1
|
Wang X, Pan S, Chen L, Liang C, Zhu Y, Zhou K, Shi X. Sijunzi decoction enhances sensitivity of colon cancer cells to NK cell destruction by modulating P53 expression. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118115. [PMID: 38580190 DOI: 10.1016/j.jep.2024.118115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sijunzi Decoction (SJZD), a traditional Chinese herbal remedy, is frequently employed in the treatment of various cancers, including colon cancer. Previous research suggests that SJZD plays a pivotal role in modulating the immune system and enhancing immunity against tumors. However, the precise role of SJZD in combating colon cancer and its potential molecular functions in regulating natural killer cells remain elusive. AIMS OF THE STUDY To elucidate the potential mechanism underlying the anticolon cancer effects of SJZD in synergy with natural killer (NK) cells through both in vivo and in vitro experiments. MATERIALS AND METHODS In vivo experiments: A subcutaneous tumor mouse model of colon cancer and in vivo NK cell depletion experiments were conducted to observe the anticolon cancer effects of SJZD. Flow cytometry assessed immune cell depletion in mouse spleens, while immunohistochemical (IHC) staining detected the expression of apoptotic genes in tumor tissues. In vitro experiments: The mechanism by which SJZD regulates the sensitization of colon cancer cells to NK cells was investigated using real-time polymerase chain reaction (RT-PCR), western blotting (WB), and co-culture experiments with NK cells. RESULTS Sijunzi Decoction (SJZD) significantly impeded tumor growth in mice; however, NK cell depletion markedly attenuated the tumor-suppressive effect of SJZD. Immunohistochemical (IHC) results indicated that SJZD increased the expression of P53, death receptor 4 (DR4), and death receptor 5 (DR5) in tumor tissues. In vitro experiments, 24 h SJZD-pretreated colon cancer cells showed a substantial elevation in P53, DR4, and DR5 levels, and the activity of colon cancer cells significantly diminished after co-culture with NK cells. These effects of SJZD were reversed with the addition of the P53 inhibitor pifithrin-α (PFT-α), resulting in reduced inhibition of colon cancer cells by NK cells. CONCLUSION SJZD enhances the levels of DR4 and DR5 through the modulation of P53 expression, consequently increasing the sensitivity of colon cancer cells to NK cell-mediated killing. These findings provide a theoretical foundation for the clinical application of SJZD in patients with colon cancer. In this study, we first investigated the effect of SJZD on subcutaneous tumor growth in mice with colon cancer using in vivo assays and assessed the impact of NK cells on the anticolon cancer effect of SJZD in vivo through NK cell depletion. In vitro experiments were conducted to explore the potential mechanism of action of SJZD in NK cell-mediated anticolon cancer effects.
Collapse
Affiliation(s)
- Xinxin Wang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Shufang Pan
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Liangyan Chen
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Chengchen Liang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Yueyi Zhu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Ke Zhou
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Xiaolan Shi
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| |
Collapse
|
2
|
Salim EI, Alabasy MM, Nashar EME, Al-Zahrani NS, Alzahrani MA, Guo Z, Beltagy DM, Shahen M. Molecular interactions between metformin and D-limonene inhibit proliferation and promote apoptosis in breast and liver cancer cells. BMC Complement Med Ther 2024; 24:185. [PMID: 38711049 PMCID: PMC11071183 DOI: 10.1186/s12906-024-04453-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 03/22/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Cancer is a fatal disease that severely affects humans. Designing new anticancer strategies and understanding the mechanism of action of anticancer agents is imperative. HYPOTHESIS/PURPOSE In this study, we evaluated the utility of metformin and D-limonene, alone or in combination, as potential anticancer therapeutics using the human liver and breast cancer cell lines HepG2 and MCF-7. STUDY DESIGN An integrated systems pharmacology approach is presented for illustrating the molecular interactions between metformin and D-limonene. METHODS We applied a systems-based analysis to introduce a drug-target-pathway network that clarifies different mechanisms of treatment. The combination treatment of metformin and D-limonene induced apoptosis in both cell lines compared with single drug treatments, as indicated by flow cytometric and gene expression analysis. RESULTS The mRNA expression of Bax and P53 genes were significantly upregulated while Bcl-2, iNOS, and Cox-2 were significantly downregulated in all treatment groups compared with normal cells. The percentages of late apoptotic HepG2 and MCF-7 cells were higher in all treatment groups, particularly in the combination treatment group. Calculations for the combination index (CI) revealed a synergistic effect between both drugs for HepG2 cells (CI = 0.14) and MCF-7 cells (CI = 0.22). CONCLUSION Our data show that metformin, D-limonene, and their combinations exerted significant antitumor effects on the cancer cell lines by inducing apoptosis and modulating the expression of apoptotic genes.
Collapse
Affiliation(s)
- Elsayed I Salim
- Department of Zoology, Research Lab of Molecular Carcinogenesis, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Mona M Alabasy
- Department of Zoology, Research Lab of Molecular Carcinogenesis, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Eman M El Nashar
- Department of Anatomy, College of Medicine, King Khalid University, Abha, 62529, Saudi Arabia
| | - Norah S Al-Zahrani
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, 62529, Saudi Arabia
| | - Mohammed A Alzahrani
- Internal Medicine Department, College of Medicine, King Khalid University, Abha, 62529, Saudi Arabia
| | - Zihu Guo
- College of Life Science, Center of Bioinformatics, Northwest A and F University, Yangling, Shaanxi, 712100, China
| | - Doha M Beltagy
- Biochemistry Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Mohamed Shahen
- Department of Zoology, Research Lab of Molecular Carcinogenesis, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
3
|
Shi R, Wang F, Fu Q, Zeng P, Chen G, Chen Z. Molecular mechanism analysis of apoptosis induced by silk fibroin peptides. Int J Biol Macromol 2024; 264:130687. [PMID: 38462112 DOI: 10.1016/j.ijbiomac.2024.130687] [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: 01/09/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 03/12/2024]
Abstract
Silk fibroin derived from silkworm cocoons exhibits excellent mechanical properties, good biocompatibility, and low immunogenicity. Previous studies showed that silk fibroin had an inhibitory effect on cells, suppressing proliferation and inducing apoptosis. However, the source of the toxicity and the mechanism of apoptosis induction are still unclear. In this study, we hypothesized that the toxicity of silk fibroin might originate from the crystalline region of the heavy chain of silk fibroin. We then verified the hypothesis and the specific induction mechanism. A target peptide segment was obtained from α-chymotrypsin. The potentially toxic mixture of silk fibroin peptides (SFPs) was separated by ion exchange, and the toxicity was tested by an MTT assay. The results showed that SFPs obtained after 4 h of enzymatic hydrolysis had significant cytotoxicity, and SFPs with isoelectric points of 4.0-6.8 (SFPα II) had a significant inhibitory effect on cell growth. LC-MS/MS analysis showed that SFPα II contained a large number of glycine-rich and alanine-rich repetitive sequence polypeptides from the heavy-chain crystallization region. A series of experiments showed that SFPα II mediated cell death through the apoptotic pathway by decreasing the expression of Bcl-2 protein and increasing the expression of Bax protein. SFPα II mainly affected the p53 pathway and the AMPK signaling pathway in HepG2 cells. SFPα II may indirectly increase the expression of Cers2 by inhibiting the phosphorylation of EGFR, which activated apoptotic signaling in the cellular mitochondrial pathway and inhibited the Akt/NF-κB pathway by increasing the expression of PPP2R2A.
Collapse
Affiliation(s)
- Ruyu Shi
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Fuping Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Qiang Fu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Peng Zeng
- The Seventh People's Hospital of Chongqing, Chongqing 400054, China
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Zhongmin Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| |
Collapse
|
4
|
Jung YJ, Muneeswaran T, Choi JS, Kim S, Han JH, Cho WS, Park JW. Modified toxic potential of multi-walled carbon nanotubes to zebrafish (Danio rerio) following a two-year incubation in water. JOURNAL OF HAZARDOUS MATERIALS 2024; 462:132763. [PMID: 37839374 DOI: 10.1016/j.jhazmat.2023.132763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
Multi-walled carbon nanotubes (MWCNTs), widely used in several industrial fields, are not readily degradable thus, persist in environmental matrices, serving as a source of environmental toxicity to organisms. However, the effects of environmental weathering on nanomaterial toxicity remain unclear. Herein, we prepared aged-MWCNTs (a-CNTs) by incubating commercial pristine-MWCNTs (p-CNTs) for two years and compared their changes in physicochemical properties and toxic effects on zebrafish. The characterization of a-CNTs by transmission electron microscopy, X-ray photoelectron spectra, Raman spectroscopy, and Fourier-transform infrared spectroscopy showed an increased surface area, pore size, structural defects, and surface oxidation than those of p-CNTs. Zebrafish were exposed to 100 mg/L p-CNT and a-CNT for four days. Subsequently, the mRNA expression of antioxidant enzymes, including cat, gst, and sod, in a-CNT group increased by 1.5- to 1.7-fold, consistent with increased expression of genes associated with inflammation (interleukin-8) and apoptosis (p53) compared to control. The higher toxicity of a-CNTs to zebrafish than p-CNT might be due to the increased oxidative potential by altered physicochemical properties. These findings provide new insights into the risk assessment and environmental management of MWCNTs in the aquatic environment. However, further testing at environmentally relevant doses, different exposure durations, and diverse weathering parameters is warranted.
Collapse
Affiliation(s)
- Youn-Joo Jung
- Environmental Exposure & Toxicology Research Center, Korea Institute of Toxicology, Jinju 52834, Republic of Korea; Joint Research Center for Alternative and Predictive Toxicology (JRC-APT), Korea Institute of Toxicology, Jinju 52834, Republic of Korea
| | - Thillaichidambaram Muneeswaran
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Jin Soo Choi
- Environmental Exposure & Toxicology Research Center, Korea Institute of Toxicology, Jinju 52834, Republic of Korea; Joint Research Center for Alternative and Predictive Toxicology (JRC-APT), Korea Institute of Toxicology, Jinju 52834, Republic of Korea
| | - Sumin Kim
- School of Applied Chemical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jong Hun Han
- School of Applied Chemical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Wan-Seob Cho
- Lab of Toxicology, Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea.
| | - June-Woo Park
- Environmental Exposure & Toxicology Research Center, Korea Institute of Toxicology, Jinju 52834, Republic of Korea; Joint Research Center for Alternative and Predictive Toxicology (JRC-APT), Korea Institute of Toxicology, Jinju 52834, Republic of Korea; Human and Environmental Toxicology Program, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea.
| |
Collapse
|
5
|
Kharboush TG, Ahmed IA, Farag AA, Kharboush T, Sayed AEDH, Abdel-Kareim AM, Al Mohaini M, Attia H, Eid RA, Zaki MSA, Al-Tabbakh ASM. Epigenetic alterations of miR-155 and global DNA methylation as potential mediators of ochratoxin A cytotoxicity and carcinogenicity in human lung fibroblasts. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:5473-5483. [PMID: 38114706 PMCID: PMC10799132 DOI: 10.1007/s11356-023-31283-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023]
Abstract
Ochratoxin A (OTA) is a well-known mycotoxin that adversely affects different human cells. Inhalational exposure to OTA and subsequent pulmonary diseases have been previously reported, yet its potential carcinogenicity and underlying molecular mechanisms have not been fully elucidated. This study aimed to evaluate the OTA-induced cytotoxicity and the epigenetic changes underlying its potential carcinogenicity in fetal lung fibroblast (WI-38) cells. OTA cytotoxicity was assessed by MTT assay; RT-qPCR was used to determine the expression of BAX, BCL-2, TP53, and miR-155, while ELISA was used for measuring 5-methyl cytosine percentage to assess global DNA methylation in OTA-treated versus control cells. WI-38 cells demonstrated sensitivity to OTA with IC50 at 22.38 μM. Though BAX and Bcl-2 were downregulated, with low BAX/BCL-2 ratio, and TP53 was upregulated, their fold changes showed decline trend with increasing OTA concentration. A significant dose-dependent miR-155 upregulation was observed, with dynamic time-related decline. Using subtoxic OTA concentrations, a significant global DNA hypermethylation with significant dose-dependent and dynamic alterations was identified. Global DNA hypermethylation and miR-155 upregulation are epigenetic mechanisms that mediate OTA toxicity on WI-38 cells. BAX downregulation, reduced BAX/BCL-2 ratio together with miR-155 upregulation indicated either the inhibition of TP53-dependent apoptosis or a tissue specific response to OTA exposure. The aforementioned OTA-induced variations present a new molecular evidence of OTA cytotoxicity and possible carcinogenicity in lung fibroblast cells.
Collapse
Affiliation(s)
- Taghrid G Kharboush
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Inas A Ahmed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
- Central Laboratory for Research, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Amina A Farag
- Department of Forensic Medicine & Clinical Toxicology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Tayseir Kharboush
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Alaa El-Din H Sayed
- Department of Zoology, Faculty of Science, Assiut University, Asyut, 71516, Egypt.
- Molecular Biology Research & Studies Institute, Assiut University, Asyut, 71516, Egypt.
| | - Amal M Abdel-Kareim
- Department of Zoology, Faculty of Science, Benha University, Benha, 13518, Egypt
| | - Mohammed Al Mohaini
- Basic Sciences Department, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, 31982, Alahsa, Saudi Arabia
- King Abdullah International Medical Research Center, 31982, Alahsa, Saudi Arabia
| | - Hend Attia
- Clinical and Chemical Pathology, School of Medicine, Newgiza University (NGU), Giza, Egypt
| | - Refaat A Eid
- Department of Pathology, College of Medicine, King Khalid University, P.O. Box 62529, Abha, Saudi Arabia
| | - Mohamed Samir A Zaki
- Department of Anatomy, College of Medicine, King Khalid University, P.O. Box 62529, Abha, Saudi Arabia
| | - Al-Shaimaa M Al-Tabbakh
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| |
Collapse
|
6
|
Wang X, Cai Y, Wu C, Liang J, Tang K, Lin Z, Chen L, Lu Y, Wang Q. Conversion of senescent cartilage into a pro-chondrogenic microenvironment with antibody-functionalized copper sulfate nanoparticles for efficient osteoarthritis therapy. J Nanobiotechnology 2023; 21:258. [PMID: 37550685 PMCID: PMC10408088 DOI: 10.1186/s12951-023-02036-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023] Open
Abstract
The development of osteoarthritis (OA) correlates with the expansion of senescent cells in cartilage, which contributes to an inflammatory microenvironment that accelerates matrix degradation and hampers cartilage generation. To address OA, we synthesized small copper sulfide nanoparticles functionalized with anti-beta-2-microglobulin antibodies (B2M-CuS NPs) that catalyze the formation of toxic •OH from H2O2 via peroxidase-like activity. These B2M-CuS NPs are specifically targeted to induce apoptosis in senescent chondrocytes while showing no toxicity toward normal chondrocytes. Furthermore, B2M-CuS NPs enhance the chondrogenesis of normal chondrocytes. Thus, B2M-CuS NPs can effectively treat OA by clearing senescent chondrocytes and promoting cartilage regeneration after intra-articular injection into the knee joints of surgery-induced OA mice. This study uses smart nanomaterials to treat OA with a synergistic strategy that both remodels senescent cartilage and creates a pro-chondrogenic microenvironment.
