1
|
Zhu Z, Feng S, Zeng A, Song L. Advances in Palmitoylation: A key Regulator of liver cancer development and therapeutic targets. Biochem Pharmacol 2025; 234:116810. [PMID: 39978688 DOI: 10.1016/j.bcp.2025.116810] [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: 10/14/2024] [Revised: 02/06/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
Liver cancer ranks as the second leading cause of cancer-related deaths globally, which remains a significant public health concern. The development of liver cancer is associated with several signaling pathways, particularly metabolic reprogramming. Protein S-palmitoylation, a type of lipid post-translational modification (PTM), involves the reversible attachment of palmitic acid to a cysteine residue through a thioester bond. This modification is found in a wide range of proteins, including enzymes, cancer promoters, tumor suppressors, and transcription factors. The palmitoylation process is catalyzed by the zinc finger DHHC-type containing (ZDHHC) protein family, while the reverse process, depalmitoylation, is facilitated by palmitoyl-protein thioesterases (PPTs). Dysregulation of palmitoylation has been linked to various cancer hallmark functions, cancer metabolism, and tumor microenvironment regulation. Currently, membrane palmitoylated protein (MPP) and PPT1 have been identified as prognostic markers and potential therapeutic targets in liver cancer. In this review, we summarize recent advances in understanding the effects of palmitoylation on liver cancer development, metastasis, and prognosis prediction, and explore potential therapeutic strategies for managing liver cancer.
Collapse
Affiliation(s)
- Zilong Zhu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China
| | - Shenghui Feng
- Intensive Care Unit, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan 610041, PR China.
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China.
| |
Collapse
|
2
|
Nie AY, Xiao ZH, Deng JL, Li N, Hao LY, Li SH, Hu XY. Bidirectional regulation of the cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon gene pathway and its impact on hepatocellular carcinoma. World J Gastrointest Oncol 2025; 17:98556. [PMID: 39958554 PMCID: PMC11755995 DOI: 10.4251/wjgo.v17.i2.98556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) ranks as the fourth leading cause of cancer-related deaths in China, and the treatment options are limited. The cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) activates the stimulator of interferon gene (STING) signaling pathway as a crucial immune response pathway in the cytoplasm, which detects cytoplasmic DNA to regulate innate and adaptive immune responses. As a potential therapeutic target, cGAS-STING pathway markedly inhibits tumor cell proliferation and metastasis, with its activation being particularly relevant in HCC. However, prolonged pathway activation may lead to an immunosuppressive tumor microenvironment, which fostering the invasion or metastasis of liver tumor cells. AIM To investigate the dual-regulation mechanism of cGAS-STING in HCC. METHODS This review was conducted according to the PRISMA guidelines. The study conducted a comprehensive search for articles related to HCC on PubMed and Web of Science databases. Through rigorous screening and meticulous analysis of the retrieved literature, the research aimed to summarize and elucidate the impact of the cGAS-STING pathway on HCC tumors. RESULTS All authors collaboratively selected studies for inclusion, extracted data, and the initial search of online databases yielded 1445 studies. After removing duplicates, the remaining 964 records were screened. Ultimately, 55 articles met the inclusion criteria and were included in this review. CONCLUSION Acute inflammation can have a few inhibitory effects on cancer, while chronic inflammation generally promotes its progression. Extended cGAS-STING pathway activation will result in a suppressive tumor microenvironment.
Collapse
Affiliation(s)
- Ai-Yu Nie
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Zhong-Hui Xiao
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Jia-Li Deng
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Na Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Li-Yuan Hao
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Sheng-Hao Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Xiao-Yu Hu
- Department of Infection, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, Sichuan Province, China
| |
Collapse
|
3
|
Vona R, Cittadini C, Ortona E, Matarrese P. Sex Disparity in Cancer: Role of Autophagy and Estrogen Receptors. Cells 2025; 14:273. [PMID: 39996745 PMCID: PMC11854201 DOI: 10.3390/cells14040273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/24/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Autophagy, a cellular process essential for maintaining homeostasis, plays a fundamental role in recycling damaged components and in adapting to stress. The dysregulation of autophagy is implicated in numerous human diseases, including cancer, where it exhibits a dual role as both a suppressor and a promoter, depending on the context and the stage of tumor development. The significant sex differences observed in autophagic processes are determined by biological factors, such as genetic makeup and sex hormones. Estrogens, through their interaction with specific receptors, modulate autophagy and influence tumor progression, therapy resistance, and the immune response to tumors. In females, the escape from X inactivation and estrogen signaling may be responsible for the advantages, in terms of lower incidence and longer survival, observed in oncology. Women often show better responses to traditional chemotherapy, while men respond better to immunotherapy. The action of sex hormones on the immune system could contribute to these differences. However, women experience more severe adverse reactions to anticancer drugs. The estrogen/autophagy crosstalk-involved in multiple aspects of the tumor, i.e., development, progression and the response to therapy-deserves an in-depth study, as it could highlight sex-specific mechanisms useful for designing innovative and gender-tailored treatments from the perspective of precision medicine.
Collapse
Affiliation(s)
- Rosa Vona
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| | | | | | - Paola Matarrese
- Center for Gender-Specific Medicine, National Institute of Health, 00161 Rome, Italy; (C.C.); (E.O.)
| |
Collapse
|
4
|
Dwyer S, Ruth J, Seidel HE, Raz AA, Chodosh LA. Autophagy is required for mammary tumor recurrence by promoting dormant tumor cell survival following therapy. Breast Cancer Res 2024; 26:143. [PMID: 39425240 PMCID: PMC11488247 DOI: 10.1186/s13058-024-01878-7] [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: 03/06/2024] [Accepted: 08/02/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Mortality from breast cancer is principally due to tumor recurrence. Recurrent breast cancers arise from the pool of residual tumor cells, termed minimal residual disease, that survive treatment and may exist in a dormant state for 20 years or more following treatment of the primary tumor. As recurrent breast cancer is typically incurable, understanding the mechanisms underlying dormant tumor cell survival is a critical priority in breast cancer research. The importance of this goal is further underscored by emerging evidence suggesting that targeting dormant residual tumor cells in early-stage breast cancer patients may be a means to prevent tumor recurrence and its associated mortality. In this regard, the role of autophagy in dormant tumor cell survival and recurrence remains unresolved, with conflicting reports of both pro-survival/recurrence-promoting and pro-death/recurrence-suppressing effects of autophagy inhibition in dormant tumor cells. Resolving this question has important clinical implications. METHODS We used genetically engineered mouse models that faithfully recapitulate key features of human breast cancer progression, including minimal residual disease, tumor dormancy, and recurrence. We used genetic and pharmacological approaches to inhibit autophagy, including treatment with chloroquine, genetic knockdown of ATG5 or ATG7, or deletion of BECN and determined their effects on dormant tumor cell survival and recurrence. RESULTS We demonstrate that the survival and recurrence of dormant mammary tumor cells following therapy is dependent upon autophagy. We find that autophagy is induced in vivo following HER2 downregulation and remains activated in dormant residual tumor cells. Using genetic and pharmacological approaches we show that inhibiting autophagy by chloroquine administration, ATG5 or ATG7 knockdown, or deletion of a single allele of the tumor suppressor Beclin 1 is sufficient to inhibit mammary tumor recurrence, and that autophagy inhibition results in the death of dormant mammary tumor cells in vivo. CONCLUSIONS Our findings demonstrate a pro-tumorigenic role for autophagy in tumor dormancy and recurrence following therapy, reveal that dormant tumor cells are uniquely reliant upon autophagy for their survival, and indicate that targeting dormant residual tumor cells by inhibiting autophagy impairs tumor recurrence. These studies identify a pharmacological target for a cellular state that is resistant to commonly used anti-neoplastic agents and suggest autophagy inhibition as an approach to reduce dormant minimal residual disease in order to prevent lethal tumor recurrence.
Collapse
Affiliation(s)
- Samantha Dwyer
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Room 614 BRB II/III, 421 Curie Boulevard, Philadelphia, PA, 19104-6160, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason Ruth
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Room 614 BRB II/III, 421 Curie Boulevard, Philadelphia, PA, 19104-6160, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hans E Seidel
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Room 614 BRB II/III, 421 Curie Boulevard, Philadelphia, PA, 19104-6160, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Amelie A Raz
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Room 614 BRB II/III, 421 Curie Boulevard, Philadelphia, PA, 19104-6160, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lewis A Chodosh
- Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Room 614 BRB II/III, 421 Curie Boulevard, Philadelphia, PA, 19104-6160, USA.
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
Wang M, Liao J, Wang J, Xu M, Cheng Y, Wei L, Huang A. HDAC2 promotes autophagy-associated HCC malignant progression by transcriptionally activating LAPTM4B. Cell Death Dis 2024; 15:593. [PMID: 39147759 PMCID: PMC11327261 DOI: 10.1038/s41419-024-06981-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 08/17/2024]
Abstract
Hepatocellular carcinoma (HCC) is a significant global health challenge. The activation of autophagy plays an essential role in promoting the proliferation and survival of cancer cells. However, the upstream regulatory network and mechanisms governing autophagy in HCC remain unclear. This study demonstrated that histone deacetylase 2 (HDAC2) regulates autophagy in HCC. Its expression was elevated in HCC tissues, and high HDAC2 expression was strongly associated with poor prognosis in individuals with HCC. Integrated in vitro and in vivo investigations confirmed that HDAC2 promotes autophagy and autophagy-related malignant progression in HCC. Mechanistically, HDAC2 bound specifically to the lysosome-associated protein transmembrane 4-β (LAPTM4B) promoter at four distinct binding sites, enhancing its transcriptional activation and driving autophagy-related malignant progression in HCC. These findings establish LAPTM4B as a direct target gene of HDAC2. Furthermore, the selective inhibitor of HDAC2 effectively alleviated the malignant development of HCC. In addition, multivariate Cox regression analysis of 105 human HCC samples revealed that HDAC2 expression is an independent predictor of HCC prognosis. This study underscores the crucial role of the HDAC2-LAPTM4B axis in regulating autophagy in the malignant evolution of HCC and highlights the potential of targeting HDAC2 to prevent and halt the malignant progression of HCC.
Collapse
Affiliation(s)
- Meifeng Wang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
| | - Jianping Liao
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
| | - Jie Wang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
| | - Meifang Xu
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
| | - Ye Cheng
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China.
| | - Aimin Huang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China.
- Institute of Oncology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, Fujian, 350004, China.
| |
Collapse
|
6
|
Li Y, Luo B, Lin X, Bai D, Li L, Gao D, Li X, Zhong X, Wei Y, Yang L, Zhu X, Han L, Tian H, Zhang R, Wang P. 20(R)-Panaxatriol enhances METTL3-mediated m 6A modification of STUB1 to inhibit autophagy and exert antitumor effects in Triple-Negative Breast Cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155537. [PMID: 38823344 DOI: 10.1016/j.phymed.2024.155537] [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: 10/21/2023] [Revised: 01/31/2024] [Accepted: 03/13/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Aberrant activation of autophagy in triple-negative breast cancer (TNBC) has led researchers to investigate potential therapeutic strategies targeting this process. The regulation of autophagy is significantly influenced by METTL3. Our previous research has shown that the Panax ginseng-derived compound, 20(R)-panaxatriol (PT), has potential as an anti-tumor agent. However, it remains unclear whether PT can modulate autophagy through METTL3 to exert its anti-tumor effects. OBJECTIVE Our objective is to investigate whether PT can regulate autophagy in TNBC cells and elucidate the molecular mechanisms. STUDY DESIGN For in vitro experiments, we employed SUM-159-PT and MDA-MB-231 cells. While in vivo experiments involved BALB/c nude mice and NOD/SCID mice. METHODS In vitro, TNBC cells were treated with PT, and cell lines with varying expression levels of METTL3 were established. We assessed the impact on tumor cell activity and autophagy by analyzing autophagic flux, Western Blot (WB), and methylation levels. In vivo, subcutaneous transplantation models were established in BALB/c nude and NOD/SCID mice to observe the effect of PT on TNBC growth. HE staining and immunofluorescence were employed to analyze histopathological changes in tumor tissues. MeRIP-seq and dual-luciferase reporter gene assays were used to identify key downstream targets. Additionally, the silencing of STIP1 Homology And U-Box Containing Protein 1 (STUB1) explored PT's effects. The mechanism of PT's action on STUB1 via METTL3 was elucidated through mRNA stability assays, mRNA alternative splicing analysis, and nuclear-cytoplasmic mRNA separation. RESULTS In both in vivo and in vitro experiments, it was discovered that PT significantly upregulates the expression of METTL3, leading to autophagy inhibition and therapeutic effects in TNBC. Simultaneously, through MeRIP-seq analysis and dual-luciferase reporter gene assays, we have demonstrated that PT modulates STUB1 via METTL3, influencing autophagy in TNBC cells. Furthermore, intriguingly, PT extends the half-life of STUB1 mRNA by enhancing its methylation modification, thereby enhancing its stability. CONCLUSION In summary, our research reveals that PT increases STUB1 m6A modification through a METTL3-mediated mechanism in TNBC cells, inhibiting autophagy and further accentuating its anti-tumor properties. Our study provides novel mechanistic insights into TNBC pathogenesis and potential drug targets for TNBC.
