1
|
Steen TV, Espinoza I, Duran C, Casadevall G, Serrano-Hervás E, Cuyàs E, Verdura S, Kemble G, Kaufmann SH, McWilliams R, Osuna S, Billadeau DD, Menendez JA, Lupu R. Fatty acid synthase (FASN) inhibition cooperates with BH3 mimetic drugs to overcome resistance to mitochondrial apoptosis in pancreatic cancer. Neoplasia 2025; 62:101143. [PMID: 39999714 PMCID: PMC11908614 DOI: 10.1016/j.neo.2025.101143] [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: 09/05/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Resistance to mitochondrial apoptosis is a major driver of chemoresistance in pancreatic ductal adenocarcinoma (PDAC). However, pharmacological manipulation of the mitochondrial apoptosis threshold in PDAC cells remains an unmet therapeutic goal. We hypothesized that fatty acid synthase inhibitors (FASNis), a family of targeted metabolic therapeutics recently entering the clinic, could lower the apoptotic threshold in chemoresistant PDAC cells and be synergistic with BH3 mimetics that neutralize anti-apoptotic proteins. Computational studies with TVB-3166 and TVB-3664, two analogues of the clinical-grade FASNi TVB-2640 (denifanstat), confirmed their uncompetitive behavior towards NADPH when bound to the FASN ketoacyl reductase domain. The extent of NADPH accumulation, a consequence of FASN inhibition, paralleled the sensitivity of PDAC cells to the apoptotic effects of TVB FASNis in conventional PDAC cell lines that naturally express varying levels of FASN. FASN inhibition dramatically increased the sensitivity of "FASN-high" expressing PDAC cells to the BCL2/BCL-XL/BCL-W inhibitor ABT-263/navitoclax and the BCL2-selective inhibitor ABT-199/venetoclax, both in vitro and in in vivo xenografted tumors. The ability of TVB FASNis to shift the balance of pro- and anti-apoptotic proteins and thereby push PDAC cells closer to the apoptotic threshold was also observed in cell lines developed from patient-derived xenografts (PDXs) representative of the classical (pancreatic) transcriptomic subtype of PDAC. Experiments in PDAC PDXs in vivo confirmed the synergistic antitumor activity of TVB-3664 with navitoclax and venetoclax, independent of the nature of the replication stress signature of patient-derived PDAC cells. The discovery that targeted inhibition of FASN is a metabolic perturbation that sensitizes PDAC cells to BH3 mimetics warrants further investigation to overcome resistance to mitochondrial apoptosis in PDAC patients.
Collapse
Affiliation(s)
- Travis Vander Steen
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ingrid Espinoza
- National Institute of Health, National Heart Lung and Blood Institute (NHLBI), Bethesda, MD 20817, USA; Lung Development and Pediatric Branch (HNH36), Bethesda, MD 20817, USA
| | - Cristina Duran
- Institut de Química Computacional i Catàlisi and Departament de Química, Universitat de Girona, Girona 17003, Spain
| | - Guillem Casadevall
- Institut de Química Computacional i Catàlisi and Departament de Química, Universitat de Girona, Girona 17003, Spain
| | - Eila Serrano-Hervás
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona 17007, Spain; Metabolism and Cancer Group,Girona Biomedical Research Institute (IDIBGI), Salt 17190, Girona, Spain
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona 17007, Spain; Metabolism and Cancer Group,Girona Biomedical Research Institute (IDIBGI), Salt 17190, Girona, Spain
| | - Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona 17007, Spain; Metabolism and Cancer Group,Girona Biomedical Research Institute (IDIBGI), Salt 17190, Girona, Spain
| | | | - Scott H Kaufmann
- Mayo Clinic Cancer Center, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA; Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Robert McWilliams
- Mayo Clinic Cancer Center, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sílvia Osuna
- Institut de Química Computacional i Catàlisi and Departament de Química, Universitat de Girona, Girona 17003, Spain; ICREA, Barcelona 08010, Spain
| | - Daniel D Billadeau
- Mayo Clinic Cancer Center, Rochester, MN 55905, USA; Division of Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA; Department of Immunology College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona 17007, Spain; Metabolism and Cancer Group,Girona Biomedical Research Institute (IDIBGI), Salt 17190, Girona, Spain.
| | - Ruth Lupu
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; Mayo Clinic Cancer Center, Rochester, MN 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
2
|
Ahn CR, Baek SH. Synergistic effects of 6-shogaol and hyperthermia on ACHN renal cancer cells: modulation of ROS and heat shock pro-teins in cancer therapy. Front Pharmacol 2025; 16:1522285. [PMID: 40051570 PMCID: PMC11882530 DOI: 10.3389/fphar.2025.1522285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Introduction Renal cancer is known for its aggressive progression and resistance to standard treatments, underscoring the need for novel therapeutic strategies. This study explores the potential of combining 6-shogaol (6-SHO), a bioactive compound derived from ginger (Zingiber officinale), with hyperthermia to enhance anticancer efficacy in ACHN renal cancer cells. Methods ACHN cells were treated with 6-SHO and exposed to hyperthermic conditions. We evaluated the combined effects on apoptosis, cell cycle arrest, and cell proliferation, as well as the role of reactive oxygen species (ROS) and heat shock proteins (HSPs) in mediating these responses. Results The combination of 6-SHO and hyperthermia significantly increased apoptosis, induced G2/M phase cell cycle arrest, and reduced cell proliferation more effectively than either treatment alone. ROS played a critical role in these effects, with modulation of HSPs and heat shock factor 1 (HSF1) further disrupting cancer cell survival mechanisms. Discussion These findings highlight the synergistic potential of 6-SHO and hyperthermia as a novel therapeutic approach in renal cancer treatment, supporting the need for further research and clinical evaluation.
Collapse
Affiliation(s)
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
3
|
Tian C, Deng S, Yang M, Bai B, Pan Y, Xie G, Zhao D, Wei L. Indole-3-carbinol and its main derivative 3,3'-diindolylmethane: Regulatory roles and therapeutic potential in liver diseases. Biomed Pharmacother 2024; 180:117525. [PMID: 39388997 DOI: 10.1016/j.biopha.2024.117525] [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: 08/05/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Indole-3-carbinol (I3C), a compound found in cruciferous vegetables, has shown significant efficacy in treating both cancerous and non-cancerous diseases. Its primary derivative, 3,3'-diindolylmethane (DIM), formed during digestion, also exhibits similar therapeutic benefits. In liver disorders, I3C and DIM exhibit dual roles by inhibiting and promoting hepatocellular carcinoma (HCC) and providing relief for nonmalignant liver diseases, such as acute liver injury (ALI), hepatic fibrosis, nonalcoholic fatty liver disease (NAFLD), and alcohol-related liver disease (ALD). Mechanistically, I3C and DIM modulate various pathophysiological processes, including cell proliferation, apoptosis, oxidative stress, and lipogenesis. This review aims to enhance researchers' understanding of the regulatory roles of I3C and DIM in these liver diseases and explore the potential of plant-derived substances in liver disease treatment.
Collapse
Affiliation(s)
- Chao Tian
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Shizhou Deng
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Research and Development Department, Guangdong Longsee Biomedical Corporation, Guangzhou 510700, China
| | - Ming Yang
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Baochen Bai
- Department of Cardiology, Peking University People's hospital, Beijing 100044, China
| | - Yi Pan
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Gangqiao Xie
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dongliang Zhao
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China.
| |
Collapse
|
4
|
Vivanco PG, Taboada P, Coelho A. The Southern European Atlantic Diet and Its Supplements: The Chemical Bases of Its Anticancer Properties. Nutrients 2023; 15:4274. [PMID: 37836558 PMCID: PMC10574233 DOI: 10.3390/nu15194274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Scientific evidence increasingly supports the strong link between diet and health, acknowledging that a well-balanced diet plays a crucial role in preventing chronic diseases such as obesity, diabetes, cardiovascular issues, and certain types of cancer. This perspective opens the door to developing precision diets, particularly tailored for individuals at risk of developing cancer. It encompasses a vast research area and involves the study of an expanding array of compounds with multilevel "omics" compositions, including genomics, transcriptomics, proteomics, epigenomics, miRNomics, and metabolomics. We review here the components of the Southern European Atlantic Diet (SEAD) from both a chemical and pharmacological standpoint. The information sources consulted, complemented by crystallographic data from the Protein Data Bank, establish a direct link between the SEAD and its anticancer properties. The data collected strongly suggest that SEAD offers an exceptionally healthy profile, particularly due to the presence of beneficial biomolecules in its foods. The inclusion of olive oil and paprika in this diet provides numerous health benefits, and scientific evidence supports the anticancer properties of dietary supplements with biomolecules sourced from vegetables of the brassica genus. Nonetheless, further research is warranted in this field to gain deeper insights into the potential benefits of the SEAD's bioactive compounds against cancer.
Collapse
Affiliation(s)
- Pablo García Vivanco
- Spanish Academy of Nutrition and Dietetics, 31006 Pamplona, Spain
- Nutrition and Digestive Working Group, Spanish Society of Clinical, Family, and Community Pharmacy (SEFAC), 28045 Madrid, Spain
| | - Pablo Taboada
- Department of Condensed Matter Physics, Faculty of Physics, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Institute of Materials-USC (IMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Alberto Coelho
- Institute of Materials-USC (IMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
5
|
Centofanti F, Buono A, Verboni M, Tomino C, Lucarini S, Duranti A, Pandolfi PP, Novelli G. Synthetic Methodologies and Therapeutic Potential of Indole-3-Carbinol (I3C) and Its Derivatives. Pharmaceuticals (Basel) 2023; 16:240. [PMID: 37259386 PMCID: PMC9960368 DOI: 10.3390/ph16020240] [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: 01/10/2023] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 01/01/2025] Open
Abstract
Indole-3-carbinol (I3C) is a natural product contained in vegetables belonging to the Brassicaceae family and has been studied in recent decades for its biological and pharmacological properties. Herein, we will analyze: (1) the biosynthetic processes and synthetic procedures through which I3C and its main derivatives have been obtained; (2) the characteristics that lead to believe that both I3C and its derivatives are responsible for several important activities-in particular, antitumor and antiviral, through insights concerning in vitro assays and in vivo tests; (3) the mechanisms of action of the most important compounds considered; (4) the potential social impact that the enhancement of the discussed molecules can have in the prevention and treatment of the pathologies' examined field-first of all, those related to respiratory tract disorders and cancer.
