glycine) [...].
Collapse
Affiliation(s)
- Pál Perjési
- Institute of Pharmaceutical Chemistry, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
32
|
Marini HR, Facchini BA, di Francia R, Freni J, Puzzolo D, Montella L, Facchini G, Ottaiano A, Berretta M, Minutoli L. Glutathione: Lights and Shadows in Cancer Patients. Biomedicines 2023; 11:2226. [PMID: 37626722 PMCID: PMC10452337 DOI: 10.3390/biomedicines11082226] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
In cases of cellular injury, there is an observed increase in the production of reactive oxygen species (ROS). When this production becomes excessive, it can result in various conditions, including cancerogenesis. Glutathione (GSH), the most abundant thiol-containing antioxidant, is fundamental to re-establishing redox homeostasis. In order to evaluate the role of GSH and its antioxi-dant effects in patients affected by cancer, we performed a thorough search on Medline and EMBASE databases for relevant clinical and/or preclinical studies, with particular regard to diet, toxicities, and pharmacological processes. The conjugation of GSH with xenobiotics, including anti-cancer drugs, can result in either of two effects: xenobiotics may lose their harmful effects, or GSH conjugation may enhance their toxicity by inducing bioactivation. While being an interesting weapon against chemotherapy-induced toxicities, GSH may also have a potential protective role for cancer cells. New studies are necessary to better explain the relationship between GSH and cancer. Although self-prescribed glutathione (GSH) implementation is prevalent among cancer patients with the intention of reducing the toxic effects of anticancer treatments and potentially preventing damage to normal tissues, this belief lacks substantial scientific evidence for its efficacy in reducing toxicity, except in the case of cisplatin-related neurotoxicity. Therefore, the use of GSH should only be considered under medical supervision, taking into account the appropriate timing and setting.
Collapse
Affiliation(s)
- Herbert Ryan Marini
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| | - Bianca Arianna Facchini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80133 Napoli, Italy;
| | - Raffaele di Francia
- Gruppo Oncologico Ricercatori Italiani (GORI-ONLUS), 33170 Pordenone, Italy;
| | - José Freni
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (J.F.); (D.P.)
| | - Domenico Puzzolo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (J.F.); (D.P.)
| | - Liliana Montella
- Division of Medical Oncology, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, 80078 Pozzuoli, Italy; (L.M.); (G.F.)
| | - Gaetano Facchini
- Division of Medical Oncology, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, 80078 Pozzuoli, Italy; (L.M.); (G.F.)
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, 80131 Napoli, Italy;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| |
Collapse
|
33
|
Yang X, Ong HW, Dickmander RJ, Smith JL, Brown JW, Tao W, Chang E, Moorman NJ, Axtman AD, Willson TM. Optimization of 3-Cyano-7-cyclopropylamino-pyrazolo[1,5-a]pyrimidines Toward the Development of an In Vivo Chemical Probe for CSNK2A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540828. [PMID: 37292607 PMCID: PMC10245575 DOI: 10.1101/2023.05.15.540828] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
3-cyano-7-cyclopropylamino-pyrazolo[1,5-a]pyrimidines, including the chemical probe SGC-CK2-1, are potent and selective inhibitors of CSNK2A in cells but have limited utility in animal models due to their poor pharmacokinetic properties. While developing analogs with reduced intrinsic clearance and the potential for sustained exposure in mice, we discovered that Phase II conjugation by GST enzymes was a major metabolic transformation in hepatocytes. A protocol for co-dosing with ethacrynic acid, a covalent reversible GST inhibitor, was developed to improve the exposure of analog 2h in mice. A double co-dosing protocol, using a combination of ethacrynic acid and irreversible P450 inhibitor 1-aminobenzotriazole increased the blood level of 2h by 40-fold at a 5 h time point.
