501
|
Zheng R, Li F, Li F, Gong A. Targeting tumor vascularization: promising strategies for vascular normalization. J Cancer Res Clin Oncol 2021; 147:2489-2505. [PMID: 34148156 DOI: 10.1007/s00432-021-03701-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022]
Abstract
Tumor recurrence after the clinical cure of tumor often results from the presence of an abnormal microenvironment, including an aberrant vasculature. The tumor microenvironment is rich in pro-angiogenic factors but lacks pro-maturation factors. Pro-angiogenic conditions in the tumor microenvironment, such as hypoxia, are double-edged swords, promoting both the repair of normal tissues and the development of an abnormal blood vessel network. The coexistence of perfusion and hypoxic zones and uneven blood vessel distribution in tumor tissues profoundly influence tumor deterioration, recurrence, and metastasis. Traditional anti-angiogenic therapies have shown limited efficacy, and promote drug resistance, and even metastasis. In contrast, vascular normalization therapy induces a more physiological-like state, leading to better outcomes and fewer side effects. Vascular normalization entails modifying the tumor vascular system to improve tumor oxygenation and substance transport, thereby contributing to improving the efficacy of radiotherapy, chemotherapy, and immunotherapy. This review mainly focuses on the process of tumor vascularization; potential therapeutic targets, including cells, metabolism, signaling pathways, and angiogenesis-related genes; and possible strategies to normalize blood vessels through regulating tumor vessel generation, the development of tumor vessels, and blood vessel fusion and pruning.
Collapse
Affiliation(s)
- Ruiqi Zheng
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Feifan Li
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Fengcen Li
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Aihua Gong
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China.
| |
Collapse
|
502
|
Tran S, Fairlie WD, Lee EF. BECLIN1: Protein Structure, Function and Regulation. Cells 2021; 10:cells10061522. [PMID: 34204202 PMCID: PMC8235419 DOI: 10.3390/cells10061522] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023] Open
Abstract
BECLIN1 is a well-established regulator of autophagy, a process essential for mammalian survival. It functions in conjunction with other proteins to form Class III Phosphoinositide 3-Kinase (PI3K) complexes to generate phosphorylated phosphatidylinositol (PtdIns), lipids essential for not only autophagy but other membrane trafficking processes. Over the years, studies have elucidated the structural, biophysical, and biochemical properties of BECLIN1, which have shed light on how this protein functions to allosterically regulate these critical processes of autophagy and membrane trafficking. Here, we review these findings and how BECLIN1’s diverse protein interactome regulates it, as well as its impact on organismal physiology.
Collapse
Affiliation(s)
- Sharon Tran
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
| | - W. Douglas Fairlie
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
- Correspondence: (W.D.F.); (E.F.L.)
| | - Erinna F. Lee
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
- Correspondence: (W.D.F.); (E.F.L.)
| |
Collapse
|
503
|
Xu S, Tang L, Liu Z, Yang K, Cheng Q. Bioinformatic Analyses Identify a Prognostic Autophagy-Related Long Non-coding RNA Signature Associated With Immune Microenvironment in Diffuse Gliomas. Front Cell Dev Biol 2021; 9:694633. [PMID: 34211979 PMCID: PMC8239411 DOI: 10.3389/fcell.2021.694633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/24/2021] [Indexed: 12/22/2022] Open
Abstract
Background Autophagy and long non-coding RNA (lncRNA) play a critical role in tumor progression and microenvironment. However, the role of autophagy-related lncRNAs (ARLs) in glioma microenvironment remains unclear. Methods A total of 988 diffuse glioma samples were extracted from TCGA and CGGA databases. Consensus clustering was applied to reveal different subgroups of diffuse gliomas. Kaplan-Meier analysis was used to evaluate survival differences between groups. The infiltration of immune cells was estimated by ssGSEA, TIMER, and CIBERSORT algorithms. The construction of ARL signature was conducted using principal component analysis. Results Consensus clustering revealed two clusters of diffuse gliomas, in which cluster 1 was associated with poor prognosis and enriched with malignant subtypes of gliomas. Moreover, cluster 1 exhibited high apoptotic and immune characteristics, and it had a low purity and high infiltration of several immune cells. The constructed ARL signature showed a promising accuracy in predicting the prognosis of glioma patients. ARL score was significantly elevated in the malignant subtype of glioma and the high ARL score indicated a poor prognosis. Besides, the high ARL score notably indicated low tumor purity and high infiltration of macrophages and neutrophils. Conclusion Our study developed and validated a novel ARL signature for the classification of diffuse glioma, which was closely associated with glioma immune microenvironment and could serve as a promising prognostic biomarker for glioma patients.
Collapse
Affiliation(s)
- Shengchao Xu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Lu Tang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Kui Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
504
|
Shi Y, Shen HM, Gopalakrishnan V, Gordon N. Epigenetic Regulation of Autophagy Beyond the Cytoplasm: A Review. Front Cell Dev Biol 2021; 9:675599. [PMID: 34195194 PMCID: PMC8237754 DOI: 10.3389/fcell.2021.675599] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved catabolic process induced under various stress conditions to protect the cell from harm and allow survival in the face of nutrient- or energy-deficient states. Regulation of autophagy is complex, as cells need to adapt to a continuously changing microenvironment. It is well recognized that the AMPK and mTOR signaling pathways are the main regulators of autophagy. However, various other signaling pathways have also been described to regulate the autophagic process. A better understanding of these complex autophagy regulatory mechanisms will allow the discovery of new potential therapeutic targets. Here, we present a brief overview of autophagy and its regulatory pathways with emphasis on the epigenetic control mechanisms.
Collapse
Affiliation(s)
- Yin Shi
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Vidya Gopalakrishnan
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nancy Gordon
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
505
|
Liao YX, Lv JY, Zhou ZF, Xu TY, Yang D, Gao QM, Fan L, Li GD, Yu HY, Liu KY. CXCR4 blockade sensitizes osteosarcoma to doxorubicin by inducing autophagic cell death via PI3K‑Akt‑mTOR pathway inhibition. Int J Oncol 2021; 59:49. [PMID: 34080667 PMCID: PMC8208619 DOI: 10.3892/ijo.2021.5229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022] Open
Abstract
Doxorubicin is one of the most frequently used chemotherapy drugs in the treatment of osteosarcoma (OS), but the emergence of chemoresistance often leads to treatment failure. C-X-C motif chemokine receptor 4 (CXCR4) has been demonstrated to regulate OS progression and metastasis. However, whether CXCR4 is also involved in OS chemoresistance and its molecular mechanisms has yet to be fully elucidated. In the present study, CXCR4-mediated autophagy for OS chemotherapy was investigated by western blot analysis, transmission electron microscopy and confocal microscopy. CXCR4 silencing enhanced doxorubicin-induced apoptosis by reducing P-glycoprotein in CXCR4+ LM8 cells, while CXCR4 overexpression promoted OS doxorubicin resistance in CXCR4−Dunn cells. Furthermore, CXCR4 silencing with or without doxorubicin increased the expression of beclin 1 and light chain 3B, and the number of autophagosomes and autolysosomes, as well as induced autophagic flux activation by suppressing the PI3K/AKT/mTOR signaling pathway. In addition, pretreatment with the autophagy inhibitor bafilomycin A1 attenuated CXCR4 abrogation-induced cell death. Finally, the CXCR4 antagonist AMD3100 synergistically reinforced the antitumor effect of doxorubicin in an orthotopic OS mouse model. Taken together, the present study revealed that CXCR4 inhibition sensitizes OS to doxorubicin by inducing autophagic cell death. Therefore, targeting the CXCR4/autophagy axis may be a promising therapeutic strategy to overcome OS chemotherapy resistance.
Collapse
Affiliation(s)
- Yu-Xin Liao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Ji-Yang Lv
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Zi-Fei Zhou
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Tian-Yang Xu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Dong Yang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Qiu-Ming Gao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Lin Fan
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Guo-Dong Li
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Hai-Yang Yu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Kai-Yuan Liu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| |
Collapse
|
506
|
Jiménez-Mora E, Gallego B, Díaz-Gago S, Lasa M, Baquero P, Chiloeches A. V600EBRAF Inhibition Induces Cytoprotective Autophagy through AMPK in Thyroid Cancer Cells. Int J Mol Sci 2021; 22:ijms22116033. [PMID: 34204950 PMCID: PMC8199856 DOI: 10.3390/ijms22116033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/17/2022] Open
Abstract
The dysregulation of autophagy is important in the development of many cancers, including thyroid cancer, where V600EBRAF is a main oncogene. Here, we analyse the effect of V600EBRAF inhibition on autophagy, the mechanisms involved in this regulation and the role of autophagy in cell survival of thyroid cancer cells. We reveal that the inhibition of V600EBRAF activity with its specific inhibitor PLX4720 or the depletion of its expression by siRNA induces autophagy in thyroid tumour cells. We show that V600EBRAF downregulation increases LKB1-AMPK signalling and decreases mTOR activity through a MEK/ERK-dependent mechanism. Moreover, we demonstrate that PLX4720 activates ULK1 and increases autophagy through the activation of the AMPK-ULK1 pathway, but not by the inhibition of mTOR. In addition, we find that autophagy blockade decreases cell viability and sensitize thyroid cancer cells to V600EBRAF inhibition by PLX4720 treatment. Finally, we generate a thyroid xenograft model to demonstrate that autophagy inhibition synergistically enhances the anti-proliferative and pro-apoptotic effects of V600EBRAF inhibition in vivo. Collectively, we uncover a new role of AMPK in mediating the induction of cytoprotective autophagy by V600EBRAF inhibition. In addition, these data establish a rationale for designing an integrated therapy targeting V600EBRAF and the LKB1-AMPK-ULK1-autophagy axis for the treatment of V600EBRAF-positive thyroid tumours.
Collapse
Affiliation(s)
- Eva Jiménez-Mora
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Beatriz Gallego
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Sergio Díaz-Gago
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Marina Lasa
- Departamento de Bioquímica-Instituto de Investigaciones Biomédicas “Alberto Sols”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28029 Madrid, Spain;
| | - Pablo Baquero
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
| | - Antonio Chiloeches
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina, Campus Universitario, Universidad de Alcalá, Alcalá de Henares, 28871 Madrid, Spain; (E.J.-M.); (B.G.); (S.D.-G.); (P.B.)
- Correspondence:
| |
Collapse
|
507
|
Liu L, Zhang J, Liu H, Shi M, Zhang J, Chen L, Huang L, Li B, Xu P. Correlation of autophagy-related genes for predicting clinical prognosis in colorectal cancer. Biomark Med 2021; 15:715-729. [PMID: 34169735 DOI: 10.2217/bmm-2020-0292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 03/19/2021] [Indexed: 12/24/2022] Open
Abstract
Aim: Autophagy plays a controversial role in cancer. The role of autophagy-related genes (ARGs) in colorectal cancer (CRC) was evaluated based on publicly available data from The Cancer Genome Atlas and the Human Autophagy Database. Materials & methods: After collecting CRC-related transcript and clinical data and a list of ARGs from public databases, the Wilcoxon test was used to identify the differentially expressed ARGs between CRC and paired normal tissues. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were used to identify the major biological properties and pathways associated with these genes. Univariate Cox regression was used to identify the prognosis-associated ARGs, and a forest plot was used to visualize the results. Kaplan-Meier analysis of the 5-year survival rate was performed. Univariate and multivariate Cox analyses were used to verify the impact of the prognosis-associated ARGs. Results: A total of 36 differentially expressed genes (16 upregulated and 20 downregulated in CRC) were obtained from among 206 ARGs. There were 53 enriched pathways, including the p53 signaling pathway, platinum drug resistance, apoptosis, EGFR tyrosine kinase inhibitor resistance and ErbB signaling pathway (p- and q-values <0.05). Kaplan-Meier analysis showed that the 5-year survival rate was 46.0% (95% CI: 0.335-0.631) and 76.0% (95% CI: 0.651-0.886) in the high- and low-risk groups, respectively. The high-risk patients had worse survival probability (p = 6.256 × 10-5). Independent-samples t-tests revealed that MAP1LC3C expression was higher in patients aged ≤65 than >65 (p = 0.022); RAB7A expression was higher in patients aged ≤65 than >65 (p = 7.31 × 10-4), higher in M1 than M0 (p = 0.042), higher in N1-3 than N0 (p = 0.002) and higher in stage III and IV than I and II (p = 0.042); risk score was higher in N1-3 than N0 (p = 0.001) and in stage III and IV than I and II (p = 0.002); and WIPI2 expression was higher in M1 than M0 (p = 0.002), higher in N1-3 than N0 (p = 2.059 × 10-7) and higher in stage III and IV than I and II (p = 2.299 × 10-7). There were no differences in risk score between males and females (p = 0.593), T1-2 and T3-4 (p = 0.082) or M0 and M1 (p = 0.072). Univariate and multivariate Cox analyses showed that RAB7A was a lower-risk gene, while MAP1LC3C, WIPI2, DAPK1, ULK3 and PELP1 were high-risk genes. Conclusion: Certain ARGs are potential prognostic molecular markers of poor prognosis in CRC. Additionally, the p53 signaling pathway, platinum drug resistance, apoptosis, EGFR tyrosine kinase inhibitor resistance and ErbB signaling pathway may be critical pathways regulated by ARGs in CRC.