Collapse
Affiliation(s)
- Federica Centofanti
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
| | - Alessandro Buono
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Michele Verboni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Carlo Tomino
- Scientific Direction—IRCCS San Raffaele Rome, 00166 Rome, Italy
| | - Simone Lucarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Pier Paolo Pandolfi
- William N. Pennington Cancer Institute, Renown Health, Nevada System of Higher Education, Reno, NV 89502, USA
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
6
|
Hussain Y, Singh J, Raza W, Meena A, Rajak S, Sinha RA, Luqman S. Purpurin ameliorates alcohol-induced hepatotoxicity by reducing ROS generation and promoting Nrf2 expression. Life Sci 2022; 309:120964. [PMID: 36115584 DOI: 10.1016/j.lfs.2022.120964] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/01/2022] [Accepted: 09/11/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION AND AIM Purpurin, a naturally occurring anthraquinone isolated from the roots of Rubia cordifolia, exhibits anti-cancer, anti-genotoxic, anti-microbial, neuromodulatory and photodynamic activity. However, purpurin's in vivo and in vitro antioxidant mechanism remains unexplored. The present study explores the anti-oxidative mechanism of purpurin under the influence of alcohol using in vivo and in vitro test systems. METHODS Mice hepatocytes and alcohol-induced liver toxicity model were used to evaluate the effect of purpurin. The non-enzymatic and enzymatic oxidative stress markers were estimated by the colorimetric method. The reactive oxygen species (ROS) were quantified in mitochondria and cells using flow cytometer. Real-time PCR and western blotting were used to quantify cytochrome 450 subtype 2E1 (CYP2E1) and Nrf2 expression in the liver tissue of mice. In silico studies were performed through receptor-ligand binding interaction. KEY FINDINGS Purpurin effectively reduced total cellular and mitochondrial ROS in primary hepatocytes and WRL-68 cells. It prevented alcohol-induced ROS-dependent biochemical and cellular insults observed by analysing the serum glutamic pyruvic transaminase (SGPT), glutamic-oxaloacetic transaminase (SGOT) levels and CYP2E1 expression in liver tissue of alcohol-administered mice. Moreover, it also restored the activity of antioxidant enzymes. Its antioxidant effect was established by glutathione and ROS-dependent mechanisms using buthionine sulfoximine and N-acetyl cysteine. Along with alcohol, purpurin up-regulated Nrf2 expression in hepatocytes. SIGNIFICANCE This work confirmed the ameliorative effect of purpurin for alcohol-induced hepatotoxicity by drabbing free radicals and curbing oxidative stress via activation of antioxidant signalling pathways.
Collapse
Affiliation(s)
- Yusuf Hussain
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Jyoti Singh
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Jawaharlal Nehru University, New Delhi 110067, India
| | - Waseem Raza
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Jawaharlal Nehru University, New Delhi 110067, India
| | - Abha Meena
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Suaib Luqman
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
7
|
Ryu BI, Kim KT. Antioxidant activity and protective effect of methyl gallate against t-BHP induced oxidative stress through inhibiting ROS production. Food Sci Biotechnol 2022; 31:1063-1072. [DOI: 10.1007/s10068-022-01120-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/29/2022] [Accepted: 06/09/2022] [Indexed: 11/04/2022] Open
|
8
|
Li H, Wang Y, Su R, Jia Y, Lai X, Su H, Fan Y, Wang Y, Xing W, Qin J. Dimethyl Fumarate Combined With Vemurafenib Enhances Anti-Melanoma Efficacy via Inhibiting the Hippo/YAP, NRF2-ARE, and AKT/mTOR/ERK Pathways in A375 Melanoma Cells. Front Oncol 2022; 12:794216. [PMID: 35141161 PMCID: PMC8820202 DOI: 10.3389/fonc.2022.794216] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/05/2022] [Indexed: 12/18/2022] Open
Abstract
Melanoma is a deadly form of skin cancer with high rates of resistance to traditional chemotherapy and radiotherapy. BRAF inhibitors (BRAFi) can achieve initial efficacy when used to treat melanoma patients, but drug resistance and relapse are common, emphasizing the need for new therapeutic strategies. Herein, we reported that combination of dimethyl fumarate (DMF) and vemurafenib (Vem) inhibited melanoma cell proliferation more significantly and induced more cell death than single agent did both in vitro and in vivo. DMF/Vem treatment induced cell death through inhibiting the expression and transcriptional activity of NRF2 thereby resulting in more reactive oxygen species (ROS) and via inhibiting the expression of YAP, a key downstream effector of Hippo pathway. DMF/Vem treatment also reduced phosphorylation of AKT, 4EBP1, P70S6K and ERK in AKT/mTOR/ERK signaling pathways. RNA-seq analysis revealed that DMF/Vem treatment specifically suppressed 4561 genes which belong to dozens of cell signaling pathways. These results indicated that DMF/Vem treatment manifested an enhanced antitumor efficacy through inhibiting multiple cell signaling pathways, and thus would be a novel promising therapeutic approach targeted for melanoma.
Collapse
Affiliation(s)
- Hongxia Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yaping Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Rina Su
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yuchen Jia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xiong Lai
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Huimin Su
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yaochun Fan
- Inner Mongolia Autonomous Region Center for Disease Control and Prevention, Hohhot, China
| | - Yuewu Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Wanjin Xing
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, College of Life Sciences, Inner Mongolia University, Hohhot, China
- *Correspondence: Wanjin Xing, ; Jianzhong Qin,
| | - Jianzhong Qin
- College of Biological Sciences and Biotechnology, Dalian University, Dalian, China
- *Correspondence: Wanjin Xing, ; Jianzhong Qin,
| |
Collapse
|
9
|
Endria Gunadi E, Wisnu Prajoko Y, Putra A. Effectiveness of Mesenchymal Stem Cells and Bovine Colostrum on Decreasing Tumor Necrosis Factor-Α Levels and Enhancement of Macrophages M2 in Remnant Liver. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Mesenchymal stem cells (MSCs) and bovine colostrum are potential therapies for the treatment of various degenerative and immune diseases.
AIM: This study aimed to analyze the effect of MSCs on levels of tumor necrosis factor-Α (TNF-α) and macrophages M2 in the liver fibrosis of Wistar rats after 50% resection.
METHODS: This study is a quasi-experimental post-test-only control group design to analyze the effect of giving bovine colostrum and MSCs to test animals on the process of regeneration of the remaining 50% liver with fibrosis. The study was conducted at the Stem Cell and Cancer Research Universitas Sultan Agung. The number of samples used was 40 male Wistar rats. The independent variables included MSC 1.000.0000 cells and bovine colostrum at a dose of 15 μL/g. Dependent variables used were macrophages M2 and levels of TNF-α ELISA.
RESULTS: TNF-α levels on day 3 were (p = 0.001), day 7 were (p = 0.01), and day 10 were (p = 0.01) in liver tissue in various study groups analyzed using ELISA on day three*. The results showed differences which were significant between the control and treatment groups (p < 0.05). The expression of CD163 marked brown in liver tissue had more expression than the control group.
CONCLUSION: The combination of MSCs and bovine colostrum can reduce TNF-α levels and significantly increase macrophages expression in the liver fibrosis of Wistar rats after 50% resection on the 3th, 7th, and 10th days.
Collapse
|
10
|
Schroeder B, Vander Steen T, Espinoza I, Venkatapoorna CMK, Hu Z, Silva FM, Regan K, Cuyàs E, Meng XW, Verdura S, Arbusà A, Schneider PA, Flatten KS, Kemble G, Montero J, Kaufmann SH, Menendez JA, Lupu R. Fatty acid synthase (FASN) regulates the mitochondrial priming of cancer cells. Cell Death Dis 2021; 12:977. [PMID: 34675185 PMCID: PMC8531299 DOI: 10.1038/s41419-021-04262-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/12/2021] [Accepted: 09/24/2021] [Indexed: 12/24/2022]
Abstract
Inhibitors of the lipogenic enzyme fatty acid synthase (FASN) have attracted much attention in the last decade as potential targeted cancer therapies. However, little is known about the molecular determinants of cancer cell sensitivity to FASN inhibitors (FASNis), which is a major roadblock to their therapeutic application. Here, we find that pharmacological starvation of endogenously produced FAs is a previously unrecognized metabolic stress that heightens mitochondrial apoptotic priming and favors cell death induction by BH3 mimetic inhibitors. Evaluation of the death decision circuits controlled by the BCL-2 family of proteins revealed that FASN inhibition is accompanied by the upregulation of the pro-death BH3-only proteins BIM, PUMA, and NOXA. Cell death triggered by FASN inhibition, which causally involves a palmitate/NADPH-related redox imbalance, is markedly diminished by concurrent loss of BIM or PUMA, suggesting that FASN activity controls cancer cell survival by fine-tuning the BH3 only proteins-dependent mitochondrial threshold for apoptosis. FASN inhibition results in a heightened mitochondrial apoptosis priming, shifting cells toward a primed-for-death state "addicted" to the anti-apoptotic protein BCL-2. Accordingly, co-administration of a FASNi synergistically augments the apoptosis-inducing activity of the dual BCL-XL/BCL-2 inhibitor ABT-263 (navitoclax) and the BCL-2 specific BH3-mimetic ABT-199 (venetoclax). FASN inhibition, however, fails to sensitize breast cancer cells to MCL-1- and BCL-XL-selective inhibitors such as S63845 and A1331852. A human breast cancer xenograft model evidenced that oral administration of the only clinically available FASNi drastically sensitizes FASN-addicted breast tumors to ineffective single-agents navitoclax and venetoclax in vivo. In summary, a novel FASN-driven facet of the mitochondrial priming mechanistically links the redox-buffering mechanism of FASN activity to the intrinsic apoptotic threshold in breast cancer cells. Combining next-generation FASNis with BCL-2-specific BH3 mimetics that directly activate the apoptotic machinery might generate more potent and longer-lasting antitumor responses in a clinical setting.