Collapse
Affiliation(s)
- Xuan Yang
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, United States
| | - Han Wee Ong
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, United States
| | - Rebekah J. Dickmander
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, United States
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Jeffery L. Smith
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | | | - William Tao
- Takeda San Diego, San Diego, CA 92121, United States
| | - Edcon Chang
- Takeda San Diego, San Diego, CA 92121, United States
| | - Nathaniel J. Moorman
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, United States
- Department of Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Alison D. Axtman
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, United States
| | - Timothy M. Willson
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
- Rapidly Emerging Antiviral Drug Development Initiative (READDI), Chapel Hill, NC 27599, United States
| |
Collapse
|
34
|
Di Giacomo C, Malfa GA, Tomasello B, Bianchi S, Acquaviva R. Natural Compounds and Glutathione: Beyond Mere Antioxidants. Antioxidants (Basel) 2023; 12:1445. [PMID: 37507985 PMCID: PMC10376414 DOI: 10.3390/antiox12071445] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
The tripeptide glutathione plays important roles in many cell processes, including differentiation, proliferation, and apoptosis; in fact, disorders in glutathione homeostasis are involved both in the etiology and in the progression of several human diseases, including cancer. Natural compounds have been found to modulate glutathione levels and function beyond their role as mere antioxidants. For example, certain compounds can upregulate the expression of glutathione-related enzymes, increase the availability of cysteine, the limiting amino acid for glutathione synthesis, or directly interact with glutathione and modulate its function. These compounds may have therapeutic potential in a variety of disease states where glutathione dysregulation is a contributing factor. On the other hand, flavonoids' potential to deplete glutathione levels could be significant for cancer treatment. Overall, while natural compounds may have potential therapeutic and/or preventive properties and may be able to increase glutathione levels, more research is needed to fully understand their mechanisms of action and their potential benefits for the prevention and treatment of several diseases. In this review, particular emphasis will be placed on phytochemical compounds belonging to the class of polyphenols, terpenoids, and glucosinolates that have an impact on glutathione-related processes, both in physiological and pathological conditions. These classes of secondary metabolites represent the most food-derived bioactive compounds that have been intensively explored and studied in the last few decades.
Collapse
Affiliation(s)
- Claudia Di Giacomo
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Giuseppe Antonio Malfa
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Barbara Tomasello
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Simone Bianchi
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Rosaria Acquaviva
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Research Centre on Nutraceuticals and Health Products (CERNUT), University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
35
|
Habas E, Akbar R, Farfar K, Arrayes N, Habas A, Rayani A, Alfitori G, Habas E, Magassabi Y, Ghazouani H, Aladab A, Elzouki AN. Malignancy diseases and kidneys: A nephrologist prospect and updated review. Medicine (Baltimore) 2023; 102:e33505. [PMID: 37058030 PMCID: PMC10101313 DOI: 10.1097/md.0000000000033505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/21/2023] [Indexed: 04/15/2023] Open
Abstract
Acute kidney injury (AKI), chronic renal failure, and tubular abnormalities represent the kidney disease spectrum of malignancy. Prompt diagnosis and treatment may prevent or reverse these complications. The pathogenesis of AKI in cancer is multifactorial. AKI affects outcomes in cancer, oncological therapy withdrawal, increased hospitalization rate, and hospital stay. Renal function derangement can be recovered with early detection and targeted therapy of cancers. Identifying patients at higher risk of renal damage and implementing preventive measures without sacrificing the benefits of oncological therapy improve survival. Multidisciplinary approaches, such as relieving obstruction, hydration, etc., are required to minimize the kidney injury rate. Different keywords, texts, and phrases were used to search Google, EMBASE, PubMed, Scopus, and Google Scholar for related original and review articles that serve the article's aim well. In this nonsystematic article, we aimed to review the published data on cancer-associated kidney complications, their pathogenesis, management, prevention, and the latest updates. Kidney involvement in cancer occurs due to tumor therapy, direct kidney invasion by tumor, or tumor complications. Early diagnosis and therapy improve the survival rate. Pathogenesis of cancer-related kidney involvement is different and complicated. Clinicians' awareness of all the potential causes of cancer-related complications is essential, and a kidney biopsy should be conducted to confirm the kidney pathologies. Chronic kidney disease is a known complication in malignancy and therapies. Hence, avoiding nephrotoxic drugs, dose standardization, and early cancer detection are mandatory measures to prevent renal involvement.