Collapse
Affiliation(s)
- Liyan Liu
- Department of Pharmacy, Jiangxi Cancer Hospital, 519 Beijing East Road, Nanchang, 330029, PR China
- Department of Pharmacy, Affiliated Cancer Hospital of Nanchang University, 519 Beijing East Road, Nanchang, 330029, PR China
- Laboratory Animal Science & Technology Center, Workstation of Academician, College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Jilin Zhang
- Department of Traditional Chinese Medicine, Jiangxi Provincial People's Hospital, 92 Aiguo Road, Nanchang, 330006, PR China
| | - Hongdong Liu
- Laboratory Animal Science & Technology Center, Workstation of Academician, College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Min Shi
- Laboratory Animal Science & Technology Center, Workstation of Academician, College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Jie Zhang
- Laboratory Animal Science & Technology Center, Workstation of Academician, College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Li Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, 611137, PR China
| | - Luqi Huang
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medical, China Academy of Chinese Medical Sciences, 16 Nanxiao Street, Dongzhimen, Dongcheng District, Beijing, 100700, PR China
| | - Bin Li
- Laboratory Animal Science & Technology Center, Workstation of Academician, College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Peng Xu
- Laboratory Animal Science & Technology Center, Workstation of Academician, College of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| |
Collapse
|
508
|
Martins WK, Silva MDND, Pandey K, Maejima I, Ramalho E, Olivon VC, Diniz SN, Grasso D. Autophagy-targeted therapy to modulate age-related diseases: Success, pitfalls, and new directions. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100033. [PMID: 34909664 PMCID: PMC8663935 DOI: 10.1016/j.crphar.2021.100033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 04/15/2021] [Accepted: 05/02/2021] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a critical metabolic process that supports homeostasis at a basal level and is dynamically regulated in response to various physiological and pathological processes. Autophagy has some etiologic implications that support certain pathological processes due to alterations in the lysosomal-degradative pathway. Some of the conditions related to autophagy play key roles in highly relevant human diseases, e.g., cardiovascular diseases (15.5%), malignant and other neoplasms (9.4%), and neurodegenerative conditions (3.7%). Despite advances in the discovery of new strategies to treat these age-related diseases, autophagy has emerged as a therapeutic option after preclinical and clinical studies. Here, we discuss the pitfalls and success in regulating autophagy initiation and its lysosome-dependent pathway to restore its homeostatic role and mediate therapeutic effects for cancer, neurodegenerative, and cardiac diseases. The main challenge for the development of autophagy regulators for clinical application is the lack of specificity of the repurposed drugs, due to the low pharmacological uniqueness of their target, including those that target the PI3K/AKT/mTOR and AMPK pathway. Then, future efforts must be conducted to deal with this scenery, including the disclosure of key components in the autophagy machinery that may intervene in its therapeutic regulation. Among all efforts, those focusing on the development of novel allosteric inhibitors against autophagy inducers, as well as those targeting autolysosomal function, and their integration into therapeutic regimens should remain a priority for the field.
Collapse
Affiliation(s)
- Waleska Kerllen Martins
- Laboratory of Cell and Membrane (LCM), Anhanguera University of São Paulo (UNIAN), Rua Raimundo Pereira de Magalhães, 3,305. Pirituba, São Paulo, 05145-200, Brazil
| | - Maryana do Nascimento da Silva
- Laboratory of Cell and Membrane (LCM), Anhanguera University of São Paulo (UNIAN), Rua Raimundo Pereira de Magalhães, 3,305. Pirituba, São Paulo, 05145-200, Brazil
| | - Kiran Pandey
- Center for Neural Science, New York University, Meyer Building, Room 823, 4 Washington Place, New York, NY, 10003, USA
| | - Ikuko Maejima
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa Machi, Maebashi, Gunma, 3718512, Japan
| | - Ercília Ramalho
- Laboratory of Cell and Membrane (LCM), Anhanguera University of São Paulo (UNIAN), Rua Raimundo Pereira de Magalhães, 3,305. Pirituba, São Paulo, 05145-200, Brazil
| | - Vania Claudia Olivon
- Laboratory of Pharmacology and Physiology, UNIDERP, Av. Ceará, 333. Vila Miguel Couto, Campo Grande, MS, 79003-010, Brazil
| | - Susana Nogueira Diniz
- Laboratory of Molecular Biology and Functional Genomics, Anhanguera University of São Paulo (UNIAN), Rua Raimundo Pereira de Magalhães, 3,305. Pirituba, São Paulo, 05145-200, Brazil
| | - Daniel Grasso
- Instituto de Estudios de la Inmunidad Humoral (IDEHU), Universidad de Buenos Aires, CONICET, Junín 954 p4, Buenos Aires, C1113AAD, Argentina
| |
Collapse
|
509
|
Hao L, Zhong YM, Tan CP, Mao ZW. Acidity-responsive phosphorescent metal complexes for cancer imaging and theranostic applications. J Organomet Chem 2021. [DOI: 10.1016/j.jorganchem.2021.121821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
510
|
Li Q, Wang L, Ji D, Bao X, Tan G, Liang X, Deng P, Pi H, Lu Y, Chen C, He M, Zhang L, Zhou Z, Yu Z, Deng A. BIX-01294, a G9a inhibitor, suppresses cell proliferation by inhibiting autophagic flux in nasopharyngeal carcinoma cells. Invest New Drugs 2021; 39:686-696. [PMID: 33387131 DOI: 10.1007/s10637-020-01053-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/15/2020] [Indexed: 10/22/2022]
Abstract
G9a, a histone methyltransferase, has been found to be upregulated in a range of tumor tissues, and contributes to tumor growth and metastasis. However, the impact of G9a inhibition as a potential therapeutic target in nasopharyngeal carcinoma (NPC) is unclear. In the present study we aimed to investigate the anti-proliferative effect of G9a inhibition in the NPC cell lines CNE1 and CNE2, and to further elucidate the molecular mechanisms underlying these effects. The expression of G9a in NPC tumor tissues was significantly higher than that in normal nasopharyngeal tissues. The pharmacological inhibition of G9a by BIX-01294 (BIX) inhibited proliferation and induced caspase-independent apoptosis in NPC cells in vitro. Treatment with BIX induced autophagosome accumulation, which exacerbated the cytotoxic activity of BIX in NPC cells. Mechanistic studies have found that BIX impairs autophagosomes by initiating autophagy in a Beclin-1-independent way, and impairs autophagic degradation by inhibiting lysosomal cathepsin D activation, leading to lysosomal dysfunction. BIX was able to suppress tumor growth, possibly by inhibiting autophagic flux; it might therefore constitute a promising candidate for NPC therapy.
Collapse
Affiliation(s)
- Qian Li
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Liuqian Wang
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Di Ji
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaomin Bao
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Guojing Tan
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaojun Liang
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yonghui Lu
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chunhai Chen
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mindi He
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lei Zhang
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhou Zhou
- Department of Environmental Medicine, Department of Emergency Medicine of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengping Yu
- Department of Occupational Health, Army Medical University (Third Military Medical University), Chongqing, China
| | - Anchun Deng
- Department of Otolaryngology Head and Neck Surgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
511
|
Construction and Validation of an Autophagy-Related Prognostic Model for Osteosarcoma Patients. JOURNAL OF ONCOLOGY 2021; 2021:9943465. [PMID: 34194501 PMCID: PMC8181090 DOI: 10.1155/2021/9943465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022]
Abstract
While the prognostic value of autophagy-related genes (ARGs) in OS patients remains scarcely known, increasing evidence is indicating that autophagy is closely associated with the development and progression of osteosarcoma (OS). Therefore, we explored the prognostic value of ARGs in OS patients and illuminate associated mechanisms in this study. When the OS patients in the training/validation cohort were stratified into high- and low-risk groups according to the risk model established using least absolute shrinkage and selection operator (LASSO) regression analysis, we observed that patients in the low-risk group possessed better prognosis (P < 0.0001). Univariate/Multivariate COX regression and subgroup analysis demonstrated that the ARGs-based risk model was an independent survival indicator for OS patients. The nomogram incorporating the risk model and clinical features exhibited excellent prognostic accuracy. GO, KEGG, and GSVA analyses collectively indicated that bone development-associated pathway mediated the contribution of ARGs to the malignance of OS. Immune infiltration analysis suggested the potential pivotal role of macrophage in OS. In summary, the risk model based on 12 ARGs possessed potent capacity in predicting the prognosis of OS patients. Our work may assist clinicians to map out more reasonable treatment strategies and facilitate individual-targeted therapy in osteosarcoma.
Collapse
|
512
|
Zhang GZ, Wu ZL, Li CY, Ren EH, Yuan WH, Deng YJ, Xie QQ. Development of a Machine Learning-Based Autophagy-Related lncRNA Signature to Improve Prognosis Prediction in Osteosarcoma Patients. Front Mol Biosci 2021; 8:615084. [PMID: 34095215 PMCID: PMC8176230 DOI: 10.3389/fmolb.2021.615084] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/29/2021] [Indexed: 12/15/2022] Open
Abstract
Background Osteosarcoma is a frequent bone malignancy in children and young adults. Despite the availability of some prognostic biomarkers, most of them fail to accurately predict prognosis in osteosarcoma patients. In this study, we used bioinformatics tools and machine learning algorithms to establish an autophagy-related long non-coding RNA (lncRNA) signature to predict the prognosis of osteosarcoma patients. Methods We obtained expression and clinical data from osteosarcoma patients in the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and Gene Expression Omnibus (GEO) databases. We acquired an autophagy gene list from the Human Autophagy Database (HADb) and identified autophagy-related lncRNAs by co-expression analyses. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the autophagy-related lncRNAs were conducted. Univariate and multivariate Cox regression analyses were performed to assess the prognostic value of the autophagy-related lncRNA signature and validate the relationship between the signature and osteosarcoma patient survival in an independent cohort. We also investigated the relationship between the signature and immune cell infiltration. Results We initially identified 69 autophagy-related lncRNAs, 13 of which were significant predictors of overall survival in osteosarcoma patients. Kaplan-Meier analyses revealed that the 13 autophagy-related lncRNAs could stratify patients based on their outcomes. Receiver operating characteristic curve analyses confirmed the superior prognostic value of the lncRNA signature compared to clinically used prognostic biomarkers. Importantly, the autophagy-related lncRNA signature predicted patient prognosis independently of clinicopathological characteristics. Furthermore, we found that the expression levels of the autophagy-related lncRNA signature were significantly associated with the infiltration levels of different immune cell subsets, including T cells, NK cells, and dendritic cells. Conclusion The autophagy-related lncRNA signature established here is an independent and robust predictor of osteosarcoma patient survival. Our findings also suggest that the expression of these 13 autophagy-related lncRNAs may promote osteosarcoma progression by regulating immune cell infiltration in the tumor microenvironment.