Collapse
Affiliation(s)
- Barbara Schroeder
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.,Mayo Clinic Cancer Center, Rochester, MN, 55905, USA.,Helmholtz Pioneer Campus, Heimholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Ingolstädter Landstraße 1 D-85764 Neuherberg, Munich, Germany
| | - Travis Vander Steen
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ingrid Espinoza
- Department of Preventive Medicine, John D. Bower School of Population Health, University of Mississippi Medical Center, Jackson, MS, 39216, USA.,Cancer Institute, School of Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Chandra M Kurapaty Venkatapoorna
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.,Department of Nutrition, Dietetics, and Hospital Management, Auburn University, Auburn, AL, 36849, USA
| | - Zeng Hu
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.,Radiation Oncology Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Fernando Martín Silva
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Kevin Regan
- Department of Experimental Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Elisabet Cuyàs
- Girona Biomedical Research Institute, 17190, Salt, Girona, Spain.,Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism & Cancer Group, Catalan Institute of Oncology, 17007, Girona, Spain
| | - X Wei Meng
- Deparment of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sara Verdura
- Girona Biomedical Research Institute, 17190, Salt, Girona, Spain.,Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism & Cancer Group, Catalan Institute of Oncology, 17007, Girona, Spain
| | - Aina Arbusà
- Girona Biomedical Research Institute, 17190, Salt, Girona, Spain.,Program Against Cancer Therapeutic Resistance (ProCURE), Metabolism & Cancer Group, Catalan Institute of Oncology, 17007, Girona, Spain
| | | | - Karen S Flatten
- Deparment of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - George Kemble
- Sagimet Biosciences (formerly 3-V Biosciences), San Mateo, CA, 94402, USA
| | - Joan Montero
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), 08028, Barcelona, Spain
| | - Scott H Kaufmann
- Mayo Clinic Cancer Center, Rochester, MN, 55905, USA.,Deparment of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Javier A Menendez
- Department of Experimental Pathology, Mayo Clinic, Rochester, MN, 55905, USA. .,Girona Biomedical Research Institute, 17190, Salt, Girona, Spain.
| | - Ruth Lupu
- Division of Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA. .,Mayo Clinic Cancer Center, Rochester, MN, 55905, USA. .,Department of Biochemistry and Molecular Biology Laboratory, Mayo Clinic Laboratory, Rochester, MN, 55905, USA.
| |
Collapse
|
11
|
Williams DE. Indoles Derived From Glucobrassicin: Cancer Chemoprevention by Indole-3-Carbinol and 3,3'-Diindolylmethane. Front Nutr 2021; 8:734334. [PMID: 34660663 PMCID: PMC8517077 DOI: 10.3389/fnut.2021.734334] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/27/2021] [Indexed: 12/22/2022] Open
Abstract
Hydrolysis of glucobrassicin by plant or bacterial myrosinase produces multiple indoles predominantly indole-3-carbinol (I3C). I3C and its major in vivo product, 3,3'-diindolylmethane (DIM), are effective cancer chemopreventive agents in pre-clinical models and show promise in clinical trials. The pharmacokinetics/pharmacodynamics of DIM have been studied in both rodents and humans and urinary DIM is a proposed biomarker of dietary intake of cruciferous vegetables. Recent clinical studies at Oregon State University show surprisingly robust metabolism of DIM in vivo with mono- and di-hydroxylation followed by conjugation with sulfate or glucuronic acid. DIM has multiple mechanisms of action, the most well-characterized is modulation of aryl hydrocarbon receptor (AHR) signaling. In rainbow trout dose-dependent cancer chemoprevention by dietary I3C is achieved when given prior to or concurrent with aflatoxin B1, polycyclic aromatic hydrocarbons, nitrosamines or direct acting carcinogens such as N-methyl-N'-nitro-nitrosoguanidine. Feeding pregnant mice I3C inhibits transplacental carcinogenesis. In humans much of the focus has been on chemoprevention of breast and prostate cancer. Alteration of cytochrome P450-dependent estrogen metabolism is hypothesized to be an important driver of DIM-dependent breast cancer prevention. The few studies done to date comparing glucobrassicin-rich crucifers such as Brussels sprouts with I3C/DIM supplements have shown the greater impact of the latter is due to dose. Daily ingestion of kg quantities of Brussels sprouts is required to produce in vivo levels of DIM achievable by supplementation. In clinical trials these supplement doses have elicited few if any adverse effects. Sulforaphane from glucoraphanin can act synergistically with glucobrassicin-derived DIM and this may lead to opportunities for combinatorial approaches (supplement and food-based) in the clinic.
Collapse
Affiliation(s)
- David E. Williams
- Department of Environmental and Molecular Toxicology, Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| |
Collapse
|
12
|
Yang TC, Yarmishyn AA, Yang YP, Lu PC, Chou SJ, Wang ML, Lin TC, Hwang DK, Chou YB, Chen SJ, Yu WK, Wang AG, Hsu CC, Chiou SH. Mitochondrial transport mediates survival of retinal ganglion cells in affected LHON patients. Hum Mol Genet 2021; 29:1454-1464. [PMID: 32277753 DOI: 10.1093/hmg/ddaa063] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/02/2020] [Accepted: 03/30/2020] [Indexed: 01/17/2023] Open
Abstract
The mutations in the genes encoding the subunits of complex I of the mitochondrial electron transport chain are the most common cause of Leber's hereditary optic neuropathy (LHON), a maternal hereditary disease characterized by retinal ganglion cell (RGC) degeneration. The characteristics of incomplete penetrance indicate that nuclear genetic and environmental factors also determine phenotypic expression of LHON. Therefore, further understanding of the role of mutant mitochondrial nicotinamide adenine dinucleotide dehydrogenase subunit proteins and nuclear genetic factors/environmental effects in the etiology of LHON is needed. In this study, we generated human-induced pluripotent stem cells (hiPSCs) from healthy control, unaffected LHON mutation carrier, and affected LHON patient. hiPSC-derived RGCs were used to study the differences between affected and unaffected carriers of mitochondrial DNA point mutation m.11778G > A in the MT-ND4 gene. We found that both mutated cell lines were characterized by increase in reactive oxygen species production, however, only affected cell line had increased levels of apoptotic cells. We found a significant increase in retrograde mitochondria and a decrease in stationary mitochondria in the affected RGC axons. In addition, the messenger RNA and protein levels of KIF5A in the LHON-affected RGCs were significantly reduced. Antioxidant N-acetyl-L-cysteine could restore the expression of KIF5A and the normal pattern of mitochondrial movement in the affected RGCs. To conclude, we found essential differences in the mutually dependent processes of oxidative stress, mitochondrial transport and apoptosis between two LHON-specific mutation carrier RGC cell lines, asymptomatic carrier and disease-affected, and identified KIF5A as a central modulator of these differences.
Collapse
Affiliation(s)
- Tien-Chun Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | | | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.,School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | - Pin-Chen Lu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Shih-Jie Chou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Mong-Lien Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.,School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Tai-Chi Lin
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - De-Kuang Hwang
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yu-Bai Chou
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Shih-Jen Chen
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Wei-Kuang Yu
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - An-Guor Wang
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Chih-Chien Hsu
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.,School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.,Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| |
Collapse
|
13
|
Cui J, Liu H, Xu S. Selenium-deficient diet induces necroptosis in the pig brain by activating TNFR1 via mir-29a-3p. Metallomics 2021; 12:1290-1301. [PMID: 32568328 DOI: 10.1039/d0mt00032a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Selenium (Se) deficiency is one of the crucial factors related to nervous system disease and necroptosis. MicroRNAs (miRNAs) play vital roles in regulating necroptosis. However, the mechanism of Se deficiency-induced necroptosis in the pig brain tissue and the role that miRNAs play in this process are unclear. Therefore, in this study, in vitro and pig models of Se deficiency were replicated, and electron microscopy, quantitative real-time polymerase chain reaction (qRT-PCR) and western blot assays were performed. The results showed that brain cells typically undergo necrotic changes, and that Se deficiency suppresses mir-29a-3p, which increases the levels of TNFRSF1A (TNFR1). Subsequently, a distinct increase in the necroptosis markers (RIPK1, RIPK3, and MLKL) and an evident decrease in caspase 8 was observed. And the expression of 10 selenoproteins was decreased. Moreover, the in vitro experiments showed that the expression of mir-29a-3p decreased as the Se content in the medium decreased and the application of an mir-29a-3p inhibitor increased the number of necrotic cells and the accumulation of ROS, and these effects were inhibited by necrostatin-1 (Nec-1) and N-acetyl-cysteine (NAC), respectively. Taken together, we proved that Se deficiency induced necroptosis both in vitro and in vivo through the targeted regulation of TNFR1 by mir-29a-3p in the pig brain.
Collapse
Affiliation(s)
- Jiawen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | | | | |
Collapse
|
14
|
BAKİ AM, VURAL P, AYDIN AF, SOLUK TEKKEŞİN M, DOĞRU-ABBASOĞLU S, UYSAL M. Effect of α-lipoic acid and N-acetylcysteine on liver oxidative stress, preneoplastic lesions induced by diethylnitrosamine plus high-fat diet. ARCHIVES OF CLINICAL AND EXPERIMENTAL MEDICINE 2021. [DOI: 10.25000/acem.830126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
15
|
Zhou Z, Cui Y, Zhang X, Zhang Y. The role of N-acetyl-cysteine (NAC) orally daily on the sperm parameters and serum hormones in idiopathic infertile men: A systematic review and meta-analysis of randomised controlled trials. Andrologia 2021; 53:e13953. [PMID: 33405232 DOI: 10.1111/and.13953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 01/10/2023] Open
Abstract
The meta-analysis was performed to access the role of N-acetyl-cysteine (NAC) orally daily on the sperm parameters and serum hormones in idiopathic infertile men. Randomised controlled trials (RCTs) were retrieved using PubMed, EMBASE and Cochrane register databases. The references of included studies were also searched. Finally, three articles including 431 infertile men were analysed. The results indicated that the NAC group had a considerable improvement in sperm concentration (mean difference [MD], 3.80; p < .00001), ejaculate volume (MD, 0.69; p = .002), sperm motility (MD, 4.69; p < .0001) and normal morphology (MD, 1.68; p = .0002) compared with the placebo group. However, in terms of serum hormones, the NAC group did not show significant difference in increasing the serum levels of testosterone (MD, 1.35; p = .21), luteinising hormone (MD, 0.82; p = .40), follicle-stimulating hormone (MD, -7.48; p = .29) and prolactin (MD, -0.34; p = .32) compared with the placebo group. In conclusion, NAC orally daily produced a greater improvement in sperm concentration, ejaculate volume, sperm motility and normal morphology for idiopathic infertile men, whereas no significant influence in serum hormones, which required more high-quality RCTs with sufficient sample sizes and statistics to prove.