Collapse
Affiliation(s)
- Yan Li
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Bingjie Luo
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Xuan Lin
- The 8th Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan 528000, PR China
| | - Donghui Bai
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Lingyu Li
- Cancer Research Institute, Jinan University, Guangzhou, Guangdong 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong,510630, PR China; College of Pharmacy, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Duan Gao
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Xiaoyun Li
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Xianxun Zhong
- The 8th Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan 528000, PR China
| | - Yaru Wei
- The 8th Clinical Medical College of Guangzhou University of Chinese Medicine, Foshan 528000, PR China
| | - Li Yang
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong,510630, PR China; College of Pharmacy, Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Xiaofeng Zhu
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong,510630, PR China
| | - Li Han
- First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Huaqin Tian
- Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong 528000, PR China.
| | - Ronghua Zhang
- Cancer Research Institute, Jinan University, Guangzhou, Guangdong 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong,510630, PR China; College of Pharmacy, Jinan University, Guangzhou, Guangdong 510630, PR China.
| | - Panpan Wang
- Cancer Research Institute, Jinan University, Guangzhou, Guangdong 510630, PR China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Jinan University, Guangzhou, Guangdong,510630, PR China; First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, PR China.
| |
Collapse
|
7
|
Yang Y, Liu X, Yang D, Li L, Li S, Lu S, Li N. Interplay of CD36, autophagy, and lipid metabolism: insights into cancer progression. Metabolism 2024; 155:155905. [PMID: 38548128 DOI: 10.1016/j.metabol.2024.155905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/17/2024] [Accepted: 03/23/2024] [Indexed: 04/05/2024]
Abstract
CD36, a scavenger receptor B2 that is dynamically distributed between cell membranes and organelle membranes, plays a crucial role in regulating lipid metabolism. Abnormal CD36 activity has been linked to a range of metabolic disorders, such as obesity, nonalcoholic fatty liver disease, insulin resistance and cardiovascular disease. CD36 undergoes various modifications, including palmitoylation, glycosylation, and ubiquitination, which greatly affect its binding affinity to various ligands, thereby triggering and influencing various biological effects. In the context of tumors, CD36 interacts with autophagy to jointly regulate tumorigenesis, mainly by influencing the tumor microenvironment. The central role of CD36 in cellular lipid homeostasis and recent molecular insights into CD36 in tumor development indicate the applicability of CD36 as a therapeutic target for cancer treatment. Here, we discuss the diverse posttranslational modifications of CD36 and their respective roles in lipid metabolism. Additionally, we delve into recent research findings on CD36 in tumors, outlining ongoing drug development efforts targeting CD36 and potential strategies for future development and highlighting the interplay between CD36 and autophagy in the context of cancer. Our aim is to provide a comprehensive understanding of the function of CD36 in both physiological and pathological processes, facilitating a more in-depth analysis of cancer progression and a better development and application of CD36-targeting drugs for tumor therapy in the near future.
Collapse
Affiliation(s)
- Yuxuan Yang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaokun Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Di Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lianhui Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Sheng Li
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Sen Lu
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Ning Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
8
|
Renna FJ, Gonzalez CD, Vaccaro MI. Decoding the Versatile Landscape of Autophagic Protein VMP1 in Cancer: A Comprehensive Review across Tissue Types and Regulatory Mechanisms. Int J Mol Sci 2024; 25:3758. [PMID: 38612567 PMCID: PMC11011780 DOI: 10.3390/ijms25073758] [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: 02/26/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Autophagy, a catabolic process orchestrating the degradation of proteins and organelles within lysosomes, is pivotal for maintaining cellular homeostasis. However, its dual role in cancer involves preventing malignant transformation while fostering progression and therapy resistance. Vacuole Membrane Protein 1 (VMP1) is an essential autophagic protein whose expression, per se, triggers autophagy, being present in the whole autophagic flux. In pancreatic cancer, VMP1-whose expression is linked to the Kirsten Rat Sarcoma Virus (KRAS) oncogene-significantly contributes to disease promotion, progression, and chemotherapy resistance. This investigation extends to breast cancer, colon cancer, hepatocellular carcinoma, and more, highlighting VMP1's nuanced nature, contingent on specific tissue contexts. The examination of VMP1's interactions with micro-ribonucleic acids (miRNAs), including miR-21, miR-210, and miR-124, enhances our understanding of its regulatory network in cancer. Additionally, this article discusses VMP1 gene fusions, especially with ribosomal protein S6 kinase B1 (RPS6KB1), shedding light on potential implications for tumor malignancy. By deciphering the molecular mechanisms linking VMP1 to cancer progression, this exploration paves the way for innovative therapeutic strategies to disrupt these pathways and potentially improve treatment outcomes.
Collapse
Affiliation(s)
- Felipe J. Renna
- Instituto de Bioquimica y Medicina Molecular Prof Alberto Boveris (IBIMOL), CONICET, Universidad de Buenos Aires, Buenos Aires C1113AAC, Argentina;
| | - Claudio D. Gonzalez
- Instituto de Investigaciones, IUC, Medicina Traslacional, Hospital Universitario CEMIC, Buenos Aires C1431FWN, Argentina;
| | - Maria I. Vaccaro
- Instituto de Bioquimica y Medicina Molecular Prof Alberto Boveris (IBIMOL), CONICET, Universidad de Buenos Aires, Buenos Aires C1113AAC, Argentina;
- Instituto de Investigaciones, IUC, Medicina Traslacional, Hospital Universitario CEMIC, Buenos Aires C1431FWN, Argentina;
| |
Collapse
|
9
|
Akbari A, Noorbakhsh Varnosfaderani SM, Haeri MS, Fathi Z, Aziziyan F, Yousefi Rad A, Zalpoor H, Nabi-Afjadi M, Malekzadegan Y. Autophagy induced by Helicobacter Pylori infection can lead to gastric cancer dormancy, metastasis, and recurrence: new insights. Hum Cell 2024; 37:139-153. [PMID: 37924488 DOI: 10.1007/s13577-023-00996-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/11/2023] [Indexed: 11/06/2023]
Abstract
According to the findings of recent research, Helicobacter Pylori (H. pylori) infection is not only the primary cause of gastric cancer (GC), but it is also linked to the spread and invasion of GC through a number of processes and factors that contribute to virulence. In this study, we discussed that H. pylori infection can increase autophagy in GC tumor cells, leading to poor prognosis in such patients. Until now, the main concerns have been focused on H. pylori's role in GC development. According to our hypothesis, however, H. pylori infection may also lead to GC dormancy, metastasis, and recurrence by stimulating autophagy. Therefore, understanding how H. pylori possess these processes through its virulence factors and various microRNAs can open new windows for providing new prevention and/or therapeutic approaches to combat GC dormancy, metastasis, and recurrence which can occur in GC patients with H. pylori infection with targeting autophagy and eradicating H. pylori infection.
Collapse
Affiliation(s)
- Abdullatif Akbari
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Melika Sadat Haeri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zeinab Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Yousefi Rad
- Department of Biochemistry, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Hamidreza Zalpoor
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | | |
Collapse
|
10
|
Mackieh R, Al-Bakkar N, Kfoury M, Okdeh N, Pietra H, Roufayel R, Legros C, Fajloun Z, Sabatier JM. Unlocking the Benefits of Fasting: A Review of its Impact on Various Biological Systems and Human Health. Curr Med Chem 2024; 31:1781-1803. [PMID: 38018193 DOI: 10.2174/0109298673275492231121062033] [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: 08/02/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 11/30/2023]
Abstract
Fasting has gained significant attention in recent years for its potential health benefits in various body systems. This review aims to comprehensively examine the effects of fasting on human health, specifically focusing on its impact on different body's physiological systems. The cardiovascular system plays a vital role in maintaining overall health, and fasting has shown promising effects in improving cardiovascular health markers such as blood pressure, cholesterol levels, and triglyceride levels. Additionally, fasting has been suggested to enhance insulin sensitivity, promote weight loss, and improve metabolic health, thus offering potential benefits to individuals with diabetes and metabolic disorders. Furthermore, fasting can boost immune function, reduce inflammation, enhance autophagy, and support the body's defense against infections, cancer, and autoimmune diseases. Fasting has also demonstrated a positive effect on the brain and nervous system. It has been associated with neuroprotective properties, improving cognitive function, and reducing the risk of neurodegenerative diseases, besides the ability of increasing the lifespan. Hence, understanding the potential advantages of fasting can provide valuable insights for individuals and healthcare professionals alike in promoting health and wellbeing. The data presented here may have significant implications for the development of therapeutic approaches and interventions using fasting as a potential preventive and therapeutic strategy.
Collapse
Affiliation(s)
- Rawan Mackieh
- Department of Biology, Faculty of Sciences, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
| | - Nadia Al-Bakkar
- Faculty of Health Sciences, College of Life Sciences, Beirut Arab University, Beirut Campus, P.O. Box 11 50 20, Riad El Solh, Beirut 11072809, Lebanon
| | - Milena Kfoury
- Department of Biology, Faculty of Sciences, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
| | - Nathalie Okdeh
- Department of Biology, Faculty of Sciences, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
| | - Hervé Pietra
- Association Esprit Jeûne & Fasting Spirit, 226, Chemin du Pélican, Toulon 83000, France
| | - Rabih Roufayel
- College of Engineering and Technology, American University of the Middle East, Hadiya, Kuwait
| | - Christian Legros
- Univ Angers, INSERM, CNRS, MITOVASC, Team 2 CarMe, SFR ICAT, Angers 49000, France
| | - Ziad Fajloun
- Department of Biology, Faculty of Sciences, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon
| | - Jean-Marc Sabatier
- Aix-- Marseille Université, CNRS, INP, Inst Neurophysiopathol, Marseille 13385, France
| |
Collapse
|
11
|
Abstract
Maintenance of protein homeostasis and organelle integrity and function is critical for cellular homeostasis and cell viability. Autophagy is the principal mechanism that mediates the delivery of various cellular cargoes to lysosomes for degradation and recycling. A myriad of studies demonstrate important protective roles for autophagy against disease. However, in cancer, seemingly opposing roles of autophagy are observed in the prevention of early tumour development versus the maintenance and metabolic adaptation of established and metastasizing tumours. Recent studies have addressed not only the tumour cell intrinsic functions of autophagy, but also the roles of autophagy in the tumour microenvironment and associated immune cells. In addition, various autophagy-related pathways have been described, which are distinct from classical autophagy, that utilize parts of the autophagic machinery and can potentially contribute to malignant disease. Growing evidence on how autophagy and related processes affect cancer development and progression has helped guide efforts to design anticancer treatments based on inhibition or promotion of autophagy. In this Review, we discuss and dissect these different functions of autophagy and autophagy-related processes during tumour development, maintenance and progression. We outline recent findings regarding the role of these processes in both the tumour cells and the tumour microenvironment and describe advances in therapy aimed at autophagy processes in cancer.
Collapse
Affiliation(s)
- Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.
| | - Noor Gammoh
- MRC Institute of Genetics & Cancer, The University of Edinburgh, Edinburgh, UK.
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK.