Collapse
Affiliation(s)
- Guang-Zhi Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,Lintao County Traditional Chinese Medicine Hospital of Gansu Province, Lintao, China
| | - Zuo-Long Wu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Chun-Ying Li
- The Fourth People's Hospital of Qinghai Province, Xining, China
| | - En-Hui Ren
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,Department of Orthopaedics, Xining First People's Hospital, Xining, China
| | - Wen-Hua Yuan
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Ya-Jun Deng
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China
| | - Qi-Qi Xie
- Affiliated Hospital of Qinghai University, Xining, China.,Affiliated Cancer Hospital of Qinghai University, Xining, China.,Breast Disease Diagnosis and Treatment Center, Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, China
| |
Collapse
|
513
|
Kang X, Chen Y, Yi B, Yan X, Jiang C, Chen B, Lu L, Sun Y, Shi R. An integrative microenvironment approach for laryngeal carcinoma: the role of immune/methylation/autophagy signatures on disease clinical prognosis and single-cell genotypes. J Cancer 2021; 12:4148-4171. [PMID: 34093817 PMCID: PMC8176413 DOI: 10.7150/jca.58076] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
The effects of methylation/autophagy-related genes (MARGs) and immune infiltration in the tumor microenvironment on the prognosis of laryngeal cancer were comprehensively explored in this study. Survival analysis screened out 126 MARGs and 10 immune cells potentially associated with the prognosis of laryngeal carcinoma. Cox and lasso regression analyses were then used to select 8 MARGs (CAPN10, DAPK2, MBTPS2, ST13, CFLAR, FADD, PEX14 and TSC2) and 2 immune cells (Eosinophil and Mast cell) to obtain the prognostic risk scoring system (pRS). The pRS was used to establish a risk prediction model for the prognosis of laryngeal cancer. The predictive ability of the prediction model was evaluated by GEO datasets and our clinical samples. Further analysis revealed that pRS is highly associated with single nucleotide polymorphism (SNP), copy number variation (CNV), immune checkpoint blockade (ICB) therapy and tumor microenvironment. Moreover, the screened pRS-related ceRNA network and circ_0002951/miR-548k/HAS2 pathway provide potential therapeutic targets and biomarkers of laryngocarcinoma. Based on the clustering results of pRS-related genes, single cells were then genotyped and revealed by integrated scRNA-seq in laryngeal cancer samples. Fibroblasts were found enriched in high risk cell clusters at the scRNA-seq level. Fibroblast-related ligand-receptor interactions were then exposed and a neural network-based deep learning model based on these pRS-related hub gene signatures was also established with a high accuracy in cell type prediction. In conclusion, the combination of single-cell and transcriptome laryngeal carcinoma landscape analyses can investigate the link between the tumor microenvironmental and prognostic characteristics.
Collapse
Affiliation(s)
- Xueran Kang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine; Ear Institute, Shanghai JiaoTong University School of Medicine; Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Yisheng Chen
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Yi
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine; Ear Institute, Shanghai JiaoTong University School of Medicine; Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Xiaojun Yan
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine; Ear Institute, Shanghai JiaoTong University School of Medicine; Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Chenyan Jiang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine; Ear Institute, Shanghai JiaoTong University School of Medicine; Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Bin Chen
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine; Ear Institute, Shanghai JiaoTong University School of Medicine; Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Lixing Lu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine; Ear Institute, Shanghai JiaoTong University School of Medicine; Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Yuxing Sun
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine; Ear Institute, Shanghai JiaoTong University School of Medicine; Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| | - Runjie Shi
- Department of Otolaryngology-Head and Neck Surgery, Shanghai ninth people's Hospital, Shanghai Jiao Tong University School of Medicine; Ear Institute, Shanghai JiaoTong University School of Medicine; Shanghai Key Laboratory of Translational Medicine on Ear and Nose diseases, Shanghai, China
| |
Collapse
|
514
|
Zada S, Hwang JS, Ahmed M, Lai TH, Pham TM, Elashkar O, Kim DR. Cross talk between autophagy and oncogenic signaling pathways and implications for cancer therapy. Biochim Biophys Acta Rev Cancer 2021; 1876:188565. [PMID: 33992723 DOI: 10.1016/j.bbcan.2021.188565] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023]
Abstract
Autophagy is a highly conserved metabolic process involved in the degradation of intracellular components including proteins and organelles. Consequently, it plays a critical role in recycling metabolic energy for the maintenance of cellular homeostasis in response to various stressors. In cancer, autophagy either suppresses or promotes cancer progression depending on the stage and cancer type. Epithelial-mesenchymal transition (EMT) and cancer metastasis are directly mediated by oncogenic signal proteins including SNAI1, SLUG, ZEB1/2, and NOTCH1, which are functionally correlated with autophagy. In this report, we discuss the crosstalk between oncogenic signaling pathways and autophagy followed by possible strategies for cancer treatment via regulation of autophagy. Although autophagy affects EMT and cancer metastasis, the overall signaling pathways connecting cancer progression and autophagy are still illusive. In general, autophagy plays a critical role in cancer cell survival by providing a minimum level of energy via self-digestion. Thus, cancer cells face nutrient limitations and challenges under stress during EMT and metastasis. Conversely, autophagy acts as a potential cancer suppressor by degrading oncogenic proteins, which are essential for cancer progression, and by removing damaged components such as mitochondria to enhance genomic stability. Therefore, autophagy activators or inhibitors represent possible cancer therapeutics. We further discuss the regulation of autophagy-dependent degradation of oncogenic proteins and its functional correlation with oncogenic signaling pathways, with potential applications in cancer therapy.
Collapse
Affiliation(s)
- Sahib Zada
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Trang Minh Pham
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Omar Elashkar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju 527-27, Republic of Korea.
| |
Collapse
|
515
|
Su H, Yang F, Fu R, Li X, French R, Mose E, Pu X, Trinh B, Kumar A, Liu J, Antonucci L, Todoric J, Liu Y, Hu Y, Diaz-Meco MT, Moscat J, Metallo CM, Lowy AM, Sun B, Karin M. Cancer cells escape autophagy inhibition via NRF2-induced macropinocytosis. Cancer Cell 2021; 39:678-693.e11. [PMID: 33740421 PMCID: PMC8119368 DOI: 10.1016/j.ccell.2021.02.016] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/17/2020] [Accepted: 02/19/2021] [Indexed: 12/29/2022]
Abstract
Many cancers, including pancreatic ductal adenocarcinoma (PDAC), depend on autophagy-mediated scavenging and recycling of intracellular macromolecules, suggesting that autophagy blockade should cause tumor starvation and regression. However, until now autophagy-inhibiting monotherapies have not demonstrated potent anti-cancer activity. We now show that autophagy blockade prompts established PDAC to upregulate and utilize an alternative nutrient procurement pathway: macropinocytosis (MP) that allows tumor cells to extract nutrients from extracellular sources and use them for energy generation. The autophagy to MP switch, which may be evolutionarily conserved and not cancer cell restricted, depends on activation of transcription factor NRF2 by the autophagy adaptor p62/SQSTM1. NRF2 activation by oncogenic mutations, hypoxia, and oxidative stress also results in MP upregulation. Inhibition of MP in autophagy-compromised PDAC elicits dramatic metabolic decline and regression of transplanted and autochthonous tumors, suggesting the therapeutic promise of combining autophagy and MP inhibitors in the clinic.
Collapse
Affiliation(s)
- Hua Su
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210000, China
| | - Fei Yang
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210000, China
| | - Rao Fu
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210000, China
| | - Xin Li
- Laboratory of Cancer ImmunoMetabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21701, USA
| | - Randall French
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego Moores Cancer Center, La Jolla, CA 92093, USA
| | - Evangeline Mose
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego Moores Cancer Center, La Jolla, CA 92093, USA
| | - Xiaohong Pu
- Department of Pathology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210000, China
| | - Brittney Trinh
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Avi Kumar
- Institute of Engineering in Medicine, Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Junlai Liu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jelena Todoric
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Laboratory Medicine, Medical University of Vienna, Vienna 1090, Austria
| | - Yuan Liu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Yinling Hu
- Laboratory of Cancer ImmunoMetabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21701, USA
| | - Maria T Diaz-Meco
- Department of Pathology and Laboratory Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jorge Moscat
- Department of Pathology and Laboratory Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Christian M Metallo
- Institute of Engineering in Medicine, Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, University of California, San Diego Moores Cancer Center, La Jolla, CA 92093, USA
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210000, China.
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
516
|
Abstract
Su et al. demonstrate that upon inhibiting autophagy, an intracellular nutrient recycling pathway, pancreatic ductal adenocarcinoma cells upregulate NRF2-mediated transcription of macropinocytosis pathway components, thereby triggering an alternate route for tumors to scavenge nutrients from extracellular sources. Accordingly, the combined inhibition of autophagy and macropinocytosis may improve cancer treatment.
Collapse
Affiliation(s)
- Gourish Mondal
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jayanta Debnath
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
517
|
Chen RZ, Yang F, Zhang M, Sun ZG, Zhang N. Cellular and Molecular Mechanisms of Pristimerin in Cancer Therapy: Recent Advances. Front Oncol 2021; 11:671548. [PMID: 34026649 PMCID: PMC8138054 DOI: 10.3389/fonc.2021.671548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Seeking an efficient and safe approach to eliminate tumors is a common goal of medical fields. Over these years, traditional Chinese medicine has attracted growing attention in cancer treatment due to its long history. Pristimerin is a naturally occurring quinone methide triterpenoid used in traditional Chinese medicine to treat various cancers. Recent studies have identified alterations in cellular events and molecular signaling targets of cancer cells under pristimerin treatment. Pristimerin induces cell cycle arrest, apoptosis, and autophagy to exhibit anti-proliferation effects against tumors. Pristimerin also inhibits the invasion, migration, and metastasis of tumor cells via affecting cell adhesion, cytoskeleton, epithelial-mesenchymal transition, cancer stem cells, and angiogenesis. Molecular factors and pathways are associated with the anti-cancer activities of pristimerin. Furthermore, pristimerin reverses multidrug resistance of cancer cells and exerts synergizing effects with other chemotherapeutic drugs. This review aims to discuss the anti-cancer potentials of pristimerin, emphasizing multi-targeted biological and molecular regulations in cancers. Further investigations and clinical trials are warranted to understand the advantages and disadvantages of pristimerin treatment much better.
Collapse
Affiliation(s)
- Run-Ze Chen
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fei Yang
- Department of Pathology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Zhang
- Department of Dermatology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhi-Gang Sun
- Department of Thoracic Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nan Zhang
- Department of Oncology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
518
|
Yi X, Duan QY, Wu FG. Low-Temperature Photothermal Therapy: Strategies and Applications. RESEARCH (WASHINGTON, D.C.) 2021; 2021:9816594. [PMID: 34041494 PMCID: PMC8125200 DOI: 10.34133/2021/9816594] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
Although photothermal therapy (PTT) with the assistance of nanotechnology has been considered as an indispensable strategy in the biomedical field, it still encounters some severe problems that need to be solved. Excessive heat can induce treated cells to develop thermal resistance, and thus, the efficacy of PTT may be dramatically decreased. In the meantime, the uncontrollable diffusion of heat can pose a threat to the surrounding healthy tissues. Recently, low-temperature PTT (also known as mild PTT or mild-temperature PTT) has demonstrated its remarkable capacity of conquering these obstacles and has shown excellent performance in bacterial elimination, wound healing, and cancer treatments. Herein, we summarize the recently proposed strategies for achieving low-temperature PTT based on nanomaterials and introduce the synthesis, characteristics, and applications of these nanoplatforms. Additionally, the combination of PTT and other therapeutic modalities for defeating cancers and the synergistic cancer therapeutic effect of the combined treatments are discussed. Finally, the current limitations and future directions are proposed for inspiring more researchers to make contributions to promoting low-temperature PTT toward more successful preclinical and clinical disease treatments.
Collapse
Affiliation(s)
- Xiulin Yi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Qiu-Yi Duan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| |
Collapse
|
519
|
Bai X, Cao X, Qu N, Huang H, Handley M, Zhang S, Shan F. Methionine enkephalin activates autophagy and stimulates tumour cell immunogenicity in human cutaneous squamous cell carcinoma. Int Immunopharmacol 2021; 96:107733. [PMID: 33965882 DOI: 10.1016/j.intimp.2021.107733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/26/2022]
Abstract
Cutaneous squamous cell carcinoma (CSCC) is a common skin tumour. Due to weak immunogenicity, recurrence is frequent after treatment. In this study, we explored the effects and mechanisms of methionine enkephalin (MENK), an endogenous opioid peptide and negative growth regulator, in CSCC. MENK inhibited A431 cell proliferation and promoted apoptosis through the opioid growth factor receptor (OGFr). Importantly, MENK also induced autophagy in CSCC and stimulated the emission of DAMPs in A431 cells, which resulted in enhanced activation of dendritic cells (DC).In conclusion, MENK provides an effective method with therapeutic potential to modulate the CSCC microenvironment by utilizing autophagy in the cancer cells.