Collapse
Affiliation(s)
- Zhongbao Zhou
- Department of Urology, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| | - Yuanshan Cui
- Department of Urology, Beijing TianTan Hospital, Capital Medical University, Beijing, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xiaoyi Zhang
- Department of Urology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yong Zhang
- Department of Urology, Beijing TianTan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Ammar MA, Abdou AMH. Benefits of N-acetylcysteine on liver functions in living donor hepatectomy. Indian J Anaesth 2020; 64:204-209. [PMID: 32346167 PMCID: PMC7179774 DOI: 10.4103/ija.ija_876_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 12/31/2019] [Accepted: 01/25/2020] [Indexed: 01/17/2023] Open
Abstract
Background and Aims: The proportion of patients undergoing living donor liver transplantation is high especially in countries without or with limited cadaver organ sharing programs. The aim of this study was to evaluate the post-hepatectomy effect of using N-Acetylcysteine (NAC) infusion in living donors undergoing donor hepatectomy. Methods: In a prospective randomised non-blinded study, 50 healthy donors were enrolled; following hepatectomy patients were randomised into 2 groups: Group NC receiving NAC 150 mg/kg diluted in 100 ml glucose 5% over 40 minutes, followed by NAC 12.5 mg/kg in 500 ml glucose 5% over 4 hours. This was followed by NAC 6.25 mg/kg for 2 post-operative days, Group C (Control group) received ringer acetate infusion at same rate for 2 days. The primary outcome was serum lactate levels. Secondary outcomes were liver function tests, serum creatinine and urine output on intensive care unit (ICU) admission (0 hr.), after 24 hours and 48 hours, length of ICU stay. Results: Our study revealed significant reduction in serum lactate in Group NC at 0, 24 and 48 hours compared to C group (P = 0.017, 0.002, 0.014). INR values showed significant reduction after 48 hours in Group NC compared to Group C (P = 0.049). Total Bilirubin, ALT, and Creatinine, urine output and ICU stay showed no statistical difference between the 2 groups. Conclusion: The NAC protocol is a safe, cost-effective tool for improvement of post hepatectomy liver function and early stabilisation of the metabolic profile.
Collapse
Affiliation(s)
- Mona A Ammar
- Department of Anesthesia and Intensive Care, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amr M Hilal Abdou
- Department of Anesthesia and Intensive Care, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
17
|
Abstract
Since current strategies for the treatment of multidrug-resistant tuberculosis (MDR-TB) and extensively drug-resistant tuberculosis (XDR-TB) have low efficacy and highly negative side effects, research on new treatments including novel drugs is essential for curing drug-resistant tuberculosis. Host-directed therapy (HDT) has become a promising idea to modulate host cell responses to enhance protective immunity against pathogens. Bazedoxifene (BZA), which belongs to a new generation of SERMs, shows the ability to inhibit the growth of M. tuberculosis in macrophages and is associated with autophagy. Our findings reveal a previously unrecognized antibacterial function of BZA. We propose that the mechanism of SERMs action in macrophages may provide a new potential measure for host-directed therapies against TB. Tuberculosis (TB) is still the leading killer caused by Mycobacterium tuberculosis infection. There is a clear need for new treatment strategy against TB. It has been reported that tamoxifen, known as a selective estrogen receptor modulator (SERM), exhibits antimycobacterial activity and inhibits M. tuberculosis growth in macrophages. However, it remains unknown whether such antimicrobial activity is a general property of all SERMs and how it works. In this study, we identified that bazedoxifene (BZA), a newer SERM, inhibits intracellular M. tuberculosis growth in macrophages. BZA treatment increases autophagosome formation and LC3B-II protein expression in M. tuberculosis-infected macrophages. We further demonstrated that the enhancement of autophagy by BZA is dependent on increased reactive oxygen species (ROS) production and associated with phosphorylation of Akt/mTOR signaling. In summary, our data reveal a previously unappreciated antimicrobial function of BZA and suggest that future investigation focusing on the mechanism of action of SERMs in macrophages may lead to new host-directed therapies against TB. IMPORTANCE Since current strategies for the treatment of multidrug-resistant tuberculosis (MDR-TB) and extensively drug-resistant tuberculosis (XDR-TB) have low efficacy and highly negative side effects, research on new treatments including novel drugs is essential for curing drug-resistant tuberculosis. Host-directed therapy (HDT) has become a promising idea to modulate host cell responses to enhance protective immunity against pathogens. Bazedoxifene (BZA), which belongs to a new generation of SERMs, shows the ability to inhibit the growth of M. tuberculosis in macrophages and is associated with autophagy. Our findings reveal a previously unrecognized antibacterial function of BZA. We propose that the mechanism of SERMs action in macrophages may provide a new potential measure for host-directed therapies against TB.
Collapse
|
18
|
Affiliation(s)
- Yabin Meng
- Department of Biomedical Engineering, School of EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
| | - Shuyan Han
- Department of Biomedical Engineering, School of EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
| | - Zhipeng Gu
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials EngineeringSichuan University Chengdu 610065 P. R. China
| | - Jun Wu
- Department of Biomedical Engineering, School of EngineeringSun Yat‐sen University Guangzhou 510006 P. R. China
| |
Collapse
|
19
|
Zuo S, Zou W, Wu RM, Yang J, Fan JN, Zhao XK, Li HY. Icariin Alleviates IL-1β-Induced Matrix Degradation By Activating The Nrf2/ARE Pathway In Human Chondrocytes. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3949-3961. [PMID: 31819369 PMCID: PMC6876636 DOI: 10.2147/dddt.s203094] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022]
Abstract
Objective Osteoarthritis (OA) is characterized by progressive matrix destruction of articular cartilage. This study aimed to investigate the potential antioxidative and chondroprotective effects and underlying mechanism of Icariin (ICA) in interleukin-1 beta (IL-1β)-induced extracellular matrix (ECM) degradation of OA cartilage. Methods Human chondrocyte cell line HC-A was treated with different doses of ICA, and then MTT assay and PI staining were used to estimate ICA-induced chondrocyte apoptosis. Intracellular ROS and superoxide dismutase (SOD) and glutathione peroxidase (GPX) were measured after treatment by IL-1β with or without ICA. The mRNA and protein expression levels of redox transcription factor Nrf2 and the downstream effector SOD-1, SOD-2, NQO-1 and HO-1 were assayed to explore the detailed mechanism by which ICA alleviates ECM degradation. Finally, to expound the role of Nrf2 in ICA-mediated chondroprotection, we specifically depleted Nrf2 in human chondrocytes and then pretreated them with ICA followed by IL-1β. Results ICA had no cytotoxic effects on human chondrocytes and 10−9 M was selected as the optimum concentration. ROS induced by IL-1β could drastically activate matrix-degrading proteases and ICA could significantly rescue the matrix degradation and excess ROS generation caused by IL-1β. We observed that ICA activated the Nrf2/ARE pathway, consequently upregulating the generation of GPX and SOD. Ablation of Nrf2 abrogated the chondroprotective and antioxidative effects of ICA in IL-1β-treated chondrocytes. Conclusion ICA alleviates IL-1β-induced matrix degradation and eliminates ROS by activating the Nrf2/ARE pathway.
Collapse
Affiliation(s)
- Shi Zuo
- Department of Hepatobiliary Surgery, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Wei Zou
- Department of Sports Medicine, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, People's Republic of China.,Department of Orthopedics, The Fourth People's Hospital of Guiyang, Guizhou, People's Republic of China
| | - Rong-Min Wu
- Department of Ultrasonography, The Maternity Hospital of Guizhou, Guiyang, Guizhou, People's Republic of China
| | - Jing Yang
- Department of Infectious Disease, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Jian-Nan Fan
- Department of Sports Medicine, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Xue-Ke Zhao
- Department of Infectious Disease, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Hai-Yang Li
- Department of Hepatobiliary Surgery, The Hospital Affiliated to Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| |
Collapse
|
20
|
Fan R, Bian M, Hu L, Liu W. A new rhodium(I) NHC complex inhibits TrxR: In vitro cytotoxicity and in vivo hepatocellular carcinoma suppression. Eur J Med Chem 2019; 183:111721. [PMID: 31577978 DOI: 10.1016/j.ejmech.2019.111721] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/27/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022]
Abstract
Thioredoxin reductase (TrxR) is often overexpressed in different types of cancer cells including hepatocellular carcinoma (HCC) cells and regarded as a target with great promise for anticancer drug research and development. Here, we have synthesized and characterized nine new designed rhodium(I) N-heterocyclic carbene (NHC) complexes. All of them were effective towards cancer cells, especially complex 1e was more active than cisplatin and manifested strong antiproliferative activity against HCC cells. In vivo anticancer studies showed that 1e significantly repressed tumor growth in an HCC nude mouse model and ameliorated liver lesions in a chronic HCC model caused by CCl4. Notably, a mechanistic study revealed that 1e can strongly inhibit TrxR system both in vitro and in vivo. Furthermore, 1e promoted intracellular ROS accumulation, damaged mitochondrial membrane potential, promoted cancer cell apoptosis and blocked the cells in the G1 phase.