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
12
|
Abstract
Cancer cells rely on lysosome-dependent degradation to recycle nutrients that serve their energetic and biosynthetic needs. Despite great interest in repurposing the antimalarial hydroxychloroquine as a lysosomal inhibitor in clinical oncology trials, the mechanisms by which hydroxychloroquine and other lysosomal inhibitors induce tumor-cell cytotoxicity remain unclear. In this issue of the JCI, Bhardwaj et al. demonstrate that DC661, a dimeric form of chloroquine that inhibits palmitoyl-protein thioesterase 1 (PPT1), promoted lysosomal lipid peroxidation, resulting in lysosomal membrane permeabilization and tumor cell death. Remarkably, this lysosomal cell death pathway elicited cell-intrinsic immunogenicity and promoted T lymphocyte-mediated tumor cell clearance. The findings provide the mechanistic foundation for the potential combined use of immunotherapy and lysosomal inhibition in clinical trials.
Collapse
|
13
|
Abeliovich H, Debnath J, Ding WX, Jackson WT, Kim DH, Klionsky DJ, Ktistakis N, Margeta M, Münz C, Petersen M, Sadoshima J, Vergne I. Where is the field of autophagy research heading? Autophagy 2023; 19:1049-1054. [PMID: 36628432 PMCID: PMC10012950 DOI: 10.1080/15548627.2023.2166301] [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] [Indexed: 01/12/2023] Open
Abstract
In this editors' corner, the section editors were asked to indicate where they see the autophagy field heading and to suggest what they consider to be key unanswered questions in their specialty area.
Collapse
Affiliation(s)
- Hagai Abeliovich
- Department of Biochemistry, Food Science and Nutrition, Hebrew University of Jerusalem, Rehovot, Israel
| | - Jayanta Debnath
- Department of Pathology, USCF School of Medicine, San Francisco, CA, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - William T. Jackson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minn, USA
| | | | | | - Marta Margeta
- Department of Pathology, USCF School of Medicine, San Francisco, CA, USA
| | - Christian Münz
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Morten Petersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Isabelle Vergne
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, University of Toulouse III-Paul Sabatier, Toulouse, France
| |
Collapse
|
14
|
Xiao C, Wu G, Chen P, Gao L, Chen G, Zhang H. Phase separation in epigenetics and cancer stem cells. Front Oncol 2022; 12:922604. [PMID: 36081552 PMCID: PMC9445202 DOI: 10.3389/fonc.2022.922604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/25/2022] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence indicates that liquid–liquid phase separation (LLPS) is the basis of the formation of membrane-less compartments in cells. This biomolecular condensate represented by phase separation may influence epigenetics in cancer stem cells (CSCs), a small subpopulation of cancer cells responding to the initiation, maintenance, metastasis, and therapy resistance of cancer. Understanding the underlying biophysical principles and the specific characteristics of biocondensates would provide insights into the precise blocking of potential tumor targets, thereby fundamentally curbing tumor occurrence, recurrence and metastasis. In this review, we summarized the key phenomenon and experimental detection of phase separation and the possibility of regulating the stemness of CSCs through phase separation. We believe that the mechanism of phase separation in CSCs will open up new avenues for the mystery of tumor formation, and modulating phase separation will be a great strategy for CSC-targeted tumor therapy.
Collapse
Affiliation(s)
- Chanchan Xiao
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hongkong-Macao Great Bay Area Geroscience Joint Laboratory (GBGJL), School of Medicine, Jinan University, Guangzhou, China
| | - Guangjie Wu
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hongkong-Macao Great Bay Area Geroscience Joint Laboratory (GBGJL), School of Medicine, Jinan University, Guangzhou, China
| | - Pengfei Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hongkong-Macao Great Bay Area Geroscience Joint Laboratory (GBGJL), School of Medicine, Jinan University, Guangzhou, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hongkong-Macao Great Bay Area Geroscience Joint Laboratory (GBGJL), School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Lijuan Gao, ; Guobing Chen, ; Hongyi Zhang,
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hongkong-Macao Great Bay Area Geroscience Joint Laboratory (GBGJL), School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Lijuan Gao, ; Guobing Chen, ; Hongyi Zhang,
| | - Hongyi Zhang
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hongkong-Macao Great Bay Area Geroscience Joint Laboratory (GBGJL), School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Lijuan Gao, ; Guobing Chen, ; Hongyi Zhang,
| |
Collapse
|
15
|
Long JS, Kania E, McEwan DG, Barthet VJA, Brucoli M, Ladds MJ, Nössing C, Ryan KM. ATG7 is a haploinsufficient repressor of tumor progression and promoter of metastasis. Proc Natl Acad Sci U S A 2022; 119:e2113465119. [PMID: 35867735 PMCID: PMC9282388 DOI: 10.1073/pnas.2113465119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 05/04/2022] [Indexed: 01/18/2023] Open
Abstract
The role of autophagy in cancer is complex. Both tumor-promoting and tumor-suppressive effects are reported, with tumor type, stage and specific genetic lesions dictating the role. This calls for analysis in models that best recapitulate each tumor type, from initiation to metastatic disease, to specifically understand the contribution of autophagy in each context. Here, we report the effects of deleting the essential autophagy gene Atg7 in a model of pancreatic ductal adenocarcinoma (PDAC), in which mutant KrasG12D and mutant Trp53172H are induced in adult tissue leading to metastatic PDAC. This revealed that Atg7 loss in the presence of KrasG12D/+ and Trp53172H/+ was tumor promoting, similar to previous observations in tumors driven by embryonic KrasG12D/+ and deletion of Trp53. However, Atg7 hemizygosity also enhanced tumor initiation and progression, even though this did not ablate autophagy. Moreover, despite this enhanced progression, fewer Atg7 hemizygous mice had metastases compared with animals wild type for this allele, indicating that ATG7 is a promoter of metastasis. We show, in addition, that Atg7+/- tumors have comparatively lower levels of succinate, and that cells derived from Atg7+/- tumors are also less invasive than those from Atg7+/+ tumors. This effect on invasion can be rescued by ectopic expression of Atg7 in Atg7+/- cells, without affecting the autophagic capacity of the cells, or by treatment with a cell-permeable analog of succinate. These findings therefore show that ATG7 has roles in invasion and metastasis that are not related to the role of the protein in the regulation of autophagy.
Collapse
Affiliation(s)
- Jaclyn S. Long
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Elżbieta Kania
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - David G. McEwan
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Valentin J. A. Barthet
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, United Kingdom
| | - Martina Brucoli
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, United Kingdom
| | - Marcus J.G.W. Ladds
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, G61 1BD, United Kingdom
| | - Christoph Nössing
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, United Kingdom
| | - Kevin M. Ryan
- Tumour Cell Death and Autophagy Laboratory, Cancer Research UK Beatson Institute, Glasgow, G61 1BD, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, United Kingdom
| |
Collapse
|
16
|
3,4-Dihydroxyphenylethanol (DPE or Hydroxytyrosol) Counteracts ERK1/2 and mTOR Activation, Pro-Inflammatory Cytokine Release, Autophagy and Mitophagy Reduction Mediated by Benzo[a]pyrene in Primary Human Colonic Epithelial Cells. Pharmaceutics 2022; 14:pharmaceutics14030663. [PMID: 35336037 PMCID: PMC8948646 DOI: 10.3390/pharmaceutics14030663] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 02/04/2023] Open
Abstract
Understanding the effects induced by carcinogens on primary colonic epithelial cells and how to counteract them might help to prevent colon cancer, which is one of the most frequent and aggressive cancers. In this study, we exposed primary human colonic epithelial cells (HCoEpC) to Benzo[a]pyrene (B[a]P) and found that it led to an increased production of pro-inflammatory cytokines and activated ERK1/2 and mTOR. These pathways are known to be involved in inflammatory bowel disease (IBD), which represents a colon cancer risk factor. Moreover, B[a]P reduced autophagy and mitophagy, processes whose dysregulation has been clearly demonstrated to predispose to cancer either by in vitro or in vivo studies. Interestingly, all the effects induced by B[a]P could be counteracted by 3,4-Dihydroxyphenylethanol (DPE or Hydroxytyrosol, H), the most powerful anti-inflammatory and antioxidant compound contained in olive oil. This study sheds light on the mechanisms that could be involved in colon carcinogenesis induced by a chemical carcinogen and identifies a safe natural product that may help to prevent them.
Collapse
|
17
|
Khotimchenko R, Bryukhovetskiy I, Khotimchenko M, Khotimchenko Y. Bioactive Compounds with Antiglioma Activity from Marine Species. Biomedicines 2021; 9:biomedicines9080886. [PMID: 34440090 PMCID: PMC8389718 DOI: 10.3390/biomedicines9080886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 11/21/2022] Open
Abstract
The search for new chemical compounds with antitumor pharmacological activity is a necessary process for creating more effective drugs for each specific malignancy type. This review presents the outcomes of screening studies of natural compounds with high anti-glioma activity. Despite significant advances in cancer therapy, there are still some tumors currently considered completely incurable including brain gliomas. This review covers the main problems of the glioma chemotherapy including drug resistance, side effects of common anti-glioma drugs, and genetic diversity of brain tumors. The main emphasis is made on the characterization of natural compounds isolated from marine organisms because taxonomic diversity of organisms in seawaters significantly exceeds that of terrestrial species. Thus, we should expect greater chemical diversity of marine compounds and greater likelihood of finding effective molecules with antiglioma activity. The review covers at least 15 classes of organic compounds with their chemical formulas provided as well as semi-inhibitory concentrations, mechanisms of action, and pharmacokinetic profiles. In conclusion, the analysis of the taxonomic diversity of marine species containing bioactives with antiglioma activity is performed noting cytotoxicity indicators and to the tumor cells in comparison with similar indicators of antitumor agents approved for clinical use as antiglioblastoma chemotherapeutics.
Collapse
Affiliation(s)
- Rodion Khotimchenko
- School of Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia; (R.K.); (I.B.); (M.K.)
| | - Igor Bryukhovetskiy
- School of Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia; (R.K.); (I.B.); (M.K.)
| | - Maksim Khotimchenko
- School of Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia; (R.K.); (I.B.); (M.K.)
| | - Yuri Khotimchenko
- School of Biomedicine, Far Eastern Federal University, 690090 Vladivostok, Russia; (R.K.); (I.B.); (M.K.)
- Laboratory of Pharmacology, A. V. Zhirmunsky National Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690950 Vladivostok, Russia
- Correspondence:
| |
Collapse
|
18
|
Li Y, Cook KL, Yu W, Jin L, Bouker KB, Clarke R, Hilakivi-Clarke L. Inhibition of Antiestrogen-Promoted Pro-Survival Autophagy and Tamoxifen Resistance in Breast Cancer through Vitamin D Receptor. Nutrients 2021; 13:nu13051715. [PMID: 34069442 PMCID: PMC8159129 DOI: 10.3390/nu13051715] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 01/07/2023] Open
Abstract
We determined how vitamin D receptor (VDR) is linked to disease outcome in estrogen receptor-positive (ER+) breast cancer patients treated with tamoxifen (TAM). Breast cancer patients (n = 581) in four different datasets were divided into those expressing higher (above median) and lower levels of VDR in pretreatment ER+ tumors. Across all datasets, TAM-treated patients with higher pretreatment tumor VDR expression exhibited significantly longer recurrence-free survival. Ingenuity pathway analysis identified autophagy and unfolded protein response (UPR) as top differentially expressed pathways between high and low VDR-expressing ER+ cancers. Activation of VDR with vitamin D (VitD), either calcitriol or its synthetic analog EB1089, sensitized MCF-7-derived, antiestrogen-resistant LCC9 human breast cancer cells to TAM, and attenuated increased UPR and pro-survival autophagy. Silencing of VDR blocked these effects through the IRE1α-JNK pathway. Further, silencing of VDR impaired sensitivity to TAM in antiestrogen-responsive LCC1 cells, and prevented the effects of calcitriol and EB1089 on UPR and autophagy. In a preclinical mouse model, dietary VitD supplementation induced VDR activation and reduced carcinogen-induced ER+ mammary tumor incidence. In addition, IRE1α-JNK signaling was downregulated and survival autophagy was inhibited in mammary tumors of VitD-supplemented mice. Thus, activation of VDR is predictive of reduced risk of breast cancer recurrence in ER+ patients, possibly by inhibiting antiestrogen-promoted pro-survival autophagy.