Collapse
Affiliation(s)
- Xueli Bai
- Department of Gynecology, The fourth Affiliated Hospital of China Medical University, 4 Chongshandong road, Huanggu district, Shenyang, Liaoning 110004, PR China; Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Xia Cao
- Department of Gynecology, The fourth Affiliated Hospital of China Medical University, 4 Chongshandong road, Huanggu district, Shenyang, Liaoning 110004, PR China
| | - Na Qu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hai Huang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Mike Handley
- Cytocm lnc, 3001 Aloma Ave. Winter Park, FL 32792, USA
| | - Shuling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Fengping Shan
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang 110122, China.
| |
Collapse
|
520
|
Zhou Y, Yang L, Xiong L, Wang K, Hou X, Li Q, Kong F, Liu X, He J. KIF11 is upregulated in colorectal cancer and silencing of it impairs tumor growth and sensitizes colorectal cancer cells to oxaliplatin via p53/GSK3β signaling. J Cancer 2021; 12:3741-3753. [PMID: 33995648 PMCID: PMC8120193 DOI: 10.7150/jca.52103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 04/21/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the most frequently diagnosed cancer of the digestive tract. Chemotherapy drugs such as oxaliplatin are frequently administered to CRC patients diagnosed with advanced or metastatic disease. A deep understanding of the molecular mechanism underlying CRC tumorigenesis and identification of optimal biomarkers for estimating chemotherapy sensitivity are essential for the treatment of CRC. Numerous members of the kinesin family are dysregulated in cancers, contributing to tumorigenesis, metastasis and drug resistance. KIF11 is a key component of the bipolar spindle and is highly expressed in several cancer types. We analyzed KIF11 expression in clinical samples by Western blotting and qRT-PCR and explored its role and mechanism in CRC growth and sensitivity to oxaliplatin via detection of the phosphorylation profile of kinases and gain-and-loss-of-function assays. We found that KIF11 was upregulated in CRC tissues and was associated with advanced clinical stage and vessel invasion and that knockdown of KIF11 led to tumor growth arrest and increased sensitivity to oxaliplatin via enhanced DNA damage and apoptosis. Mechanistically, aberrantly activated p53 signaling or possibly deactivated GSK3β signaling was responsible for KIF11 knockdown-mediated effects in CRC cells. Thus, our data firmly demonstrated that KIF11 could serve as a potential oncogene and proper biomarker for assessing oxaliplatin sensitivity in CRC.
Collapse
Affiliation(s)
- Yan Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Leping Yang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Li Xiong
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Kunpeng Wang
- Department of General Surgery, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang 318000, China
| | - Xuyang Hou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qinglong Li
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Fanhua Kong
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xi Liu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jun He
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
521
|
Devis-Jauregui L, Eritja N, Davis ML, Matias-Guiu X, Llobet-Navàs D. Autophagy in the physiological endometrium and cancer. Autophagy 2021; 17:1077-1095. [PMID: 32401642 PMCID: PMC8143243 DOI: 10.1080/15548627.2020.1752548] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 03/18/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved catabolic process and a major cellular pathway for the degradation of long-lived proteins and cytoplasmic organelles. An increasing body of evidence has unveiled autophagy as an indispensable biological function that helps to maintain normal tissue homeostasis and metabolic fitness that can also lead to severe consequences for the normal cellular functioning when altered. Recent accumulating data point to autophagy as a key player in a wide variety of physiological and pathophysiological conditions in the human endometrium, one of the most proficient self-regenerating tissues in the human body and an instrumental player in placental species reproductive function. The current review highlights the most recent findings regarding the process of autophagy in the normal and cancerous endometrial tissue. Current research efforts aiming to therapeutically exploit autophagy and the methodological approaches used are discussed.Abbreviations: 3-MA: 3-methyladenine; ACACA (acetyl-CoA carboxylase alpha); AICAR: 5-aminoimidazole-4-carboximide riboside; AKT: AKT serine/threonine kinase; AMPK: AMP-activated protein kinase; ATG: autophagy related; ATG12: autophagy related 12; ATG16L1: autophagy related 16 like 1; ATG3: autophagy related 3; ATG4C: autophagy related 4C cysteine peptidase; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG9: autophagy related 9; Baf A1: bafilomycin A1; BAX: BCL2 associated X, apoptosis regulator; BCL2: BCL2 apoptosis regulator; BECN1: beclin 1; CACNA1D: calcium voltage-gated channel subunit alpha1 D; CASP3: caspase 3; CASP7: caspase 7; CASP8: caspase 8; CASP9: caspase 9; CD44: CD44 molecule (Indian blood group); CDH1: cadherin 1; CDKN1A: cyclin dependent kinase inhibitor 1A; CDKN2A: cyclin dependent kinase inhibitor 2A; CMA: chaperone-mediated autophagy; CQ: chloroquine; CTNNB1: catenin beta 1; DDIT3: DNA damage inducible transcript 3; EC: endometrial cancer; EGFR: epidermal growth factor receptor; EH: endometrial hyperplasia; EIF4E: eukaryotic translation initiation factor 4E; EPHB2/ERK: EPH receptor B2; ER: endoplasmic reticulum; ERBB2: er-b2 receptor tyrosine kinase 2; ERVW-1: endogenous retrovirus group W member 1, envelope; ESR1: estrogen receptor 1; FSH: follicle-stimulating hormone; GCG/GLP1: glucagon; GFP: green fluorescent protein; GIP: gastric inhibitory polypeptide; GLP1R: glucagon-like peptide-1 receptor; GLS: glutaminase; H2AX: H2A.X variant histone; HIF1A: hypoxia inducible factor 1 alpha; HMGB1: high mobility group box 1; HOTAIR: HOX transcript antisense RNA; HSPA5: heat shock protein family A (HSP70) member 5; HSPA8: heat shock protein family A (HSP70) member 8; IGF1: insulin like growth factor 1; IL27: interleukin 27; INS: insulin; ISL: isoliquiritigenin; KRAS: KRAS proto-oncogene, GTPase; LAMP2: lysosomal-associated membrane protein 2; lncRNA: long-non-coding RNA; MAP1LC3A/LC3A: microtubule associated protein 1 light chain 3 alpha; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAPK8: mitogen-activated protein kinase 8; MAPK9: mitogen-activated protein kinase 9; MPA: medroxyprogesterone acetate; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; MYCBP: MYC-binding protein; NFE2L2: nuclear factor, erythroid 2 like 2; NFKB: nuclear factor kappa B; NFKBIA: NFKB inhibitor alpha; NK: natural killer; NR5A1: nuclear receptor subfamily 5 group A member 1; PARP1: poly(ADP-ribose) polymerase 1; PAX2: paired box 2; PDK1: pyruvate dehydrogenase kinase 1; PDX: patient-derived xenograft; PIK3C3/Vps34: phosphatidylinositol 3-kinase catalytic subunit type 3; PIK3CA: phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; PIK3R1: phosphoinositide-3-kinase regulatory subunit 1; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; PPD: protopanaxadiol; PRKCD: protein kinase C delta; PROM1/CD133: prominin 1; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; PTEN: phosphatase and tensin homolog; RB1CC1/FIP200: RB1 inducible coiled-coil 1; RFP: red fluorescent protein; RPS6KB1/S6K1: ribosomal protein S6 kinase B1; RSV: resveratrol; SGK1: serum/glucocorticoid regulated kinase 1; SGK3: serum/glucocorticoid regulated kinase family member 3; SIRT: sirtuin; SLS: stone-like structures; SMAD2: SMAD family member 2; SMAD3: SMAD family member 3; SQSTM1: sequestosome 1; TALEN: transcription activator-like effector nuclease; TGFBR2: transforming growth factor beta receptor 2; TP53: tumor protein p53; TRIB3: tribbles pseudokinase 3; ULK1: unc-51 like autophagy activating kinase 1; ULK4: unc-51 like kinase 4; VEGFA: vascular endothelial growth factor A; WIPI2: WD repeat domain, phosphoinositide interacting 2; XBP1: X-box binding protein 1; ZFYVE1: zinc finger FYVE domain containing 1.
Collapse
Affiliation(s)
- Laura Devis-Jauregui
- Laboratory of Precision Medicine, Oncobell Program. Bellvitge Biomedical Research Institute (IDIBELL), Gran via De l’Hospitalet, Barcelona, Spain
| | - Núria Eritja
- Department of Pathology-Hospital Universitari Arnau De Vilanova, Universitat De Lleida, IRBLLEIDA, CIBERONC, Lleida, Spain
| | - Meredith Leigh Davis
- Institute of Genetic Medicine-International Centre for Life, Newcastle University. Central Parkway, Newcastle upon Tyne, UK
| | - Xavier Matias-Guiu
- Laboratory of Precision Medicine, Oncobell Program. Bellvitge Biomedical Research Institute (IDIBELL), Gran via De l’Hospitalet, Barcelona, Spain
- Department of Pathology-Hospital Universitari Arnau De Vilanova, Universitat De Lleida, IRBLLEIDA, CIBERONC, Lleida, Spain
- Department of Pathology-Hospital, Universitari De Bellvitge, Barcelona, Spain
| | - David Llobet-Navàs
- Laboratory of Precision Medicine, Oncobell Program. Bellvitge Biomedical Research Institute (IDIBELL), Gran via De l’Hospitalet, Barcelona, Spain
| |
Collapse
|
522
|
Abstract
Autophagy is a regulated mechanism that removes unnecessary or dysfunctional cellular components and recycles metabolic substrates. In response to stress signals in the tumour microenvironment, the autophagy pathway is altered in tumour cells and immune cells - thereby differentially affecting tumour progression, immunity and therapy. In this Review, we summarize our current understanding of the immunologically associated roles and modes of action of the autophagy pathway in cancer progression and therapy, and discuss potential approaches targeting autophagy to enhance antitumour immunity and improve the efficacy of current cancer therapy.
Collapse
Affiliation(s)
- Houjun Xia
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Weiping Zou
- Department of Surgery, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Graduate Program in Immunology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
- Graduate Program in Cancer Biology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
523
|
Yang S, Zhou Y, Zhang X, Wang L, Fu J, Zhao X, Yang L. The prognostic value of an autophagy-related lncRNA signature in hepatocellular carcinoma. BMC Bioinformatics 2021; 22:217. [PMID: 33910497 PMCID: PMC8080392 DOI: 10.1186/s12859-021-04123-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/05/2021] [Indexed: 02/08/2023] Open
Abstract
Background lncRNA may be involved in the occurrence, metastasis, and chemical reaction of hepatocellular carcinoma (HCC) through various pathways associated with autophagy. Therefore, it is urgent to reveal more autophagy-related lncRNAs, explore these lncRNAs’ clinical significance, and find new targeted treatment strategies. Methods The corresponding data of HCC patients and autophagy genes were obtained from the TCGA database, and the human autophagy database respectively. Based on the co-expression and Cox regression analysis to construct prognostic prediction signature. Results Finally, a signature containing seven autophagy-related lncRNAs (PRRT3-AS1, RP11-479G22.8, RP11-73M18.8, LINC01138, CTD-2510F5.4, CTC-297N7.9, RP11-324I22.4) was constructed. Based on the risk score of signature, Overall survival (OS) curves show that the OS of high-risk patients is significantly lower than that of low-risk patients (P = 2.292e−10), and the prognostic prediction accuracy of risk score (AUC = 0.786) is significantly higher than that of ALBI (0.532), child_pugh (0.573), AFP (0.5751), and AJCC_stage (0.631). Moreover, multivariate Cox analysis and Nomogram of risk score are indicated that the 1-year and 3-year survival rates of patients are obviously accuracy by the combined analysis of the risk score, child_pugh, age, M_stage, and Grade (The AUC of 1- and 3-years are 0.87, and 0.855). Remarkably, the 7 autophagy-related lncRNAs may participate in Spliceosome, Cell cycle, RNA transport, DNA replication, and mRNA surveillance pathway and be related to the biological process of RNA splicing and mRNA splicing. Conclusion In conclusion, the 7 autophagy-related lncRNAs might be promising prognostic and therapeutic targets for HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04123-6.
Collapse
Affiliation(s)
- Shiming Yang
- Shandong Second Provincial General Hospital, Shandong, China
| | - Yaping Zhou
- Shihezi University School of Medicine, Shihezi, China.,Clinical Laboratory Diagnosis Center, General Hospital of Xinjiang Military Region, Xinjiang, China
| | - Xiangxin Zhang
- Shihezi University School of Medicine, Shihezi, China.,The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Lu Wang
- Department of Medical Laboratory Science, Xinjiang Bayingoleng Mongolian Autonomous Prefecture People's Hospital, Xinjiang, China.
| | - Jianfeng Fu
- Clinical Laboratory Diagnosis Center, General Hospital of Xinjiang Military Region, Xinjiang, China
| | - Xiaotong Zhao
- Shihezi University School of Medicine, Shihezi, China.,The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, China
| | - Liu Yang
- Shihezi University School of Medicine, Shihezi, China. .,The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, China.
| |
Collapse
|
524
|
The Regulating Effect of Autophagy-Related MiRNAs in Kidney, Bladder, and Prostate Cancer. JOURNAL OF ONCOLOGY 2021; 2021:5510318. [PMID: 33976697 PMCID: PMC8084683 DOI: 10.1155/2021/5510318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/26/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
Autophagy is a treatment target for many disorders, including cancer, and its specific role is becoming increasingly well known. In tumors, researchers pay attention to microribonucleic acids (miRNAs) with regulatory effects to develop more effective therapeutic drugs for autophagy and find new therapeutic targets. Various studies have shown that autophagy-related miRNAs play an irreplaceable role in different tumors, such as miR-495, miR-30, and miR-101. These miRNAs are associated with autophagy resistance in gastric cancer, non-small cell lung cancer, and cervical cancer. In recent years, autophagy-related miRNAs have also been reported to play a role in autophagy in urinary system tumors. This article reviews the regulatory effects of autophagy-related miRNAs in kidney, bladder, and prostate cancer and provides new ideas for targeted therapy of the three major tumors of the urinary system.
