1
|
Chowdhury P, Dey Talukdar P, Mukherjee P, Dey D, Chatterji U, Sengupta S. Hemin-induced reactive oxygen species triggers autophagy-dependent macrophage differentiation and pro-inflammatory responses in THP-1 cells. Exp Cell Res 2024; 442:114216. [PMID: 39182663 DOI: 10.1016/j.yexcr.2024.114216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/18/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
The toxic effect of oxidized-heme, also known as hemin, is implicated in developing adverse clinical outcome in various hematolytic diseases. To simulate and reconstruct the molecular events associated with hemin exposure on circulating monocytes, we employed a THP-1 cell line based in vitro model. Flow cytometry and Western blot analyses were subsequently applied. Hemin-treated THP-1 produced ROS in a dose-dependent manner which resulted in 10-30 % of cell death primarily through apoptosis. Surviving cells induced autophagy which too was ROS-dependent, as revealed by application of N-acetyl-L-cysteine. Hemin-mediated autophagy promoted differentiation of CD14+ THP-1 cells into CD11b+ macrophages. Application of 3-methyladenine, reinforced that differentiation of THP-1 was an autophagy-dependent process. It was revealed that despite a higher polarization towards M2-macrophage, synthesis of pro-inflammatory cytokines namely TNF-α, IL-1A, IL-2, IL-8 and IL-17A predominated. IL-6, a pleiotropic cytokine, was also elevated. It may thus be surmised that hemin-induced pro-inflammatory response in THP-1 is downstream to ROS-dependent autophagy and monocyte differentiation. This finding is translationally meaningful as hemin is already approved by FDA for amelioration of acute porphyria and is actively considered as a therapeutic agent for other diseases. This study underscores the need of further research untangling the reciprocal regulation of inflammatory signaling and autophagy under oxidative stress.
Collapse
Affiliation(s)
- Pramita Chowdhury
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Priyanka Dey Talukdar
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Pritha Mukherjee
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Debangana Dey
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Urmi Chatterji
- Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India
| | - Sanghamitra Sengupta
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019, West Bengal, India.
| |
Collapse
|
2
|
Steinmetz T, Thomas J, Reimann L, Himmelreich AK, Schulz SR, Golombek F, Castiglione K, Jäck HM, Brodesser S, Warscheid B, Mielenz D. Identification of TFG- and Autophagy-Regulated Proteins and Glycerophospholipids in B Cells. J Proteome Res 2024; 23:1615-1633. [PMID: 38649144 PMCID: PMC11077586 DOI: 10.1021/acs.jproteome.3c00713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 04/25/2024]
Abstract
Autophagy supervises the proteostasis and survival of B lymphocytic cells. Trk-fused gene (TFG) promotes autophagosome-lysosome flux in murine CH12 B cells, as well as their survival. Hence, quantitative proteomics of CH12tfgKO and WT B cells in combination with lysosomal inhibition should identify proteins that are prone to lysosomal degradation and contribute to autophagy and B cell survival. Lysosome inhibition via NH4Cl unexpectedly reduced a number of proteins but increased a large cluster of translational, ribosomal, and mitochondrial proteins, independent of TFG. Hence, we propose a role for lysosomes in ribophagy in B cells. TFG-regulated proteins include CD74, BCL10, or the immunoglobulin JCHAIN. Gene ontology (GO) analysis reveals that proteins regulated by TFG alone, or in concert with lysosomes, localize to mitochondria and membrane-bound organelles. Likewise, TFG regulates the abundance of metabolic enzymes, such as ALDOC and the fatty acid-activating enzyme ACOT9. To test consequently for a function of TFG in lipid metabolism, we performed shotgun lipidomics of glycerophospholipids. Total phosphatidylglycerol is more abundant in CH12tfgKO B cells. Several glycerophospholipid species with similar acyl side chains, such as 36:2 phosphatidylethanolamine and 36:2 phosphatidylinositol, show a dysequilibrium. We suggest a role for TFG in lipid homeostasis, mitochondrial functions, translation, and metabolism in B cells.