Collapse
|
19
|
Singhal SK, Byun JS, Park S, Yan T, Yancey R, Caban A, Hernandez SG, Hewitt SM, Boisvert H, Hennek S, Bobrow M, Ahmed MSU, White J, Yates C, Aukerman A, Vanguri R, Bareja R, Lenci R, Farré PL, De Siervi A, Nápoles AM, Vohra N, Gardner K. Kaiso (ZBTB33) subcellular partitioning functionally links LC3A/B, the tumor microenvironment, and breast cancer survival. Commun Biol 2021; 4:150. [PMID: 33526872 PMCID: PMC7851134 DOI: 10.1038/s42003-021-01651-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 12/29/2020] [Indexed: 12/30/2022] Open
Abstract
The use of digital pathology for the histomorphologic profiling of pathological specimens is expanding the precision and specificity of quantitative tissue analysis at an unprecedented scale; thus, enabling the discovery of new and functionally relevant histological features of both predictive and prognostic significance. In this study, we apply quantitative automated image processing and computational methods to profile the subcellular distribution of the multi-functional transcriptional regulator, Kaiso (ZBTB33), in the tumors of a large racially diverse breast cancer cohort from a designated health disparities region in the United States. Multiplex multivariate analysis of the association of Kaiso’s subcellular distribution with other breast cancer biomarkers reveals novel functional and predictive linkages between Kaiso and the autophagy-related proteins, LC3A/B, that are associated with features of the tumor immune microenvironment, survival, and race. These findings identify effective modalities of Kaiso biomarker assessment and uncover unanticipated insights into Kaiso’s role in breast cancer progression. Through automated image analysis, Singhal et al quantify nuclear versus cytoplasmic distribution of the Kaiso transcription factor in breast cancer patient tissue. They find that Kaiso distribution correlates with breast cancer subtype and overall survival, and discover a link between cytoplasmic Kaiso and autophagy marker LC3.
Collapse
Affiliation(s)
- Sandeep K Singhal
- Department of Pathology, School of Medicine and Health Sciences, Department of Computer Science, School of Electrical Engineering and Computer Science, University of North Dakota, Grand Forks, ND, USA
| | - Jung S Byun
- Division of Intramural Research, National Institutes of Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Samson Park
- Division of Intramural Research, National Institutes of Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Tingfen Yan
- Division of Intramural Research, National Institutes of Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA.,National Institutes of Genome Research, National Institutes of Health, Bethesda, MD, USA
| | - Ryan Yancey
- Department of Pathology and Cell Biology, Columbia University Irvine Medical Center, New York, NY, USA
| | - Ambar Caban
- Department of Pathology and Cell Biology, Columbia University Irvine Medical Center, New York, NY, USA
| | - Sara Gil Hernandez
- Division of Intramural Research, National Institutes of Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Centers for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Md Shakir Uddin Ahmed
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Al, USA
| | - Jason White
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Al, USA
| | - Clayton Yates
- Department of Biology and Center for Cancer Research, Tuskegee University, Tuskegee, Al, USA
| | - Andrew Aukerman
- Department of Pathology and Cell Biology, Columbia University Irvine Medical Center, New York, NY, USA
| | - Rami Vanguri
- Department of Pathology and Cell Biology, Columbia University Irvine Medical Center, New York, NY, USA
| | - Rohan Bareja
- Department Computer Science Department, Columbia University, New York, NY, USA
| | - Romina Lenci
- Department of Pathology and Cell Biology, Columbia University Irvine Medical Center, New York, NY, USA
| | - Paula Lucia Farré
- Laboratorio de Oncologıa Molecular y Nuevos Blancos Terapeuticos, Instituto de Biologıa y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Adriana De Siervi
- Laboratorio de Oncologıa Molecular y Nuevos Blancos Terapeuticos, Instituto de Biologıa y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - Anna María Nápoles
- Division of Intramural Research, National Institutes of Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD, USA
| | - Nasreen Vohra
- Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Kevin Gardner
- Department of Pathology and Cell Biology, Columbia University Irvine Medical Center, New York, NY, USA.
| |
Collapse
|
20
|
Courtois S, Haykal M, Bodineau C, Sifré E, Azzi-Martin L, Ménard A, Mégraud F, Lehours P, Durán RV, Varon C, Bessède E. Autophagy induced by Helicobacter pylori infection is necessary for gastric cancer stem cell emergence. Gastric Cancer 2021; 24:133-144. [PMID: 32940810 DOI: 10.1007/s10120-020-01118-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The main cause of gastric cancer is the infection by the bacterium Helicobacter pylori which induces a chronic inflammation and an epithelial-to-mesenchymal transition (EMT) leading to the emergence of cells with cancer stem cell (CSC) properties. However, the underlying mechanisms have not been fully characterized. Moreover, H. pylori modulates the host cell autophagic process, but a few studies have investigated the role of this process in tumoral transformation. The aim of this study was to determine whether H. pylori-induced autophagy has a role in CSC emergence. METHODS Autophagic flux in response to H. pylori infection was characterized in AGS cell line expressing the tandem-tagged mCherry-GFP-LC3 protein and using a ratiometric flow cytometry analysis. Then, AGS and MKN45 cell lines were treated with bafilomycin or chloroquine, two pharmaceutical well-known inhibitors of autophagy, and different EMT and CSC characteristics were analyzed. RESULTS First, a co-expression of the gastric CSC marker CD44 and the autophagic marker LC3 in mice and human stomach tissues infected with H. pylori was observed. Then, we demonstrated in vitro that H. pylori was able to activate the autophagy process with a reduced autophagic flux. Finally, infected cells were treated with autophagy inhibitors, which reduced (i) appearance of mesenchymal phenotypes and migration ability related to EMT and (ii) CD44 expression as well as tumorsphere formation capacities reflecting CSC properties. CONCLUSION In conclusion, all these data show that H. pylori-induced autophagy is implicated in gastric CSC emergence and could represent an interesting therapeutic target.
Collapse
Affiliation(s)
- Sarah Courtois
- Univ. Bordeaux, INSERM, BaRITOn, U1053, F-33000, Bordeaux, France
| | - Maria Haykal
- Univ. Bordeaux, INSERM, BaRITOn, U1053, F-33000, Bordeaux, France
| | - Clément Bodineau
- Centro Andaluz de Biología Molecular Y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Américo Vespucio 24, 41092, Sevilla, Spain.,Institut Européen de Chimie et Biologie, INSERM U1218, University of Bordeaux, Pessac, France
| | - Elodie Sifré
- Univ. Bordeaux, INSERM, BaRITOn, U1053, F-33000, Bordeaux, France
| | | | - Armelle Ménard
- Univ. Bordeaux, INSERM, BaRITOn, U1053, F-33000, Bordeaux, France
| | - Francis Mégraud
- Univ. Bordeaux, INSERM, BaRITOn, U1053, F-33000, Bordeaux, France.,French National Reference Center for Campylobacters and Helicobacters (CNRCH), University Hospital of Bordeaux, Place Amelie Raba Leon, 33076, Bordeaux, France
| | - Philippe Lehours
- Univ. Bordeaux, INSERM, BaRITOn, U1053, F-33000, Bordeaux, France.,French National Reference Center for Campylobacters and Helicobacters (CNRCH), University Hospital of Bordeaux, Place Amelie Raba Leon, 33076, Bordeaux, France
| | - Raúl V Durán
- Centro Andaluz de Biología Molecular Y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Américo Vespucio 24, 41092, Sevilla, Spain
| | - Christine Varon
- Univ. Bordeaux, INSERM, BaRITOn, U1053, F-33000, Bordeaux, France
| | - Emilie Bessède
- Univ. Bordeaux, INSERM, BaRITOn, U1053, F-33000, Bordeaux, France. .,French National Reference Center for Campylobacters and Helicobacters (CNRCH), University Hospital of Bordeaux, Place Amelie Raba Leon, 33076, Bordeaux, France.
| |
Collapse
|
21
|
Wechman SL, Emdad L, Sarkar D, Das SK, Fisher PB. Vascular mimicry: Triggers, molecular interactions and in vivo models. Adv Cancer Res 2020; 148:27-67. [PMID: 32723566 DOI: 10.1016/bs.acr.2020.06.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Vascular mimicry is induced by a wide array of genes with functions related to cancer stemness, hypoxia, angiogenesis and autophagy. Vascular mimicry competent (VM-competent) cells that form de novo blood vessels are common in solid tumors facilitating tumor cell survival and metastasis. VM-competent cells display increased levels of vascular mimicry selecting for stem-like cells in an O2-gradient-dependent manner in deeply hypoxic tumor regions, while also aiding in maintaining tumor cell metabolism and stemness. Three of the principal drivers of vascular mimicry are EphA2, Nodal and HIF-1α, however, directly or indirectly many of these molecules affect VE-Cadherin (VE-Cad), which forms gap-junctions to bind angiogenic blood vessels together. During vascular mimicry, the endothelial-like functions of VM-competent cancer stem cells co-opt VE-Cad to bind cancer cells together to create cancer cell-derived blood conducting vessels. This process potentially compensates for the lack of access to blood and nutrient in avascular tumors, simultaneously providing nutrients and enhancing cancer invasion and metastasis. Current evidence also supports that vascular mimicry promotes cancer malignancy and metastasis due to the cooperation of oncogenic signaling molecules driving cancer stemness and autophagy. While a number of currently used cancer therapeutics are effective inhibitors of vascular mimicry, developing a new class of vascular mimicry specific inhibitors could allow for the treatment of angiogenesis-resistant tumors, inhibit cancer metastasis and improve patient survival. In this review, we describe the principal vascular mimicry pathways in addition to emphasizing the roles of hypoxia, autophagy and select proangiogenic oncogenes in this process.
Collapse
Affiliation(s)
- Stephen L Wechman
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
22
|
Zhang D, Zhao X, Zhang D, Gao S, Xue X, Fu J. Hyperoxia reduces STX17 expression and inhibits the autophagic flux in alveolar type II epithelial cells in newborn rats. Int J Mol Med 2020; 46:773-781. [PMID: 32467992 PMCID: PMC7307846 DOI: 10.3892/ijmm.2020.4617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 05/05/2020] [Indexed: 12/20/2022] Open
Abstract
Supplemental oxygen therapy can be life-saving for premature infants. Our previous study revealed a defect in the autophagic flux in the lung tissues of neonatal rats with hyperoxia-induced bronchopulmonary dysplasia (BPD), but the underlying mechanism remains unknown. Moreover, there are few innovative treatments that can completely alter the course of BPD. The present study examined the expression of Syntaxin 17 (STX17), a protein necessary for autophago-some-lysosome binding, in alveolar type II (AT-II) epithelial cells of neonatal rats with BPD. Neonatal Sprague-Dawley rats were randomly exposed to elevated O2 levels [fraction of inspired oxygen (FiO2), 0.8; model group] or normal room air (FiO2, 0.21; control group), and the expression levels of STX17, autophagy-related [Microtubule-associated protein 1A/1B-light chain 3B (LC3B)-II, p62, lysosomal-associated membrane protein 1)] and apoptosis-related (cleaved caspase3) mRNA and proteins were examined in lung tissues. Moreover, the expression levels of the aforementioned proteins were measured in isolated primary AT-II cells cultured in vitro under hyperoxic conditions in the presence or absence of pharmacological modulators of autophagy. Transmission electron microscopy identified that AT-II cell apoptosis and autophagosome aggregation were elevated in the lungs of BPD rats compared with control rats on postnatal day 7. STX17 mRNA and protein expression levels were decreased in lung tissue and isolated AT-II cells as early as postnatal day 3 in BPD rats, while the expression levels of LC3B-II, p62 and cleaved caspase3 were increased, reaching a peak on postnatal day 7. This early reduction in STX17 expression, followed by increased expression in autophagy- and apoptosis-related proteins, was also observed in isolated AT-II cells exposed to hyperoxia in vitro. However, treatment with the autophagy inducers rapamycin or LiCl eliminated the hyperoxia-induced reduction in STX17, partially restored the autophagy flux and increased the survival of AT-II cells exposed to hyperoxia. Collectively, these results indicated that STX17 expression in AT-II cells was reduced in the early stages of BPD in neonatal rats and may be related to the subsequent hyperoxia-induced block in autophagic flux.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xinyi Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dingning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Siyang Gao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
23
|
Agrotis A, Ketteler R. On ATG4B as Drug Target for Treatment of Solid Tumours-The Knowns and the Unknowns. Cells 2019; 9:cells9010053. [PMID: 31878323 PMCID: PMC7016753 DOI: 10.3390/cells9010053] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 12/13/2022] Open
Abstract
Autophagy is an evolutionary conserved stress survival pathway that has been shown to play an important role in the initiation, progression, and metastasis of multiple cancers; however, little progress has been made to date in translation of basic research to clinical application. This is partially due to an incomplete understanding of the role of autophagy in the different stages of cancer, and also to an incomplete assessment of potential drug targets in the autophagy pathway. While drug discovery efforts are on-going to target enzymes involved in the initiation phase of the autophagosome, e.g., unc51-like autophagy activating kinase (ULK)1/2, vacuolar protein sorting 34 (Vps34), and autophagy-related (ATG)7, we propose that the cysteine protease ATG4B is a bona fide drug target for the development of anti-cancer treatments. In this review, we highlight some of the recent advances in our understanding of the role of ATG4B in autophagy and its relevance to cancer, and perform a critical evaluation of ATG4B as a druggable cancer target.