Collapse
|
525
|
He Q, Li Z, Yin J, Li Y, Yin Y, Lei X, Zhu W. Prognostic Significance of Autophagy-Relevant Gene Markers in Colorectal Cancer. Front Oncol 2021; 11:566539. [PMID: 33937013 PMCID: PMC8081889 DOI: 10.3389/fonc.2021.566539] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 03/22/2021] [Indexed: 12/22/2022] Open
Abstract
Background Colorectal cancer (CRC) is a common malignant solid tumor with an extremely low survival rate after relapse. Previous investigations have shown that autophagy possesses a crucial function in tumors. However, there is no consensus on the value of autophagy-associated genes in predicting the prognosis of CRC patients. This work screens autophagy-related markers and signaling pathways that may participate in the development of CRC, and establishes a prognostic model of CRC based on autophagy-associated genes. Methods Gene transcripts from the TCGA database and autophagy-associated gene data from the GeneCards database were used to obtain expression levels of autophagy-associated genes, followed by Wilcox tests to screen for autophagy-related differentially expressed genes. Then, 11 key autophagy-associated genes were identified through univariate and multivariate Cox proportional hazard regression analysis and used to establish prognostic models. Additionally, immunohistochemical and CRC cell line data were used to evaluate the results of our three autophagy-associated genes EPHB2, NOL3, and SNAI1 in TCGA. Based on the multivariate Cox analysis, risk scores were calculated and used to classify samples into high-risk and low-risk groups. Kaplan-Meier survival analysis, risk profiling, and independent prognosis analysis were carried out. Receiver operating characteristic analysis was performed to estimate the specificity and sensitivity of the prognostic model. Finally, GSEA, GO, and KEGG analysis were performed to identify the relevant signaling pathways. Results A total of 301 autophagy-related genes were differentially expressed in CRC. The areas under the 1-year, 3-year, and 5-year receiver operating characteristic curves of the autophagy-based prognostic model for CRC were 0.764, 0.751, and 0.729, respectively. GSEA analysis of the model showed significant enrichment in several tumor-relevant pathways and cellular protective biological processes. The expression of EPHB2, IL-13, MAP2, RPN2, and TRAF5 was correlated with microsatellite instability (MSI), while the expression of IL-13, RPN2, and TRAF5 was related to tumor mutation burden (TMB). GO analysis showed that the 11 target autophagy genes were chiefly enriched in mRNA processing, RNA splicing, and regulation of the mRNA metabolic process. KEGG analysis showed enrichment mainly in spliceosomes. We constructed a prognostic risk assessment model based on 11 autophagy-related genes in CRC. Conclusion A prognostic risk assessment model based on 11 autophagy-associated genes was constructed in CRC. The new model suggests directions and ideas for evaluating prognosis and provides guidance to choose better treatment strategies for CRC.
Collapse
Affiliation(s)
- Qinglian He
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Ziqi Li
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Jinbao Yin
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Yuling Li
- Department of Pathology, Dongguan People's Hospital, Southern Medical University, Dongguan, China
| | - Yuting Yin
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Xue Lei
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Wei Zhu
- Department of Pathology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
526
|
Li Y, Li Y, Yin J, Wang C, Yang M, Gu J, He M, Xu H, Fu W, Zhang W, Ru Y, Liu X, Li Y, Xin Y, Gao H, Xie X, Gao Y. A mitophagy inhibitor targeting p62 attenuates the leukemia-initiation potential of acute myeloid leukemia cells. Cancer Lett 2021; 510:24-36. [PMID: 33862150 DOI: 10.1016/j.canlet.2021.04.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/06/2021] [Accepted: 04/04/2021] [Indexed: 12/20/2022]
Abstract
There has been an increasing focus on the tumorigenic potential of leukemia initiating cells (LICs) in acute myeloid leukemia (AML). Despite the important role of selective autophagy in the life-long maintenance of hematopoietic stem cells (HSCs), cancer progression, and chemoresistance, the relationship between LICs and selective autophagy remains to be fully elucidated. Sequestosome 1 (SQSTM1), also known as p62, is a selective autophagy receptor for the degradation of ubiquitinated substrates, and its loss impairs leukemia progression in AML mouse models. In this study, we evaluated the underlying mechanisms of mitophagy in the survival of LICs with XRK3F2, a p62-ZZ inhibitor. We demonstrated that XRK3F2 selectively impaired LICs but spared normal HSCs in both mouse and patient-derived tumor xenograft (PDX) AML models. Mechanistically, we observed that XRK3F2 blocked mitophagy by inhibiting the binding of p62 with defective mitochondria. Our study not only evaluated the effectiveness and safety of XRK3F2 in LICs, but also demonstrated that mitophagy plays an indispensable role in the survival of LICs during AML development and progression, which can be impaired by blocking p62.
Collapse
Affiliation(s)
- Yinghui Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yafang Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Jingjing Yin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Chaoqun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Jiali Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Mei He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Hui Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Weichao Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yongxin Ru
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Xiaolei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Ying Li
- Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yue Xin
- Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Huier Gao
- Department of Pharmacy, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300192, China
| | - Xiangqun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, NIH National Center of Excellence for Computational Drug Abuse Research, Drug Discovery Institute; Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, United States.
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| |
Collapse
|
527
|
Li XL, Zhou J, Xia CJ, Min H, Lu ZK, Chen ZR. PRIMA‑1 met induces autophagy in colorectal cancer cells through upregulation of the mTOR/AMPK‑ULK1‑Vps34 signaling cascade. Oncol Rep 2021; 45:86. [PMID: 33846805 PMCID: PMC8025076 DOI: 10.3892/or.2021.8037] [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: 10/27/2020] [Accepted: 03/05/2021] [Indexed: 12/26/2022] Open
Abstract
p53‑reactivation and induction of massive apoptosis‑1, APR‑017 methylated (PRIMA‑1met; APR246) targets mutant p53 to restore its wild‑type structure and function. It was previously demonstrated that PRIMA‑1met effectively inhibited the growth of colorectal cancer (CRC) cells in a p53‑independent manner, and distinctly induced apoptosis by upregulating Noxa in p53‑mutant cell lines. The present study including experiments of western blotting, acridine orange staining and transmission electron microscopy revealed that PRIMA‑1met induced autophagy in CRC cells independently of p53 status. Importantly, PRIMA‑1met not only promoted autophagic vesicle (AV) formation and AV‑lysosome fusion, but also increased lysosomal degradation. Furthermore, Cell Counting Kit‑8 assay, colony formation assay and small interfering RNA transfection were performed to investigate the underling mechanisms. The study indicated that activation of the mTOR/AMPK‑ULK1‑Vps34 autophagic signaling cascade was key for PRIMA‑1met‑induced autophagy. Additionally, autophagy served a crucial role in the inhibitory effect of PRIMA‑1met in cells harboring wild‑type p53, which was closely associated with the increased expression of Noxa. Taken together, the results determined the effect of PRIMA‑1met on autophagy, and further revealed mechanistic insights into different CRC cell lines. It was concluded that PRIMA‑1met‑based therapy may be an effective strategy for CRC treatment.
Collapse
Affiliation(s)
- Xiao-Lan Li
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore 117599, Republic of Singapore
| | - Chen-Jing Xia
- Department of Gastroenterology, Traditional Chinese Medicine Hospital of Kunshan, Suzhou, Jiangsu 215300, P.R. China
| | - Han Min
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Zhong-Kai Lu
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Zhi-Rong Chen
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| |
Collapse
|
528
|
Reis LB, Filippi-Chiela EC, Ashton-Prolla P, Visioli F, Rosset C. The paradox of autophagy in Tuberous Sclerosis Complex. Genet Mol Biol 2021; 44:e20200014. [PMID: 33821877 PMCID: PMC8022228 DOI: 10.1590/1678-4685-gmb-2020-0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/17/2021] [Indexed: 12/21/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disorder caused by germline mutations in TSC1 or TSC2 genes, which leads to the hyperactivation of the mTORC1 pathway, an important negative regulator of autophagy. This leads to the development of hamartomas in multiple organs. The variability in symptoms presents a challenge for the development of completely effective treatments for TSC. One option is the treatment with mTORC1 inhibitors, which are targeted to block cell growth and restore autophagy. However, the therapeutic effect of rapamycin seems to be more efficient in the early stages of hamartoma development, an effect that seems to be associated with the paradoxical role of autophagy in tumor establishment. Under normal conditions, autophagy is directly inhibited by mTORC1. In situations of bioenergetics stress, mTORC1 releases the Ulk1 complex and initiates the autophagy process. In this way, autophagy promotes the survival of established tumors by supplying metabolic precursors during nutrient deprivation; paradoxically, excessive autophagy has been associated with cell death in some situations. In spite of its paradoxical role, autophagy is an alternative therapeutic strategy that could be explored in TSC. This review compiles the findings related to autophagy and the new therapeutic strategies targeting this pathway in TSC.
Collapse
Affiliation(s)
- Larissa Brussa Reis
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| | - Eduardo C Filippi-Chiela
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul (UFRGS), Instituto de Ciências Básicas da Saúde, Departamento de Ciências Morfológicas, Porto Alegre, RS, Brazil
| | - Patricia Ashton-Prolla
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil.,Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Genética Médica, Porto Alegre, RS, Brazil
| | - Fernanda Visioli
- Universidade Federal do Rio Grande do Sul, Faculdade de Odontologia, Departamento de Patologia Oral, Porto Alegre, RS, Brazil
| | - Clévia Rosset
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
| |
Collapse
|
529
|
Miller DR, Thorburn A. Autophagy and organelle homeostasis in cancer. Dev Cell 2021; 56:906-918. [PMID: 33689692 PMCID: PMC8026727 DOI: 10.1016/j.devcel.2021.02.010] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/11/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022]
Abstract
Beginning with the earliest studies of autophagy in cancer, there have been indications that autophagy can both promote and inhibit cancer growth and progression; autophagy regulation of organelle homeostasis is similarly complicated. In this review we discuss pro- and antitumor effects of organelle-targeted autophagy and how this contributes to several hallmarks of cancer, such as evading cell death, genomic instability, and altered metabolism. Typically, the removal of damaged or dysfunctional organelles prevents tumor development but can also aid in proliferation or drug resistance in established tumors. By better understanding how organelle-specific autophagy takes place and can be manipulated, it may be possible to go beyond the brute-force approach of trying to manipulate all autophagy in order to improve therapeutic targeting of this process in cancer.
Collapse
Affiliation(s)
- Dannah R Miller
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
530
|
Tyutyunyk-Massey L, Sun Y, Dao N, Ngo H, Dammalapati M, Vaidyanathan A, Singh M, Haqqani S, Haueis J, Finnegan R, Deng X, Kirberger SE, Bos PD, Bandyopadhyay D, Pomerantz WCK, Pommier Y, Gewirtz DA, Landry JW. Autophagy-Dependent Sensitization of Triple-Negative Breast Cancer Models to Topoisomerase II Poisons by Inhibition of the Nucleosome Remodeling Factor. Mol Cancer Res 2021; 19:1338-1349. [PMID: 33811160 DOI: 10.1158/1541-7786.mcr-20-0743] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 02/23/2021] [Accepted: 03/29/2021] [Indexed: 11/16/2022]
Abstract
Epigenetic regulators can modulate the effects of cancer therapeutics. To further these observations, we discovered that the bromodomain PHD finger transcription factor subunit (BPTF) of the nucleosome remodeling factor (NURF) promotes resistance to doxorubicin, etoposide, and paclitaxel in the 4T1 breast tumor cell line. BPTF functions in promoting resistance to doxorubicin and etoposide, but not paclitaxel, and may be selective to cancer cells, as a similar effect was not observed in embryonic stem cells. Sensitization to doxorubicin and etoposide with BPTF knockdown (KD) was associated with increased DNA damage, topoisomerase II (TOP2) crosslinking and autophagy; however, there was only a modest increase in apoptosis and no increase in senescence. Sensitization to doxorubicin was confirmed in vivo with the syngeneic 4T1 breast tumor model using both genetic and pharmacologic inhibition of BPTF. The effects of BPTF inhibition in vivo are autophagy dependent, based on genetic autophagy inhibition. Finally, treatment of 4T1, 66cl4, 4T07, MDA-MB-231, but not ER-positive 67NR and MCF7 breast cancer cells with the selective BPTF bromodomain inhibitor, AU1, recapitulates genetic BPTF inhibition, including in vitro sensitization to doxorubicin, increased TOP2-DNA crosslinks and DNA damage. Taken together, these studies demonstrate that BPTF provides resistance to the antitumor activity of TOP2 poisons, preventing the resolution of TOP2 crosslinking and associated autophagy. These studies suggest that BPTF can be targeted with small-molecule inhibitors to enhance the effectiveness of TOP2-targeted cancer chemotherapeutic drugs. IMPLICATIONS: These studies suggest NURF can be inhibited pharmacologically as a viable strategy to improve chemotherapy effectiveness.