Collapse
Affiliation(s)
- Rong Fan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mianli Bian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lihong Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wukun Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
21
|
Jannatifar R, Parivar K, Roodbari NH, Nasr-Esfahani MH. Effects of N-acetyl-cysteine supplementation on sperm quality, chromatin integrity and level of oxidative stress in infertile men. Reprod Biol Endocrinol 2019; 17:24. [PMID: 30771790 PMCID: PMC6377938 DOI: 10.1186/s12958-019-0468-9] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/08/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Infertile men have higher levels of semen reactive oxygen species (ROS) than fertile men. High levels of semen ROS can cause sperm dysfunction, sperm DNA damage and reduced male reproductive potential. This study investigated the effects of supplementation with N-acetyl-cysteine (NAC) on the sperm quality, chromatin integrity and levels of oxidative stress in infertile men. METHODS The study was carried out in the unit of ACECR Infertility Research Center, Qom, Iran. The patients consisted of 50 infertile men with asthenoteratozoospermia who received NAC (600 mg/d) orally for 3 months, after which they were compared with pre-treatment status. Semen was analyzed according to WHO (2010), followed by the assessment of protamine content [chromomycin A3 (CMA3)] and DNA integrity [terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)]. Oxidative stress markers, i.e. total antioxidant capacity (TAC) and malondialdehyde (MDA), as well as hormonal profile (LH, FSH, Testosterone and Prolactin) were determined by ELISA kit. RESULTS After NAC treatment, patients' sperm count and motility increased significantly whereas abnormal morphology, DNA fragmentation and protamine deficiency showed significant decreases compared to pre-treatment levels (P < 0.05). Hormonal profile improvement was associated with lowered FSH and LH levels and increased amount of testosterone (P < 0.05). TAC significantly increased and MDA decreased with an inverse significant correlation between TAC and MDA (P < 0.05). CONCLUSION NAC oral supplementation may improve sperm parameters and oxidative/antioxidant status in infertile males.
Collapse
Affiliation(s)
- Rahil Jannatifar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Kazem Parivar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nasim Hayati Roodbari
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Reproductive Biotechnology, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Biotechnology, Isfahan, Iran.
| |
Collapse
|
22
|
Sun Y, Miao H, Ma S, Zhang L, You C, Tang F, Yang C, Tian X, Wang F, Luo Y, Lin X, Wang H, Li C, Li Z, Yu H, Liu X, Xiao Y, Gong Y, Zhang J, Quan H, Xie C. FePt-Cys nanoparticles induce ROS-dependent cell toxicity, and enhance chemo-radiation sensitivity of NSCLC cells in vivo and in vitro. Cancer Lett 2018; 418:27-40. [PMID: 29331422 DOI: 10.1016/j.canlet.2018.01.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/31/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022]
Abstract
FePt-Cys nanoparticles (FePt-Cys NPs) have been well used in many fields, despite their poor solubility and stability. We synthetized a cysteine surface modified FePt NPs, which exhibited good solubility, stability and biocompatibility. We explored the insight mechanisms of the antitumor effects of this new nanoparticle system in lung cancer cells. In the in vitro study, FePt-Cys NPs induced a reactive oxygen species (ROS) burst, which suppressed the antioxidant protein expression and induced cell apoptosis. Furthermore, FePt-Cys NPs prevented the migration and invasion of H1975 and A549 cells. These changes were correlated with a dramatic decrease in MMP-2/9 expression and enhanced the cellular attachment. We demonstrated that FePt-Cys NPs promoted the effects of chemo-radiation through activation of the caspase system and impairment of DNA damage repair. In the in vivo study, no severe allergies or drug-related deaths were observed and FePt-Cys NPs showed a synergistic effect with cisplatin and radiation. In conclusion, with good safety and efficacy, FePt-Cys NPs could therefore be potential sensitizers for chemoradiotherapy.
Collapse
Affiliation(s)
- Yingming Sun
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongtao Miao
- Key Laboratory of Artificial Micro- and Nano-Structures of the Ministry of Education and Center for Electronic Microscopy and Department of Physics, Wuhan University, Wuhan, China
| | - Shijing Ma
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lei Zhang
- Key Laboratory of Artificial Micro- and Nano-Structures of the Ministry of Education and Center for Electronic Microscopy and Department of Physics, Wuhan University, Wuhan, China
| | - Chengcheng You
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fang Tang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Cui Yang
- Key Laboratory of Artificial Micro- and Nano-Structures of the Ministry of Education and Center for Electronic Microscopy and Department of Physics, Wuhan University, Wuhan, China
| | - Xiaoli Tian
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Feng Wang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Luo
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiangjie Lin
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hui Wang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chunyang Li
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhijun Li
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongnv Yu
- Central Laboratory of Xinhua Hospital of Dalian University, Department of Medical Oncology, Xinhua Hospital of Dalian University, Dalian, China
| | - Xuefeng Liu
- The Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington DC, USA
| | - Yu Xiao
- Department of Urology, Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junhong Zhang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hong Quan
- Key Laboratory of Artificial Micro- and Nano-Structures of the Ministry of Education and Center for Electronic Microscopy and Department of Physics, Wuhan University, Wuhan, China.
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China; Center for Medical Science Research, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
23
|
Kemp R, Mole J, Gomez D. Current evidence for the use of N-acetylcysteine following liver resection. ANZ J Surg 2017; 88:E486-E490. [PMID: 29132195 DOI: 10.1111/ans.14295] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND N-acetylcysteine (NAC) has many uses in medicine; notable in the management of paracetamol toxicity, acute liver failure and liver surgery. The aim of this review was to critically appraise the published literature for the routine use of NAC in liver resection surgery. METHODS An electronic search was performed of EBSCOhost (Medline and CINAHL database), PubMed and the Cochrane Library for the period 1990-2016. MeSH headings: 'acetyl-cysteine', 'liver resection' and 'hepatectomy' were used to identify all relevant articles published in English. RESULTS Following the search criteria used, three articles were included. Two of these studies were randomized controlled trials. All the studies collated data on morbidity and mortality. All three studies did not show a significant difference in overall complications rates in patients that underwent hepatic resection that had NAC infusion compared with patients that did not. In one study, NAC administration was associated with a higher frequency of grade A post-hepatectomy liver failure. In another study, a significantly higher incidence of delirium was observed in the NAC group, which led to the trial to be terminated early. CONCLUSION The current published data do not support the routine use of NAC following liver resection.
Collapse
Affiliation(s)
- Richard Kemp
- Department of Anaesthesia, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Jonathan Mole
- Department of Anaesthesia, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Dhanny Gomez
- Department of Hepatobiliary and Pancreatic Surgery, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, UK.,NIHR Nottingham Digestive Disease Biomedical Research Unit, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
24
|
Ester-Modified Cyclometalated Iridium(III) Complexes as Mitochondria-Targeting Anticancer Agents. Sci Rep 2016; 6:38954. [PMID: 27958338 PMCID: PMC5154195 DOI: 10.1038/srep38954] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/14/2016] [Indexed: 12/11/2022] Open
Abstract
Organometallic iridium complexes are potent anticancer candidates which act through different mechanisms from cisplatin-based chemotherapy regimens. Here, ten phosphorescent cyclometalated iridium(III) complexes containing 2,2'-bipyridine-4,4'-dicarboxylic acid and its diester derivatives as ligands are designed and synthesized. The modification by ester group, which can be hydrolysed by esterase, facilitates the adjustment of drug-like properties. The quantum yields and emission lifetimes are influenced by variation of the ester substituents on the Ir(III) complexes. The cytotoxicity of these Ir(III) complexes is correlated with the length of their ester groups. Among them, 4a and 4b are found to be highly active against a panel of cancer cells screened, including cisplatin-resistant cancer cells. Mechanism studies in vitro indicate that they undergo hydrolysis of ester bonds, accumulate in mitochondria, and induce a series of cell-death related events mediated by mitochondria. Furthermore, 4a and 4b can induce pro-death autophagy and apoptosis simultaneously. Our study indicates that ester modification is a simple and feasible strategy to enhance the anticancer potency of Ir(III) complexes.
Collapse
|
25
|
Santoro AM, Monaco I, Attanasio F, Lanza V, Pappalardo G, Tomasello MF, Cunsolo A, Rizzarelli E, De Luigi A, Salmona M, Milardi D. Copper(II) ions affect the gating dynamics of the 20S proteasome: a molecular and in cell study. Sci Rep 2016; 6:33444. [PMID: 27633879 PMCID: PMC5025780 DOI: 10.1038/srep33444] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/09/2016] [Indexed: 12/14/2022] Open
Abstract
Due to their altered metabolism cancer cells are more sensitive to proteasome inhibition or changes of copper levels than normal cells. Thus, the development of copper complexes endowed with proteasome inhibition features has emerged as a promising anticancer strategy. However, limited information is available about the exact mechanism by which copper inhibits proteasome. Here we show that Cu(II) ions simultaneously inhibit the three peptidase activities of isolated 20S proteasomes with potencies (IC50) in the micromolar range. Cu(II) ions, in cell-free conditions, neither catalyze red-ox reactions nor disrupt the assembly of the 20S proteasome but, rather, promote conformational changes associated to impaired channel gating. Notably, HeLa cells grown in a Cu(II)-supplemented medium exhibit decreased proteasome activity. This effect, however, was attenuated in the presence of an antioxidant. Our results suggest that if, on one hand, Cu(II)-inhibited 20S activities may be associated to conformational changes that favor the closed state of the core particle, on the other hand the complex effect induced by Cu(II) ions in cancer cells is the result of several concurring events including ROS-mediated proteasome flooding, and disassembly of the 26S proteasome into its 20S and 19S components.