Collapse
|
24
|
Matthew-Onabanjo AN, Janusis J, Mercado-Matos J, Carlisle AE, Kim D, Levine F, Cruz-Gordillo P, Richards R, Lee MJ, Shaw LM. Beclin 1 Promotes Endosome Recruitment of Hepatocyte Growth Factor Tyrosine Kinase Substrate to Suppress Tumor Proliferation. Cancer Res 2019; 80:249-262. [PMID: 31744816 DOI: 10.1158/0008-5472.can-19-1555] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/01/2019] [Accepted: 11/13/2019] [Indexed: 12/24/2022]
Abstract
Beclin 1 has nonautophagic functions that include its ability to regulate endocytic receptor trafficking. However, the contribution of this function to tumor suppression is poorly understood. Here, we provide in vivo evidence that Beclin 1 suppresses tumor proliferation by regulating the endocytic trafficking and degradation of the EGFR and transferrin (TFR1) receptors. Beclin 1 promoted endosomal recruitment of hepatocyte growth factor tyrosine kinase substrate (HRS), which was necessary for sorting surface receptors to intraluminal vesicles for signal silencing and lysosomal degradation. In tumors with low Beclin 1 expression, endosomal HRS recruitment was diminished and receptor function was sustained. Collectively, our results demonstrate a novel role for Beclin 1 in impeding tumor growth by coordinating the regulation of key growth factor and nutrient receptors. These data provide an explanation for how low levels of Beclin 1 facilitate tumor proliferation and contribute to poor cancer outcomes. SIGNIFICANCE: Beclin 1 controls the trafficking fate of growth regulatory receptors to suppress tumor proliferation.
Collapse
Affiliation(s)
- Asia N Matthew-Onabanjo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts.,Medical Scientist Training Program, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jenny Janusis
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jose Mercado-Matos
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts.,Medical Scientist Training Program, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Anne E Carlisle
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Dohoon Kim
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Fayola Levine
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Peter Cruz-Gordillo
- Medical Scientist Training Program, University of Massachusetts Medical School, Worcester, Massachusetts.,Program in Systems Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Ryan Richards
- Program in Systems Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Michael J Lee
- Program in Systems Biology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Leslie M Shaw
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts.
| |
Collapse
|
25
|
Yin C, Zhang H, Liu X, Zhang H, Zhang Y, Bai X, Wang L, Li H, Li X, Zhang S, Zhang L, Zhang Y. Downregulated MCOLN1 Attenuates The Progression Of Non-Small-Cell Lung Cancer By Inhibiting Lysosome-Autophagy. Cancer Manag Res 2019; 11:8607-8617. [PMID: 31576167 PMCID: PMC6765329 DOI: 10.2147/cmar.s216538] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/06/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives Autophagy plays various roles in non-small-cell lung cancer (NSCLC). MCOLN1, a reactive oxygen species sensor, can regulate autophagy via lysosomal Ca(2+); however, the role of MCOLN1 in NSCLC is largely unknown. This study aimed to explore the effects of MCOLN1 on proliferation, invasion and migration in NSCLC and the underling mechanisms. Materials and methods The tissues of NSCLC patients were collected, then MCOLN1 expression in tumor and adjacent tissues was measured and its relationship with pathological staging was analyzed. The Cell Counting Kit-8 (CCK-8) assay, wound healing assay and transwell migration assay were used to evaluate the proliferation, migration and invasion ability, respectively. Live-cell imaging and transmission electron microscopy (TEM) were used to observe autophagic flux and autolysosomes. Results It was found that MCOLN1 expression was significantly decreased in human NSCLC tissues compared with normal lung tissues while more MCOLN1 in stage III–IV was shown than stage I–II, indicating that MCOLN1 increased along with the progression of NSCLC. Furthermore, CCK-8 assay, wound healing assay and transwell migration assay confirmed that the inhibition of MCOLN1 suppressed NSCLC cells proliferation migration and invasion. Overexpression of MCOLN1 promoted autophagy in A549 and H1299 cells with increased LC3-II/I, lamp1 expression and autolysosomes as well as autophagic flux shown by live-cell imaging and TEM. Conclusion Our study shows that downregulated MCOLN1 reduced lysosome-autophagy activity contributing to inhibited tumor progression, which reveals a novel role of MCOLN1 in NSCLC, and targeting MCOLN1 may be a therapeutic potential for NSCLC. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/-I_WZ7bSq9s
Collapse
Affiliation(s)
- Chuntong Yin
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Han Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Xin Liu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Haiying Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Yue Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Xue Bai
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Lei Wang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Huimin Li
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Xia Li
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Shuqian Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| | - Linyou Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, People's Republic of China
| | - Yong Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, People's Republic of China
| |
Collapse
|
26
|
Du FY, Zhou QF, Sun WJ, Chen GL. Targeting cancer stem cells in drug discovery: Current state and future perspectives. World J Stem Cells 2019; 11:398-420. [PMID: 31396368 PMCID: PMC6682504 DOI: 10.4252/wjsc.v11.i7.398] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/18/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023] Open
Abstract
In recent decades, cancer stem cells (CSCs) have been increasingly identified in many malignancies. CSC-related signaling pathways and their functions provide new strategies for treating cancer. The aberrant activation of related signaling pathways (e.g., Wnt, Notch, and Hedgehog pathways) has been linked to multiple types of malignant tumors, which makes these pathways attractive targets for cancer therapy. CSCs display many characteristic features, such as self-renewal, differentiation, high tumorigenicity, and drug resistance. Therefore, there is an urgent need to develop new therapeutic strategies to target these pathways to control stem cell replication, survival, and differentiation. Notable crosstalk occurs among different signaling pathways and potentially leads to compensatory escape. Therefore, multitarget inhibitors will be one of the main methods to overcome the drug resistance of CSCs. Many small molecule inhibitors of components of signaling pathways in CSCs have entered clinical trials, and some inhibitors, such as vismodegib, sonidegib, and glasdegib, have been approved. Tumor cells are susceptible to sonidegib and vismodegib resistance due to mutations in the Smo protein. The signal transducers and activators of transcription 3 (STAT3) inhibitor BBI608 is being evaluated in a phase III trial for a variety of cancers. Structural derivatives of BBI608 are the main focus of STAT3 inhibitor development, which is another strategy for CSC therapy. In addition to the potential pharmacological inhibitors targeting CSC-related signaling pathways, other methods of targeting CSCs are available, such as nano-drug delivery systems, mitochondrion targeting, autophagy, hyperthermia, immunotherapy, and CSC microenvironment targeting. In addition, we summarize the latest advances in the clinical development of agents targeting CSC-related signaling pathways and other methods of targeting CSCs.
Collapse
Affiliation(s)
- Fang-Yu Du
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning Province, China
| | - Qi-Fan Zhou
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning Province, China
| | - Wen-Jiao Sun
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning Province, China
| | - Guo-Liang Chen
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning Province, China
| |
Collapse
|
27
|
Das SK, Sarkar D, Emdad L, Fisher PB. MDA-9/Syntenin: An emerging global molecular target regulating cancer invasion and metastasis. Adv Cancer Res 2019; 144:137-191. [PMID: 31349898 DOI: 10.1016/bs.acr.2019.03.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With few exceptions, metastasis is the terminal stage of cancer with limited therapeutic options. Metastasis consists of numerous phenotypic and genotypic alterations of cells that are directly and indirectly induced by multiple intrinsic (cellular) and extrinsic (micro-environmental) factors. To metastasize, a cancer cell often transitions from an epithelial to mesenchymal morphology (EMT), modifies the extracellular matrix, forms emboli and survives in the circulation, escapes immune surveillance, adheres to sites distant from the initial tumor and finally develops a blood supply (angiogenesis) and colonizes in a secondary niche (a micrometastasis). Scientific advances have greatly enhanced our understanding of the precise molecular and genetic changes, operating independently or collectively, that lead to metastasis. This review focuses on a unique gene, melanoma differentiation associated gene-9 (also known as Syntenin-1; Syndecan Binding Protein (sdcbp); mda-9/syntenin), initially cloned and characterized from metastatic human melanoma and shown to be a pro-metastatic gene. In the last two decades, our comprehension of the diversity of actions of MDA-9/Syntenin on cellular phenotype has emerged. MDA-9/Sytenin plays pivotal regulatory roles in multiple signaling cascades and orchestrates both metastatic and non-metastatic events. Considering the relevance of this gene in controlling cancer invasion and metastasis, approaches have been developed to uniquely and selectively target this gene. We also provide recent updates on strategies that have been successfully employed in targeting MDA-9/Syntenin resulting in profound pre-clinical anti-cancer activity.
Collapse
Affiliation(s)
- Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
28
|
Zhan L, Yao S, Sun S, Su Q, Li J, Wei B. NLRC5 and autophagy combined as possible predictors in patients with endometriosis. Fertil Steril 2019; 110:949-956. [PMID: 30316442 DOI: 10.1016/j.fertnstert.2018.06.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/25/2018] [Accepted: 06/17/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To investigate the levels of NLRC5 and autophagy in women with leiomyoma and endometriosis and the correlation between NLRC5 level and autophagy level. DESIGN Case-control study. SETTING Clinics. PATIENT(S) Sixty-five patients were recruited: 30 women with endometriosis were compared with 35 women with leiomyoma. INTERVENTION(S) Endometriosis was definitively diagnosed during surgery by laparoscopy or laparotomy and was confirmed by histopathological evaluation (n=30). Secretory phase ectopic endometrium tissues and eutopic endometrium tissues were obtained from 30 women with endometriosis. Control endometrium tissues were collected at hysterectomy from 35 women with leiomyoma. Immunohistochemical staining of NLRC5, LC3, Beclin1 and P62 were performed. MAIN OUTCOME MEASURE(S) A semiquantitative analysis was performed. Correlations between NLRC5 level and LC3, Beclin1, P62 levels were compared. RESULT(S) The expressions of NLRC5 and P62 in the ectopic and eutopic endometrium of endometriosis groups were significantly higher than that in the endometrium of leiomyoma group. And their expressions in ectopic endometrium were significantly up-regulated compared to the eutopic endometrium. The expressions of LC3 and Beclin1 were down-regulated in the ectopic and eutopic endometrium of endometriosis groups compared to the leiomyoma group. LC3 and Beclin1 levels were lower in ectopic endometrium than in the eutopic endometrium. There is a negative correlation between NLRC5 level and LC3, Beclin1 levels. There is a positive correlation between NLRC5 level and P62 level. CONCLUSION(S) There is a negative correlation between NLRC5 level and autophagy level. NLRC5 and autophagy combined may as promising predictors in patients with endometriosis.