Collapse
Affiliation(s)
- Liliya Tyutyunyk-Massey
- VCU Massey Cancer Center, Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Yilun Sun
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, NIH, Bethesda, Maryland
| | - Nga Dao
- VCU Massey Cancer Center, Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Hannah Ngo
- VCU Massey Cancer Center, Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Mallika Dammalapati
- VCU Massey Cancer Center, Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Ashish Vaidyanathan
- VCU Massey Cancer Center, Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Manjulata Singh
- VCU Massey Cancer Center, Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Syed Haqqani
- VCU Massey Cancer Center, Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Joshua Haueis
- VCU Massey Cancer Center, Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Ryan Finnegan
- VCU Massey Cancer Center, Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Xiaoyan Deng
- VCU Massey Cancer Center, Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Steve E Kirberger
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota
| | - Paula D Bos
- VCU Massey Cancer Center, Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Dipankar Bandyopadhyay
- VCU Massey Cancer Center, Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | | | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, NIH, Bethesda, Maryland
| | - David A Gewirtz
- VCU Massey Cancer Center, Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Joseph W Landry
- VCU Massey Cancer Center, Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia.
| |
Collapse
|
531
|
Abstract
Cells use mitophagy to remove dysfunctional or excess mitochondria, frequently in response to imposed stresses, such as hypoxia and nutrient deprivation. Mitochondrial cargo receptors (MCR) induced by these stresses target mitochondria to autophagosomes through interaction with members of the LC3/GABARAP family. There are a growing number of these MCRs, including BNIP3, BNIP3L, FUNDC1, Bcl2-L-13, FKBP8, Prohibitin-2, and others, in addition to mitochondrial protein targets of PINK1/Parkin phospho-ubiquitination. There is also an emerging link between mitochondrial lipid signaling and mitophagy where ceramide, sphingosine-1-phosphate, and cardiolipin have all been shown to promote mitophagy. Here, we review the upstream signaling mechanisms that regulate mitophagy, including components of the mitochondrial fission machinery, AMPK, ATF4, FoxOs, Sirtuins, and mtDNA release, and address the significance of these pathways for stress responses in tumorigenesis and metastasis. In particular, we focus on how mitophagy modulators intersect with cell cycle control and survival pathways in cancer, including following ECM detachment and during cell migration and metastasis. Finally, we interrogate how mitophagy affects tissue atrophy during cancer cachexia and therapy responses in the clinic.
Collapse
Affiliation(s)
- Logan P Poole
- The Ben May Department for Cancer Research, The Gordon Center for Integrative Sciences, W-338, The University of Chicago, 929 E 57th Street, Chicago, IL, 60637, USA
- The Committee on Cancer Biology, The University of Chicago, Chicago, USA
| | - Kay F Macleod
- The Ben May Department for Cancer Research, The Gordon Center for Integrative Sciences, W-338, The University of Chicago, 929 E 57th Street, Chicago, IL, 60637, USA.
- The Committee on Cancer Biology, The University of Chicago, Chicago, USA.
| |
Collapse
|
532
|
Zhong J, Fang L, Chen R, Xu J, Guo D, Guo C, Guo C, Chen J, Chen C, Wang X. Polysaccharides from sporoderm-removed spores of Ganoderma lucidum induce apoptosis in human gastric cancer cells via disruption of autophagic flux. Oncol Lett 2021; 21:425. [PMID: 33850566 PMCID: PMC8025153 DOI: 10.3892/ol.2021.12686] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/02/2021] [Indexed: 12/24/2022] Open
Abstract
The sporoderm-broken spores of Ganoderma lucidum (G. lucidum) polysaccharide (BSGLP) have been demonstrated to inhibit carcinogenesis in several types of cancer. However, to the best of our knowledge, the anticancer effects of polysaccharides extracted from the newly developed sporoderm-removed spores of G. lucidum (RSGLP) have not been assessed. The present study first compared the anticancer effects of RSGLP and BSGLP in three gastric cancer cell lines and it was found that RSGLP was more potent than BSGLP in decreasing gastric cancer cell viability. RSGLP significantly induced apoptosis in AGS cells, accompanied by downregulation of Bcl-2 and pro-caspase-3 expression levels, and upregulation of cleaved-PARP. Furthermore, RSGLP increased LC3-II and p62 expression, indicative of induction of autophagy and disruption of autophagic flux in AGS cells. These results were further verified by combined treatment of AGS cells with the late-stage autophagy inhibitor chloroquine, or early-stage autophagy inducer rapamycin. Adenoviral transfection with mRFP-GFP-LC3 further confirmed that autophagic flux was inhibited by RSGLP in AGS cells. Finally, the present study demonstrated that the RSGLP-induced autophagy and disruption of autophagic flux disruption was, at least in part, responsible for RSGLP-induced apoptosis in AGS cells. The results of the present study demonstrated for the first time that RSGLP is more effective than BSGLP in inhibiting gastric cancer cell viability, and RSGLP may serve as a promising autophagy inhibitor in the management of gastric cancer.
Collapse
Affiliation(s)
- Jiayi Zhong
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China.,Department of Pharmacy, Wenling Maternal and Child Health Care Hospital, Taizhou, Zhejiang 317500, P.R. China
| | - Liu Fang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Rong Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Jing Xu
- Zhejiang Engineering Research Center of Rare Medicinal Plants, Wuyi, Zhejiang 321200, P.R. China
| | - Dandan Guo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Chengjie Guo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Cuiling Guo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Jiajun Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Chaojie Chen
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Xingya Wang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
533
|
Jiang T, Chen X, Ren X, Yang JM, Cheng Y. Emerging role of autophagy in anti-tumor immunity: Implications for the modulation of immunotherapy resistance. Drug Resist Updat 2021; 56:100752. [PMID: 33765484 DOI: 10.1016/j.drup.2021.100752] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapies such as CAR-T cell transfer and antibody-targeted therapy have produced promising clinical outcomes in patients with advanced and metastatic cancer that are resistant to conventional therapies. However, with increasing use of cancer immunotherapy in clinical treatment, multiple therapy-resistance mechanisms have gradually emerged. The tumor microenvironment (TME), an integral component of cancer, can significantly influence the therapeutic response. Thus, it is worth exploring the potential of TME in modulating therapy resistance, in the hope to devise novel strategies to reinforcing anti-cancer treatments such as immunotherapy. As a crucial recycling process in the complex TME, the role of autophagy in tumor immunity has been increasingly appreciated. Firstly, autophagy in tumor cells can affect their immune response through modulating MHC-I-antigen complexes, thus modulating immunogenic tumor cell death, changing functions of immune cells via secretory autophagy, reducing the NK- and CTL-mediated cell lysis and degradation of immune checkpoint proteins. Secondly, autophagy is critical for the differentiation, maturation and survival of immune cells in the TME and can significantly affect the immune function of these cells, thereby regulating the anti-tumor immune response. Thirdly, alteration of autophagic activity in stromal cells, especially in fibroblasts, can reconstruct the three-dimensional stromal environment and metabolic reprogramming in the TME. A number of studies have demonstrated that optimal induction or inhibition of autophagy may lead to effective therapeutic regimens when combined with immunotherapy. This review discusses the important roles of autophagy in tumor cells, immune cells and stromal cells in the context of tumor immunity, and the potential of combining the autophagy-based therapy with immunotherapy as novel therapeutic approaches against cancer.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xisha Chen
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xingcong Ren
- Department of Toxicology and Cancer Biology, Department of Pharmacology, and Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Jin-Ming Yang
- Department of Toxicology and Cancer Biology, Department of Pharmacology, and Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.
| | - Yan Cheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
534
|
Chong ZX, Yeap SK, Ho WY. Regulation of autophagy by microRNAs in human breast cancer. J Biomed Sci 2021; 28:21. [PMID: 33761957 PMCID: PMC7992789 DOI: 10.1186/s12929-021-00715-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/25/2021] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is the most common solid cancer that affects female population globally. MicroRNAs (miRNAs) are short non-coding RNAs that can regulate post-transcriptional modification of multiple downstream genes. Autophagy is a conserved cellular catabolic activity that aims to provide nutrients and degrade un-usable macromolecules in mammalian cells. A number of in vitro, in vivo and clinical studies have reported that some miRNAs could modulate autophagy activity in human breast cancer cells, and these would influence human breast cancer progression and treatment response. Therefore, this review was aimed to discuss the roles of autophagy-regulating miRNAs in influencing breast cancer development and treatment response. The review would first introduce autophagy types and process, followed by the discussion of the roles of different miRNAs in modulating autophagy in human breast cancer, and to explore how would this miRNA-autophagy regulatory process affect the disease progression or treatment response. Lastly, the potential applications and challenges of utilizing autophagy-regulating miRNAs as breast cancer biomarkers and novel therapeutic agents would be discussed.
Collapse
Affiliation(s)
- Zhi Xiong Chong
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, 43900, Sepang, Selangor, Malaysia
| | - Wan Yong Ho
- Faculty of Science and Engineering, University of Nottingham Malaysia, 43500, Semenyih, Selangor, Malaysia.
| |
Collapse
|
535
|
Functional Genetic Variants in ATG10 Are Associated with Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13061344. [PMID: 33809750 PMCID: PMC8002222 DOI: 10.3390/cancers13061344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/05/2021] [Accepted: 03/12/2021] [Indexed: 01/07/2023] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is a hematological neoplasm with a very poor survival rate. To date, diagnostic tools to monitor individuals at higher risk of developing AML are scarce. Single nucleotide polymorphisms (SNPs) have emerged as good candidates for disease prevention. AML is characterized by altered autophagy, a vital mechanism to remove and recycle unnecessary or dysfunctional cellular components. ATG10 is one of the autophagy core genes involved in the autophagosome formation. We hypothesize that SNPs located in regulatory regions of the ATG10 gene could predispose individuals to AML development. We therefore genotyped three SNPs within the ATG10 locus. We identified the ATG10rs3734114 as a potential risk factor for developing AML, whereas the ATG10rs1864182 was associated with decreased risk. These findings highlight ATG10 as a key regulator of susceptibility to AML. Furthermore, we believe that ATG10 SNPs could be exploited in the clinical setting as an AML prevention strategy. Abstract Acute myeloid leukemia (AML) is the most common acute leukemia, characterized by a heterogeneous genetic landscape contributing, among others, to the occurrence of metabolic reprogramming. Autophagy, a key player on metabolism, plays an essential role in AML. Here, we examined the association of three potentially functional genetic polymorphisms in the ATG10 gene, central for the autophagosome formation. We screened a multicenter cohort involving 309 AML patients and 356 healthy subjects for three ATG10 SNPs: rs1864182T>G, rs1864183C>T and rs3734114T>C. The functional consequences of the ATG10 SNPs in its canonical function were investigated in vitro using peripheral blood mononuclear cells from a cohort of 46 healthy individuals. Logistic regression analysis adjusted for age and gender revealed that patients carrying the ATG10rs1864182G allele showed a significantly decreased risk of developing AML (OR [odds ratio] = 0.58, p = 0.001), whereas patients carrying the homozygous ATG10rs3734114C allele had a significantly increased risk of developing AML (OR = 2.70, p = 0.004). Functional analysis showed that individuals carrying the ATG10rs1864182G allele had decreased autophagy when compared to homozygous major allele carriers. Our results uncover the potential of screening for ATG10 genetic variants in AML prevention strategies, in particular for subjects carrying other AML risk factors such as elderly individuals with clonal hematopoiesis of indeterminate potential.