Collapse
Affiliation(s)
- Anna Maria Santoro
- Istituto di Biostrutture e Bioimmagini - CNR Sede di Catania, Via P. Gaifami, 9- 95126 Catania, Italy
| | - Irene Monaco
- Istituto di Biostrutture e Bioimmagini - CNR Sede di Catania, Via P. Gaifami, 9- 95126 Catania, Italy
- Fondazione RiMed, Via Bandiera 11, 90133, Palermo, Italy
| | - Francesco Attanasio
- Istituto di Biostrutture e Bioimmagini - CNR Sede di Catania, Via P. Gaifami, 9- 95126 Catania, Italy
| | - Valeria Lanza
- Istituto di Biostrutture e Bioimmagini - CNR Sede di Catania, Via P. Gaifami, 9- 95126 Catania, Italy
| | - Giuseppe Pappalardo
- Istituto di Biostrutture e Bioimmagini - CNR Sede di Catania, Via P. Gaifami, 9- 95126 Catania, Italy
| | - Marianna Flora Tomasello
- Istituto di Biostrutture e Bioimmagini - CNR Sede di Catania, Via P. Gaifami, 9- 95126 Catania, Italy
| | - Alessandra Cunsolo
- Dipartimento di Scienze Chimiche, Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Enrico Rizzarelli
- Istituto di Biostrutture e Bioimmagini - CNR Sede di Catania, Via P. Gaifami, 9- 95126 Catania, Italy
- Dipartimento di Scienze Chimiche, Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Ada De Luigi
- IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Via Giuseppe La Masa 19, 20156, Milano, Italy
| | - Mario Salmona
- IRCCS-Istituto di Ricerche Farmacologiche “Mario Negri”, Via Giuseppe La Masa 19, 20156, Milano, Italy
| | - Danilo Milardi
- Istituto di Biostrutture e Bioimmagini - CNR Sede di Catania, Via P. Gaifami, 9- 95126 Catania, Italy
| |
Collapse
|
26
|
Grendar J, Ouellet JF, McKay A, Sutherland FR, Bathe OF, Ball CG, Dixon E. Effect of N-acetylcysteine on liver recovery after resection: A randomized clinical trial. J Surg Oncol 2016; 114:446-50. [PMID: 27302646 DOI: 10.1002/jso.24312] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/16/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND OBJECTIVES Liver failure following hepatic resection is a multifactorial complication. In experimental studies, infusion of N-acetylcysteine (NAC) can minimize hepatic parenchymal injury. METHODS Patients undergoing liver resection were randomized to postoperative care with or without NAC. No blinding was performed. Overall complication rate was the primary outcome; liver failure, length of stay, and mortality were secondary outcomes. Due to safety concerns, a premature multivariate analysis was performed and included within the model randomization to NAC, preoperative ASA, extent of resection, and intraoperative vascular occlusion as factors. RESULTS Two hundred and six patients were randomized (110 to conventional therapy; 96 to NAC). No significant differences were noted in overall complications (32.7% and 45.7%, P = 0.06) or hepatic failure (3.6% and 5.4%, P = 0.537) between treatment groups. There was significantly more delirium within the NAC group (2.7% and 9.8%, P < 0.05) that caused early trial termination. In multivariate analysis, only randomization to NAC (OR = 2.21, 95%CI = 1.16-4.19) and extensive resections (OR = 2.28, 95%CI = 1.22-4.29) were predictive of postoperative complications. CONCLUSIONS Patients randomized to postoperative NAC received no benefit. There was a trend toward a higher rate of overall complications and a significantly higher rate of delirium in the NAC group. J. Surg. Oncol. 2016;114:446-450. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jan Grendar
- Providence Portland Medical Center, Portland, Oregon
| | - Jean F Ouellet
- CHU-Pavillon Hopital de l'Enfant-Jesus, Quebec City, Quebec, Canada
| | - Andrew McKay
- Department of Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Oliver F Bathe
- Division of General Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Chad G Ball
- Division of General Surgery, University of Calgary, Calgary, Alberta, Canada
| | - Elijah Dixon
- Division of General Surgery, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
27
|
Intracellular ROS mediates gas plasma-facilitated cellular transfection in 2D and 3D cultures. Sci Rep 2016; 6:27872. [PMID: 27296089 PMCID: PMC4906281 DOI: 10.1038/srep27872] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 05/26/2016] [Indexed: 12/18/2022] Open
Abstract
This study reports the potential of cold atmospheric plasma (CAP) as a versatile tool
for delivering oligonucleotides into mammalian cells. Compared to lipofection and
electroporation methods, plasma transfection showed a better uptake efficiency and
less cell death in the transfection of oligonucleotides. We demonstrated that the
level of extracellular aqueous reactive oxygen species (ROS) produced by gas plasma
is correlated with the uptake efficiency and that this is achieved through an
increase of intracellular ROS levels and the resulting increase in cell membrane
permeability. This finding was supported by the use of ROS scavengers, which reduced
CAP-based uptake efficiency. In addition, we found that cold atmospheric plasma
could transfer oligonucleotides such as siRNA and miRNA into cells even in 3D
cultures, thus suggesting the potential for unique applications of CAP beyond those
provided by standard transfection techniques. Together, our results suggest that
cold plasma might provide an efficient technique for the delivery of siRNA and miRNA
in 2D and 3D culture models.
Collapse
|
28
|
Singh NP, Singh UP, Rouse M, Zhang J, Chatterjee S, Nagarkatti PS, Nagarkatti M. Dietary Indoles Suppress Delayed-Type Hypersensitivity by Inducing a Switch from Proinflammatory Th17 Cells to Anti-Inflammatory Regulatory T Cells through Regulation of MicroRNA. THE JOURNAL OF IMMUNOLOGY 2015; 196:1108-22. [PMID: 26712945 DOI: 10.4049/jimmunol.1501727] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 11/22/2015] [Indexed: 12/14/2022]
Abstract
Aryl hydrocarbon receptor (AhR) has been shown to have profound influence on T cell differentiation, and use of distinct AhR ligands has shown that whereas some ligands induce regulatory T cells (Tregs), others induce Th17 cells. In the present study, we tested the ability of dietary AhR ligands (indole-3-carbinol [I3C] and 3,3'-diindolylmethane [DIM]) and an endogenous AhR ligand, 6-formylindolo(3,2-b)carbazole (FICZ), on the differentiation and functions of Tregs and Th17 cells. Treatment of C57BL/6 mice with indoles (I3C or DIM) attenuated delayed-type hypersensitivity (DTH) response to methylated BSA and generation of Th17 cells while promoting Tregs. In contrast, FICZ exacerbated the DTH response and promoted Th17 cells. Indoles decreased the induction of IL-17 but promoted IL-10 and Foxp3 expression. Also, indoles caused reciprocal induction of Tregs and Th17 cells only in wild-type (AhR(+/+)) but not in AhR knockout (AhR(-/-)) mice. Upon analysis of microRNA (miR) profile in draining lymph nodes of mice with DTH, treatment with I3C and DIM decreased the expression of several miRs (miR-31, miR-219, and miR-490) that targeted Foxp3, whereas it increased the expression of miR-495 and miR-1192 that were specific to IL-17. Interestingly, treatment with FICZ had precisely the opposite effects on these miRs. Transfection studies using mature miR mimics of miR-490 and miR-1192 that target Foxp3 and IL-17, respectively, or scrambled miR (mock) or inhibitors confirmed that these miRs specifically targeted Foxp3 and IL-17 genes. Our studies demonstrate, to our knowledge for the first time, that the ability of AhR ligands to regulate the differentiation of Tregs versus Th17 cells may depend on miR signature profile.
Collapse
Affiliation(s)
- Narendra P Singh
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208
| | - Udai P Singh
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208
| | - Michael Rouse
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208
| | - Jiajia Zhang
- Department of Epidemiology and Biostatistics, University of South Carolina, Columbia, SC 29208; and
| | - Saurabh Chatterjee
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC 29208
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208;
| |
Collapse
|
29
|
Bonet-Ponce L, Saez-Atienzar S, da Casa C, Sancho-Pelluz J, Barcia JM, Martinez-Gil N, Nava E, Jordan J, Romero FJ, Galindo MF. Rotenone Induces the Formation of 4-Hydroxynonenal Aggresomes. Role of ROS-Mediated Tubulin Hyperacetylation and Autophagic Flux Disruption. Mol Neurobiol 2015; 53:6194-6208. [PMID: 26558631 DOI: 10.1007/s12035-015-9509-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/22/2015] [Indexed: 01/13/2023]
Abstract
Oxidative stress causes cellular damage by (i) altering protein stability, (ii) impairing organelle function, or (iii) triggering the formation of 4-HNE protein aggregates. The catabolic process known as autophagy is an antioxidant cellular response aimed to counteract these stressful conditions. Therefore, autophagy might act as a cytoprotective response by removing impaired organelles and aggregated proteins. In the present study, we sought to understand the role of autophagy in the clearance of 4-HNE protein aggregates in ARPE-19 cells under rotenone exposure. Rotenone induced an overproduction of reactive oxygen species (ROS), which led to an accumulation of 4-HNE inclusions, and an increase in the number of autophagosomes. The latter resulted from a disturbed autophagic flux rather than an activation of the autophagic synthesis pathway. In compliance with this, rotenone treatment induced an increase in LC3-II while upstream autophagy markers such as Beclin- 1, Vsp34 or Atg5-Atg12, were decreased. Rotenone reduced the autophagosome-to-lysosome fusion step by increasing tubulin acetylation levels through a ROS-mediated pathway. Proof of this is the finding that the free radical scavenger, N-acetylcysteine, restored autophagy flux and reduced rotenone-induced tubulin hyperacetylation. Indeed, this dysfunctional autophagic response exacerbates cell death triggered by rotenone, since 3-methyladenine, an autophagy inhibitor, reduced cell mortality, while rapamycin, an inductor of autophagy, caused opposite effects. In summary, we shed new light on the mechanisms involved in the autophagic responses disrupted by oxidative stress, which take place in neurodegenerative diseases such as Huntington or Parkinson diseases, and age-related macular degeneration.
Collapse
Affiliation(s)
- Luis Bonet-Ponce
- Facultad de Medicina y Odontología, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Sara Saez-Atienzar
- Facultad de Medicina y Odontología, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain.,Unidad de Neuropsicofarmacología Traslacional, Complejo Hospitalario Universitario de Albacete, Albacete, Spain.,Grupo de Neurofarmacología, Dpto. Ciencias Médicas. Facultad de Medicina de Albacete, IDINE, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Carmen da Casa
- Grupo de Neurofarmacología, Dpto. Ciencias Médicas. Facultad de Medicina de Albacete, IDINE, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Javier Sancho-Pelluz
- Facultad de Medicina y Odontología, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Jorge M Barcia
- Facultad de Medicina y Odontología, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Natalia Martinez-Gil
- Facultad de Medicina y Odontología, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Eduardo Nava
- Grupo de Neurofarmacología, Dpto. Ciencias Médicas. Facultad de Medicina de Albacete, IDINE, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Joaquín Jordan
- Grupo de Neurofarmacología, Dpto. Ciencias Médicas. Facultad de Medicina de Albacete, IDINE, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Francisco J Romero
- Facultad de Medicina y Odontología, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Maria F Galindo
- Unidad de Neuropsicofarmacología Traslacional, Complejo Hospitalario Universitario de Albacete, Albacete, Spain.
| |
Collapse
|
30
|
Kawano Y, Ohta M, Iwashita Y, Komori Y, Inomata M, Kitano S. Effects of the dihydrolipoyl histidinate zinc complex against carbon tetrachloride-induced hepatic fibrosis in rats. Surg Today 2015; 44:1744-50. [PMID: 24121950 DOI: 10.1007/s00595-013-0749-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/22/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE This study investigated the effects of an antioxidant, dihydrolipoyl histidinate zinc complex (DHLHZn), on the hepatic fibrosis in the carbon tetrachloride (CCl4) rat model. METHODS The animals were divided into three groups: control, CCl4, and CCl4+DHLHZn. A histological assessment of the liver fibrosis was performed using stained liver samples. The oxidative stress and antioxidant levels were evaluated by measuring the malondialdehyde (MDA) and glutathione (GSH) levels in the liver. In addition, cultured human hepatic stellate cells (LI90) were exposed to antimycin-A (AMA) and divided into four groups: control, DHLHZn, AMA, and AMA+DHLHZn. The effects of DHLHZn on AMA-induced fibrosis were evaluated by measuring the expression of transforming growth factor (TGF)-β1 and collagen α1 (I). RESULTS The hepatic fibrosis in the CCl4+DHLHZn group was attenuated compared to that in the CCl4 group. The MDA levels in the CCl4+DHLHZn group were significantly lower than those of the CCl4 group, whereas the GSH levels in the CCl4+DHLHZn group were significantly higher than those of the CCl4 group. Furthermore, the relative mRNA expression of TGF-β1 and collagen α1 (I) in the AMA+DHLHZn group was significantly lower than that in the AMA group. CONCLUSION DHLHZn may attenuate the hepatic fibrosis induced by CCl4 by decreasing the degree of oxidative stress.