Collapse
Affiliation(s)
- Xianming Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Yu Cai
- Precision Medicine in Oncology (PrMiO), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Cuixi Wu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiamin Liang
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kangning Tang
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Zefeng Lin
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Lingling Chen
- Department of Orthopedics, General Hospital of Southern Theater Command of PLA, Guangzhou, Guangdong, China
| | - Yao Lu
- Department of Joint and Orthopedics, Orthopedic Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Qing Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Dong X, Zhang Y, Sun Y, Nan Q, Li M, Ma L, Zhang L, Luo J, Qi Y, Miao Y. Promoter hypermethylation and comprehensive regulation of ncRNA lead to the down-regulation of ZNF880, providing a new insight for the therapeutics and research of colorectal cancer. BMC Med Genomics 2023; 16:148. [PMID: 37370088 PMCID: PMC10294494 DOI: 10.1186/s12920-023-01571-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The human genome encodes more than 350 kinds of Krüppel-associated box (KRAB) domain-containing zinc-finger proteins (KZFPs), KRAB-type ZNF transcription factor family (KZNF) plays a vital role in gene regulatory networks. The KZNF family members include a large number of highly homologous genes, gene subtypes and pseudogenes, and their expression has a high degree of tissue specificity and precision. Due to the high complexity of its regulatory network, the KZNF gene family has not been researched in sufficient, and the role of its members in the occurrence of cancer is mostly unexplored. In this study, ZNF880 was significantly associated with overall survival (OS) and disease-free survival (DFS) in colorectal carcinoma (CRC) patients. Low ZNF880 expression resulted in shorter OS and DFS. Combined with Colon adenocarcinoma (COAD) and Rectum adenocarcinoma (READ) data collection in the TCGA database, we found that ZNF880 was significantly down-regulated in CRC. Further analysis of the sequence variation of ZNF880 in CRC showed that ZNF880 accumulated a large number of SNV in the C2H2 domain and KRAB domain, while promoter region of ZNF880 also showed high methylation in COAD and READ. Combined with the Cbioportal and TIMER databases, the expression of mutant ZNF880 was significantly lower in COAD compared to the wild type. Simultaneously, the lncRNA-miRNA-ZNF880 ceRNA regulatory network was constructed through co-expression and miRNAs target gene prediction, demonstrating the precision of the ZNF880 regulatory network. In addition, the decreased expression of ZNF880 caused the significant immune infiltration decreases of CD8 + cells in COAD. In contrast, the immune infiltration of CD4 + cells and macrophages in COAD is positively correlated with ZNF880. Finally, through protein-protein interaction (PPI) network analysis and transcription factor target gene prediction, we screened out the genes most likely to be related to the function of ZNF880. CENPK, IFNGR2, REC8 and ZBTB17 were identified as the most closely functioning genes with ZNF880, which may indicate that ZNF880 has important links with the formation of cell centromere, tumor immunity, cell cycle and other pathways closely related to the occurrence of CRC. These studies show that the down-regulation of ZNF880 gene is closely related to CRC, and the targeted change of the expression of its regulatory molecules (miRNA and lncRNA) may be a new perspective for CRC treatment.
Collapse
Affiliation(s)
- Xiangqian Dong
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China
| | - Yinghui Zhang
- Department of Gastroenterology, Affiliated Hospital of Yunnan University, Kunming, 650021, China
| | - Yang Sun
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China
| | - Qiong Nan
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China
| | - Maojuan Li
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China
| | - Lanqing Ma
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China
| | - Lei Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China
| | - Juan Luo
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China
| | - Yating Qi
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China
| | - Yinglei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, NO.295 Xichang Road, Kunming, 650032, P.R. China.
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, 650032, China.
| |
Collapse
|
8
|
Moawadh MS, Mir R, Tayeb FJ, Asim O, Ullah MF. Molecular Evaluation of the Impact of Polymorphic Variants in Apoptotic ( Bcl-2/Bax) and Proinflammatory Cytokine ( TNF-α/IL-8) Genes on the Susceptibility and Progression of Myeloproliferative Neoplasms: A Case-Control Biomarker Study. Curr Issues Mol Biol 2023; 45:3933-3952. [PMID: 37232720 DOI: 10.3390/cimb45050251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The regulation of apoptosis (the programmed cell death) is dependent on the crucial involvement of BCL2 and BAX. The Bax-248G>A and Bcl-2-938 C>A polymorphic variations in the promoter sequences of the Bax and Bcl-2 gene have been recently associated with low Bax expression, progression to advanced stages, treatment resistance, and shortened overall survival rate in some hematological malignancies, including chronic myeloid leukemia (CML) and other myeloproliferative neoplasms. Chronic inflammation has been linked to various stages of carcinogenesis wherein pro-inflammatory cytokines play diverse roles in influencing cancer microenvironment leading to cell invasion and cancer progression. Cytokines such as TNF-α and IL-8 have been implicated in cancer growth in both solid and hematological malignancies with studies showing their elevated levels in patients. Genomic approaches have in recent years provided significant knowledge with the regard to the association of certain SNPs (single nucleotide polymerphisms) either in a gene or its promoter that can influence its expression, with the risk and susceptibility to human diseases including cancer. This study has investigated the consequences of promoter SNPs in apoptosis genes Bax-248G>A (rs4645878)/Bcl-2-938C>A (rs2279115) and pro-inflammatory cytokines TNF-α rs1800629 G>A/IL-8 rs4073 T>A on the risk and susceptibility towards hematological cancers. The study design has 235 individuals both male and female enrolled as subjects that had 113 cases of MPDs (myeloproliferative disorders) and 122 healthy individuals as controls. The genotyping studies were conducted through ARMS PCR (amplification-refractory mutation system PCR). The Bcl-2-938 C>A polymorphism showed up in 22% of patients in the study, while it was observed in only 10% of normal controls. This difference in genotype and allele frequency between the two groups was significant (p = 0.025). Similarly, the Bax-248G>A polymorphism was detected in 6.48% of the patients and 4.54% of the normal controls, with a significant difference in genotype and allele frequency between the groups (p = 0.048). The results suggest that the Bcl-2-938 C>A variant is linked to an elevated risk of MPDs in the codominant, dominant, and recessive inheritance models. Moreover, the study indicated allele A as risk allele which can significantly increase the risk of MPDs unlike the C allele. In case of Bax gene covariants, these were associated with an increased risk of MPDs in the codominant inheritance model and dominant inheritance model. It was found that the allele A significantly enhanced the risk of MPDs unlike the G allele. The frequencies of IL-8 rs4073 T>A in patients was found to be TT (16.39%), AT (36.88%) and AA (46.72%), compared to controls who were more likely to have frequencies of TT (39.34%), AT (37.70%) and AA (22.95%) as such, respectively. There was a notable overrepresentation of the AA genotype and GG homozygotes among patients compared to controls in TNF-α polymorphic variants, with 6.55% of patients having the AA genotype and 84% of patients being GG homozygotes, compared to 1.63% and 69%, respectively in controls. The data from the current study provide partial but important evidence that polymorphisms in apoptotic genes Bcl-2-938C>A and Bax-248G>A and pro-inflammatory cytokines IL-8 rs4073 T>A and TNF-α G>A may help predict the clinical outcomes of patients and determine the significance of such polymorphic variations in the risk of myeloproliferative diseases and their role as prognostic markers in disease management using a case-control study approach.
Collapse
Affiliation(s)
- Mamdoh S Moawadh
- Department of Medical Laboratory Technology (FAMS), University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Rashid Mir
- Department of Medical Laboratory Technology (FAMS), University of Tabuk, Tabuk 71491, Saudi Arabia
- Division of Molecular Biology, Prince Fahd Chair for Biomedical Research, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Faris J Tayeb
- Department of Medical Laboratory Technology (FAMS), University of Tabuk, Tabuk 71491, Saudi Arabia
- Community College, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Orooba Asim
- Division of Molecular Biology, Prince Fahd Chair for Biomedical Research, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Mohammad Fahad Ullah
- Department of Medical Laboratory Technology (FAMS), University of Tabuk, Tabuk 71491, Saudi Arabia
| |
Collapse
|
9
|
Vitale I, Pietrocola F, Guilbaud E, Aaronson SA, Abrams JM, Adam D, Agostini M, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Aqeilan RI, Arama E, Baehrecke EH, Balachandran S, Bano D, Barlev NA, Bartek J, Bazan NG, Becker C, Bernassola F, Bertrand MJM, Bianchi ME, Blagosklonny MV, Blander JM, Blandino G, Blomgren K, Borner C, Bortner CD, Bove P, Boya P, Brenner C, Broz P, Brunner T, Damgaard RB, Calin GA, Campanella M, Candi E, Carbone M, Carmona-Gutierrez D, Cecconi F, Chan FKM, Chen GQ, Chen Q, Chen YH, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Ciliberto G, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Daugaard M, Dawson TM, Dawson VL, De Maria R, De Strooper B, Debatin KM, Deberardinis RJ, Degterev A, Del Sal G, Deshmukh M, Di Virgilio F, Diederich M, Dixon SJ, Dynlacht BD, El-Deiry WS, Elrod JW, Engeland K, Fimia GM, Galassi C, Ganini C, Garcia-Saez AJ, Garg AD, Garrido C, Gavathiotis E, Gerlic M, Ghosh S, Green DR, Greene LA, Gronemeyer H, Häcker G, Hajnóczky G, Hardwick JM, Haupt Y, He S, Heery DM, Hengartner MO, Hetz C, Hildeman DA, Ichijo H, Inoue S, Jäättelä M, Janic A, Joseph B, Jost PJ, Kanneganti TD, Karin M, Kashkar H, Kaufmann T, Kelly GL, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Kluck R, Krysko DV, Kulms D, Kumar S, Lavandero S, Lavrik IN, Lemasters JJ, Liccardi G, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Luedde T, MacFarlane M, Madeo F, Malorni W, Manic G, Mantovani R, Marchi S, Marine JC, Martin SJ, Martinou JC, Mastroberardino PG, Medema JP, Mehlen P, Meier P, Melino G, Melino S, Miao EA, Moll UM, Muñoz-Pinedo C, Murphy DJ, Niklison-Chirou MV, Novelli F, Núñez G, Oberst A, Ofengeim D, Opferman JT, Oren M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pentimalli F, Pereira DM, Pervaiz S, Peter ME, Pinton P, Porta G, Prehn JHM, Puthalakath H, Rabinovich GA, Rajalingam K, Ravichandran KS, Rehm M, Ricci JE, Rizzuto R, Robinson N, Rodrigues CMP, Rotblat B, Rothlin CV, Rubinsztein DC, Rudel T, Rufini A, Ryan KM, Sarosiek KA, Sawa A, Sayan E, Schroder K, Scorrano L, Sesti F, Shao F, Shi Y, Sica GS, Silke J, Simon HU, Sistigu A, Stephanou A, Stockwell BR, Strapazzon F, Strasser A, Sun L, Sun E, Sun Q, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Troy CM, Turk B, Urbano N, Vandenabeele P, Vanden Berghe T, Vander Heiden MG, Vanderluit JL, Verkhratsky A, Villunger A, von Karstedt S, Voss AK, Vousden KH, Vucic D, Vuri D, Wagner EF, Walczak H, Wallach D, Wang R, Wang Y, Weber A, Wood W, Yamazaki T, Yang HT, Zakeri Z, Zawacka-Pankau JE, Zhang L, Zhang H, Zhivotovsky B, Zhou W, Piacentini M, Kroemer G, Galluzzi L. Apoptotic cell death in disease-Current understanding of the NCCD 2023. Cell Death Differ 2023; 30:1097-1154. [PMID: 37100955 PMCID: PMC10130819 DOI: 10.1038/s41418-023-01153-w] [Citation(s) in RCA: 112] [Impact Index Per Article: 112.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 04/28/2023] Open
Abstract
Apoptosis is a form of regulated cell death (RCD) that involves proteases of the caspase family. Pharmacological and genetic strategies that experimentally inhibit or delay apoptosis in mammalian systems have elucidated the key contribution of this process not only to (post-)embryonic development and adult tissue homeostasis, but also to the etiology of multiple human disorders. Consistent with this notion, while defects in the molecular machinery for apoptotic cell death impair organismal development and promote oncogenesis, the unwarranted activation of apoptosis promotes cell loss and tissue damage in the context of various neurological, cardiovascular, renal, hepatic, infectious, neoplastic and inflammatory conditions. Here, the Nomenclature Committee on Cell Death (NCCD) gathered to critically summarize an abundant pre-clinical literature mechanistically linking the core apoptotic apparatus to organismal homeostasis in the context of disease.
Collapse
Affiliation(s)
- Ilio Vitale
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy.