Collapse
Affiliation(s)
- Elmukhtar Habas
- Facharzt Internal Medicine, Facharzt Nephrology, Medical Department, Hamad General Hospital, Doha, Qatar
| | - Raza Akbar
- Medical Department, Hamad General Hospital, Doha, Qatar
| | - Kalifa Farfar
- Facharzt Internal Medicine, Medical Department, Alwakra General Hospital, Alwakra, Qatar
| | - Nada Arrayes
- Medical Education Fellow, Lincoln Medical School, University of Lincoln, Lincoln, UK
| | - Aml Habas
- Hematology-Oncology Department, Tripoli Children Hospital, Tripoli, Libya
| | - Amnna Rayani
- Facharzt Pediatric, Facharzt Hemotoncology, Hematology-Oncology Department, Tripoli Children Hospital, Tripoli, Libya
| | | | - Eshrak Habas
- Medical Department, Tripoli Central Hospital, University of Tripoli, Tripoli, Libya
| | | | - Hafidh Ghazouani
- Quality Department, Senior Epidemiologist, Hamad Medical Corporation, Doha, Qatar
| | | | | |
Collapse
|
36
|
Kupreienko O, Pouliou F, Konstandinidis K, Axarli I, Douni E, Papageorgiou AC, Labrou NE. Inhibition Analysis and High-Resolution Crystal Structure of Mus musculus Glutathione Transferase P1-1. Biomolecules 2023; 13:biom13040613. [PMID: 37189361 DOI: 10.3390/biom13040613] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023] Open
Abstract
Multidrug resistance is a significant barrier that makes anticancer therapies less effective. Glutathione transferases (GSTs) are involved in multidrug resistance mechanisms and play a significant part in the metabolism of alkylating anticancer drugs. The purpose of this study was to screen and select a lead compound with high inhibitory potency against the isoenzyme GSTP1-1 from Mus musculus (MmGSTP1-1). The lead compound was selected following the screening of a library of currently approved and registered pesticides that belong to different chemical classes. The results showed that the fungicide iprodione [3-(3,5-dichlorophenyl)-2,4-dioxo-N-propan-2-ylimidazolidine-1-carboxamide] exhibited the highest inhibition potency (ΙC50 = 11.3 ± 0.5 μΜ) towards MmGSTP1-1. Kinetics analysis revealed that iprodione functions as a mixed-type inhibitor towards glutathione (GSH) and non-competitive inhibitor towards 1-chloro-2,4-dinitrobenzene (CDNB). X-ray crystallography was used to determine the crystal structure of MmGSTP1-1 at 1.28 Å resolution as a complex with S-(p-nitrobenzyl)glutathione (Nb-GSH). The crystal structure was used to map the ligand-binding site of MmGSTP1-1 and to provide structural data of the interaction of the enzyme with iprodione using molecular docking. The results of this study shed light on the inhibition mechanism of MmGSTP1-1 and provide a new compound as a potential lead structure for future drug/inhibitor development.
Collapse
|
37
|
Heritable Risk and Protective Genetic Components of Glaucoma Medication Non-Adherence. Int J Mol Sci 2023; 24:ijms24065636. [PMID: 36982708 PMCID: PMC10058353 DOI: 10.3390/ijms24065636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Glaucoma is the leading cause of irreversible blindness, affecting 76 million globally. It is characterized by irreversible damage to the optic nerve. Pharmacotherapy manages intraocular pressure (IOP) and slows disease progression. However, non-adherence to glaucoma medications remains problematic, with 41–71% of patients being non-adherent to their prescribed medication. Despite substantial investment in research, clinical effort, and patient education protocols, non-adherence remains high. Therefore, we aimed to determine if there is a substantive genetic component behind patients’ glaucoma medication non-adherence. We assessed glaucoma medication non-adherence with prescription refill data from the Marshfield Clinic Healthcare System’s pharmacy dispensing database. Two standard measures were calculated: the medication possession ratio (MPR) and the proportion of days covered (PDC). Non-adherence on each metric was defined as less than 80% medication coverage over 12 months. Genotyping was done using the Illumina HumanCoreExome BeadChip in addition to exome sequencing on the 230 patients (1) to calculate the heritability of glaucoma medication non-adherence and (2) to identify SNPs and/or coding variants in genes associated with medication non-adherence. Ingenuity pathway analysis (IPA) was utilized to derive biological meaning from