Collapse
Affiliation(s)
- Lei Zhan
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Shun Yao
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Shiying Sun
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Qian Su
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Jun Li
- School of Pharmacy, Anhui Key Laboratory of Bioactivity of Natural Products, Anhui Medical University, Hefei, People's Republic of China
| | - Bing Wei
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China.
| |
Collapse
|
29
|
Palmisano NJ, Meléndez A. Autophagy in C. elegans development. Dev Biol 2019; 447:103-125. [PMID: 29709599 PMCID: PMC6204124 DOI: 10.1016/j.ydbio.2018.04.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 03/19/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
Autophagy involves the sequestration of cytoplasmic contents in a double-membrane structure referred to as the autophagosome and the degradation of its contents upon delivery to lysosomes. Autophagy activity has a role in multiple biological processes during the development of the nematode Caenorhabditis elegans. Basal levels of autophagy are required to remove aggregate prone proteins, paternal mitochondria, and spermatid-specific membranous organelles. During larval development, autophagy is required for the remodeling that occurs during dauer development, and autophagy can selectively degrade components of the miRNA-induced silencing complex, and modulate miRNA-mediated silencing. Basal levels of autophagy are important in synapse formation and in the germ line, to promote the proliferation of proliferating stem cells. Autophagy activity is also required for the efficient removal of apoptotic cell corpses by promoting phagosome maturation. Finally, autophagy is also involved in lipid homeostasis and in the aging process. In this review, we first describe the molecular complexes involved in the process of autophagy, its regulation, and mechanisms for cargo recognition. In the second section, we discuss the developmental contexts where autophagy has been shown to be important. Studies in C. elegans provide valuable insights into the physiological relevance of this process during metazoan development.
Collapse
Affiliation(s)
- Nicholas J Palmisano
- Biology Department, Queens College, CUNY, Flushing, NY, USA; Biology Ph.D. Program, The Graduate Center of the City University of New York, NK, USA
| | - Alicia Meléndez
- Biology Department, Queens College, CUNY, Flushing, NY, USA; Biology Ph.D. Program, The Graduate Center of the City University of New York, NK, USA; Biochemistry Ph.D. Program, The Graduate Center of the City University of New York, NY, USA.
| |
Collapse
|
30
|
Cvetanova B, Shen YC, Shyur LF. Cumingianoside A, a Phyto-Triterpenoid Saponin Inhibits Acquired BRAF Inhibitor Resistant Melanoma Growth via Programmed Cell Death. Front Pharmacol 2019; 10:30. [PMID: 30745871 PMCID: PMC6360185 DOI: 10.3389/fphar.2019.00030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/11/2019] [Indexed: 01/01/2023] Open
Abstract
Mutated proto-oncogene BRAF is a bona fide therapeutic target for melanomas. Regrettably, melanoma acquires resistance to BRAF inhibitors, e.g., vemurafenib (PLX4032) casting doubt on this promising melanoma targeted therapy. In this study, we explored the bioactivity of triterpenoid saponin cumingianoside A (CUMA), isolated from leaves and twigs of Dysoxylum cumingianum against PLX4032-resistant BRAFV 600E mutant melanoma A375-R in vitro and in vivo. Our data show that CUMA treatment inhibited A375-R melanoma cell proliferation in a time- and dose-dependent manner. CUMA also suppressed the activity of CDK1/cyclin B1 complex and led to G2/M-phase arrest of A375-R cells. Furthermore, CUMA treatment resulted in induction of apoptosis as shown by the increased activation of caspase 3 and caspase 7, and the proteolytic cleavage of poly(ADP-ribose) polymerase (PARP). We also observed that CUMA induced autophagy-like activity in A375-R cells, as shown by the increased expression of autophagy-related genes and increased formation of autophagosomes. Moreover, we found that CUMA treatment induced ER stress response and co-treatment with an ER stress inhibitor (4-PBA) could attenuate apoptosis induced by CUMA. Importantly, orally administered CUMA as a single agent or in combination with PLX4032 exhibited strong tumor growth inhibition in a PLX4032-resistant A375-R xenograft mouse model, and with little toxicity. This is the first report to explore the anti-tumor activity of CUMA in vitro and in vivo mechanistically, and our results imply that this triterpenoid saponin may be suitable for development into an anti-melanoma agent.
Collapse
Affiliation(s)
- Biljana Cvetanova
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Ya-Ching Shen
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Lie-Fen Shyur
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan.,Ph.D. Program in Translational Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
31
|
Zhang Y, Whaley-Connell AT, Sowers JR, Ren J. Autophagy as an emerging target in cardiorenal metabolic disease: From pathophysiology to management. Pharmacol Ther 2018; 191:1-22. [PMID: 29909238 PMCID: PMC6195437 DOI: 10.1016/j.pharmthera.2018.06.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 06/05/2018] [Indexed: 12/16/2022]
Abstract
Although advances in medical technology and health care have improved the early diagnosis and management for cardiorenal metabolic disorders, the prevalence of obesity, insulin resistance, diabetes, hypertension, dyslipidemia, and kidney disease remains high. Findings from numerous population-based studies, clinical trials, and experimental evidence have consolidated a number of theories for the pathogenesis of cardiorenal metabolic anomalies including resistance to the metabolic action of insulin, abnormal glucose and lipid metabolism, oxidative and nitrosative stress, endoplasmic reticulum (ER) stress, apoptosis, mitochondrial damage, and inflammation. Accumulating evidence has recently suggested a pivotal role for proteotoxicity, the unfavorable effects of poor protein quality control, in the pathophysiology of metabolic dysregulation and related cardiovascular complications. The ubiquitin-proteasome system (UPS) and autophagy-lysosomal pathways, two major although distinct cellular clearance machineries, govern protein quality control by degradation and clearance of long-lived or damaged proteins and organelles. Ample evidence has depicted an important role for protein quality control, particularly autophagy, in the maintenance of metabolic homeostasis. To this end, autophagy offers promising targets for novel strategies to prevent and treat cardiorenal metabolic diseases. Targeting autophagy using pharmacological or natural agents exhibits exciting new strategies for the growing problem of cardiorenal metabolic disorders.
Collapse
Affiliation(s)
- Yingmei Zhang
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| | - Adam T Whaley-Connell
- Research Service, Harry S Truman Memorial Veterans' Hospital, University of Missouri-Columbia School of Medicine, Columbia, MO, USA; Diabetes and Cardiovascular Center, Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, MO, USA
| | - James R Sowers
- Research Service, Harry S Truman Memorial Veterans' Hospital, University of Missouri-Columbia School of Medicine, Columbia, MO, USA; Diabetes and Cardiovascular Center, Department of Medicine, University of Missouri-Columbia School of Medicine, Columbia, MO, USA
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA.
| |
Collapse
|
32
|
Lin HH, Chung Y, Cheng CT, Ouyang C, Fu Y, Kuo CY, Chi KK, Sadeghi M, Chu P, Kung HJ, Li CF, Limesand KH, Ann DK. Autophagic reliance promotes metabolic reprogramming in oncogenic KRAS-driven tumorigenesis. Autophagy 2018; 14:1481-1498. [PMID: 29956571 PMCID: PMC6135591 DOI: 10.1080/15548627.2018.1450708] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 02/28/2018] [Accepted: 03/06/2018] [Indexed: 12/13/2022] Open
Abstract
Defects in basal autophagy limit the nutrient supply from recycling of intracellular constituents. Despite our understanding of the prosurvival role of macroautophagy/autophagy, how nutrient deprivation, caused by compromised autophagy, affects oncogenic KRAS-driven tumor progression is poorly understood. Here, we demonstrate that conditional impairment of the autophagy gene Atg5 (atg5-KO) extends the survival of KRASG12V-driven tumor-bearing mice by 38%. atg5-KO tumors spread more slowly during late tumorigenesis, despite a faster onset. atg5-KO tumor cells displayed reduced mitochondrial function and increased mitochondrial fragmentation. Metabolite profiles indicated a deficiency in the nonessential amino acid asparagine despite a compensatory overexpression of ASNS (asparagine synthetase), key enzyme for de novo asparagine synthesis. Inhibition of either autophagy or ASNS reduced KRASG12V-driven tumor cell proliferation, migration, and invasion, which was rescued by asparagine supplementation or knockdown of MFF (mitochondrial fission factor). Finally, these observations were reflected in human cancer-derived data, linking ASNS overexpression with poor clinical outcome in multiple cancers. Together, our data document a widespread yet specific asparagine homeostasis control by autophagy and ASNS, highlighting the previously unrecognized role of autophagy in suppressing the metabolic barriers of low asparagine and excessive mitochondrial fragmentation to permit malignant KRAS-driven tumor progression.
Collapse
Affiliation(s)
- H. Helen Lin
- Department of Diabetes and Metabolic Diseases Research
| | - Yiyin Chung
- Department of Diabetes and Metabolic Diseases Research
| | | | | | - Yong Fu
- Department of Diabetes and Metabolic Diseases Research
| | | | - Kevin K. Chi
- Department of Diabetes and Metabolic Diseases Research
| | | | | | - Hsing-Jien Kung
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, Sacramento, CA USA
| | - Chien-Feng Li
- Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan
| | | | - David K. Ann
- Department of Diabetes and Metabolic Diseases Research
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA
| |
Collapse
|
33
|
Abstract
Obesity poses a severe threat to human health, including the increased prevalence of hypertension, insulin resistance, diabetes mellitus, cancer, inflammation, sleep apnoea and other chronic diseases. Current therapies focus mainly on suppressing caloric intake, but the efficacy of this approach remains poor. A better understanding of the pathophysiology of obesity will be essential for the management of obesity and its complications. Knowledge gained over the past three decades regarding the aetiological mechanisms underpinning obesity has provided a framework that emphasizes energy imbalance and neurohormonal dysregulation, which are tightly regulated by autophagy. Accordingly, there is an emerging interest in the role of autophagy, a conserved homeostatic process for cellular quality control through the disposal and recycling of cellular components, in the maintenance of cellular homeostasis and organ function by selectively ridding cells of potentially toxic proteins, lipids and organelles. Indeed, defects in autophagy homeostasis are implicated in metabolic disorders, including obesity, insulin resistance, diabetes mellitus and atherosclerosis. In this Review, the alterations in autophagy that occur in response to nutrient stress, and how these changes alter the course of obesogenesis and obesity-related complications, are discussed. The potential of pharmacological modulation of autophagy for the management of obesity is also addressed.
Collapse
Affiliation(s)
- Yingmei Zhang
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri-Columbia School of Medicine, Columbia, MO, USA
| | - Jun Ren
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| |
Collapse
|
34
|
Vera-Ramirez L, Vodnala SK, Nini R, Hunter KW, Green JE. Autophagy promotes the survival of dormant breast cancer cells and metastatic tumour recurrence. Nat Commun 2018; 9:1944. [PMID: 29789598 PMCID: PMC5964069 DOI: 10.1038/s41467-018-04070-6] [Citation(s) in RCA: 307] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 03/29/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer recurrence after initial diagnosis and treatment is a major cause of breast cancer (BC) mortality, which results from the metastatic outbreak of dormant tumour cells. Alterations in the tumour microenvironment can trigger signalling pathways in dormant cells leading to their proliferation. However, processes involved in the initial and the long-term survival of disseminated dormant BC cells remain largely unknown. Here we show that autophagy is a critical mechanism for the survival of disseminated dormant BC cells. Pharmacologic or genetic inhibition of autophagy in dormant BC cells results in significantly decreased cell survival and metastatic burden in mouse and human 3D in vitro and in vivo preclinical models of dormancy. In vivo experiments identify autophagy gene autophagy-related 7 (ATG7) to be essential for autophagy activation. Mechanistically, inhibition of the autophagic flux in dormant BC cells leads to the accumulation of damaged mitochondria and reactive oxygen species (ROS), resulting in cell apoptosis.
Collapse
Affiliation(s)
- Laura Vera-Ramirez
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Suman K Vodnala
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ryan Nini
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kent W Hunter
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Jeffrey E Green
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
35
|
|
36
|
MDA-9/Syntenin regulates protective autophagy in anoikis-resistant glioma stem cells. Proc Natl Acad Sci U S A 2018; 115:5768-5773. [PMID: 29760085 DOI: 10.1073/pnas.1721650115] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Glioma stem cells (GSCs) comprise a small subpopulation of glioblastoma multiforme cells that contribute to therapy resistance, poor prognosis, and tumor recurrence. Protective autophagy promotes resistance of GSCs to anoikis, a form of programmed cell death occurring when anchorage-dependent cells detach from the extracellular matrix. In nonadherent conditions, GSCs display protective autophagy and anoikis-resistance, which correlates with expression of melanoma differentiation associated gene-9/Syntenin (MDA-9) (syndecan binding protein; SDCBP). When MDA-9 is suppressed, GSCs undergo autophagic death supporting the hypothesis that MDA-9 regulates protective autophagy in GSCs under anoikis conditions. MDA-9 maintains protective autophagy through phosphorylation of BCL2 and by suppressing high levels of autophagy through EGFR signaling. MDA-9 promotes these changes by modifying FAK and PKC signaling. Gain-of-function and loss-of-function genetic approaches demonstrate that MDA-9 regulates pEGFR and pBCL2 expression through FAK and pPKC. EGFR signaling inhibits autophagy markers (ATG5, Lamp1, LC3B), helping to maintain protective autophagy, and along with pBCL2 maintain survival of GSCs. In the absence of MDA-9, this protective mechanism is deregulated; EGFR no longer maintains protective autophagy, leading to highly elevated and sustained levels of autophagy and consequently decreased cell survival. In addition, pBCL2 is down-regulated in the absence of MDA-9, leading to cell death in GSCs under conditions of anoikis. Our studies confirm a functional link between MDA-9 expression and protective autophagy in GSCs and show that inhibition of MDA-9 reverses protective autophagy and induces anoikis and cell death in GSCs.