- Candiolo Cancer Institute, FPO -IRCCS, Candiolo, Italy.
| | - Federico Pietrocola
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dieter Adam
- Institut für Immunologie, Kiel University, Kiel, Germany
| | - Massimiliano Agostini
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Patrizia Agostinis
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Emad S Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
- BIOGEM, Avellino, Italy
| | - Ivano Amelio
- Division of Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - David W Andrews
- Sunnybrook Research Institute, Toronto, ON, Canada
- Departments of Biochemistry and Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Rami I Aqeilan
- Hebrew University of Jerusalem, Lautenberg Center for Immunology & Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Jerusalem, Israel
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniele Bano
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Nickolai A Barlev
- Department of Biomedicine, Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Jiri Bartek
- Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, School of Medicine, Louisiana State University Health New Orleans, New Orleans, LA, USA
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Francesca Bernassola
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Mathieu J M Bertrand
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Marco E Bianchi
- Università Vita-Salute San Raffaele, School of Medicine, Milan, Italy and Ospedale San Raffaele IRCSS, Milan, Italy
| | | | - J Magarian Blander
- Department of Medicine, Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | | | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Pediatric Hematology and Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Medical Faculty, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Carl D Bortner
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| | - Pierluigi Bove
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Patricia Boya
- Centro de Investigaciones Biologicas Margarita Salas, CSIC, Madrid, Spain
| | - Catherine Brenner
- Université Paris-Saclay, CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l'oncogénèse pour de nouvelles approches thérapeutiques, Villejuif, France
| | - Petr Broz
- Department of Immunobiology, University of Lausanne, Epalinges, Vaud, Switzerland
| | - Thomas Brunner
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Rune Busk Damgaard
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michelangelo Campanella
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
- UCL Consortium for Mitochondrial Research, London, UK
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | - Francesco Cecconi
- Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, Copenhagen, Denmark
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francis K-M Chan
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Guo-Qiang Chen
- State Key Lab of Oncogene and its related gene, Ren-Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Youhai H Chen
- Shenzhen Institute of Advanced Technology (SIAT), Shenzhen, Guangdong, China
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Cidlowski
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, USA
| | - Aaron Ciechanover
- The Technion-Integrated Cancer Center, The Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Marcus Conrad
- Helmholtz Munich, Institute of Metabolism and Cell Death, Neuherberg, Germany
| | - Juan R Cubillos-Ruiz
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Mads Daugaard
- Department of Urologic Sciences, Vancouver Prostate Centre, Vancouver, BC, Canada
| | - Ted M Dawson
- Institute for Cell Engineering and the Departments of Neurology, Neuroscience and Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Institute for Cell Engineering and the Departments of Neurology, Neuroscience and Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruggero De Maria
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Bart De Strooper
- VIB Centre for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- The Francis Crick Institute, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Ralph J Deberardinis
- Howard Hughes Medical Institute and Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexei Degterev
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Giannino Del Sal
- Department of Life Sciences, University of Trieste, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park-Padriciano, Trieste, Italy
- IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | | | - Marc Diederich
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Brian D Dynlacht
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, NY, USA
| | - Wafik S El-Deiry
- Division of Hematology/Oncology, Brown University and the Lifespan Cancer Institute, Providence, RI, USA
- Legorreta Cancer Center at Brown University, The Warren Alpert Medical School, Brown University, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - John W Elrod
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Kurt Engeland
- Molecular Oncology, University of Leipzig, Leipzig, Germany
| | - Gian Maria Fimia
- Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases 'L. Spallanzani' IRCCS, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Carlo Ganini
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
- Biochemistry Laboratory, Dermopatic Institute of Immaculate (IDI) IRCCS, Rome, Italy
| | - Ana J Garcia-Saez
- CECAD, Institute of Genetics, University of Cologne, Cologne, Germany
| | - Abhishek D Garg
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Carmen Garrido
- INSERM, UMR, 1231, Dijon, France
- Faculty of Medicine, Université de Bourgogne Franche-Comté, Dijon, France
- Anti-cancer Center Georges-François Leclerc, Dijon, France
| | - Evripidis Gavathiotis
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY, USA
| | - Motti Gerlic
- Department of Clinical Microbiology and Immunology, Sackler school of Medicine, Tel Aviv university, Tel Aviv, Israel
| | - Sourav Ghosh
- Department of Neurology and Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lloyd A Greene
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Hinrich Gronemeyer
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, U1258, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Georg Häcker
- Faculty of Medicine, Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - György Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Departments of Molecular Microbiology and Immunology, Pharmacology, Oncology and Neurology, Johns Hopkins Bloomberg School of Public Health and School of Medicine, Baltimore, MD, USA
| | - Ygal Haupt
- VITTAIL Ltd, Melbourne, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sudan He
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China
| | - David M Heery
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
- Buck Institute for Research on Aging, Novato, CA, USA
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, The University of Tokyo, Tokyo, Japan
| | - Satoshi Inoue
- National Cancer Center Research Institute, Tokyo, Japan
| | - Marja Jäättelä
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ana Janic
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Bertrand Joseph
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Philipp J Jost
- Clinical Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | | | - Michael Karin
- Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Hamid Kashkar
- CECAD Research Center, Institute for Molecular Immunology, University of Cologne, Cologne, Germany
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Richard N Kitsis
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, NY, USA
- Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, New York, NY, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY, USA
| | | | - Ruth Kluck
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Dmitri V Krysko
- Cell Death Investigation and Therapy Lab, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Dagmar Kulms
- Department of Dermatology, Experimental Dermatology, TU-Dresden, Dresden, Germany
- National Center for Tumor Diseases Dresden, TU-Dresden, Dresden, Germany
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Sergio Lavandero
- Universidad de Chile, Facultad Ciencias Quimicas y Farmaceuticas & Facultad Medicina, Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Inna N Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University, Magdeburg, Germany
| | - John J Lemasters
- Departments of Drug Discovery & Biomedical Sciences and Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Gianmaria Liccardi
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Stuart A Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Richard A Lockshin
- Department of Biology, Queens College of the City University of New York, Flushing, NY, USA
- St. John's University, Jamaica, NY, USA
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Heinrich Heine University, Duesseldorf, Germany
| | - Marion MacFarlane
- Medical Research Council Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Field of Excellence BioHealth - University of Graz, Graz, Austria
| | - Walter Malorni
- Center for Global Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gwenola Manic
- IIGM - Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy
- Candiolo Cancer Institute, FPO -IRCCS, Candiolo, Italy
| | - Roberto Mantovani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
| | - Saverio Marchi
- Department of Clinical and Molecular Sciences, Marche Polytechnic University, Ancona, Italy
| | - Jean-Christophe Marine
- VIB Center for Cancer Biology, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Jean-Claude Martinou
- Department of Cell Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Pier G Mastroberardino
- Department of Molecular Genetics, Rotterdam, the Netherlands
- IFOM-ETS The AIRC Institute for Molecular Oncology, Milan, Italy
- Department of Life, Health, and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Patrick Mehlen
- Apoptosis, Cancer, and Development Laboratory, Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon1, Lyon, France
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Gerry Melino
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Sonia Melino
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Edward A Miao
- Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Ute M Moll
- Department of Pathology and Stony Brook Cancer Center, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Cristina Muñoz-Pinedo
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Daniel J Murphy
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | | | - Flavia Novelli
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, The University of Michigan, Ann Arbor, MI, USA
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Dimitry Ofengeim
- Rare and Neuroscience Therapeutic Area, Sanofi, Cambridge, MA, USA
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Moshe Oren
- Department of Molecular Cell Biology, The Weizmann Institute, Rehovot, Israel
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine and Howard Hughes Medical Institute, New York, NY, USA
| | - Theocharis Panaretakis
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of GU Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | | | - David M Pereira
- REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Shazib Pervaiz
- Department of Physiology, YLL School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore
- National University Cancer Institute, NUHS, Singapore, Singapore
- ISEP, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - Marcus E Peter
- Department of Medicine, Division Hematology/Oncology, Northwestern University, Chicago, IL, USA
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Giovanni Porta
- Center of Genomic Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin 2, Ireland
| | - Hamsa Puthalakath
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina. Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | - Kodi S Ravichandran
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Cell Clearance, Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Equipe labellisée Ligue Contre le Cancer, Nice, France
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Nirmal Robinson
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, Australia
| | - Cecilia M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Barak Rotblat
- Department of Life sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
- The NIBN, Beer Sheva, Israel
| | - Carla V Rothlin
- Department of Immunobiology and Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge, UK
| | - Thomas Rudel
- Microbiology Biocentre, University of Würzburg, Würzburg, Germany
| | - Alessandro Rufini
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milano, Italy
- University of Leicester, Leicester Cancer Research Centre, Leicester, UK
| | - Kevin M Ryan
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Kristopher A Sarosiek
- John B. Little Center for Radiation Sciences, Harvard School of Public Health, Boston, MA, USA
- Department of Systems Biology, Lab of Systems Pharmacology, Harvard Program in Therapeutics Science, Harvard Medical School, Boston, MA, USA
- Department of Environmental Health, Molecular and Integrative Physiological Sciences Program, Harvard School of Public Health, Boston, MA, USA
| | - Akira Sawa
- Johns Hopkins Schizophrenia Center, Johns Hopkins University, Baltimore, MD, USA
| | - Emre Sayan
- Faculty of Medicine, Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Kate Schroder
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Luca Scorrano
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
| | - Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, NJ, USA
| | - Feng Shao
- National Institute of Biological Sciences, Beijing, PR China
| | - Yufang Shi
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Giuseppe S Sica
- Department of Surgical Science, University Tor Vergata, Rome, Italy
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Antonella Sistigu
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Brent R Stockwell
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Flavie Strapazzon
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Univ Lyon, Univ Lyon 1, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyogène CNRS, INSERM, Lyon, France
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Liming Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Sun
- Laboratory of Cell Engineering, Institute of Biotechnology, Beijing, China
- Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China
| | - Gyorgy Szabadkai
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, UK
| | - Stephen W G Tait
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Daolin Tang
- Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
- Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Carol M Troy
- Departments of Pathology & Cell Biology and Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, J. Stefan Institute, Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Nicoletta Urbano
- Department of Oncohaematology, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Methusalem Program, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Achucarro Center for Neuroscience, IKERBASQUE, Bilbao, Spain
- School of Forensic Medicine, China Medical University, Shenyang, China
- State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Andreas Villunger
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
- The Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences (OeAW), Vienna, Austria
- The Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD), Vienna, Austria
| | - Silvia von Karstedt
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Anne K Voss
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA
| | - Daniela Vuri
- Department of Experimental Medicine, University of Rome Tor Vergata, TOR, Rome, Italy
| | - Erwin F Wagner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Henning Walczak
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
- CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, London, UK
| | - David Wallach
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Ruoning Wang
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Ying Wang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Achim Weber
- University of Zurich and University Hospital Zurich, Department of Pathology and Molecular Pathology, Zurich, Switzerland
- University of Zurich, Institute of Molecular Cancer Research, Zurich, Switzerland
| | - Will Wood
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Huang-Tian Yang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Zahra Zakeri
- Queens College and Graduate Center, City University of New York, Flushing, NY, USA
| | - Joanna E Zawacka-Pankau
- Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
- Department of Biochemistry, Laboratory of Biophysics and p53 protein biology, Medical University of Warsaw, Warsaw, Poland
| | - Lin Zhang
- Department of Pharmacology & Chemical Biology, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Wenzhao Zhou
- Laboratory of Cell Engineering, Institute of Biotechnology, Beijing, China
- Research Unit of Cell Death Mechanism, 2021RU008, Chinese Academy of Medical Science, Beijing, China
| | - Mauro Piacentini
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Mafi A, Rezaee M, Hedayati N, Hogan SD, Reiter RJ, Aarabi MH, Asemi Z. Melatonin and 5-fluorouracil combination chemotherapy: opportunities and efficacy in cancer therapy. Cell Commun Signal 2023; 21:33. [PMID: 36759799 PMCID: PMC9912526 DOI: 10.1186/s12964-023-01047-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/14/2023] [Indexed: 02/11/2023] Open
Abstract
Combined chemotherapy is a treatment method based on the simultaneous use of two or more therapeutic agents; it is frequently necessary to produce a more effective treatment for cancer patients. Such combined treatments often improve the outcomes over that of the monotherapy approach, as the drugs synergistically target critical cell signaling pathways or work independently at different oncostatic sites. A better prognosis has been reported in patients treated with combination therapy than in patients treated with single drug chemotherapy. In recent decades, 5-fluorouracil (5-FU) has become one of the most widely used chemotherapy agents in cancer treatment. This medication, which is soluble in water, is used as the first line of anti-neoplastic agent in the treatment of several cancer types including breast, head and neck, stomach and colon cancer. Within the last three decades, many studies have investigated melatonin as an anti-cancer agent; this molecule exhibits various functions in controlling the behavior of cancer cells, such as inhibiting cell growth, inducing apoptosis, and inhibiting invasion. The aim of this review is to comprehensively evaluate the role of melatonin as a complementary agent with 5-FU-based chemotherapy for cancers. Additionally, we identify the potential common signaling pathways by which melatonin and 5-FU interact to enhance the efficacy of the combined therapy. Video abstract.
Collapse
Affiliation(s)
- Alireza Mafi
- grid.411036.10000 0001 1498 685XDepartment of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Malihe Rezaee
- grid.411600.2School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran ,grid.411705.60000 0001 0166 0922Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Neda Hedayati
- grid.411746.10000 0004 4911 7066School of Medicine, Iran University of Medical Science, Tehran, Islamic Republic of Iran
| | - Sara Diana Hogan
- grid.8993.b0000 0004 1936 9457Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Russel J. Reiter
- grid.43582.380000 0000 9852 649XDepartment of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX USA
| | - Mohammad-Hossein Aarabi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| |
Collapse
|
11
|
Li QZ, Zhou ZR, Hu CY, Li XB, Chang YZ, Liu Y, Wang YL, Zhou XW. Recent advances of bioactive proteins/polypeptides in the treatment of breast cancer. Food Sci Biotechnol 2023; 32:265-282. [PMID: 36619215 PMCID: PMC9808697 DOI: 10.1007/s10068-022-01233-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/24/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023] Open
Abstract
Proteins do not only serve as nutrients to fulfill the demand for food, but also are used as a source of bioactive proteins/polypeptides for regulating physical functions and promoting physical health. Female breast cancer has the highest incidence in the world and is a serious threat to women's health. Bioactive proteins/polypeptides exert strong anti-tumor effects and exhibit inhibition of multiple breast cancer cells. This review discussed the suppressing effects of bioactive proteins/polypeptides on breast cancer in vitro and in vivo, and their mechanisms of migration and invasion inhibition, apoptosis induction, and cell cycle arrest. This may contribute to providing a basis for the development of bioactive proteins/polypeptides for the treatment of breast cancer. Graphical abstract
Collapse
Affiliation(s)
- Qi-Zhang Li
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Food and Biological Engineering, Hubei University of Technology, No.28, Nanli Road, Wuhan, 430068 Hubei People’s Republic of China
- School of Agriculture and Biology, and Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240 People’s Republic of China
| | - Ze-Rong Zhou
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Food and Biological Engineering, Hubei University of Technology, No.28, Nanli Road, Wuhan, 430068 Hubei People’s Republic of China
| | - Cui-Yu Hu
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Key Laboratory of Industrial Microbiology, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Food and Biological Engineering, Hubei University of Technology, No.28, Nanli Road, Wuhan, 430068 Hubei People’s Republic of China
| | - Xian-Bin Li
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, Guangdong 510006 People’s Republic of China
| | - Yu-Zhou Chang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH 43210 USA
| | - Yan Liu
- School of Agriculture and Biology, and Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240 People’s Republic of China
| | - Yu-Liang Wang
- School of Agriculture and Biology, and Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240 People’s Republic of China
| | - Xuan-Wei Zhou
- School of Agriculture and Biology, and Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240 People’s Republic of China
| |
Collapse
|
12
|
Li D, Luo D, Hu S, Zhao H, Peng B. Syringic Acid Suppressed Proliferation, Invasion, and Migration via Inhibition of Matrix Metalloproteinases Expression on Glioblastoma Cells by Promoting Apoptosis. Curr Pharm Biotechnol 2023; 24:310-316. [PMID: 35570553 DOI: 10.2174/1389201023666220513100129] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/20/2022] [Accepted: 03/02/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Human brain tumor glioblastoma (GBM) is the most hostile malignancy, currently lacking a successful cure and good prognosis. OBJECTIVE To examine the anticancer effects of syringic acid (SA) on human cancer GBM cells. METHODOLOGY The different doses of SA were added to GBM cells to study its effect on viability, invasion, relocation, apoptosis, and mRNA and protein levels. Hence, we explored the antiproliferative, anti-invasive, and apoptotic activity of SA on GBM human U-251 cells. RESULTS MTT assay and live/dead assay revealed the anti-proliferative activity of SA on U-251 glioma cells. Apoptotic activity of SA was shown by DAPI staining, caspase-3, Bax, and Bcl-2 mRNA expressions. The cell cycle regulation was also confirmed by reducing the mRNA expression of cyclinD1, CDK4, and CDK6. Treatment of SA with U-251 cells suppressed MMPs expressions and enhanced TIMPs protein levels. CONCLUSION Our findings put forward that SA could prevent GBM cells' invasion and relocation. SA is an ideal neuroprotective agent for controlling brain malignancy.