Collapse
|
536
|
Autophagy Triggers Tamoxifen Resistance in Human Breast Cancer Cells by Preventing Drug-Induced Lysosomal Damage. Cancers (Basel) 2021; 13:cancers13061252. [PMID: 33809171 PMCID: PMC7999102 DOI: 10.3390/cancers13061252] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Endocrine therapy with tamoxifen or other endocrine drugs represents the standard treatment for estrogen receptor-positive breast cancer. In spite of effectiveness of this therapy, onset of drug resistance worsens the prognosis of about 30% of patients. Autophagy has recently been proposed as a key player of drug resistance, but the underlying mechanisms are not completely understood. In this research, the authors investigate how autophagy triggers drug resistance in breast cancer cells. The results evidence that tamoxifen affects lysosome integrity, which suggests that this effect may contribute to the anticancer activity of this drug. Activation of autophagy and overexpression of iron-binding proteins synergize in protecting the lysosomal compartment, restraining drug effectiveness in breast cancer cells. According to these results, tamoxifen-resistant cells show an increased autophagic flux and overexpress iron-binding proteins. These findings indicate that screening for the level of iron-binding proteins may help to identify patients at risk for developing drug resistance. Abstract Endocrine resistance is a major complication during treatment of estrogen receptor-positive breast cancer. Although autophagy has recently gained increasing consideration among the causative factors, the link between autophagy and endocrine resistance remains elusive. Here, we investigate the autophagy-based mechanisms of tamoxifen resistance in MCF7 cells. Tamoxifen (Tam) triggers autophagy and affects the lysosomal compartment of MCF7 cells, such that activated autophagy supports disposal of tamoxifen-damaged lysosomes by lysophagy. MCF7 cells resistant to 5 µM tamoxifen (MCF7-TamR) have a higher autophagic flux and an enhanced resistance to Tam-induced lysosomal alterations compared to parental cells, which suggests a correlation between the two events. MCF7-TamR cells overexpress messenger RNAs (mRNAs) for metallothionein 2A and ferritin heavy chain, and they are re-sensitized to Tam by inhibition of autophagy. Overexpressing these proteins in parental MCF7 cells protects lysosomes from Tam-induced damage and preserves viability, while inhibiting autophagy abrogates lysosome protection. Consistently, we also demonstrate that other breast cancer cells that overexpress selected mRNAs encoding iron-binding proteins are less sensitive to Tam-induced lysosomal damage when autophagy is activated. Collectively, our data demonstrate that autophagy triggers Tam resistance in breast cancer cells by favoring the lysosomal relocation of overexpressed factors that restrain tamoxifen-induced lysosomal damage.
Collapse
|
537
|
Abstract
Autophagy is an evolutionarily conserved process necessary to maintain cell homeostasis in response to various forms of stress such as nutrient deprivation and hypoxia as well as functioning to remove damaged molecules and organelles. The role of autophagy in cancer varies depending on the stage of cancer. Cancer therapeutics can also simultaneously evoke cancer cell senescence and ploidy increase. Both cancer cell senescence and polyploidization are reversible by depolyploidization giving rise to the progeny. Autophagy activation may be indispensable for cancer cell escape from senescence/polyploidy. As cancer cell polyploidy is proposed to be involved in cancer origin, the role of autophagy in polyploidization/depolyploidization of senescent cancer cells seems to be crucial. Accordingly, this review is an attempt to understand the complicated interrelationships between reversible cell senescence/polyploidy and autophagy.
Collapse
|
538
|
Lee XC, Werner E, Falasca M. Molecular Mechanism of Autophagy and Its Regulation by Cannabinoids in Cancer. Cancers (Basel) 2021; 13:cancers13061211. [PMID: 33802014 PMCID: PMC7999886 DOI: 10.3390/cancers13061211] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary This review examines the complex function of autophagy in malignancy and explores its regulation by cannabinoids in different cancers. Autophagy is an important process in the maintenance of cellular homeostasis, through the degradation and recycling of cytoplasmic constituents. The action of autophagy is highly dependent on tumour stage and type and the receptors with which ligands interact. Cannabinoids are growingly being acknowledged for their anticancer activities and are known to stimulate several mechanisms such as apoptosis and autophagy. Better understanding the mechanism of action behind autophagy and its regulation by cannabinoids will allow the development of novel cancer therapeutics. Abstract Autophagy is a “self-degradation” process whereby malfunctioned cytoplasmic constituents and protein aggregates are engulfed by a vesicle called the autophagosome, and subsequently degraded by the lysosome. Autophagy plays a crucial role in sustaining protein homeostasis and can be an alternative source of energy under detrimental circumstances. Studies have demonstrated a paradoxical function for autophagy in cancer, displaying both tumour suppressive and tumour promotive roles. In early phases of tumour development autophagy promotes cancer cell death. In later phases, autophagy enables cancer cells to survive and withstand therapy. Cannabinoids, which are derivatives of the Cannabis sativa L. plant, have shown to be associated with autophagy induction in cells. There is an emerging interest in studying the signalling pathways involved in cannabinoid-induced autophagy and their potential application in anticancer therapies. In this review, the molecular mechanisms involved in the autophagy degradation process will be discussed. This review also highlights a role for autophagy in cancer progression, with cannabinoid-induced autophagy presenting a novel strategy for anticancer therapy.
Collapse
|
539
|
Recent Advances in Understanding the Role of Autophagy in Paediatric Brain Tumours. Diagnostics (Basel) 2021; 11:diagnostics11030481. [PMID: 33803216 PMCID: PMC8000899 DOI: 10.3390/diagnostics11030481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a degradative process occurring in eukaryotic cells to maintain homeostasis and cell survival. After stressful conditions including nutrient deprivation, hypoxia or drugs administration, autophagy is induced to counteract pathways that could lead to cell death. In cancer, autophagy plays a paradoxical role, acting both as tumour suppressor—by cleaning cells from damaged organelles and inhibiting inflammation or, alternatively, by promoting genomic stability and tumour adaptive response—or as a pro-survival mechanism to protect cells from stresses such as chemotherapy. Neural-derived paediatric solid tumours represent a variety of childhood cancers with unique anatomical location, cellular origins, and clinical presentation. These tumours are a leading cause of morbidity and mortality among children and new molecular diagnostics and therapies are necessary for longer survival and reduced morbidity. Here, we review advances in our understanding of how autophagy modulation exhibits antitumor properties in experimental models of paediatric brain tumours, i.e., medulloblastoma (MB), ependymoma (EPN), paediatric low-grade and high-grade gliomas (LGGs, HGGs), atypical teratoid/rhabdoid tumours (ATRTs), and retinoblastoma (RB). We also discuss clinical perspectives to consider how targeting autophagy may be relevant in these specific paediatric tumours.
Collapse
|
540
|
Yu J, Mao W, Xu B, Chen M. Construction and validation of an autophagy-related long noncoding RNA signature for prognosis prediction in kidney renal clear cell carcinoma patients. Cancer Med 2021; 10:2359-2369. [PMID: 33650306 PMCID: PMC7982638 DOI: 10.1002/cam4.3820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/30/2021] [Accepted: 02/18/2021] [Indexed: 12/18/2022] Open
Abstract
Purpose The purpose of this study was to identify autophagy‐associated long noncoding RNAs (ARlncRNAs) using the kidney renal clear cell carcinoma (KIRC) patient data from The Cancer Genome Atlas (TCGA) database and to construct a prognostic risk‐related ARlncRNAs signature to accurately predict the prognosis of KIRC patients. Methods The KIRC patient data were originated from TCGA database and were classified into a training set and testing set. Seven prognostic risk‐related ARlncRNAs, identified using univariate, lasso, and multivariate Cox regression analysis, were used to construct prognostic risk‐related signatures. Kaplan–Meier curves and receiver operating characteristic (ROC) curves as well as independent prognostic factor analysis and correlation analysis with clinical characteristics were utilized to evaluate and verify the specificity and sensitivity of the signature in training set and testing set, respectively. Two nomograms were established to predict the probable 1‐, 3‐, and 5‐year survival of the KIRC patients. In addition, the lncRNA‐mRNA co‐expression network was constructed and Gene Ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to identify biological functions of ARlncRNAs. Results We constructed and verified a prognostic risk‐related ARlncRNAs signature in training set and testing set, respectively. We found the survival time of KIRC patients with low‐risk scores was significantly better than those with high‐risk scores in training set and testing set. ROC curves suggested that the area under the ROC (AUC) value for prognostic risk score signature was 0.81 in training set and 0.705 in testing set. And AUC values corresponding to 1‐, 3‐, and 5 years of OS were 0.809, 0.753, and 0.794 in training set and 0.698, 0.682, and 0.754 in testing set, respectively. We established the two nomograms that confirmed high C‐index and accomplished good prediction accuracy. Conclusions We constructed a prognostic risk‐related ARlncRNAs signature that could accurately predict the prognosis of KIRC patients.
Collapse
Affiliation(s)
- JunJie Yu
- Department of medical college, Southeast University, Nanjing, China
| | - WeiPu Mao
- Department of medical college, Southeast University, Nanjing, China
| | - Bin Xu
- Department of Urology, Southeast University Zhongda hospital, Nanjing, China
| | - Ming Chen
- Department of Urology, Southeast University Zhongda hospital, Nanjing, China
| |
Collapse
|
541
|
Leidal AM, Debnath J. Emerging roles for the autophagy machinery in extracellular vesicle biogenesis and secretion. FASEB Bioadv 2021; 3:377-386. [PMID: 33977236 PMCID: PMC8103724 DOI: 10.1096/fba.2020-00138] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Autophagy classically functions to maintain cell health during stressful conditions by targeting cytosolic components for degradation and recycling via lysosomal pathways. However, accumulating evidence also supports roles for autophagy-related genes (ATGs) in non-degradative processes including cellular secretion. Here, we review emerging roles for the autophagy machinery in regulating extracellular vesicle loading and secretion and discuss how functional coupling of these pathways may impact normal physiology and disease.
Collapse
Affiliation(s)
- Andrew M Leidal
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center University of California San Francisco San Francisco CA USA
| | - Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center University of California San Francisco San Francisco CA USA
| |
Collapse
|
542
|
Mukhopadhyay S, Vander Heiden MG, McCormick F. The Metabolic Landscape of RAS-Driven Cancers from biology to therapy. NATURE CANCER 2021; 2:271-283. [PMID: 33870211 PMCID: PMC8045781 DOI: 10.1038/s43018-021-00184-x] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Our understanding of how the RAS protein family, and in particular mutant KRAS promote metabolic dysregulation in cancer cells has advanced significantly over the last decade. In this Review, we discuss the metabolic reprogramming mediated by oncogenic RAS in cancer, and elucidating the underlying mechanisms could translate to novel therapeutic opportunities to target metabolic vulnerabilities in RAS-driven cancers.
Collapse
Affiliation(s)
- Suman Mukhopadhyay
- National Cancer Institute RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Frank McCormick
- National Cancer Institute RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
543
|
Lin C, Blessing AM, Pulliam TL, Shi Y, Wilkenfeld SR, Han JJ, Murray MM, Pham AH, Duong K, Brun SN, Shaw RJ, Ittmann MM, Frigo DE. Inhibition of CAMKK2 impairs autophagy and castration-resistant prostate cancer via suppression of AMPK-ULK1 signaling. Oncogene 2021; 40:1690-1705. [PMID: 33531625 PMCID: PMC7935762 DOI: 10.1038/s41388-021-01658-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 12/18/2020] [Accepted: 01/14/2021] [Indexed: 01/30/2023]
Abstract
Previous work has suggested androgen receptor (AR) signaling mediates prostate cancer progression in part through the modulation of autophagy. However, clinical trials testing autophagy inhibition using chloroquine derivatives in men with castration-resistant prostate cancer (CRPC) have yet to yield promising results, potentially due to the side effects of this class of compounds. We hypothesized that identification of the upstream activators of autophagy in prostate cancer could highlight alternative, context-dependent targets for blocking this important cellular process during disease progression. Here, we used molecular, genetic, and pharmacological approaches to elucidate an AR-mediated autophagy cascade involving Ca2+/calmodulin-dependent protein kinase kinase 2 (CAMKK2; a kinase with a restricted expression profile), 5'-AMP-activated protein kinase (AMPK), and Unc-51 like autophagy activating kinase 1 (ULK1), but independent of canonical mechanistic target of rapamycin (mTOR) activity. Increased CAMKK2-AMPK-ULK1 signaling correlated with disease progression in genetic mouse models and patient tumor samples. Importantly, CAMKK2 disruption impaired tumor growth and prolonged survival in multiple CRPC preclinical mouse models. Similarly, an inhibitor of AMPK-ULK1 blocked autophagy, cell growth, and colony formation in prostate cancer cells. Collectively, our findings converge to demonstrate that AR can co-opt the CAMKK2-AMPK-ULK1 signaling cascade to promote prostate cancer by increasing autophagy. Thus, this pathway may represent an alternative autophagic target in CRPC.