any significant genes in aggregate. Over 12 months, 59% of patients were found to be non-adherent as measured by the MPR80, and 67% were non-adherent as measured by the PDC80. Genome-wide complex trait analysis (GCTA) suggested that 57% (MPR80) and 48% (PDC80) of glaucoma medication non-adherence could be attributed to a genetic component. Missense mutations in TTC28, KIAA1731, ADAMTS5, OR2W3, OR10A6, SAXO2, KCTD18, CHCHD6, and UPK1A were all found to be significantly associated with glaucoma medication non-adherence by whole exome sequencing after Bonferroni correction (p < 10−3) (PDC80). While missense mutations in TINAG, CHCHD6, GSTZ1, and SEMA4G were found to be significantly associated with medication non-adherence by whole exome sequencing after Bonferroni correction (p < 10−3) (MPR80). The same coding SNP in CHCHD6 which functions in Alzheimer’s disease pathophysiology was significant by both measures and increased risk for glaucoma medication non-adherence by three-fold (95% CI, 1.62–5.8). Although our study was underpowered for genome-wide significance, SNP rs6474264 within ZMAT4 (p = 5.54 × 10–6) was found to be nominally significant, with a decreased risk for glaucoma medication non-adherence (OR, 0.22; 95% CI, 0.11–0.42)). IPA demonstrated significant overlap, utilizing, both standard measures including opioid signaling, drug metabolism, and synaptogenesis signaling. CREB signaling in neurons (which is associated with enhancing the baseline firing rate for the formation of long-term potentiation in nerve fibers) was shown to have protective associations. Our results suggest a substantial heritable genetic component to glaucoma medication non-adherence (47–58%). This finding is in line with genetic studies of other conditions with a psychiatric component (e.g., post-traumatic stress disorder (PTSD) or alcohol dependence). Our findings suggest both risk and protective statistically significant genes/pathways underlying glaucoma medication non-adherence for the first time. Further studies investigating more diverse populations with larger sample sizes are needed to validate these findings.
Collapse
|
38
|
Barkhordari A, Jafari-Gharabaghlou D, Turk Z, Zarghami N. Potential Anti-Cancer Effect of Helenalin as a Natural Bioactive Compound on the Growth and Telomerase Gene Expression in Breast Cancer Cell Line. Asian Pac J Cancer Prev 2023; 24:133-140. [PMID: 36708561 PMCID: PMC10152844 DOI: 10.31557/apjcp.2023.24.1.133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Indexed: 05/04/2023] Open
Abstract
OBJECTIVE The telomerase gene is overexpressed in the majority of tumors and cancers compared to normal and healthy cells, and on the other hand, this enzymatic protein is overactive, therefore, the telomerase enzyme is considered a primary target for diagnostic and therapeutic purposes in most cancers. This has been hypothesized that Helenalin has anti-telomerase activity in a wide range of cancers and Tumor tissues. In this study, we investigated the inhibitory effect of helenalin extract on telomerase gene expression in the T47D breast cancer cell line. METHODS We used the MTT assay to evaluate the cytotoxic effect of different concentrations of helenalin on the T47D breast cancer cell line at 24, 48, and 72 hours. Besides, the expression of the hTERT gene in T47D cell lines treated with 1.0 and 5.0 µM helenalin after 24, 48, and 72 h incubation times was investigated through real-time PCR. RESULTS According to the MTT assay, the inhibitory effect of helenalin on T47D cell proliferation is time and dose-dependent. Moreover, the results of Real-time PCR showed that exposure of T47D cell lines to helenalin led to a significant Decreasing in the expressional values of the hTERT gene as a time and dose-dependent procedure compared with the control group (P ≤ 0.05). CONCLUSION These preliminary results demonstrated the cytotoxic potential of helenalin through inhibition of hTERT against T47D breast cancer cells.
Collapse
Affiliation(s)
- Amin Barkhordari
- Cellular and Molecular Research Center, Grash University of Medical Sciences, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeynep Turk
- Department of Medicine, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| | - Nosratollah Zarghami
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|