Collapse
Affiliation(s)
- Dan Li
- Department of Neurosurgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Dongdong Luo
- Department of Neurosurgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Su Hu
- Department of Neurosurgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Hailin Zhao
- Department of Neurosurgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Biao Peng
- Department of Neurosurgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| |
Collapse
|
13
|
TPGS loaded triphenyltin (IV) micelles induced apoptosis by upregulating p53 in breast cancer cells and inhibit tumor progression in T-cell lymphoma bearing mice. Life Sci 2022; 308:120937. [PMID: 36088999 DOI: 10.1016/j.lfs.2022.120937] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/27/2022] [Accepted: 09/04/2022] [Indexed: 10/31/2022]
Abstract
AIMS Currently, breast cancer is one of the most frequently diagnosed and the second leading cause of cancer related deaths in women worldwide. Our present study aimed to investigate the major mechanistic effects of micelles (TSD-30-F, TSD-34-F) on breast cancer cells as well as their antitumor efficacy in in vivo DL bearing BALB/c mice. METHODS Apoptotic death by micelles was investigated by mitochondrial aggregation, membrane potential and DNA fragmentation assay in MCF-7 and MDA-MB-231 cells. Molecular mode of action of micelles were determined by RT-PCR and western blot analysis, drug-ligand interaction was analyzed by in silico methods, while, in vivo antitumor activity was investigated by Kaplen-Meier survival curve, T/C value, body weight and belly size of BALB/c mice. KEY FINDINGS TSD-30-F and TSD-34-F micelles displayed significant apoptotic induction. At molecular level, TSD-30 and TSD-34 micelles showed up-regulation of p53, Bax, Bak, Caspase-3 and down-regulation of Bcl-2 genes as well as proteins in tested breast cancer cells. In silico analysis revealed that TSD-30 and TSD-34 showed efficient binding affinity with p53, Caspase-3, Bax and Bcl-2 proteins. Significant in vivo antitumor efficacy was exhibited by the micelles formulations by increasing life span with reduced bodyweight and belly size growth pattern in BALB/c mice compared to DTX-F micelles. SIGNIFICANCE Our results suggest that triphenyltin (IV) micelles could be a very promising therapeutic candidate for treatment of breast cancer patients and occupy a new place in targeted breast cancer therapeutic.
Collapse
|
14
|
Mihanfar A, Yousefi B, Ghazizadeh Darband S, Sadighparvar S, Kaviani M, Majidinia M. Melatonin increases 5-flurouracil-mediated apoptosis of colorectal cancer cells through enhancing oxidative stress and downregulating survivin and XIAP. ACTA ACUST UNITED AC 2021; 11:253-261. [PMID: 34631487 PMCID: PMC8494259 DOI: 10.34172/bi.2021.36] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 05/28/2020] [Accepted: 07/04/2020] [Indexed: 12/12/2022]
Abstract
![]()
Introduction: Colorectal cancer (CRC) is one of the most lethal human malignancies with a global alarming rate of incidence. The development of resistance against common chemotherapeutics such as 5-fluorouracil (5-FU) remains a big burden for CRC therapy. Therefore, we investigated the effects of melatonin on the increasing 5-FU- mediated apoptosis and its underlying mechanism in SW-480 CRC cell line.
Methods: The effects of melatonin and 5- FU, alone or in combination, on cell proliferation were evaluated using an MTT assay. Further, Annexin-V Flow cytometry was used for determining the effects of melatonin and 5-FU on the apoptosis of SW-480 cell lines. The expression levels of Bax, Bcl-2, pro-caspase-3/activated caspase 3, X-linked inhibitor of apoptosis proteins (XIAP), and survivin were measured after 48 hours incubation with drugs. Cellular levels of reactive oxygen species (ROS), catalase, superoxide dismutase and glutathione peroxidase were also evaluated.
Results: Melatonin and 5-FU significantly decreased the cell proliferation of SW-480 cells. Combination of 5-FU with melatonin significantly decreased the IC50 value of 5-FU from 100 μM to 50 μM. Moreover, combination therapy increased intracellular levels of ROS and suppressed antioxidant enzymatic activities (P < 0.05). Treatment with either melatonin or 5-FU resulted in the induction of apoptosis in comparison to control (P > 0.05). XIAP and survivin expression levels potently decreased after combination treatment with melatonin and 5-FU (P < 0.05).
Conclusion: We demonstrated that melatonin exerts a reversing effect on the resistance to apoptosis by targeting oxidative stress, XIAP and survivin in CRC cells. Therefore, more studies need for better understanding of underlying mechanisms for beneficial effects of combination of melatonin and 5-FU.
Collapse
Affiliation(s)
- Ainaz Mihanfar
- Student Research Community, Urmia University of Medical Sciences, Urmia, Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Shirin Sadighparvar
- Neurophysiology Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Mojtaba Kaviani
- School of Nutrition and Dietetics, Acadia University, Wolfville, Nova Scotia, Canada
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
15
|
Anjum S, Hashim M, Malik SA, Khan M, Lorenzo JM, Abbasi BH, Hano C. Recent Advances in Zinc Oxide Nanoparticles (ZnO NPs) for Cancer Diagnosis, Target Drug Delivery, and Treatment. Cancers (Basel) 2021; 13:4570. [PMID: 34572797 PMCID: PMC8468934 DOI: 10.3390/cancers13184570] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is regarded as one of the most deadly and mirthless diseases and it develops due to the uncontrolled proliferation of cells. To date, varieties of traditional medications and chemotherapies have been utilized to fight tumors. However, their immense drawbacks, such as reduced bioavailability, insufficient supply, and significant adverse effects, make their use limited. Nanotechnology has evolved rapidly in recent years and offers a wide spectrum of applications in the healthcare sectors. Nanoscale materials offer strong potential for curing cancer as they pose low risk and fewer complications. Several metal oxide NPs are being developed to diagnose or treat malignancies, but zinc oxide nanoparticles (ZnO NPs) have remarkably demonstrated their potential in the diagnosis and treatment of various types of cancers due to their biocompatibility, biodegradability, and unique physico-chemical attributes. ZnO NPs showed cancer cell specific toxicity via generation of reactive oxygen species and destruction of mitochondrial membrane potential, which leads to the activation of caspase cascades followed by apoptosis of cancerous cells. ZnO NPs have also been used as an effective carrier for targeted and sustained delivery of various plant bioactive and chemotherapeutic anticancerous drugs into tumor cells. In this review, at first we have discussed the role of ZnO NPs in diagnosis and bio-imaging of cancer cells. Secondly, we have extensively reviewed the capability of ZnO NPs as carriers of anticancerous drugs for targeted drug delivery into tumor cells, with a special focus on surface functionalization, drug-loading mechanism, and stimuli-responsive controlled release of drugs. Finally, we have critically discussed the anticancerous activity of ZnO NPs on different types of cancers along with their mode of actions. Furthermore, this review also highlights the limitations and future prospects of ZnO NPs in cancer theranostic.
Collapse
Affiliation(s)
- Sumaira Anjum
- Department of Biotechnology, Kinnaird College for Women, Jail Road, Lahore 54000, Pakistan; (M.H.); (S.A.M.); (M.K.)
| | - Mariam Hashim
- Department of Biotechnology, Kinnaird College for Women, Jail Road, Lahore 54000, Pakistan; (M.H.); (S.A.M.); (M.K.)
| | - Sara Asad Malik
- Department of Biotechnology, Kinnaird College for Women, Jail Road, Lahore 54000, Pakistan; (M.H.); (S.A.M.); (M.K.)
| | - Maha Khan
- Department of Biotechnology, Kinnaird College for Women, Jail Road, Lahore 54000, Pakistan; (M.H.); (S.A.M.); (M.K.)
| | - José M. Lorenzo
- Centro Tecnológico de la Carne de Galicia, Avenida de Galicia 4, Parque Tecnológico de Galicia, 32900 San Cibrao das Viñas, Ourense, Spain;
- Área de Tecnología de los Alimentos, Facultad de Ciencias de Ourense, Universidad de Vigo, 32004 Ourense, Spain
| | - Bilal Haider Abbasi
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 15320, Pakistan;
| | - Christophe Hano
- Laboratoire de Biologie des Ligneux et des Grandes Cultures, INRAE USC1328, Eure & Loir Campus, University of Orleans, 28000 Chartres, France;
| |
Collapse
|
16
|
Song X, Luo Y, Ma L, Hu X, Simal-Gandara J, Wang LS, Bajpai VK, Xiao J, Chen F. Recent trends and advances in the epidemiology, synergism, and delivery system of lycopene as an anti-cancer agent. Semin Cancer Biol 2021; 73:331-346. [PMID: 33794344 DOI: 10.1016/j.semcancer.2021.03.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Xunyu Song
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China
| | - Yinghua Luo
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China
| | - Lingjun Ma
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China
| | - Xiaosong Hu
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E-32004 Ourense, Spain
| | - Li-Shu Wang
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Vivek K Bajpai
- Department of Energy and Materials Engineering, Dongguk University, 30 Pildong-ro 1-gil, Seoul 04620, Republic of Korea
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Food Science and Technology, University of Vigo - Ourense Campus, E-32004 Ourense, Spain.
| | - Fang Chen
- College of Food Science and Nutritional Engineering, National Engineering Research Centre for Fruit and Vegetable Processing, Key Laboratory of Fruits and Vegetables Processing, Ministry of Agriculture, Engineering Research Centre for Fruits and Vegetables Processing, Ministry of Education, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
17
|
Akhigbe RE, Hamed MA, Odetayo AF, Akhigbe TM, Ajayi AF, Ajibogun FAH. Omega-3 fatty acid rescues ischaemia/perfusion-induced testicular and sperm damage via modulation of lactate transport and xanthine oxidase/uric acid signaling. Biomed Pharmacother 2021; 142:111975. [PMID: 34343894 DOI: 10.1016/j.biopha.2021.111975] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/14/2021] [Accepted: 07/26/2021] [Indexed: 12/27/2022] Open
Abstract
This study aimed to explore the potential antioxidant, anti-inflammatory, and anti-apoptotic effects of omega 3 fatty acid (Ω-3) in a rat model of testicular torsion/detorsion (T/D). Under ketamine/xylazine anaesthesia, age-matched adult male Wistar rats of comparable weight underwent sham-operation or testicular torsion by fixing the left testis rotated at 720° for two and half hours. After detorsion, animals were treated with either olive oil as vehicle or Ω-3 subcutaneously for three days. On post-operative day 3, rats were culled and the ipsilateral and contralateral testes, as well as obtained blood samples, were analyzed. Our findings revealed that T/D led to significant poor weight gain, distorted gross anatomy, and cytoarchitecture of the testes, low sperm quality, redox imbalance, and inflammation of the ipsilateral and contralateral testes. This was accompanied by reduced circulatory testosterone, a decline in testicular lactate metabolism and transport, upregulation of xanthine oxidase/uric acid signaling, and increased testicular DNA fragmentation. Administration of Ω-3 attenuated T/D-induced damage to the testes and sperm cells with a significant rise in the level of serum testosterone. Enhancement of lactate transport and down-regulation of xanthine oxidase/uric acid signaling by Ω-3 may be beneficial in protecting against T/D-related oxido-inflammatory damage and male infertility.
Collapse
Affiliation(s)
- R E Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; Reproductive Biology and Toxicology Research Laboratories, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Department of Chemical Sciences, Kings University, Odeomu, Osun, Nigeria.
| | - M A Hamed
- Reproductive Biology and Toxicology Research Laboratories, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Buntai Medical and Diagnostic Laboratories, Osogbo, Nigeria.
| | - A F Odetayo
- Reproductive Biology and Toxicology Research Laboratories, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Department of Physiology, University of Ilorin, Ilorin, Kwara State, Nigeria.
| | - T M Akhigbe
- Reproductive Biology and Toxicology Research Laboratories, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Department of Crop Production and Soil Science, Faculty of Agricultural Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - A F Ajayi
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - F A H Ajibogun
- Bioresources Development Centre, National Biotechnology Development Agency, Ministry of Science and Technology, Ogbomoso, Oyo State, Nigeria.
| |
Collapse
|
18
|
Valente LJ, Tarangelo A, Li AM, Naciri M, Raj N, Boutelle AM, Li Y, Mello SS, Bieging-Rolett K, DeBerardinis RJ, Ye J, Dixon SJ, Attardi LD. p53 deficiency triggers dysregulation of diverse cellular processes in physiological oxygen. J Cell Biol 2021; 219:152074. [PMID: 32886745 PMCID: PMC7594498 DOI: 10.1083/jcb.201908212] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 06/17/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
The mechanisms by which TP53, the most frequently mutated gene in human cancer, suppresses tumorigenesis remain unclear. p53 modulates various cellular processes, such as apoptosis and proliferation, which has led to distinct cellular mechanisms being proposed for p53-mediated tumor suppression in different contexts. Here, we asked whether during tumor suppression p53 might instead regulate a wide range of cellular processes. Analysis of mouse and human oncogene-expressing wild-type and p53-deficient cells in physiological oxygen conditions revealed that p53 loss concurrently impacts numerous distinct cellular processes, including apoptosis, genome stabilization, DNA repair, metabolism, migration, and invasion. Notably, some phenotypes were uncovered only in physiological oxygen. Transcriptomic analysis in this setting highlighted underappreciated functions modulated by p53, including actin dynamics. Collectively, these results suggest that p53 simultaneously governs diverse cellular processes during transformation suppression, an aspect of p53 function that would provide a clear rationale for its frequent inactivation in human cancer.