Collapse
|
37
|
Zarkesh M, Zadeh-Vakili A, Azizi F, Fanaei SA, Foroughi F, Hedayati M. The Association of BRAF V600E Mutation With Tissue Inhibitor of Metalloproteinase-3 Expression and Clinicopathological Features in Papillary Thyroid Cancer. Int J Endocrinol Metab 2018; 16:e56120. [PMID: 29868127 PMCID: PMC5972213 DOI: 10.5812/ijem.56120] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 11/18/2017] [Accepted: 01/07/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Papillary thyroid cancer (PTC) is the most common endocrine malignancy. The aim of this study was to investigate the association of tissue inhibitor metalloproteinase-3 (TIMP3) mRNA and protein levels in thyroid tissues, based on BRAF V600E status with the clinicopathologic characteristics of PTC. METHODS A total of 60 fresh frozen tissue samples of PTC patients (15 male and 45 female) were collected during thyroidectomy. All clinicopathological information was obtained and samples were reviewed as well as confirmed by a pathologist; exon 15 of the BRAF gene was genotyped by sequencing, TIMP3 mRNA level was assessed using SYBR-Green Real-Time PCR, and TIMP3 protein level was measured using ELISA. RESULTS Of 60 cases, BRAF mutation was found in 24 (40%). Larger tumor size and higher lymph node metastasis frequency were observed, significant in BRAF (+), compared to the BRAF (-) PTC group (P = 0.039 and P = 0.03, respectively). No significant difference was seen in the tumoral tissues of the TIMP3 mRNA level in BRAF (+), compared to BRAF (-) PTC samples. However, the mean TIMP3 protein level was significantly lower in tumoral tissues, compared to matched non-tumoral tissues in BRAF (+) PTC (P=0.003); TIMP3 protein level was significantly lower in tumoral tissues compared to matched non-tumoral tissues in BRAF (+), in subjects who had no lymph node metastasis and also in subjects with lymph node metastasis in both BRAF positive and negative PTC cases. CONCLUSION Our results showed that BRAF mutation was associated with a larger tumor size, higher frequency of lymph node metastasis, and lower TIMP3 protein levels. Lower TIMP3 protein level was associated with the lymph node metastasis in PTC patients.
Collapse
Affiliation(s)
- Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Azita Zadeh-Vakili
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - S Ahmad Fanaei
- Association Professor of General Surgery, Erfan Hospital, Tehran, IR Iran
| | - Forough Foroughi
- Department of Pathology, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Mehdi Hedayati, Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, IR Iran. Tel: +98-2122432500, Fax: +98-2122416264, E-mail:
| |
Collapse
|
38
|
Abstract
Subtraction hybridization identified genes displaying differential expression as metastatic human melanoma cells terminally differentiated and lost tumorigenic properties by treatment with recombinant fibroblast interferon and mezerein. This approach permitted cloning of multiple genes displaying enhanced expression when melanoma cells terminally differentiated, called melanoma differentiation associated (mda) genes. One mda gene, mda-7, has risen to the top of the list based on its relevance to cancer and now inflammation and other pathological states, which based on presence of a secretory sequence, chromosomal location, and an IL-10 signature motif has been named interleukin-24 (MDA-7/IL-24). Discovered in the early 1990s, MDA-7/IL-24 has proven to be a potent, near ubiquitous cancer suppressor gene capable of inducing cancer cell death through apoptosis and toxic autophagy in cancer cells in vitro and in preclinical animal models in vivo. In addition, MDA-7/IL-24 embodied profound anticancer activity in a Phase I/II clinical trial following direct injection with an adenovirus (Ad.mda-7; INGN-241) in tumors in patients with advanced cancers. In multiple independent studies, MDA-7/IL-24 has been implicated in many pathological states involving inflammation and may play a role in inflammatory bowel disease, psoriasis, cardiovascular disease, rheumatoid arthritis, tuberculosis, and viral infection. This review provides an up-to-date review on the multifunctional gene mda-7/IL-24, which may hold potential for the therapy of not only cancer, but also other pathological states.
Collapse
|
39
|
Kuo CJ, Hansen M, Troemel E. Autophagy and innate immunity: Insights from invertebrate model organisms. Autophagy 2018; 14:233-242. [PMID: 29130360 DOI: 10.1080/15548627.2017.1389824] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Macroautophagy/autophagy is a fundamental intracellular degradation process with multiple roles in immunity, including direct elimination of intracellular microorganisms via 'xenophagy.' In this review, we summarize studies from the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans that highlight the roles of autophagy in innate immune responses to viral, bacterial, and fungal pathogens. Research from these genetically tractable invertebrates has uncovered several conserved immunological paradigms, such as direct targeting of intracellular pathogens by xenophagy and regulation of autophagy by pattern recognition receptors in D. melanogaster. Although C. elegans has no known pattern recognition receptors, this organism has been particularly useful in understanding many aspects of innate immunity. Indeed, work in C. elegans was the first to show xenophagic targeting of microsporidia, a fungal pathogen that infects all animals, and to identify TFEB/HLH-30, a helix-loop-helix transcription factor, as an evolutionarily conserved regulator of autophagy gene expression and host tolerance. Studies in C. elegans have also highlighted the more recently appreciated relationship between autophagy and tolerance to extracellular pathogens. Studies of simple, short-lived invertebrates such as flies and worms will continue to provide valuable insights into the molecular mechanisms by which autophagy and immunity pathways intersect and their contribution to organismal survival. Abbreviations Atg autophagy related BECN1 Beclin 1 CALCOCO2 calcium binding and coiled-coil domain 2 Cry5B crystal toxin 5B Daf abnormal dauer formation DKF-1 D kinase family-1 EPG-7 Ectopic P Granules-7 FuDR fluorodeoxyuridine GFP green fluorescent protein HLH-30 Helix Loop Helix-30 Imd immune deficiency ins-18 INSulin related-18; LET-363, LEThal-363 lgg-1 LC3, GABARAP and GATE-16 family-1 MAPK mitogen-activated protein kinase MATH the meprin and TRAF homology MTOR mechanistic target of rapamycin NBR1 neighbor of BRCA1 gene 1 NFKB nuclear factor of kappa light polypeptide gene enhancer in B cells NOD nucleotide-binding oligomerization domain containing OPTN optineurin PAMPs pathogen-associated molecular patterns Park2 Parkinson disease (autosomal recessive, juvenile) 2, parkin pdr-1 Parkinson disease related PFTs pore-forming toxins PGRP peptidoglycan-recognition proteins PIK3C3 phosphatidylinositol 3- kinase catalytic subunit type 3 pink-1 PINK (PTEN-I induced kinase) homolog PRKD protein kinase D; PLC, phospholipase C PRKN parkin RBR E3 ubiquitin protein ligase PRRs pattern-recognition receptors PtdIns3P phosphatidylinositol-3-phosphate rab-5 RAB family-5 RB1CC1 RB1-inducible coiled-coil 1 RNAi RNA interference sqst SeQueSTosome related SQSTM1 sequestosome 1 TBK1 TANK-binding kinase 1 TFEB transcription factor EB TGFB/TGF-β transforming growth factor beta TLRs toll-like receptors unc-51 UNCoordinated-51 VPS vacuolar protein sorting; VSV, vesicular stomatitis virus VSV-G VSV surface glycoprotein G Wipi2 WD repeat domain, phosphoinositide interacting 2.
Collapse
Affiliation(s)
- Cheng-Ju Kuo
- a Division of Biological Sciences , University of California , San Diego, La Jolla , CA , USA.,b Institute of Basic Medical Sciences , College of Medicine , National Cheng Kung University , Tainan , Taiwan
| | - Malene Hansen
- c Sanford Burnham Prebys Medical Discovery Institute, Program of Development, Aging and Regeneration , La Jolla , CA , USA
| | - Emily Troemel
- a Division of Biological Sciences , University of California , San Diego, La Jolla , CA , USA
| |
Collapse
|
40
|
Abstract
Cancer is a daunting global problem confronting the world's population. The most frequent therapeutic approaches include surgery, chemotherapy, radiotherapy, and more recently immunotherapy. In the case of chemotherapy, patients ultimately develop resistance to both single and multiple chemotherapeutic agents, which can culminate in metastatic disease which is a major cause of patient death from solid tumors. Chemoresistance, a primary cause of treatment failure, is attributed to multiple factors including decreased drug accumulation, reduced drug-target interactions, increased populations of cancer stem cells, enhanced autophagy activity, and reduced apoptosis in cancer cells. Reprogramming tumor cells to undergo drug-induced apoptosis provides a promising and powerful strategy for treating resistant and recurrent neoplastic diseases. This can be achieved by downregulating dysregulated antiapoptotic factors or activation of proapoptotic factors in tumor cells. A major target of dysregulation in cancer cells that can occur during chemoresistance involves altered expression of Bcl-2 family members. Bcl-2 antiapoptotic molecules (Bcl-2, Bcl-xL, and Mcl-1) are frequently upregulated in acquired chemoresistant cancer cells, which block drug-induced apoptosis. We presently overview the potential role of Bcl-2 antiapoptotic proteins in the development of cancer chemoresistance and overview the clinical approaches that use Bcl-2 inhibitors to restore cell death in chemoresistant and recurrent tumors.
Collapse
|
41
|
Bonavida B. Linking Autophagy and the Dysregulated NFκB/ SNAIL/YY1/RKIP/PTEN Loop in Cancer: Therapeutic Implications. Crit Rev Oncog 2018; 23:307-320. [PMID: 30311562 PMCID: PMC6370039 DOI: 10.1615/critrevoncog.2018027212] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The role of autophagy in the pathogenesis of various cancers has been well documented in many reports. Autophagy in cancer cells regulates cell proliferation, viability, invasion, epithelial-to-mesenchymal transition (EMT), metastasis, and responses to chemotherapeutic and immunotherapeutic treatment strategies. These manifestations are the result of various regulatory gene products that govern autophagic, biochemical, and molecular mechanisms. In several human cancer cell models, the presence of a dysregulated circuit-namely, NFκB/SNAIL/YY1/RKIP/PTEN-that plays a major role in the regulation of tumor cell unique characteristics just listed for autophagy-regulated activities. Accordingly, the autophagic mechanism and the dysregulated circuit in cancer cells share many of the same properties and activities. Thus, it has been hypothesized that there must exist a biochemical/molecular link between the two. The present review describes the link and the association of each gene product of the dysregulated circuit with the autophagic mechanism and delineates the presence of crosstalk. Crosstalk between autophagy and the dysregulated circuit is significant and has important implications in the development of targeted therapies aimed at either autophagy or the dysregulated gene products in cancer cells.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90025-1747,
| |
Collapse
|
42
|
Wechman SL, Pradhan AK, DeSalle R, Das SK, Emdad L, Sarkar D, Fisher PB. New Insights Into Beclin-1: Evolution and Pan-Malignancy Inhibitor Activity. Adv Cancer Res 2017; 137:77-114. [PMID: 29405978 DOI: 10.1016/bs.acr.2017.11.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autophagy is a functionally conserved self-degradation process that facilitates the survival of eukaryotic life via the management of cellular bioenergetics and maintenance of the fidelity of genomic DNA. The first known autophagy inducer was Beclin-1. Beclin-1 is expressed in multicellular eukaryotes ranging throughout plants to animals, comprising a nonmonophyllic group, as shown in this report via aggressive BLAST searches. In humans, Beclin-1 is a haploinsuffient tumor suppressor as biallelic deletions have not been observed in patient tumors clinically. Therefore, Beclin-1 fails the Knudson hypothesis, implicating expression of at least one Beclin-1 allele is essential for cancer cell survival. However, Beclin-1 is frequently monoallelically deleted in advanced human cancers and the expression of two Beclin-1 allelles is associated with greater anticancer effects. Overall, experimental evidence suggests that Beclin-1 inhibits tumor formation, angiogenesis, and metastasis alone and in cooperation with the tumor suppressive molecules UVRAG, Bif-1, Ambra1, and MDA-7/IL-24 via diverse mechanisms of action. Conversely, Beclin-1 is upregulated in cancer stem cells (CSCs), portending a role in cancer recurrence, and highlighting this molecule as an intriguing molecular target for the treatment of CSCs. Many aspects of Beclin-1's biological effects remain to be studied. The consequences of these BLAST searches on the molecular evolution of Beclin-1, and the eukaryotic branches of the tree of life, are discussed here in greater detail with future inquiry focused upon protist taxa. Also in this review, the effects of Beclin-1 on tumor suppression and cancer malignancy are discussed. Beclin-1 holds significant promise for the development of novel targeted cancer therapeutics and is anticipated to lead to a many advances in our understanding of eukaryotic evolution, multicellularity, and even the treatment of CSCs in the coming decades.