Collapse
|
31
|
P2X(7) receptor in the kidneys of diabetic rats submitted to aerobic training or to N-acetylcysteine supplementation [corrected]. PLoS One 2014; 9:e97452. [PMID: 24940871 PMCID: PMC4062402 DOI: 10.1371/journal.pone.0097452] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 04/20/2014] [Indexed: 12/22/2022] Open
Abstract
Previous studies in our laboratory showed that N-acetylcysteine supplementation or aerobic training reduced oxidative stress and the progression of diabetic nephropathy in rats. The P2X7 receptor is up-regulated in pathological conditions, such as diabetes mellitus. This up-regulation is related to oxidative stress and induces tissue apoptosis or necrosis. The aim of the present study is to assess the role of P2X7 receptor in the kidneys of diabetic rats submitted to aerobic training or N-acetylcysteine supplementation. Diabetes was induced in male Wistar rats by streptozotocin (60 mg/kg, i.v.) and the training was done on a treadmill; N-acetylcysteine was given in the drinking water (600 mg/L). By confocal microscopy, as compared to control, the kidneys of diabetic rats showed increased P2×7 receptor expression and a higher activation in response to 2′(3′)-O-(4-benzoylbenzoyl) adenosine5'–triphosphate (specific agonist) and adenosine triphosphate (nonspecific agonist) (all p<0.05). All these alterations were reduced in diabetic rats treated with N-acetylcysteine, exercise or both. We also observed measured proteinuria and albuminuria (early marker of diabetic nephropathy) in DM groups. Lipoperoxidation was strongly correlated with P2X7 receptor expression, which was also correlated to NO•, thus associating this receptor to oxidative stress and kidney lesion. We suggest that P2X7 receptor inhibition associated with the maintenance of redox homeostasis could be useful as coadjuvant treatment to delay the progression of diabetic nephropathy.
Collapse
|
32
|
Mandal AK, Ghosh D, Sarkar S, Ghosh A, Swarnakar S, Das N. Nanocapsulated quercetin downregulates rat hepatic MMP-13 and controls diethylnitrosamine-induced carcinoma. Nanomedicine (Lond) 2014; 9:2323-37. [PMID: 24593002 DOI: 10.2217/nnm.14.11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIMS The aims of our work were to investigate the controlling role and the efficacy of nanocapsulated quercetin drug delivery system on the decrement of inflammatory mediators such as MMP-13 in diethyl nitrosamine (DEN)-induced hepatocarcinogenesis. MATERIALS & METHODS Hepatocellular carcinoma was developed in the Swiss albino rats by the exposure of DEN. DEN administration caused the generation of reactive oxygen species, upregulation of TNF-α, IL-6, activation of MMP-13, severe oxidative damage, hyperplastic nodules with preneoplastic lesions and the histopathological changes in rat liver. RESULTS & CONCLUSION Nanocapsulated quercetin treatment restricted all alterations in DEN-mediated development of hepatocarcinogenesis. Therefore, it may be concluded that nanocapsulated quercetin may be accepted as a potent therapeutic formulation in preventing DEN-mediated hepatocarcinogenesis.
Collapse
Affiliation(s)
- Ardhendu Kumar Mandal
- Drug Development/Diagnostics & Biotechnology Division, Indian Institute of Chemical Biology, 4, Raja SC Mullick Road, Jadavpur, Kolkata - 700032, India
| | | | | | | | | | | |
Collapse
|
33
|
Cao JJ, Picklo MJ. N-acetylcysteine supplementation decreases osteoclast differentiation and increases bone mass in mice fed a high-fat diet. J Nutr 2014; 144:289-96. [PMID: 24381219 DOI: 10.3945/jn.113.185397] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Obesity induced by high-fat (HF) diets increases bone resorption, decreases trabecular bone mass, and reduces bone strength in various animal models. This study investigated whether N-acetylcysteine (NAC), an antioxidant and a glutathione precursor, alters glutathione status and mitigates bone microstructure deterioration in mice fed an HF diet. Forty-eight 6-wk-old male C57BL/6 mice were randomly assigned to 4 treatment groups (n = 12 per group) and fed either a normal-fat [NF (10% energy as fat)] or an HF (45% energy as fat) diet ad libitum with or without NAC supplementation at 1 g/kg diet for 17 wk. Compared with the NF groups, mice in the HF groups had higher body weight, greater serum leptin concentrations and osteoclast differentiation, and lower trabecular bone volume, trabecular number, and connectivity density (P < 0.05). NAC supplementation increased the serum-reduced glutathione concentration and bone volume and decreased osteoclast differentiation in HF-fed mice (P < 0.05). We further demonstrated that osteoclast differentiation was directly regulated by glutathione status. NAC treatment of murine macrophage RAW 264.7 cells in vitro increased glutathione status and decreased osteoclast formation. These results show that NAC supplementation increases the bone mass of obese mice induced by an HF diet through elevating glutathione status and decreasing bone resorption.
Collapse
Affiliation(s)
- Jay J Cao
- USDA, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND
| | | |
Collapse
|
34
|
Ho CY, Weng CJ, Jhang JJ, Cheng YT, Huang SM, Yen GC. Diallyl sulfide as a potential dietary agent to reduce TNF-α- and histamine-induced proinflammatory responses in A7r5 cells. Mol Nutr Food Res 2014; 58:1069-78. [DOI: 10.1002/mnfr.201300617] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 11/20/2013] [Accepted: 11/24/2013] [Indexed: 12/15/2022]
Affiliation(s)
- Cheng-Ying Ho
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
| | - Chia-Jui Weng
- Graduate Institute of Applied Living Science; Tainan University of Technology; Yongkang Distric; Tainan City Taiwan
| | - Jhih-Jia Jhang
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
| | - Yu-Ting Cheng
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
| | - Shang-Ming Huang
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology; National Chung Hsing University; Taichung Taiwan
- Agricultural Biotechnology Center; National Chung Hsing University; Taichung Taiwan
| |
Collapse
|
35
|
Li L, Hu X, Xia Y, Xiao G, Zheng P, Wang C. Linkage of oxidative stress and mitochondrial dysfunctions to spontaneous culture degeneration in Aspergillus nidulans. Mol Cell Proteomics 2013; 13:449-61. [PMID: 24345786 DOI: 10.1074/mcp.m113.028480] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Filamentous fungi including mushrooms frequently and spontaneously degenerate during subsequent culture maintenance on artificial media, which shows the loss or reduction abilities of asexual sporulation, sexuality, fruiting, and production of secondary metabolites, thus leading to economic losses during mass production. To better understand the underlying mechanisms of fungal degeneration, the model fungus Aspergillus nidulans was employed in this study for comprehensive analyses. First, linkage of oxidative stress to culture degeneration was evident in A. nidulans. Taken together with the verifications of cell biology and biochemical data, a comparative mitochondrial proteome analysis revealed that, unlike the healthy wild type, a spontaneous fluffy sector culture of A. nidulans demonstrated the characteristics of mitochondrial dysfunctions. Relative to the wild type, the features of cytochrome c release, calcium overload and up-regulation of apoptosis inducing factors evident in sector mitochondria suggested a linkage of fungal degeneration to cell apoptosis. However, the sector culture could still be maintained for generations without the signs of growth arrest. Up-regulation of the heat shock protein chaperones, anti-apoptotic factors and DNA repair proteins in the sector could account for the compromise in cell death. The results of this study not only shed new lights on the mechanisms of spontaneous degeneration of fungal cultures but will also provide alternative biomarkers to monitor fungal culture degeneration.