Collapse
Affiliation(s)
- Liz J Valente
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Amy Tarangelo
- Department of Biology, Stanford University, Stanford, CA
| | - Albert Mao Li
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Marwan Naciri
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA.,École Normale Supérieure de Lyon, Université Claude Bernard Lyon I, Université de Lyon, Lyon, France
| | - Nitin Raj
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Anthony M Boutelle
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Yang Li
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Stephano Spano Mello
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA.,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY
| | - Kathryn Bieging-Rolett
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jiangbin Ye
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
19
|
Kulbay M, Bernier-Parker N, Bernier J. The role of the DFF40/CAD endonuclease in genomic stability. Apoptosis 2021; 26:9-23. [PMID: 33387146 DOI: 10.1007/s10495-020-01649-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 12/18/2022]
Abstract
Maintenance of genomic stability in cells is primordial for cellular integrity and protection against tumor progression. Many factors such as ultraviolet light, oxidative stress, exposure to chemical reagents, particularly mutagens and radiation, can alter the integrity of the genome. Thus, human cells are equipped with many mechanisms that prevent these irreversible lesions in the genome, as DNA repair pathways, cell cycle checkpoints, and telomeric function. These mechanisms activate cellular apoptosis to maintain DNA stability. Emerging studies have proposed a new protein in the maintenance of genomic stability: the DNA fragmentation factor (DFF). The DFF40 is an endonuclease responsible of the oligonucleosomal fragmentation of the DNA during apoptosis. The lack of DFF in renal carcinoma cells induces apoptosis without oligonucleosomal fragmentation, which poses a threat to genetic information transfer between cancerous and healthy cells. In this review, we expose the link between the DFF and genomic instability as the source of disease development.
Collapse
Affiliation(s)
- Merve Kulbay
- INRS - Centre Armand-Frappier-Santé-Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada
- Department of Medicine, Université de Montréal, 2900 Blvd. Edouard Montpetit, Montreal, QC, Canada
| | - Nathan Bernier-Parker
- Toronto Animal Health Partners Emergency and Specialty Hospital, 1 Scarsdale Road, North York, ON, M3B 2R2, Canada
| | - Jacques Bernier
- INRS - Centre Armand-Frappier-Santé-Biotechnologie, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada.
| |
Collapse
|
20
|
Torrealba N, Rodríguez-Berriguete G, Vera R, Fraile B, Olmedilla G, Martínez-Onsurbe P, Sánchez-Chapado M, Paniagua R, Royuela M. Homeostasis: apoptosis and cell cycle in normal and pathological prostate. Aging Male 2020; 23:335-345. [PMID: 29730957 DOI: 10.1080/13685538.2018.1470233] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Prostatic diseases such as hyperplasia and cancer are a consequence of glandular aging due to the loss of homeostasis. Glandular homeostasis is guaranteed by the delicate balance between production and cell death. Both cell renewal and apoptosis are part of this delicate balance. We will explore the predictive capacity for biochemical progression, following prostatectomy, of some members of the Bcl-2 family and of proteins involved in cell cycle inhibition in conjunction with established classical markers. The expression of Bcl-2, Bcl-xL, Mcl-1, Bax, Bim, Bad, PUMA, Noxa, p21, p27, Rb and p53 were analyzed by immunochemistry in 86 samples of radical prostatectomy and correlated with each of the markers established clinicopathological tests using statistical tests such as Sperman, Kaplan-Meier curves, unifactorial Cox, and multifactorial. The most relevant results are: (1) Positive correlation between: p27 with clinical T stage; and PUMA with pathological T stage; (2) Negative correlation between: Bcl-2 with clinical T stage, Bcl-xL with survival, Noxa and pRb with Gleason score.Our results suggest that the expression of Bcl-2, Bcl-xL, PUMA, Noxa, p27, and Rb were related to some of the classic markers established to predict biochemical progression after prostatectomy.
Collapse
Affiliation(s)
- Norelia Torrealba
- Department of Biomedicine and Biotechnology, University of Alcalá, Alcala de Henares, Spain
| | | | - Raúl Vera
- Department of Biomedicine and Biotechnology, University of Alcalá, Alcala de Henares, Spain
| | - Benito Fraile
- Department of Biomedicine and Biotechnology, University of Alcalá, Alcala de Henares, Spain
| | - Gabriel Olmedilla
- Department of Pathology, Príncipe de Asturias Hospital, Alcalá de Henares, Madrid, Spain
| | - Pilar Martínez-Onsurbe
- Department of Pathology, Príncipe de Asturias Hospital, Alcalá de Henares, Madrid, Spain
| | | | - Ricardo Paniagua
- Department of Biomedicine and Biotechnology, University of Alcalá, Alcala de Henares, Spain
| | - Mar Royuela
- Department of Biomedicine and Biotechnology, University of Alcalá, Alcala de Henares, Spain
| |
Collapse
|
21
|
Somade OT, Ajayi BO, Olunaike OE, Jimoh LA. Hepatic oxidative stress, up-regulation of pro-inflammatory cytokines, apoptotic and oncogenic markers following 2-methoxyethanol administrations in rats. Biochem Biophys Rep 2020; 24:100806. [PMID: 32913901 PMCID: PMC7472863 DOI: 10.1016/j.bbrep.2020.100806] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 08/18/2020] [Accepted: 08/18/2020] [Indexed: 01/29/2023] Open
Abstract
2-methoxyethanol (2-ME) is an organic solvent widely used in the manufacture of brake fluids, paints, resins, varnish, nail polish, acetate cellulose, wood coloring, and as a plasticizer in plastics manufacturing. We therefore, investigated its effect on the liver, in a time-course study in male Wistar rats. Animals were orally administered 50 mg/kg body weight of 2-ME for a period of 7, 14, and 21 days. Following 7 days of administration of 2-ME, there was a significant increase in the level of Bax, c-Myc, K-Ras, TNF-α, IL-1β, IL-6, MDA and GPx activity, while the levels of Bcl-2, NO and GSH were significantly reduced compared with control. At the end of 14 days exposure, Bcl-2, and GSH levels, as well as GST activity, were significantly decreased, while levels of Bax, c-Myc, K-Ras, caspase-3, TNF-α, IL-1β, IL-6, MDA and NO were significantly increased compared with control. After 21 days of 2-ME administration, Bcl-2, IL-10, and GSH levels, as well as SOD and GST activities, were significantly decreased, while levels of Bax, c-Myc, K-Ras, caspase-3, p53, TNF-α, IL-1β, IL-6, MDA and NO were significantly increased compared with control. Lastly, liver histopathology confirmed and corroborated the biochemical findings reported above. We therefore, advised that exposures to 2-ME should be strictly avoided as it could trigger hepatic damage through the disorganization of the antioxidant system, up-regulation of inflammatory, apoptotic, and oncogenic markers in rats.
Collapse
Key Words
- 2-methoxyethanol
- Apoptosis
- Bax, Bcl-2 associated X
- Bcl-2, B-cell lymphoma 2
- CAT, catalase
- GPx, glutathione peroxidase
- GSH, reduced glutathione
- GST, glutathione S-transferase
- IL-10, interleukin 10
- IL-1β, interleukin-1 beta
- IL-6, interleukin-6
- Inflammation
- K-Ras, Kirsten rat sarcoma viral oncogene
- Liver
- MDA, malondialdehyde
- NO, nitric oxide
- Oncogenes
- Oxidative stress
- SOD, superoxide dismutase
- TNF-α, tumor necrosis factor alpha
- c-Myc, myelocytomatosis
- p53, tumor suppressor protein
Collapse
Affiliation(s)
- Oluwatobi T. Somade
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - Babajide O. Ajayi
- Department of Chemical Sciences, Faculty of Natural Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Oyinkansola E. Olunaike
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - Latifah A. Jimoh
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| |
Collapse
|
22
|
Ajayi AF, Akhigbe RE. In vivo exposure to codeine induces reproductive toxicity: role of HER2 and p53/Bcl-2 signaling pathway. Heliyon 2020; 6:e05589. [PMID: 33294712 PMCID: PMC7695972 DOI: 10.1016/j.heliyon.2020.e05589] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/17/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023] Open
Abstract
Several studies have implicated codeine use in the aetiopathogenesis of male infertility. The purpose of this study was to investigate the role of HER2, Ki67, oestrogen and p53/Bcl-2 signaling pathways and the possible outcome of codeine cessation on codeine-induced reproductive toxicity. Thirty adult male Wistar rats of comparable ages and weights were randomly allocated into 5 groups. The control animals received distilled water per os (p.o), while animals in the low-dose (LDC) and high dose (HDC) codeine-treated groups received 2 and 5 mg/kg/day of codeine respectively p.o for 6 weeks. The animals in the low-dose codeine recovery (LDC-R) and high-dose codeine recovery (HDC-R) groups received treatment as LDC and HDC respectively followed by another drug-free six weeks, recovery period. Cessation of codeine exposure led to a partial reversal of codeine-induced poor sperm quality, reduced litter size and weight, increased oxidative testicular injury, testicular apoptosis, and testicular DNA damage caused by codeine administration. Codeine-induced gonado-spermotoxicity was associated with a reduction of circulatory testosterone, suppression of testicular HER2, Ki67, and Bcl-2 expression, down-regulation of oestrogen signaling, and upregulation of testicular caspase 3 activities and p53 signaling pathway. Conclusion: Upregulation of oestrogen signaling associated with enhanced testicular HER2 and Ki67 expression during the recovery period is seemingly beneficial in protecting against codeine-related testicular injury and infertility.
Collapse
Affiliation(s)
- A F Ajayi
- Department of Physiology, College of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria
| | - R E Akhigbe
- Department of Physiology, College of Medicine, Ladoke Akintola University of Technology, Ogbomoso, Oyo, Nigeria.,Reproductive Biology and Toxicology Research Laboratories, Oasis of Grace Hospital, Osogbo, Osun, Nigeria.,Department of Chemical Sciences, Kings University, Odeomu, Osun, Nigeria
| |
Collapse
|
23
|
Zhang Y, Li P, Rong J, Ge Y, Hu C, Bai X, Shi W. Small molecule CDS-3078 induces G 2/M phase arrest and mitochondria-mediated apoptosis in HeLa cells. Exp Ther Med 2020; 20:284. [PMID: 33209128 PMCID: PMC7668142 DOI: 10.3892/etm.2020.9414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
The tumor suppressor p53 serves important roles in cell cycle arrest and apoptosis, and its activation increases the sensitivity of cancer cells to radiotherapy or chemotherapy. In the present study, the small molecule 2-[1-(4-(benzyloxy)phenyl)-3-oxoisoindolin-2-yl)-2-(4-methoxyphenyl)] acetic acid (CDS-3078) significantly increased p53 mRNA expression levels in a dose-dependent manner. Treatment with CDS-3078 increased p53 expression levels and p53-mediated activation of its downstream target genes in HeLa cells. Additionally, p53+/+ HeLa cells treated with CDS-3078 presented with dysfunctional mitochondria, as indicated by the decrease in Bcl-2 levels, the increase in Bcl-2 homologous antagonist killer and the increase in cytochrome c release from the mitochondria to the cytoplasm. The present results suggested that CDS-3078 treatment significantly induced G2/M phase cell cycle arrest. Therefore, CDS-3078 administration induced apoptosis via p53-mediated cell cycle arrest, causing mitochondrial dysfunction and resulting in apoptotic cell death in cervical cancer cells. Collectively, the present results suggested that CDS-3078 may be a potential anticancer agent.
Collapse
Affiliation(s)
- Yuanxin Zhang
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, P.R. China
| | - Pengcheng Li
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, P.R. China
| | - Jiamin Rong
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, P.R. China
| | - Yakun Ge
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, P.R. China
| | - Chenming Hu
- The Center for Combinatorial Chemistry and Drug Discovery, College of Pharmacy, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Xu Bai
- The Center for Combinatorial Chemistry and Drug Discovery, College of Pharmacy, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Wei Shi
- Key Laboratory for Molecular Enzymology and Engineering of The Ministry of Education, School of Life Science, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
24
|
Boskabadi SH, Balanezhad SZ, Neamati A, Tabrizi MH. The green-synthesized zinc oxide nanoparticle as a novel natural apoptosis inducer in human breast (MCF7 and MDA-MB231) and colon (HT-29) cancer cells. INORG NANO-MET CHEM 2020. [DOI: 10.1080/24701556.2020.1808991] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
| | | | - Ali Neamati
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | |
Collapse
|
25
|
Gui F, Zhang Y, Wan J, Zhan X, Yao Y, Li Y, Haddock AN, Shi J, Guo J, Chen J, Zhu X, Edenfield BH, Zhuang L, Hu C, Wang Y, Mukhopadhyay D, Radisky ES, Zhang L, Lugea A, Pandol SJ, Bi Y, Ji B. Trypsin activity governs increased susceptibility to pancreatitis in mice expressing human PRSS1R122H. J Clin Invest 2020; 130:189-202. [PMID: 31550238 DOI: 10.1172/jci130172] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022] Open
Abstract
Currently, an effective targeted therapy for pancreatitis is lacking. Hereditary pancreatitis (HP) is a heritable, autosomal-dominant disorder with recurrent acute pancreatitis (AP) progressing to chronic pancreatitis (CP) and a markedly increased risk of pancreatic cancer. In 1996, mutations in PRSS1 were linked to the development of HP. Here, we developed a mouse model by inserting a full-length human PRSS1R122H gene, the most commonly mutated gene in human HP, into mice. Expression of PRSS1R122H protein in the pancreas markedly increased stress signaling pathways and exacerbated AP. After the attack of AP, all PRSS1R122H mice had disease progression to CP, with similar histologic features as those observed in human HP. By comparing PRSS1R122H mice with PRSS1WT mice, as well as enzymatically inactivated Dead-PRSS1R122H mice, we unraveled that increased trypsin activity is the mechanism for R122H mutation to sensitize mice to the development of pancreatitis. We further discovered that trypsin inhibition, in combination with anticoagulation therapy, synergistically prevented progression to CP in PRSS1R122H mice. These animal models help us better understand the complex nature of this disease and provide powerful tools for developing and testing novel therapeutics for human pancreatitis.
Collapse
Affiliation(s)
- Fu Gui
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Yuebo Zhang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jianhua Wan
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Xianbao Zhan
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Yao Yao
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Yinghua Li
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ashley N Haddock
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Ji Shi
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jia Guo
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jiaxiang Chen
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Xiaohui Zhu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Lu Zhuang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Cheng Hu
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ying Wang
- Department of Biochemistry and Molecular Biology
| | | | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Aurelia Lugea
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Stephen J Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yan Bi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Baoan Ji
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
26
|
Augert A, Mathsyaraja H, Ibrahim AH, Freie B, Geuenich MJ, Cheng PF, Alibeckoff SP, Wu N, Hiatt JB, Basom R, Gazdar A, Sullivan LB, Eisenman RN, MacPherson D. MAX Functions as a Tumor Suppressor and Rewires Metabolism in Small Cell Lung Cancer. Cancer Cell 2020; 38:97-114.e7. [PMID: 32470392 PMCID: PMC7363581 DOI: 10.1016/j.ccell.2020.04.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Small cell lung cancer (SCLC) is a highly aggressive and lethal neoplasm. To identify candidate tumor suppressors we applied CRISPR/Cas9 gene inactivation screens to a cellular model of early-stage SCLC. Among the top hits was MAX, the obligate heterodimerization partner for MYC family proteins that is mutated in human SCLC. Max deletion increases growth and transformation in cells and dramatically accelerates SCLC progression in an Rb1/Trp53-deleted mouse model. In contrast, deletion of Max abrogates tumorigenesis in MYCL-overexpressing SCLC. Max deletion in SCLC resulted in derepression of metabolic genes involved in serine and one-carbon metabolism. By increasing serine biosynthesis, Max-deleted cells exhibit resistance to serine depletion. Thus, Max loss results in metabolic rewiring and context-specific tumor suppression.