Collapse
Affiliation(s)
- Stephen L Wechman
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Anjan K Pradhan
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Rob DeSalle
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, United States
| | - Swadesh K Das
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Luni Emdad
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Devanand Sarkar
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
43
|
She T, Feng J, Lian S, Li R, Zhao C, Song G, Luo J, Dawuti R, Cai S, Qu L, Shou C. Sarsaparilla (Smilax Glabra Rhizome) Extract Activates Redox-Dependent ATM/ATR Pathway to Inhibit Cancer Cell Growth by S Phase Arrest, Apoptosis, and Autophagy. Nutr Cancer 2017; 69:1281-1289. [PMID: 29111814 DOI: 10.1080/01635581.2017.1362447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Tiantian She
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Junnan Feng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Shenyi Lian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ruobing Li
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Chuanke Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Guoliang Song
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Jie Luo
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Rouxianguli Dawuti
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Shaoqing Cai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Like Qu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chengchao Shou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
44
|
Blagoev KB, Wilkerson J, Burotto M, Kim C, Espinal-Domínguez E, García-Alfonso P, Alimchandani M, Miettinen M, Blanco-Codesido M, Fojo T. Neutral evolution of drug resistant colorectal cancer cell populations is independent of their KRAS status. PLoS One 2017; 12:e0175484. [PMID: 28981524 PMCID: PMC5628783 DOI: 10.1371/journal.pone.0175484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/27/2017] [Indexed: 01/13/2023] Open
Abstract
Emergence of tumor resistance to an anti-cancer therapy directed against a putative target raises several questions including: (1) do mutations in the target/pathway confer resistance? (2) Are these mutations pre-existing? (3) What is the relative fitness of cells with/without the mutation? We addressed these questions in patients with metastatic colorectal cancer (mCRC). We conducted an exhaustive review of published data to establish a median doubling time for CRCs and stained a cohort of CRCs to document mitotic indices. We analyzed published data and our own data to calculate rates of growth (g) and regression (d, decay) of tumors in patients with CRC correlating these results with the detection of circulating MT-KRAS DNA. Additionally we estimated mathematically the caloric burden of such tumors using data on mitotic and apoptotic indices. We conclude outgrowth of cells harboring intrinsic or acquired MT-KRAS cannot explain resistance to anti-EGFR (epidermal growth factor receptor) antibodies. Rates of tumor growth with panitumumab are unaffected by presence/absence of MT-KRAS. While MT-KRAS cells may be resistant to anti-EGFR antibodies, WT-KRAS cells also rapidly bypass this blockade suggesting inherent resistance mechanisms are responsible and a neutral evolution model is most appropriate. Using the above clinical data on tumor doubling times and mitotic and apoptotic indices we estimated the caloric intake required to support tumor growth and suggest it may explain in part cancer-associated cachexia.
Collapse
Affiliation(s)
- Krastan B. Blagoev
- Physics of Living Systems, National Science Foundation, Arlington, Virginia, United States of America
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| | - Julia Wilkerson
- Medical Oncology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, United States of America
| | - Mauricio Burotto
- Departamento de Oncologia, Clinica Alemana de Santiago, Santiago, Chile
| | - Chul Kim
- Medical Oncology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, United States of America
| | | | - Pilar García-Alfonso
- Departamento de Oncologia Medica, Gregorio Marañon University Hospital, Madrid, Spain
| | - Meghna Alimchandani
- Center for Biologics Evaluation and Research, US Food and Drug Administration (USFDA), Silver Spring, Maryland, United States of America
| | - Markku Miettinen
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, United States of America
| | | | - Tito Fojo
- Division of Hematology and Oncology, Department of Medicine, Columbia University, New York and James J. Peters VA Medical Center, Bronx, New York, United States of America
| |
Collapse
|
45
|
Abstract
Tumor-associated inflammation is predictive of poor prognosis and drives a variety of tumorigenic phenotypes, including tumor proliferation and survival, angiogenesis, invasiveness, and metastasis. Here, we review mammalian data addressing the interaction of macroautophagy/autophagy with key signaling cascades associated with tumor inflammation. Although our understanding of this area remains incomplete, certain inflammatory pathways have emerged as important mediators of the crosstalk between autophagy and inflammation in tumors. Consistent with the multifaceted roles for autophagy in tumor cells, results to date support the hypothesis that inflammatory pathways can suppress or induce autophagy in a context-dependent manner; in turn, autophagy suppresses or promotes inflammation in cancers. Furthermore, emerging data suggest that autophagy may influence cytokine production and secretion via diverse mechanisms, which has implications for the immune and inflammatory microenvironment in tumors.
Collapse
Affiliation(s)
- Teresa Monkkonen
- a Department of Pathology and Helen Diller Family Comprehensive Cancer Center , University of California San Francisco , San Francisco , CA , USA
| | - Jayanta Debnath
- a Department of Pathology and Helen Diller Family Comprehensive Cancer Center , University of California San Francisco , San Francisco , CA , USA
| |
Collapse
|
46
|
Monitoring Autophagy Immunohistochemically and Ultrastructurally during Human Head and Neck Carcinogenesis. Relationship with the DNA Damage Response Pathway. Int J Mol Sci 2017; 18:ijms18091920. [PMID: 28880214 PMCID: PMC5618569 DOI: 10.3390/ijms18091920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/01/2017] [Accepted: 09/03/2017] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a catabolic process that preserves cellular homeostasis. Its exact role during carcinogenesis is not completely defined. Specifically in head and neck cancer, such information from clinical settings that comprise the whole spectrum of human carcinogenesis is very limited. Towards this direction, we examined the in situ status of the autophagy-related factors, Beclin-1, microtubule-associated protein 1 light chain 3, member B (LC3B) and sequestosome 1/p62 (p62) in clinical material covering all histopathological stages of human head and neck carcinogenesis. This material is unique as each panel of lesions is derived from the same patient and moreover we have previously assessed it for the DNA damage response (DDR) activation status. Since Beclin-1, LC3B and p62 reflect the nucleation, elongation and degradation stages of autophagy, respectively, their combined immunohistochemical (IHC) expression profiles could grossly mirror the autophagic flux. This experimental approach was further corroborated by ultrastructural analysis, applying transmission electron microscopy (TEM). The observed Beclin-1/LC3B/p62 IHC patterns, obtained from serial sections analysis, along with TEM findings are suggestive of a declined authophagic activity in preneoplastic lesions that was restored in full blown cancers. Correlating these findings with DDR status in the same pathological stages are indicative of: (i) an antitumor function of autophagy in support to that of DDR, possibly through energy deprivation in preneoplastic stages, thus preventing incipient cancer cells from evolving; and (ii) a tumor-supporting role in the cancerous stage.
Collapse
|
47
|
An oasis in the desert of cancer chemotherapeutic resistance: The enlightenment from reciprocal crosstalk between signaling pathways of UPR and autophagy in cancers. Biomed Pharmacother 2017; 92:972-981. [DOI: 10.1016/j.biopha.2017.05.132] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/28/2017] [Accepted: 05/28/2017] [Indexed: 12/21/2022] Open
|
48
|
Kimmelman AC, White E. Autophagy and Tumor Metabolism. Cell Metab 2017; 25:1037-1043. [PMID: 28467923 PMCID: PMC5604466 DOI: 10.1016/j.cmet.2017.04.004] [Citation(s) in RCA: 628] [Impact Index Per Article: 78.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 03/19/2017] [Accepted: 04/05/2017] [Indexed: 02/08/2023]
Abstract
Autophagy is a critical cellular process that generally protects cells and organisms from stressors such as nutrient deprivation. In addition to its role in normal physiology, autophagy plays a role in pathological processes such as cancer. Indeed, there has been substantial work exploring the complex and context-dependent role of autophagy in cancer. One of the emerging themes is that in certain cancer types, autophagy is important to support tumor growth; therefore, inhibiting autophagy as a therapeutic approach is actively being tested in clinical trials. A key mechanism of how autophagy promotes the growth and survival of various cancers is its ability to support cellular metabolism. The diverse metabolic fuel sources that can be produced by autophagy provide tumors with metabolic plasticity and can allow them to thrive in what can be an austere microenvironment. Therefore, understanding how autophagy can fuel cellular metabolism will enable more effective combinatorial therapeutic strategies.
Collapse
Affiliation(s)
- Alec C Kimmelman
- Perlmutter Cancer Center, Department of Radiation Oncology, NYU Medical School, New York, NY 10016, USA.
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA; Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
49
|
Kellner R, De la Concepcion JC, Maqbool A, Kamoun S, Dagdas YF. ATG8 Expansion: A Driver of Selective Autophagy Diversification? TRENDS IN PLANT SCIENCE 2017; 22:204-214. [PMID: 28038982 DOI: 10.1016/j.tplants.2016.11.015] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 11/24/2016] [Accepted: 11/28/2016] [Indexed: 05/18/2023]
Abstract
Selective autophagy is a conserved homeostatic pathway that involves engulfment of specific cargo molecules into specialized organelles called autophagosomes. The ubiquitin-like protein ATG8 is a central player of the autophagy network that decorates autophagosomes and binds to numerous cargo receptors. Although highly conserved across eukaryotes, ATG8 diversified from a single protein in algae to multiple isoforms in higher plants. We present a phylogenetic overview of 376 ATG8 proteins across the green plant lineage that revealed family-specific ATG8 clades. Because these clades differ in fixed amino acid polymorphisms, they provide a mechanistic framework to test whether distinct ATG8 clades are functionally specialized. We propose that ATG8 expansion may have contributed to the diversification of selective autophagy pathways in plants.
Collapse
Affiliation(s)
- Ronny Kellner
- The Sainsbury Laboratory, Norwich Research Park, Norwich NR4 7UH, UK; Max Planck Institute for Plant Breeding Research, Carl-von-Linné-Weg 10, Cologne 50829, Germany
| | - Juan Carlos De la Concepcion
- The Sainsbury Laboratory, Norwich Research Park, Norwich NR4 7UH, UK; John Innes Centre, Department of Biological Chemistry, Norwich Research Park, Norwich NR4 7UH, UK
| | - Abbas Maqbool
- John Innes Centre, Department of Biological Chemistry, Norwich Research Park, Norwich NR4 7UH, UK
| | - Sophien Kamoun
- The Sainsbury Laboratory, Norwich Research Park, Norwich NR4 7UH, UK.
| | - Yasin F Dagdas
- The Sainsbury Laboratory, Norwich Research Park, Norwich NR4 7UH, UK; The Gregor Mendel Institute of Molecular Plant Biology, Dr. Bohr-Gasse 3, 1030, Vienna, Austria.
| |
Collapse
|
50
|
Gaballah HH, Gaber RA, Mohamed DA. Apigenin potentiates the antitumor activity of 5-FU on solid Ehrlich carcinoma: Crosstalk between apoptotic and JNK-mediated autophagic cell death platforms. Toxicol Appl Pharmacol 2017; 316:27-35. [DOI: 10.1016/j.taap.2016.12.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/17/2016] [Accepted: 12/19/2016] [Indexed: 12/22/2022]
|