Collapse
Affiliation(s)
- Lin Li
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|
36
|
Martinez-Outschoorn UE, Curry JM, Ko YH, Lin Z, Tuluc M, Cognetti D, Birbe RC, Pribitkin E, Bombonati A, Pestell RG, Howell A, Sotgia F, Lisanti MP. Oncogenes and inflammation rewire host energy metabolism in the tumor microenvironment: RAS and NFκB target stromal MCT4. Cell Cycle 2013; 12:2580-97. [PMID: 23860378 PMCID: PMC3865048 DOI: 10.4161/cc.25510] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Here, we developed a model system to evaluate the metabolic effects of oncogene(s) on the host microenvironment. A matched set of "normal" and oncogenically transformed epithelial cell lines were co-cultured with human fibroblasts, to determine the "bystander" effects of oncogenes on stromal cells. ROS production and glucose uptake were measured by FACS analysis. In addition, expression of a panel of metabolic protein biomarkers (Caveolin-1, MCT1, and MCT4) was analyzed in parallel. Interestingly, oncogene activation in cancer cells was sufficient to induce the metabolic reprogramming of cancer-associated fibroblasts toward glycolysis, via oxidative stress. Evidence for "metabolic symbiosis" between oxidative cancer cells and glycolytic fibroblasts was provided by MCT1/4 immunostaining. As such, oncogenes drive the establishment of a stromal-epithelial "lactate-shuttle", to fuel the anabolic growth of cancer cells. Similar results were obtained with two divergent oncogenes (RAS and NFκB), indicating that ROS production and inflammation metabolically converge on the tumor stroma, driving glycolysis and upregulation of MCT4. These findings make stromal MCT4 an attractive target for new drug discovery, as MCT4 is a shared endpoint for the metabolic effects of many oncogenic stimuli. Thus, diverse oncogenes stimulate a common metabolic response in the tumor stroma. Conversely, we also show that fibroblasts protect cancer cells against oncogenic stress and senescence by reducing ROS production in tumor cells. Ras-transformed cells were also able to metabolically reprogram normal adjacent epithelia, indicating that cancer cells can use either fibroblasts or epithelial cells as "partners" for metabolic symbiosis. The antioxidant N-acetyl-cysteine (NAC) selectively halted mitochondrial biogenesis in Ras-transformed cells, but not in normal epithelia. NAC also blocked stromal induction of MCT4, indicating that NAC effectively functions as an "MCT4 inhibitor". Taken together, our data provide new strategies for achieving more effective anticancer therapy. We conclude that oncogenes enable cancer cells to behave as selfish "metabolic parasites", like foreign organisms (bacteria, fungi, viruses). Thus, we should consider treating cancer like an infectious disease, with new classes of metabolically targeted "antibiotics" to selectively starve cancer cells. Our results provide new support for the "seed and soil" hypothesis, which was first proposed in 1889 by the English surgeon, Stephen Paget.
Collapse
|
37
|
Mesenchymal stem cells restore CCl4-induced liver injury by an antioxidative process. Cell Biol Int 2013; 36:1267-74. [PMID: 23035905 DOI: 10.1042/cbi20110634] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We have investigated BM (bone marrow)-derived MSCs (mesenchymal stem cells) for the treatment of liver injury. It was hypothesized that MSC-mediated resolution of liver injury could occur through an antioxidative process. After being injected with CCl4 (carbon tetrachloride), mice were injected with syngenic BM-derived MSCs or normal saline. Oxidative stress activity of the MSCs was determined by the analysis of ROS (reactive oxygen species) and SOD (superoxide dismutase) activity. In addition, cytoprotective genes of the liver tissue were assessed by real-time PCR and ARE (antioxidant-response element) reporter assay. Up-regulated ROS of CCl4-treated liver cells was attenuated by co-culturing with MSCs. Suppression of SOD by adding an SOD inhibitor decreased the effect of MSCs on injured liver cells. MSCs significantly increased SOD activity and inhibited ROS production in the injured liver. The gene expression levels of Hmox-1 (haem oxygenase-1), BI-1 (Bax inhibitor-1), HGF (hepatocyte growth factor), GST (glutathione transferase) and Nrf2 (nuclear factor-erythoid 2 p45 subunit-related factor 20), attenuated by CCl4, were increased up to basal levels after MSC transplantation. In addition, MSCs induced an ARE, shown by luciferase activity, which represented a cytoprotective response in the injured liver. Evidence of a new cytoprotective effect is shown in which MSCs promote an antioxidant response and supports the potential of using MSC transplantation as an effective treatment modality for liver disease.
Collapse
|
38
|
Ni Z, Dai X, Wang B, Ding W, Cheng P, Xu L, Lian J, He F. Natural Bcl-2 inhibitor (-)- gossypol induces protective autophagy via reactive oxygen species-high mobility group box 1 pathway in Burkitt lymphoma. Leuk Lymphoma 2013; 54:2263-8. [PMID: 23398207 DOI: 10.3109/10428194.2013.775437] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
(-)- Gossypol, a natural inhibitor of anti-apoptotic Bcl-2 proteins, has presented an effective anti-tumor activity in numerous preclinical trials. More and more evidence in vivo and in vitro validates that (-)- gossypol can dramatically suppress cell proliferation and induce cell death in hematological malignancies. However, the detailed mechanisms are not well known. In the present study, we showed that treatment with (-)- gossypol stimulated reactive oxygen species (ROS) generation and induced autophagy in Burkitt lymphoma cells. Antioxidant N-acetyl-cysteine (NAC) pretreatment attenuated (-)- gossypol-induced autophagy. Furthermore, (-)- gossypol treatment increased the translocation of high mobility group box 1 (HMGB1) from nuclei to cytoplasm, which can be suppressed by NAC pretreatment. NAC pretreatment also dramatically enhanced (-)- gossypol-induced apoptosis and total cell death. These results indicate that (-)- gossypol induces a protective autophagy in Burkitt lymphoma cells, partly due to ROS induction and cytosolic translocation of HMGB1. Antioxidants may serve as potent chemosensitizers to enhance cell death through blocking (-)- gossypol-induced autophagy.
Collapse
Affiliation(s)
- Zhenhong Ni
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University , Chongqing , China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Cheng P, Ni Z, Dai X, Wang B, Ding W, Rae Smith A, Xu L, Wu D, He F, Lian J. The novel BH-3 mimetic apogossypolone induces Beclin-1- and ROS-mediated autophagy in human hepatocellular carcinoma [corrected] cells. Cell Death Dis 2013; 4:e489. [PMID: 23392177 PMCID: PMC3734844 DOI: 10.1038/cddis.2013.17] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Apogossypolone (ApoG2), a novel derivative of gossypol, exhibits superior antitumor activity in Bcl-2 transgenic mice, and induces autophagy in several cancer cells. However, the detailed mechanisms are not well known. In the present study, we showed that ApoG2 induced autophagy through Beclin-1- and reactive oxygen species (ROS)-dependent manners in human hepatocellular carcinoma (HCC) cells. Incubating the HCC cell with ApoG2 abrogated the interaction of Beclin-1 and Bcl-2/xL, stimulated ROS generation, increased phosphorylation of ERK and JNK, and HMGB1 translocation from the nucleus to cytoplasm while suppressing mTOR. Moreover, inhibition of the ROS-mediated autophagy by antioxidant N-acetyl-cysteine (NAC) potentiates ApoG2-induced apoptosis and cell killing. Our results show that ApoG2 induced protective autophagy in HCC cells, partly due to ROS generation, suggesting that antioxidant may serve as a potential chemosensitizer to enhance cancer cell death through blocking ApoG2-stimulated autophagy. Our novel insights may facilitate the rational design of clinical trials for Bcl-2-targeted cancer therapy.
Collapse
Affiliation(s)
- P Cheng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hayashi H, Shimamoto K, Taniai E, Ishii Y, Morita R, Suzuki K, Shibutani M, Mitsumori K. Liver tumor promoting effect of omeprazole in rats and its possible mechanism of action. J Toxicol Sci 2012; 37:491-501. [PMID: 22687989 DOI: 10.2131/jts.37.491] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Omeprazole (OPZ), a proton pump inhibitor, is a cytochrome P450 (CYP) 1A1/2 inducer. Some CYP1A inducers are known to have liver tumor promoting effects in rats and the ability to enhance oxidative stress. In this study, we performed a two-stage liver carcinogenesis bioassay in rats to examine the tumor promoting effect of OPZ (Experiment 1) and to clarify a possible mechanism of action (Experiment 2). In Experiment 1, male F344 rats were subjected to a two-third partial hepatectomy, and treated with 0, 138 or 276 mg/kg OPZ by oral gavage once a day for six weeks after an intraperitoneal injection of N-diethylnitrosamine (DEN). Liver weights significantly increased in the DEN+OPZ groups, and the number and area of glutathione S-transferase placental form (GST-P) positive foci significantly increased in the DEN+276 mg/kg OPZ group. In Experiment 2, the same experiment as Experiment 1 was performed, but the dosage of OPZ was 0 or 276 mg/kg. The number and area of GST-P positive foci as well as liver weights significantly increased in the DEN+276 mg/kg OPZ group. The number of proliferative cell nuclear antigen (PCNA)-positive cells also significantly increased in the same group. Real-time RT-PCR showed that the expression of AhR battery genes including Cyp1a1, Cyp1a2, Ugt1a6 and Nqo1, and Nrf2 battery genes including Gpx2, Yc2, Akr7a3, Aldh1a1 Me1 and Ggt1 were significantly upregulated in this group. However, the production of microsomal reactive oxygen species (ROS) and formation of thiobarbituric acid-reactive substances (TBARS) decreased, and 8-hydroxydeoxyguanosine (8-OHdG) content remained unchanged in this group. These results indicate that OPZ, CYP1A inducer, is a liver tumor promoter in rats, but oxidative stress is not involved in the liver tumor promoting effect of OPZ.
Collapse
Affiliation(s)
- Hitomi Hayashi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Krajka-Kuźniak V, Szaefer H, Ignatowicz E, Adamska T, Baer-Dubowska W. Beetroot juice protects against N-nitrosodiethylamine-induced liver injury in rats. Food Chem Toxicol 2012; 50:2027-33. [PMID: 22465004 DOI: 10.1016/j.fct.2012.03.062] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 03/16/2012] [Accepted: 03/17/2012] [Indexed: 01/27/2023]
Abstract
Red beetroot, a common ingredient of diet, is a rich source of a specific class of antioxidants, betalains. Our previous studies have shown the protective role of beetroot juice against carcinogen induced oxidative stress in rats. The aim of this study was to examine the effect of long term feeding (28 days) with beetroot juice on phase I and phase II enzymes, DNA damage and liver injury induced by hepatocarcinogenic N-nitrosodiethylamine (NDEA). Long term feeding with beetroot juice decreased the activities of enzymatic markers of cytochrome P450, CYP1A1/1A2 and CYP2E1. NDEA treatment also reduced the activities of these enzymes, but increased the activity of CYP2B. Moreover, combined treatment with beetroot juice and NDEA enhanced significantly CYP2B only. Modulation of P450 enzyme activities was accompanied by changes in the relevant proteins levels. Increased level and activity of NQO1 was the most significant change among phase II enzymes. Beetroot juice reduced the DNA damage increased as the result of NDEA treatment, as well as the biomarkers of liver injury. Collectively, these results confirm the protective effect of beetroot juice against oxidative damage shown in our previous studies and indicate that metabolic alterations induced by beetroot feeding may protect against liver damage.
Collapse
|