Collapse
Affiliation(s)
- Arnaud Augert
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Haritha Mathsyaraja
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ali H Ibrahim
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Brian Freie
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Michael J Geuenich
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Quest University Canada, 3200 University Boulevard, Squamish, BC V8B 0N8, Canada
| | - Pei-Feng Cheng
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Sydney P Alibeckoff
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Nan Wu
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Joseph B Hiatt
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Ryan Basom
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA
| | - Adi Gazdar
- University of Texas, Southwestern, USA, 6000 Harry Hines Boulevard, Dallas, TX 75235, USA
| | - Lucas B Sullivan
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA; Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Robert N Eisenman
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
27
|
Stallinger A, Kretschmer N, Kleinegger F, Brvar L, Liegl-Atzwanger B, Prokesch A, Durchschein C, Bauer R, Deutsch A, Rinner B. β,β-Dimethylacrylshikonin Induces Apoptosis in Melanoma Cell Lines by NOXA Upregulation. JOURNAL OF NATURAL PRODUCTS 2020; 83:305-315. [PMID: 31961147 DOI: 10.1021/acs.jnatprod.9b00719] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Melanoma is the most aggressive form of skin cancer, with high metastasis rates and poor prognosis. Survival rates and possible therapies depend on the state of the tumor and its mutational profile. BRAF and NRAS are the most frequent driver mutations. Currently, there is no efficient therapy for NRAS-mutated or late-stage melanoma. In this study, the therapeutic potential of β,β-dimethylacrylshikonin (DMAS) was investigated on melanoma. The influence of DMAS was determined in five different melanoma cell lines with different mutational profiles. The effects of this compound on cell viability, apoptosis, and gene and protein expression were examined. The results obtained were validated in vivo. DMAS significantly reduced the viability of several melanoma cell lines in a concentration- and time-dependent manner. Furthermore, DMAS induced caspase-3-dependent apoptosis via NOXA upregulation, as confirmed by NOXA knockdown experiments. This is the first time that NOXA-dependent apoptosis was shown with respect to a shikonin derivative and melanoma. Additionally, tumor regression and necrosis under DMAS treatment were demonstrated in vivo. Importantly, BRAF as well as NRAS-mutated metastatic human melanoma cell lines were treated successfully in vitro and in vivo. Taken together, DMAS showed promising results and is worthy of further study.
Collapse
Affiliation(s)
- Alexander Stallinger
- Division of Biomedical Research , Medical University of Graz , 8036 Graz , Austria
| | - Nadine Kretschmer
- Division of Biomedical Research , Medical University of Graz , 8036 Graz , Austria
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy , University of Graz , 8010 Graz , Austria
| | - Florian Kleinegger
- Diagnostic and Research Institute of Pathology , Medical University of Graz , 8010 Graz , Austria
| | - Luka Brvar
- Division of Biomedical Research , Medical University of Graz , 8036 Graz , Austria
| | | | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism & Aging , Medical University of Graz , 8010 Graz , Austria
- Division of Cell Biology, Histology and Embryology , Medical University of Graz , 8010 Graz , Austria
| | - Christin Durchschein
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy , University of Graz , 8010 Graz , Austria
| | - Rudolf Bauer
- Institute of Pharmaceutical Sciences, Department of Pharmacognosy , University of Graz , 8010 Graz , Austria
| | - Alexander Deutsch
- Division of Hematology, Medical University of Graz , 8036 Graz , Austria
| | - Beate Rinner
- Division of Biomedical Research , Medical University of Graz , 8036 Graz , Austria
| |
Collapse
|
28
|
Huang Q, Chen Y, Hao L, Zhou R, Li Y, Li Q, Zhu B, Cai X. Pegylated carbon nitride nanosheets for enhanced reactive oxygen species generation and photodynamic therapy under hypoxic conditions. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 25:102167. [PMID: 32006685 DOI: 10.1016/j.nano.2020.102167] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/22/2019] [Accepted: 01/02/2020] [Indexed: 02/05/2023]
Abstract
The application of photodynamic therapy (PDT) is of ever-increasing importance in the treatment of malignant tumors; however, there are several major constraints that make it impossible to achieve optimal therapeutic effects. Our objective is to develop a novel photosensitizing drug for skin cancer. In the experiment, we fabricated four-arm-poly ethylene glycol modified amino-rich graphite phase carbon nitride nanosheets (AGCN-PEG), which have good stability in physiological solution and show selective accumulation in tumor cells. Under hypoxic conditions, the AGCN-PEG induced PDT can effectively inhibit growth on A431 human epidermoid carcinoma cells in vivo and in vitro. What's more, after being combined with TMPyP4, the therapeutic effect of AGCN-PEG was greatly improved.
Collapse
Affiliation(s)
- Qian Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Implant Dentistry, Stomatologic Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yang Chen
- Department of Liver Surgery& Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Liying Hao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanjing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qirong Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bofeng Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Forensic Genetics, School of Forensic Medicine, Southern Medical University, Guangzhou, PR China
| | - Xiaoxiao Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
29
|
Salari S, Neamati A, Tabrizi MH, Seyedi SMR. Green‐synthesized Zinc oxide nanoparticle, an efficient safe anticancer compound for human breast MCF7 cancer cells. Appl Organomet Chem 2019. [DOI: 10.1002/aoc.5417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Sahar Salari
- Department of Biology, Mashhad BranchIslamic Azad University Mashhad Iran
| | - Ali Neamati
- Department of Biology, Mashhad BranchIslamic Azad University Mashhad Iran
| | | | | |
Collapse
|
30
|
Baluchi I, Anani H, Hassanshahi G, Fatemi A, Khalilabadi RM. The effect of maslinic acid on apoptotic genes in u266 multiple myeloma cell line. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Zhang Y, Ge Y, Shi W, Hu C, Bai X, Rong J, Yu M. CDS-1548 induces apoptosis in HeLa cells by activating caspase 3. Oncol Lett 2019; 18:1881-1887. [PMID: 31423257 PMCID: PMC6614674 DOI: 10.3892/ol.2019.10511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 05/17/2019] [Indexed: 11/16/2022] Open
Abstract
Cervical cancer continues to be a threat to female health globally. In the present study, the potential anticancer activity of 2-[2-hydroxyl-1-(4-methoxy phenyl) ethyl]-3-(4-benzyloxy phenyl) isoindolin-1-one (CDS-1548), was evaluated in HeLa cells. CDS-1548 is an organic small-molecule compound characterized by two chiral centers, with the nuclear parent 1H-isoindolin-1-one. CDS-1548 administration significantly elevated the transcriptional activity of p53 and its downstream target genes in a dose-dependent manner. Additionally, CDS-1548 treatment increased the expression levels of p53 and mouse double minute 2 homolog, as well as inducing apoptosis in HeLa cells. Furthermore, CDS-1548 treatment downregulated the expression of B-cell lymphoma 2, upregulated Bcl-2 homologous antagonist killer, promoted the release of cytochrome c from mitochondria to cytoplasm, and activated the production of caspase 3 and 9. Collectively, these results suggested that CDS-1548 inhibited HeLa cell proliferation by promoting G2/M cell cycle phase arrest and inducting of mitochondria-mediated apoptosis.
Collapse
Affiliation(s)
- Yuanxin Zhang
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, P.R. China
| | - Yakun Ge
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, P.R. China
| | - Wei Shi
- Key Laboratory for Molecular Enzymology and Engineering of The Ministry of Education, School of Life Science, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Chenming Hu
- The Center for Combinatorial Chemistry and Drug Discovery, College of Pharmacy, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Xu Bai
- The Center for Combinatorial Chemistry and Drug Discovery, College of Pharmacy, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jimin Rong
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, P.R. China
| | - Ming Yu
- College of Biology and Food Engineering, Jilin Institute of Chemical Technology, Jilin, Jilin 132022, P.R. China
| |
Collapse
|
32
|
Xu YR, Yang WX. Roles of three Es-Caspases during spermatogenesis and Cadmium-induced apoptosis in Eriocheir sinensis. Aging (Albany NY) 2019; 10:1146-1165. [PMID: 29851651 PMCID: PMC5990378 DOI: 10.18632/aging.101454] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/18/2018] [Indexed: 01/08/2023]
Abstract
Functions of Caspases remain obscure in Crustacea. We studied the existence and participations of apoptosis-related factors in Eriocheir sinensis testis. Three Es-Caspases (Es-Caspase 3/ 7/ 8) in E. sinensis were cloned and characterized. We observed that three es-caspases mRNA had specific expression patterns during spermiogenesis, with weak signal around the nucleus and invaginated acrosomal vesicle in early-stage spermatids, became stronger in middle-stage, finally focused on the acrosomal tube and nucleus in mature sperm. We then investigated the immunostaining intensity and positional alterations of Es-Caspase 3, Es-Caspase 8 and p53 during spermatogenesis, which were correlated with the differential tendencies of cells to undergo apoptosis and specific organelles shaping processes. After apoptotic induction by Cadmium, Es-Caspase 8 increased gradually, while Es-Caspase 3 increased firstly and then decreased, Es-p53 initially decreased and then increased. These results implies that Es-Caspase 3/ Es-Caspase 8/ p53 may play roles in Cadmium-induced apoptosis during spermatogenesis, and Caspase 8-Caspase 3-p53 pathway may interact with extrinsic or intrinsic pathways to regulate the destiny of sperm cells. Our study revealed the indispensable roles of Caspases during spermatogenesis and the possible molecular interactions in response to the Cadmium-induced apoptosis in E. sinensis, which filled the gap of apoptotic mechanisms of crustacean.
Collapse
Affiliation(s)
- Ya-Ru Xu
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
33
|
Xu J, Li Y, Hu H. Effects of lycopene on ovarian cancer cell line SKOV3 in vitro: Suppressed proliferation and enhanced apoptosis. Mol Cell Probes 2019; 46:101419. [PMID: 31279748 DOI: 10.1016/j.mcp.2019.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/30/2019] [Accepted: 07/03/2019] [Indexed: 11/26/2022]
Abstract
AIMS This study aims to investigate the influences of lycopene on ovarian cancer cells SKOV3 in vitro and the corresponding mechanism. METHODS SKOV3 cultures were divided into four groups and treated with lycopene at 0 (the control group), 1 × 10-6, 1 × 10-5 and 1 × 10-4 mol/L respectively. The proliferation rate of SKOV3 cells was determined using MTT colorimetric assay, and the apoptosis rate of SKOV3 cells was measured using flow cytometry. Western blotting was used to detect the expressions of Bax and Bcl-2. RESULTS When compared with the control group, the proliferation rates of SKOV3 cells in the three lycopene groups were significantly lower (P < 0.05) while the apoptosis rates were significantly higher (P < 0.05). After lycopene treatment, the protein expression of Bax (an apoptotic protein) in SKOV3 cells increased significantly, and the protein expression of Bcl-2 (an anti-apoptotic protein) decreased significantly. CONCLUSION Lycopene could inhibit the proliferation of SKOV3 cells and enhance their apoptosis in vitro. The induction of cell apoptosis might be mediated by up-regulating Bax expression and down-regulating Bcl-2 expression.
Collapse
Affiliation(s)
- Jian Xu
- Department of Gynaecology, Ji'nan Central Hospital, Ji'nan, Shandong, 250000, China
| | - Yuanfang Li
- Department of Reproduction, Ji'nan Central Hospital, Ji'nan, Shandong, 250000, China
| | - Hongyan Hu
- Department of Interventional Medicine, Ji'nan Central Hospital, Ji'nan, Shandong, 250000, China.
| |
Collapse
|
34
|
Human Amniotic Membrane and Its Anti-cancer Mechanism: a Good Hope for Cancer Therapy. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s42399-019-00090-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
35
|
Rahimi Kalateh Shah Mohammad G, Seyedi SMR, Karimi E, Homayouni-Tabrizi M. The cytotoxic properties of zinc oxide nanoparticles on the rat liver and spleen, and its anticancer impacts on human liver cancer cell lines. J Biochem Mol Toxicol 2019; 33:e22324. [PMID: 30951608 DOI: 10.1002/jbt.22324] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 02/11/2019] [Accepted: 03/15/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Due to their unique properties including cellular uptake and the delivery efficiency to biological systems, nanoparticles are used in various preclinical and clinical applications. The aim of this study was to investigate the toxicity impacts of zinc oxide nanoparticles (ZnO-NPs) on morphology and functionality of the rat's liver and spleen and illustrated its safe-therapeutic doses. METHODS The 28 female Swiss albino rats (180-220 g) and two human hepatocyte cell lines (HepG2 and HUH7) were designed as an in vivo and in vitro study, respectively. Samples were treated with certain doses of ZnO-NPs. The rat's liver morphology and functionality and apoptotic genes expression profile (Bax, Bcl-2, and P53) were analyzed to detect the cytotoxicity and antitumor impacts of ZnO-NPs, respectively. RESULTS The results showed a positive significant association between the increasing doses of ZnO-NPs and alanine aminotransferase/aspartate aminotransferase values. Moreover, a meaningful correlation was detected between the rat's liver and spleen weight and ZnO-NPs doses. Furthermore, the histopathological analysis of rat's liver showed the individual cytotoxic properties of ZnO-NPs. Finally, the positive significant correlation was detected among the expression of Bax and P53 genes with ZnO-NPs. In addition, the negative correlation was demonstrated between the expression of Bcl-2 and ZnO-NPs. CONCLUSION In general, in the current study, the antitumor effects of ZnO-NPs were confirmed by the enhancement of P53 and Bax genes expression profile, which are indicated the apoptotic induction in HUH7 cell line. Moreover, we introduced a safe-clinical ZnO-NPs dosage, have antitumor effects.