Collapse
Affiliation(s)
- Chenchu Lin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Alicia M Blessing
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Thomas L Pulliam
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Yan Shi
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
| | - Sandi R Wilkenfeld
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jenny J Han
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mollianne M Murray
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander H Pham
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
| | - Kevin Duong
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Sonja N Brun
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Reuben J Shaw
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael M Ittmann
- Departments of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Houston, TX, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA.
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA.
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
544
|
Machado ER, Annunziata I, van de Vlekkert D, Grosveld GC, d’Azzo A. Lysosomes and Cancer Progression: A Malignant Liaison. Front Cell Dev Biol 2021; 9:642494. [PMID: 33718382 PMCID: PMC7952443 DOI: 10.3389/fcell.2021.642494] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/08/2021] [Indexed: 01/04/2023] Open
Abstract
During primary tumorigenesis isolated cancer cells may undergo genetic or epigenetic changes that render them responsive to additional intrinsic or extrinsic cues, so that they enter a transitional state and eventually acquire an aggressive, metastatic phenotype. Among these changes is the alteration of the cell metabolic/catabolic machinery that creates the most permissive conditions for invasion, dissemination, and survival. The lysosomal system has emerged as a crucial player in this malignant transformation, making this system a potential therapeutic target in cancer. By virtue of their ubiquitous distribution in mammalian cells, their multifaced activities that control catabolic and anabolic processes, and their interplay with other organelles and the plasma membrane (PM), lysosomes function as platforms for inter- and intracellular communication. This is due to their capacity to adapt and sense nutrient availability, to spatially segregate specific functions depending on their position, to fuse with other compartments and with the PM, and to engage in membrane contact sites (MCS) with other organelles. Here we review the latest advances in our understanding of the role of the lysosomal system in cancer progression. We focus on how changes in lysosomal nutrient sensing, as well as lysosomal positioning, exocytosis, and fusion perturb the communication between tumor cells themselves and between tumor cells and their microenvironment. Finally, we describe the potential impact of MCS between lysosomes and other organelles in propelling cancer growth and spread.
Collapse
Affiliation(s)
- Eda R. Machado
- Department of Genetics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Ida Annunziata
- Department of Genetics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | | | - Gerard C. Grosveld
- Department of Genetics, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Alessandra d’Azzo
- Department of Genetics, St. Jude Children’s Research Hospital, Memphis, TN, United States
- Department of Anatomy and Neurobiology, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
545
|
Li Q, Ni Y, Zhang L, Jiang R, Xu J, Yang H, Hu Y, Qiu J, Pu L, Tang J, Wang X. HIF-1α-induced expression of m6A reader YTHDF1 drives hypoxia-induced autophagy and malignancy of hepatocellular carcinoma by promoting ATG2A and ATG14 translation. Signal Transduct Target Ther 2021; 6:76. [PMID: 33619246 PMCID: PMC7900110 DOI: 10.1038/s41392-020-00453-8] [Citation(s) in RCA: 246] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/15/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022] Open
Abstract
N6-methyladenosine (m6A), and its reader protein YTHDF1, play a pivotal role in human tumorigenesis by affecting nearly every stage of RNA metabolism. Autophagy activation is one of the ways by which cancer cells survive hypoxia. However, the possible involvement of m6A modification of mRNA in hypoxia-induced autophagy was unexplored in human hepatocellular carcinoma (HCC). In this study, specific variations in YTHDF1 expression were detected in YTHDF1-overexpressing, -knockout, and -knockdown HCC cells, HCC organoids, and HCC patient-derived xenograft (PDX) murine models. YTHDF1 expression and hypoxia-induced autophagy were significantly correlated in vitro; significant overexpression of YTHDF1 in HCC tissues was associated with poor prognosis. Multivariate cox regression analysis identified YTHDF1 expression as an independent prognostic factor in patients with HCC. Multiple HCC models confirmed that YTHDF1 deficiency inhibited HCC autophagy, growth, and metastasis. Luciferase reporter assays and chromatin immunoprecipitation demonstrated that HIF-1α regulated YTHDF1 transcription by directly binding to its promoter region under hypoxia. The results of methylated RNA immunoprecipitation sequencing, proteomics, and polysome profiling indicated that YTHDF1 contributed to the translation of autophagy-related genes ATG2A and ATG14 by binding to m6A-modified ATG2A and ATG14 mRNA, thus facilitating autophagy and autophagy-related malignancy of HCC. Taken together, HIF-1α-induced YTHDF1 expression was associated with hypoxia-induced autophagy and autophagy-related HCC progression via promoting translation of autophagy-related genes ATG2A and ATG14 in a m6A-dependent manner. Our findings suggest that YTHDF1 is a potential prognostic biomarker and therapeutic target for patients with HCC.
Collapse
Affiliation(s)
- Qing Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China.,School of Medicine, Southeast University, Nanjing, China
| | - Yong Ni
- Department of Hepatopancreatobiliary Surgery, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Liren Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Runqiu Jiang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, People's Republic of China.,Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jing Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hong Yang
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yuanchang Hu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Jiannan Qiu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China.
| | - Jinhai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China. .,School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
546
|
Long noncoding RNA H19 contributes to the proliferation and autophagy of glioma cells through mTOR/ULK1 pathway. Neuroreport 2021; 32:352-358. [PMID: 33661803 DOI: 10.1097/wnr.0000000000001602] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Long noncoding RNA (LncRNA) H19 has been proven to be involved in many kinds of cancers including glioma, and a previous study has shown an autophagy regulation of H19. The mammalian target of rapamycin (mTOR) signaling pathway plays a key role in autophagy and Unc-51 like autophagy activating kinase 1 (ULK1) is also thought to be involved in autophagy signaling. In our study, we investigated the role of mTOR/ULK1 autophagy signaling in the H19-mediated promotion of glioma proliferation. Human glioma cells U87 and U251 and normal human astrocytes HA1800 were used in the study. First, the expression of H19 was determined in U87, U251, and HA1800 cells. Then, the cell proliferation and migration of glioma cells were detected, while the protein levels of main molecules of the mTOR/ULK1 pathway and autophagy-related proteins were also examined. Rapamycin, an inhibitor of mTOR, was used to further study the role of H19 in autophagy. We observed that overexpressed H19 promoted the proliferation and migration in glioma cells. The autophagy of U87 cells was suppressed when H19 was overexpressed and enhanced when H19 was silenced. H19 overexpression inhibited mTOR phosphorylation and promoted ULK1 phosphorylation. H19 promoted proliferation, migration, and autophagy by regulating mTOR signaling. In conclusion, we validate that H19 contributes to the proliferation and autophagy of glioma cells through the mTOR/ULK1 pathway.
Collapse
|
547
|
Li Y, Wang Y, Yu X, Yu T, Zheng X, Chu Q. Radix Tetrastigma Inhibits the Non-Small Cell Lung Cancer via Bax/Bcl-2/Caspase-9/Caspase-3 Pathway. Nutr Cancer 2021; 74:320-332. [PMID: 33586527 DOI: 10.1080/01635581.2021.1881569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lung cancer with high mortality is regarded as a challenging global problem with unsatisfied curative effects. Clinically, the chemotherapy drugs are often faced with side-effects and tumor resistance. Radix Tetrastigma (RT) is a traditional Chinese herb and now regarded as a kind of functional food. In this study, A549-bearing nude mice control was adopted to evaluate the anti-tumor capacity of RT. Results demonstrated that RT showed excellent anti-tumor ability with no side-effect on mice compared to chemotherapy drug (5-Fu).Further studies proved that RT down-regulated the proliferation-related proteins (PCNA, Ki67) and vascular endothelial growth factor (VEGF). Additionally, RT up-regulated the ratio of Bax/Bcl-2, which caused the over-expression of Caspase-9, leading to the activation of downstream protein caspase-3, eventually resulting in apoptosis of A549 in solid tumor. These results together suggest that RT inhibits the non-small cell lung cancer (NSCLC) via Bax/Bcl2/Caspase-9/Caspase-3 pathway. Furthermore, the anti-A549 abilities of the main flavonoid components from RT were compared, and kaempferol-3-O-rutinoside would play a role in RT's outstanding anti-NSCLC ability.
Collapse
Affiliation(s)
- Yonglu Li
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, People's Republic of China.,Zhejiang Key Laboratory for Agro-food Processing, Zhejiang University, Hangzhou, People's Republic of China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Yaxuan Wang
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, People's Republic of China.,Zhejiang Key Laboratory for Agro-food Processing, Zhejiang University, Hangzhou, People's Republic of China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Xin Yu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, People's Republic of China.,Zhejiang Key Laboratory for Agro-food Processing, Zhejiang University, Hangzhou, People's Republic of China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Ting Yu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, People's Republic of China.,Zhejiang Key Laboratory for Agro-food Processing, Zhejiang University, Hangzhou, People's Republic of China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, People's Republic of China.,Zhejiang Key Laboratory for Agro-food Processing, Zhejiang University, Hangzhou, People's Republic of China.,Fuli Institute of Food Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Qiang Chu
- State Key Laboratory of Silicon Materials, School of Materials Science and Engineering, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
548
|
Wang L, Ning S. TRIMming Type I Interferon-Mediated Innate Immune Response in Antiviral and Antitumor Defense. Viruses 2021; 13:279. [PMID: 33670221 PMCID: PMC7916971 DOI: 10.3390/v13020279] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
The tripartite motif (TRIM) family comprises at least 80 members in humans, with most having ubiquitin or SUMO E3 ligase activity conferred by their N-terminal RING domain. TRIMs regulate a wide range of processes in ubiquitination- or sumoylation-dependent manners in most cases, and fewer as adaptors. Their roles in the regulation of viral infections, autophagy, cell cycle progression, DNA damage and other stress responses, and carcinogenesis are being increasingly appreciated, and their E3 ligase activities are attractive targets for developing specific immunotherapeutic strategies for immune diseases and cancers. Given their importance in antiviral immune response, viruses have evolved sophisticated immune escape strategies to subvert TRIM-mediated mechanisms. In this review, we focus on their regulation of IFN-I-mediated innate immune response, which plays key roles in antiviral and antitumor defense.
Collapse
Affiliation(s)
- Ling Wang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA;
- Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Shunbin Ning
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA;
- Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
549
|
Seven new prenylated flavanones from the roots of Sophora flavescens and their anti-proliferative activities. Bioorg Chem 2021; 109:104716. [PMID: 33607362 DOI: 10.1016/j.bioorg.2021.104716] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 01/07/2023]
Abstract
Aiming to discover potent anti-proliferative agents from the roots of Sophora flavescens, seven new prenylated flavanones were isolated, along with 16 known compounds. Their structures were elucidated by interpretation of their spectroscopic data (1D and 2D NMR, UV, IR, CD, and HRESIMS) and comparison to literature data. In the in vitro assay, 21 showed anti-proliferative activity against human hepatoma cells (HepG2). Studies of its mechanism revealed that 21 could significantly activate autophagic flux and trigger ROS release in HepG2 cells. Western blot experiments demonstrated that 21 could activate the key signaling protein of autophagy and ROS, while it does not affect the main protein of the apoptosis signaling pathway. These results suggested that 21 mediates its anti-proliferative effects through autophagic cell death, which is apoptosis-independent.
Collapse
|
550
|
Chaetocochin J, an epipolythiodioxopiperazine alkaloid, induces apoptosis and autophagy in colorectal cancer via AMPK and PI3K/AKT/mTOR pathways. Bioorg Chem 2021; 109:104693. [PMID: 33609914 DOI: 10.1016/j.bioorg.2021.104693] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/26/2021] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is the third commonly diagnosed malignancy and the second leading cause of cancer death worldwide. Development of novel chemotherapeutics is crucial. Natural products are the main source of drug discovery, and epipolythiodioxopiperazine (ETP) alkaloids are one kind of them have been reported to have potent biological activities. In the present study, we first isolated Chaetocochin J (CJ), an ETP alkaloid from the secondary metabolites of Chaetomium sp, and studied the anti-CRC activity and mechanism of it. The results showed that CJ exhibits potent proliferation inhibition effect, its IC50 to CRC cells are around 0.5 µM. CJ also induces apoptosis of CRC cells in a dose-dependent manner, and this effect is stronger than topotecan. In addition, CJ treatment triggers autophagic flux in CRC cells, inhibition of autophagy by chloroquine didn't affect CJ-induced apoptosis and growth inhibition, suggesting CJ may simultaneously induced apoptosis and autophagy in CRC cells. We further explored the mechanism of action, and found that CJ exerts its anti-CRC function via AMPK and PI3K/AKT/mTOR pathways and further regulation of their downstream signaling cascade in CRC cells, including apoptosis and autophagy. These data potently suggest that CJ may be a potential drug candidate for CRC treatment.
Collapse
|