Collapse
Affiliation(s)
| | | | - Ehsan Karimi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | |
Collapse
|
36
|
Niu N, Li Z, Zhu M, Sun H, Yang J, Xu S, Zhao W, Song R. Effects of nuclear respiratory factor‑1 on apoptosis and mitochondrial dysfunction induced by cobalt chloride in H9C2 cells. Mol Med Rep 2019; 19:2153-2163. [PMID: 30628711 PMCID: PMC6390059 DOI: 10.3892/mmr.2019.9839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 09/28/2018] [Indexed: 01/31/2023] Open
Abstract
Hypoxia-induced apoptosis occurs in various diseases. Cobalt chloride (CoCl2) is a hypoxia mimic agent that is frequently used in studies investigating the mechanisms of hypoxia. Nuclear respiratory factor-1 (NRF-1) is a transcription factor with an important role in the expression of mitochondrial respiratory and mitochondria-associated genes. However, few studies have evaluated the effects of NRF-1 on apoptosis, particularly with regard to damage caused by CoCl2. In the present study, the role of NRF-1 in mediating CoCl2-induced apoptosis was investigated using cell viability analysis, flow cytometry, fluorescence imaging, western blotting analysis, energy metabolism analysis and reverse transcription-quantitative polymerase chain reaction. The present results revealed that the apoptosis caused by CoCl2 could be alleviated by NRF-1. Furthermore, overexpression of NRF-1 increased the expression of B-cell lymphoma-2, hypoxia inducible factor-1α and NRF-2. Also, cell damage induced by CoCl2 may be associated with depolarization of mitochondrial membrane potential, and NRF-1 suppressed this effect. Notably, the oxygen consumption rate (OCR) was reduced in CoCl2-treated cells, whereas overexpression of NRF-1 enhanced the OCR, suggesting that NRF-1 had protective effects. In summary, the present study demonstrated that NRF-1 protected against CoCl2-induced apoptosis, potentially by strengthening mitochondrial function to resist CoCl2-induced damage to H9C2 cells. The results of the present study provide a possible way for the investigation of myocardial diseases.
Collapse
Affiliation(s)
- Nan Niu
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Zihua Li
- School of Pharmacy, Tsinghua University, Beijing 100084, P.R. China
| | - Mingxing Zhu
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Hongli Sun
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Jihui Yang
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Shimei Xu
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Wei Zhao
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Rong Song
- Department of Critical Care Medicine, The Fifth Hospital of the Chinese People's Liberation Army, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| |
Collapse
|
37
|
Tee SH. Mechanism diagrams and abstraction-by-aggregation. STUDIES IN HISTORY AND PHILOSOPHY OF BIOLOGICAL AND BIOMEDICAL SCIENCES 2018; 71:17-25. [PMID: 30318277 DOI: 10.1016/j.shpsc.2018.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 08/01/2018] [Accepted: 10/01/2018] [Indexed: 06/08/2023]
Abstract
Mechanism diagrams exhibit visually the organized parts and operations of a biological mechanism. A mechanism diagram can facilitate mechanistic research by providing a mechanistic explanation of the phenomenon of interest. Much research has been focusing on the mechanistic explanation and the explanatory mechanistic models. As a specific type of scientific diagram, a simple mechanism diagram can be explanatory by drawing on the rich explanatory resources of non-depicted background knowledge. The relationship between the visually depicted and the background knowledge is underexplored. It is unclear how the non-depicted background knowledge of a mechanism diagram contributes to providing a better-informed explanation of the phenomenon of interest in biological sciences. With the aim to explore this relationship, I articulate that a mechanism diagram provides a mechanistic explanation by a process called abstraction-by-aggregation. Through visual cues, the unified relevant background knowledge provides an epistemic access to a better-informed explanation.
Collapse
Affiliation(s)
- Sim-Hui Tee
- Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, 43900, Sepang, Selangor, Malaysia.
| |
Collapse
|
38
|
Xing Z, He Z, Wang S, Yan Y, Zhu H, Gao Y, Zhao Y, Zhang L. Ameliorative effects and possible molecular mechanisms of action of fibrauretine from Fibraurea recisa Pierre on d-galactose/AlCl 3-mediated Alzheimer's disease. RSC Adv 2018; 8:31646-31657. [PMID: 35548215 PMCID: PMC9085853 DOI: 10.1039/c8ra05356a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/26/2018] [Indexed: 12/23/2022] Open
Abstract
Fibrauretine is one of the main active ingredients from the rattan stems of Fibraurea recisa Pierre It exhibits a series of significant pharmacological effects. The present study aimed to evaluate the potential anti Alzheimer's disease (AD) effects of fibrauretine on a d-galactose/AlCl3-induced mouse model, and the underlying mechanisms of action were further investigated for the first time. Our results showed that pretreatment with fibrauretine significantly improved the ability of spatial short-term working memory in the model mice during the Y-maze test, as well as the abilities of spatial learning and memory during the Morris water maze. The levels of brain tissue amyloid (Aβ), P-Tau, Tau and acetylcholinesterase (AchE) were evidently increased in d-galactose/AlCl3-intoxicated mice, and these effects were reversed by fibrauretine. In contrast, a significant increase in the levels of the neurotransmitter acetylcholine (Ach) and choline acetyl transferase (ChAT) was observed in the fibrauretine-treated groups compared with the model group. Neuronal oxidative stress, evidenced by increased malondialdehyde (MDA) and nitric oxide (NO) levels and a decline in glutathione (GSH), catalase (CAT) and superoxide dismutase (SOD) activity, was significantly alleviated by fibrauretine pretreatment. The suppression of the neuroinflammatory response by fibrauretine was realized not only by the decrease in the levels of tumour necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the brain tissues and by the enzyme-linked immunosorbent assay (ELISA) but also by the protein expression levels of nuclear factor-κB (NF-κB), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS), which were measured by immunohistochemistry and western blotting. In addition, the protein expression levels of inflammatory factors interleukin-33 (IL-33) and ST2 in the brain tissues were detected by immunohistochemistry. Furthermore, the effects of western blotting demonstrated that the administration of fibrauretine significantly suppressed the protein expression levels of caspase-3, cleaved caspase-3, and Bax and increased the protein expression levels of Bcl-2, and the results of the H&E and TUNEL assay all suggested the inhibition of apoptosis in the neurons. The results clearly suggest that the underlying molecular mechanisms of action of the fibrauretine-mediated alleviation of d-galactose/AlCl3-induced Alzheimer's disease may involve antioxidant, anti-inflammatory, and anti-apoptotic effects.
Collapse
Affiliation(s)
- Zhiheng Xing
- Jilin Agricultural University Changchun 130118 Jilin China +86 431 84533358 +86 431 84533358
| | - Zhongmei He
- Jilin Agricultural University Changchun 130118 Jilin China +86 431 84533358 +86 431 84533358
- College of Chinese Medicinal Materials Changchun 130118 Jilin China
| | - Shuning Wang
- Jilin Agricultural University Changchun 130118 Jilin China +86 431 84533358 +86 431 84533358
| | - Yu Yan
- Jilin Agricultural University Changchun 130118 Jilin China +86 431 84533358 +86 431 84533358
| | - Hongyan Zhu
- Jilin Agricultural University Changchun 130118 Jilin China +86 431 84533358 +86 431 84533358
- College of Chinese Medicinal Materials Changchun 130118 Jilin China
| | - Yugang Gao
- Jilin Agricultural University Changchun 130118 Jilin China +86 431 84533358 +86 431 84533358
- College of Chinese Medicinal Materials Changchun 130118 Jilin China
| | - Yan Zhao
- Jilin Agricultural University Changchun 130118 Jilin China +86 431 84533358 +86 431 84533358
- College of Chinese Medicinal Materials Changchun 130118 Jilin China
| | - Lianxue Zhang
- Jilin Agricultural University Changchun 130118 Jilin China +86 431 84533358 +86 431 84533358
- College of Chinese Medicinal Materials Changchun 130118 Jilin China
| |
Collapse
|
39
|
Guha P, Gardell J, Darpolor J, Cunetta M, Lima M, Miller G, Espat NJ, Junghans RP, Katz SC. STAT3 inhibition induces Bax-dependent apoptosis in liver tumor myeloid-derived suppressor cells. Oncogene 2018; 38:533-548. [PMID: 30158673 DOI: 10.1038/s41388-018-0449-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 05/29/2018] [Accepted: 06/19/2018] [Indexed: 11/09/2022]
Abstract
Immunosuppressive myeloid-derived suppressor cells (MDSC) subvert antitumor immunity and limit the efficacy of chimeric antigen receptor T cells (CAR-T). Previously, we reported that the GM-CSF/JAK2/STAT3 axis drives liver-associated MDSC (L-MDSC) proliferation and blockade of this axis rescued antitumor immunity. We extended these findings in our murine liver metastasis (LM) model, by treating tumor-bearing mice with STAT3 inhibitors (STATTIC or BBI608) to further our understanding of how STAT3 drives L-MDSC suppressive function. STAT3 inhibition caused significant reduction of tumor burden as well as L-MDSC frequencies due to decrease in pSTAT3 levels. L-MDSC isolated from STATTIC or BBI608-treated mice had significantly reduced suppressive function. STAT3 inhibition of L-MDSC was associated with enhanced antitumor activity of CAR-T. Further investigation demonstrated activation of apoptotic signaling pathways in L-MDSC following STAT3 inhibition as evidenced by an upregulation of the pro-apoptotic proteins Bax, cleaved caspase-3, and downregulation of the anti-apoptotic protein Bcl-2. Accordingly, there was also a decrease of pro-survival markers, pErk and pAkt, and an increase in pro-death marker, Fas, with activation of downstream JNK and p38 MAPK. These findings represent a previously unrecognized link between STAT3 inhibition and Fas-induced apoptosis of MDSCs. Our findings suggest that inhibiting STAT3 has potential clinical application for enhancing the efficacy of CAR-T cells in LM through modulation of L-MDSC.
Collapse
Affiliation(s)
- Prajna Guha
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Jillian Gardell
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Josephine Darpolor
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Marissa Cunetta
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Matthew Lima
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - George Miller
- New York University School of Medicine, New York, NY, USA
| | - N Joseph Espat
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA.,Department of Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Richard P Junghans
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Steven C Katz
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA. .,Department of Surgery, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
40
|
Yabasin IB, Sanches JGP, Ibrahim MM, Huidan J, Williams W, Lu ZL, Wen Q. Cisatracurium Retards Cell Migration and Invasion Upon Upregulation of p53 and Inhibits the Aggressiveness of Colorectal Cancer. Front Physiol 2018; 9:941. [PMID: 30108509 PMCID: PMC6079220 DOI: 10.3389/fphys.2018.00941] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is reported to be the third and fourth, most diagnosed and cause of cancer associated deaths respectively. In 2012 for instance, about 1.4 million new cases were reported, and approximately 700,000 deaths recorded. Survival from CRC is dependent on the stage at which it is diagnosed coupled with appropriate surgical and medical intervention. Cisatracurium is widely used for skeletal muscle relaxation during abdominal surgeries, including bowel and colon surgeries. Recent studies reported that cisatracurium inhibits progression of human cancer cells, however, the mechanisms leading to the inhibition are yet to be completely understood. To elucidate mechanisms resulting particularly in tumor cell growth and metastasis, we developed ex vivo and in in vivo xenograft models of CRC. Cisatracurium caused upregulation of p53 and its down-stream genes and proteins known to regulate proliferation and metastasis in vitro and in vivo. Genomic analyses of CRC following cisatracurium treatment revealed moderate to high DNA damage, while functional analyses demonstrated significant tumor cells growth regression, as well as repression of migration and invasion. Importantly, cisatracurium increased E-Cadherin and CALD-1 but decreased SNAI-1 and SLUG levels in vitro and in vivo. Together, the findings demonstrate that elevation of p53 upon cisatracurium-induced genomic injury, represent a potential mechanism by which cisatracurium result in the suppression of CRC progression and metastasis.
Collapse
Affiliation(s)
- Iddrisu B Yabasin
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | | | - Mohammed M Ibrahim
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Jin Huidan
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Walana Williams
- Department of Microbiology and Immunology, Dalian Medical University, Dalian, China
| | - Zhi-Li Lu
- Department of Ophthalmology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qingping Wen
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
41
|
Abbastabar M, Kheyrollah M, Azizian K, Bagherlou N, Tehrani SS, Maniati M, Karimian A. Multiple functions of p27 in cell cycle, apoptosis, epigenetic modification and transcriptional regulation for the control of cell growth: A double-edged sword protein. DNA Repair (Amst) 2018; 69:63-72. [PMID: 30075372 DOI: 10.1016/j.dnarep.2018.07.008] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 07/19/2018] [Accepted: 07/19/2018] [Indexed: 01/27/2023]
Abstract
The cell cycle is controlled by precise mechanisms to prevent malignancies such as cancer, and the cell needs these tight and advanced controls. Cyclin dependent kinase inhibitor p27 (also known as KIP1) is a factor that inhibits the progression of the cell cycle by using specific molecular mechanisms. The inhibitory effect of p27 on the cell cycle is mediated by CDKs inhibition. Other important functions of p27 include cell proliferation, cell differentiation and apoptosis. Post- translational modification of p27 by phosphorylation and ubiquitination respectively regulates interaction between p27 and cyclin/CDK complex and degradation of p27. In this review, we focus on the multiple function of p27 in cell cycle regulation, apoptosis, epigenetic modifications and post- translational modification, and briefly discuss the mechanisms and factors that have important roles in p27 functions.
Collapse
Affiliation(s)
- Maryam Abbastabar
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Kheyrollah
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Khalil Azizian
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Nazanin Bagherlou
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Sadra Samavarchi Tehrani
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Mahmood Maniati
- Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ansar Karimian
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Cancer & Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran; Student Research Committee, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
42
|
Sharma VK, Raimondi V, Ruggero K, Pise-Masison CA, Cavallari I, Silic-Benussi M, Ciminale V, D'Agostino DM. Expression of miR-34a in T-Cells Infected by Human T-Lymphotropic Virus 1. Front Microbiol 2018; 9:832. [PMID: 29780367 PMCID: PMC5945834 DOI: 10.3389/fmicb.2018.00832] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/12/2018] [Indexed: 12/19/2022] Open
Abstract
Human T-lymphotropic virus 1 (HTLV-1) immortalizes T-cells and is the causative agent of adult T-cell leukemia/lymphoma (ATLL). HTLV-1 replication and transformation are governed by multiple interactions between viral regulatory proteins and host cell factors that remain to be fully elucidated. The present study investigated the impact of HTLV-1 infection on the expression of miR-34a, a microRNA whose expression is downregulated in many types of cancer. Results of RT-PCR assays showed that five out of six HTLV-1-positive cell lines expressed higher levels of miR-34a compared to normal PBMC or purified CD4+ T-cells. ATLL cell line ED, which did not express miR-34a, showed methylation of the miR-34a promoter. Newly infected PBMC and samples from 10 ATLL patients also showed a prominent increase in miR-34a expression compared to PBMC controls. The primary miR-34a transcript expressed in infected cell line C91PL contained binding motifs for NF-κB and p53. Pharmacological inhibition of NF-κB with Bay 11-7082 indicated that this pathway contributes to sustain miR-34a levels in infected cells. Treatment of infected cell lines with the p53 activator nutlin-3a resulted in a further increase in miR-34a levels, thus confirming it as a transcriptional target of p53. Nutlin-3a-treated cells showed downregulation of known miR-34a targets including the deacetylase SIRT1, which was accompanied by increased acetylation of p53, a substrate of SIRT1. Transfection of C91PL cells with a miR-34a mimic also led to downregulation of mRNA targets including SIRT1 as well as the pro-apoptotic factor BAX. Unlike nutlin-3a, the miR-34a mimic did not cause cell cycle arrest or reduce cell viability. On the other hand, sequestration of miR-34a with a sponge construct resulted in an increase in death of C91PL cells. These findings provide evidence for a functional role for miR-34a in fine-tuning the expression of target genes that influence the turnover of HTLV-1-infected cells.
Collapse
Affiliation(s)
- Varun K Sharma
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | | - Katia Ruggero
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Cynthia A Pise-Masison
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | | | | | - Vincenzo Ciminale
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy.,Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | | |
Collapse
|
43
|
Ozer E, Canda MS, Ulukus C, Guray M, Erbayraktar S. Expression of BCL-2, BAX and P53 Proteins in Pituitary Adenomas an Immunohistochemical Study. TUMORI JOURNAL 2018; 89:54-9. [PMID: 12729363 DOI: 10.1177/030089160308900112] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and Background Although pituitary adenomas are usually benign lesions, their growth rate is highly variable and unpredictable. Apoptosis appears to be an important process in neoplastic lesions. The purpose of this study was to investigate the expression of apoptosis-related proteins including Bcl-2, bax and p53 in pituitary adenomas and its correlation with hormone function, tumor size, local control, and proliferative activity. Study Design The expression of Bcl-2, Bax and p53 proteins and hormonal function were determined in formalin-fixed, paraffin-embedded tissue from 41 untreated pituitary adenomas using immunohistochemistry. The patients were followed for a median of 60 months (range, 12 to 95). Patient charts were reviewed to record tumor recurrence and size. Tumor proliferative activity was assessed by immunohistochemistry using Ki-67 antibody. Results Of 41 pituitary adenomas, 26 (63%) were hormone-secreting and 15 (37%) non-functioning, 34 (83%) were macroadenoma and 7 (17%) microadenoma, and 15 (37%) showed local relapse. Six (14%) adenomas were of low proliferative activity, whereas the others (86%) were non-proliferative. Immunohistochemically, 31 adenomas (75%) showed bcl-2 positivity, 37 (90%) bax positivity, and 7 (17%) p53 positivity. Statistical analysis revealed that Bcl-2 protein expression significantly diminished in prolactin-secreting and non-functioning adenomas (P = 0.005 and P = 0.006, respectively), and increased in growth hormone-secreting adenomas (P = 0.003). In addition, expression of bax protein significantly decreased in recurrent tumors, in contrast to p53 protein, which showed a significant increase (P = 0.03 and P = 0.002, respectively). Conclusions We think that apoptosis-related proteins such as Bcl-2, Bax and p53 may be significantly related to hormone function and local control in pituitary adenomas.
Collapse
Affiliation(s)
- Erdener Ozer
- Department of Pathology, Dokuz Eylul University, School of Medicine, Inciralti, Izmir, Turkey.
| | | | | | | | | |
Collapse
|
44
|
Ryu H, Nam KY, Kim JS, Hwang SG, Song JY, Ahn J. The small molecule AU14022 promotes colorectal cancer cell death via p53-mediated G2/M-phase arrest and mitochondria-mediated apoptosis. J Cell Physiol 2018; 233:4666-4676. [DOI: 10.1002/jcp.26234] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/05/2017] [Indexed: 11/10/2022]
Affiliation(s)
- Hwani Ryu
- Division of Radiation Cancer Research; Korea Institute of Radiological and Medical Sciences; Seoul Korea
| | - Ky-Youb Nam
- Research Center; Pharos I&BT Co., Ltd.; Anyang Korea
| | - Jae Sung Kim
- Division of Radiation Cancer Research; Korea Institute of Radiological and Medical Sciences; Seoul Korea
| | - Sang-Gu Hwang
- Division of Radiation Cancer Research; Korea Institute of Radiological and Medical Sciences; Seoul Korea
| | - Jie-Young Song
- Division of Radiation Cancer Research; Korea Institute of Radiological and Medical Sciences; Seoul Korea
| | - Jiyeon Ahn
- Division of Radiation Cancer Research; Korea Institute of Radiological and Medical Sciences; Seoul Korea
| |
Collapse
|
45
|
Kinzel L, Ernst A, Orth M, Albrecht V, Hennel R, Brix N, Frey B, Gaipl US, Zuchtriegel G, Reichel CA, Blutke A, Schilling D, Multhoff G, Li M, Niyazi M, Friedl AA, Winssinger N, Belka C, Lauber K. A novel HSP90 inhibitor with reduced hepatotoxicity synergizes with radiotherapy to induce apoptosis, abrogate clonogenic survival, and improve tumor control in models of colorectal cancer. Oncotarget 2017; 7:43199-43219. [PMID: 27259245 PMCID: PMC5190018 DOI: 10.18632/oncotarget.9774] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 05/24/2016] [Indexed: 12/20/2022] Open
Abstract
The chaperone heat shock protein 90 (HSP90) crucially supports the maturation, folding, and stability of a variety of client proteins which are of pivotal importance for the survival and proliferation of cancer cells. Consequently, targeting of HSP90 has emerged as an attractive strategy of anti-cancer therapy, and it appears to be particularly effective in the context of molecular sensitization towards radiotherapy as has been proven in preclinical models of different cancer entities. However, so far the clinical translation has largely been hampered by suboptimal pharmacological properties and serious hepatotoxicity of first- and second-generation HSP90 inhibitors. Here, we report on NW457, a novel radicicol-derived member of the pochoxime family with reduced hepatotoxicity, how it inhibits the DNA damage response and how it synergizes with ionizing irradiation to induce apoptosis, abrogate clonogenic survival, and improve tumor control in models of colorectal cancer in vitro and in vivo.
Collapse
Affiliation(s)
- Linda Kinzel
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Anne Ernst
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Valerie Albrecht
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Roman Hennel
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nikko Brix
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gabriele Zuchtriegel
- Department of Otorhinolaryngology, Head and Neck Surgery, and Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christoph A Reichel
- Department of Otorhinolaryngology, Head and Neck Surgery, and Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology at the Center for Clinical Veterinary Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Minglun Li
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Anna A Friedl
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nicolas Winssinger
- Department of Organic Chemistry, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Claus Belka
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
46
|
Toraih EA, Aly NM, Abdallah HY, Al-Qahtani SA, Shaalan AA, Hussein MH, Fawzy MS. MicroRNA-target cross-talks: Key players in glioblastoma multiforme. Tumour Biol 2017; 39:1010428317726842. [PMID: 29110584 DOI: 10.1177/1010428317726842] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The role of microRNAs in brain cancer is still naive. Some act as oncogene and others as tumor suppressors. Discovery of efficient biomarkers is mandatory to debate that aggressive disease. Bioinformatically selected microRNAs and their targets were investigated to evaluate their putative signature as diagnostic and prognostic biomarkers in primary glioblastoma multiforme. Expression of a panel of seven microRNAs (hsa-miR-34a, hsa-miR-16, hsa-miR-17, hsa-miR-21, hsa-miR-221, hsa-miR-326, and hsa-miR-375) and seven target genes ( E2F3, PI3KCA, TOM34, WNT5A, PDCD4, DFFA, and EGFR) in 43 glioblastoma multiforme specimens were profiled compared to non-cancer tissues via quantitative reverse transcription-polymerase chain reaction. Immunohistochemistry staining for three proteins (VEGFA, BAX, and BCL2) was performed. Gene enrichment analysis identified the biological regulatory functions of the gene panel in glioma pathway. MGMT ( O-6-methylguanine-DNA methyltransferase) promoter methylation was analyzed for molecular subtyping of tumor specimens. Our data demonstrated a significant upregulation of five microRNAs (hsa-miR-16, hsa-miR-17, hsa-miR-21, hsa-miR-221, and hsa-miR-375), three genes ( E2F3, PI3KCA, and Wnt5a), two proteins (VEGFA and BCL2), and downregulation of hsa-miR-34a and three other genes ( DFFA, PDCD4, and EGFR) in brain cancer tissues. Receiver operating characteristic analysis revealed that miR-34a (area under the curve = 0.927) and miR-17 (area under the curve = 0.900) had the highest diagnostic performance, followed by miR-221 (area under the curve = 0.845), miR-21 (area under the curve = 0.836), WNT5A (area under the curve = 0.809), PDCD4 (area under the curve = 0.809), and PI3KCA (area under the curve = 0.800). MGMT promoter methylation status was associated with high miR-221 levels. Moreover, patients with VEGFA overexpression and downregulation of TOM34 and BAX had poor overall survival. Nevertheless, miR-17, miR-221, and miR-326 downregulation were significantly associated with high recurrence rate. Multivariate analysis by hierarchical clustering classified patients into four distinct groups based on gene panel signature. In conclusion, the explored microRNA-target dysregulation could pave the road toward developing potential therapeutic strategies for glioblastoma multiforme. Future translational and functional studies are highly recommended to better understand the complex bio-molecular signature of this difficult-to-treat tumor.
Collapse
Affiliation(s)
- Eman Ali Toraih
- 1 Genetics Unit, Histology and Cell Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Nagwa Mahmoud Aly
- 2 Department of Medical Biochemistry, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Hoda Y Abdallah
- 1 Genetics Unit, Histology and Cell Biology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Saeed Awad Al-Qahtani
- 3 Department of Physiology, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Aly Am Shaalan
- 4 Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.,5 Department of Anatomy and Histology, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | | | - Manal Said Fawzy
- 2 Department of Medical Biochemistry, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.,7 Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
47
|
Kaiser AM, Attardi LD. Deconstructing networks of p53-mediated tumor suppression in vivo. Cell Death Differ 2017; 25:93-103. [PMID: 29099489 DOI: 10.1038/cdd.2017.171] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/18/2017] [Accepted: 08/31/2017] [Indexed: 02/07/2023] Open
Abstract
The transcription factor p53 is a vital tumor suppressor. Upon activation by diverse stresses including oncogene activation, DNA damage, hypoxia and nutrient deprivation, p53 activates a panoply of target genes and orchestrates numerous downstream responses that suppress tumorigenesis. Although early studies of p53 suggested that its ability to induce cell cycle arrest, senescence and apoptosis programs accounted for its tumor-suppressor activity, more recent studies have challenged this notion. Moreover, p53 regulates a suite of additional processes, such as metabolism, stem cell function, invasion and metastasis. The processes p53 coordinately regulates to enact tumor suppression, and how such regulation occurs, thus remain elusive. In this review, we will summarize our current knowledge of p53-mediated tumor-suppressive mechanisms gleaned from in vivo studies in mouse models.
Collapse
Affiliation(s)
- Alyssa M Kaiser
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura D Attardi
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
48
|
Zheng B, Wang J, Pan H, Chen H, Ji W, Liao Z, Gong X, Wang H, Chang J. A visual guide to gene/optothermal synergy therapy nanosystem using tungsten oxide. J Colloid Interface Sci 2017; 506:460-470. [DOI: 10.1016/j.jcis.2017.07.077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 11/17/2022]
|
49
|
Aberrant expression of translationally controlled tumor protein (TCTP) can lead to radioactive susceptibility and chemosensitivity in lung cancer cells. Oncotarget 2017; 8:101922-101935. [PMID: 29254214 PMCID: PMC5731924 DOI: 10.18632/oncotarget.21747] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/29/2017] [Indexed: 01/15/2023] Open
Abstract
Translationally controlled tumor protein (TCTP) is an evolutionally highly conserved protein which has been implicated as a biomarker for cancer cell reversion although the mechanism is not very clear. This makes it a potential target for cancer therapy. P53 tumor suppressor protein is important in regulating cell growth, it can induce either growth arrest or programmed cell death (apoptosis). TCTP and P53 has been reported that can regulate the protein level of each other. Here we proved that TCTP is a malignancy state keeper in lung cancer and lower level of TCTP protein made cells more sensitive to stressful condition. No obvious difference has been observed from wildtype and the TCTP knockdown lung cancer cells (A549) when located in the normal circumstances. While under the stressful condition, the existence of higher protein level of TCTP can protect cells from apoptosis. TCTP and P53 formed a feedback signal pathway and through it to regulate the downstream Akt signal pathways to make the lung cancer cells keep a higher metabolism level and protect cancer cells from apoptosis induced by outside stress.
Collapse
|
50
|
Zhang C, Chen W, Zhang X, Zheng Y, Yu F, Liu Y, Wang Y. Grape seed proanthocyanidins induce mitochondrial pathway-mediated apoptosis in human colorectal carcinoma cells. Oncol Lett 2017; 14:5853-5860. [PMID: 29113217 PMCID: PMC5661607 DOI: 10.3892/ol.2017.6992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 08/17/2017] [Indexed: 01/22/2023] Open
Abstract
Grape seed proanthocyanidins (GSPs) have been reported to possess a wide array of pharmacological and biochemical properties. Recently, GSPs have been reported to inhibit various types of colorectal cancer; however, the mechanism(s) involved remain unclear. The present study investigated the effects of GSPs on HCT-116 human colorectal carcinoma cell line. Exposure of these cells to GSPs for 48 h resulted in a significant concentration-dependent inhibition of cell viability. Further investigation indicated that GSPs induced apoptosis of these cells. Analyses of mRNA expression levels using reverse transcription-quantitative polymerase chain reaction and protein expression levels by western blotting revealed that this was associated with increased expression levels of p53 tumor suppressor protein, cytochrome c, and pro-apoptotic proteins, apoptosis regulator Bax (Bax) and Bcl-2 homologous antagonist/killer. Furthermore, decreased expression levels of the anti-apoptotic protein, B cell lymphoma-2 and activation of caspase-2, caspase-3 and caspase-9 were demonstrated. GSP-induced loss of mitochondrial membrane potential was also detected by JC-1 assay. These findings suggested that GSPs induced colon cancer cell apoptosis via the mitochondrial signaling pathway. This provided evidence indicating that GSPs may provide potential chemotherapeutic agents for colorectal cancer.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China.,Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, Fujian 361021, P.R. China
| | - Weili Chen
- Department of Chemistry and Physics, Heihe University, Heihe, Heilongjiang 164300, P.R. China
| | - Xuhao Zhang
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin 130031, P.R. China
| | - Yanbing Zheng
- Department of Chemistry and Physics, Heihe University, Heihe, Heilongjiang 164300, P.R. China
| | - Fengli Yu
- Department of Chemistry and Physics, Heihe University, Heihe, Heilongjiang 164300, P.R. China
| | - Yulong Liu
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|