Collapse
Affiliation(s)
- Tobit
D. Steinmetz
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Jana Thomas
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Lena Reimann
- CIBSS
Centre for Integrative Biological Signalling Studies, University of Freiburg, D-79104 Freiburg, Germany
| | - Ann-Kathrin Himmelreich
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Sebastian R. Schulz
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Florian Golombek
- Chair
of Bioprocess Engineering, Technical Faculty, FAU Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Kathrin Castiglione
- Chair
of Bioprocess Engineering, Technical Faculty, FAU Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Hans-Martin Jäck
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
- FAU
Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| | - Susanne Brodesser
- Cologne
Excellence Cluster on Cellular Stress Responses in Aging-associated
Diseases (CECAD), University of Köln, D-50931 Köln, Germany
| | - Bettina Warscheid
- CIBSS
Centre for Integrative Biological Signalling Studies, University of Freiburg, D-79104 Freiburg, Germany
- Department
of Biochemistry, Theodor Boveri-Institute, Biocenter, University of Würzburg, D-97074 Würzburg, Germany
| | - Dirk Mielenz
- Division
of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Zentrum, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, D-91054 Erlangen, Germany
- FAU
Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, D-91054 Erlangen, Germany
| |
Collapse
|
3
|
Mirabdali S, Ghafouri K, Farahmand Y, Gholizadeh N, Yazdani O, Esbati R, Hajiagha BS, Rahimi A. The role and function of autophagy through signaling and pathogenetic pathways and lncRNAs in ovarian cancer. Pathol Res Pract 2024; 253:154899. [PMID: 38061269 DOI: 10.1016/j.prp.2023.154899] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 01/24/2024]
Abstract
Lysosomal-driven autophagy is a tightly controlled cellular catabolic process that breaks down and recycles broken or superfluous cell parts. It is involved in several illnesses, including cancer, and is essential in preserving cellular homeostasis. Autophagy prevents DNA mutation and cancer development by actively eliminating pro-oxidative mitochondria and protein aggregates from healthy cells. Oncosuppressor and oncogene gene mutations cause dysregulation of autophagy. Increased autophagy may offer cancer cells a pro-survival advantage when oxygen and nutrients are scarce and resistance to chemotherapy and radiation. This finding justifies the use of autophagy inhibitors in addition to anti-neoplastic treatments. Excessive autophagy levels can potentially kill cells. The diagnosis and treatment of ovarian cancer present many difficulties due to its complexity and heterogeneity. Understanding the role of autophagy, a cellular process involved in the breakdown and recycling of cellular components, in ovarian cancer has garnered increasing attention in recent years. Of particular note is the increasing amount of data indicating a close relationship between autophagy and ovarian cancer. Autophagy either promotes or restricts tumor growth in ovarian cancer. Dysregulation of autophagy signaling pathways in ovarian cancers can affect the development, metastasis, and response to tumor treatment. The precise mechanism underlying autophagy concerning ovarian cancer remains unclear, as does the role autophagy plays in ovarian carcinoma. In this review, we tried to encapsulate and evaluate current findings in investigating autophagy in ovarian cancer.
Collapse
Affiliation(s)
- Seyedsaber Mirabdali
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kimia Ghafouri
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yalda Farahmand
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Gholizadeh
- Department of Dermatology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Omid Yazdani
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Romina Esbati
- Department of Medicine, Shahid Beheshti University, Tehran, Iran
| | - Bahareh Salmanian Hajiagha
- Department of Cellular and Molecular Biology, Faculty of Basic Science, Tehran East Branch, Islamic Azad University, Tehran, Iran.
| | - Asiye Rahimi
- Faculty of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Huang Y, Li S, He S, Li Y, He Q, Wu Y. Chlamydia psittaci inclusion membrane protein CPSIT_0842 induces macrophage apoptosis through MAPK/ERK-mediated autophagy. Int J Biochem Cell Biol 2023; 157:106376. [PMID: 36716815 DOI: 10.1016/j.biocel.2023.106376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/20/2022] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
Chlamydia psittaci is a multi-host zoonotic pathogen, which mainly infects poultry and inflicts an appreciable economic burden on the livestock farming industry. C. psittaci inclusion membrane proteins are uniquely positioned at the host-pathogen interface and are important virulence proteins. We have previously confirmed that Incs regulate host cell survival to help Chlamydia sp. evade host-cell-mediated defense mechanisms. However, the role of the Inc, CPSIT_0842, in the regulation of cell death following the establishment of persistent C. psittaci infection remains unknown. This study explored the effect of CPSIT_0842 on the crosstalk between the autophagic and apoptotic pathways in macrophages. Results showed that CPSIT_0842 initiated autophagy and blocked autophagic flux in human macrophages, as indicated by autophagy-related protein LC3-II, Beclin-1, and p62 upregulation, autophagosome accumulation, and lysosomal protein LAMP1 diminution. We also showed that the disruption of autophagic flux had a regulatory effect on CPSIT_0842-induced apoptosis. Moreover, the suppression of autophagy initiation by 3-methyladenine attenuated CPSIT_0842-induced apoptosis. By contrast, the induction of autophagic flux by rapamycin did not significantly affect CPSIT_0842-induced apoptosis. Taken together, these findings demonstrate that CPSIT_0842 induced macrophage apoptosis by initiating incomplete autophagy through the MAPK/ERK/mTOR signaling pathway, which may be instrumental to the ability of C. psittaci to evade the host innate immune response and establish persistent infection. The improved understanding of the autophagic and cell death pathways triggered upon bacterial inclusion will likely help in the development of novel treatment strategies for chlamydia infection.
Collapse
Affiliation(s)
- Yanru Huang
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001, Hunan, China
| | - Sijia Li
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001, Hunan, China
| | - Siqin He
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001, Hunan, China
| | - Yumeng Li
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001, Hunan, China; Department of Clinical Laboratory, The First Affiliated Hospital of University of South China, Hengyang 421000, Hunan, China
| | - Qingzhi He
- School of Biotechnology, Guilin Medical University, Guilin 541199, China
| | - Yimou Wu
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
5
|
Campellone KG, Lebek NM, King VL. Branching out in different directions: Emerging cellular functions for the Arp2/3 complex and WASP-family actin nucleation factors. Eur J Cell Biol 2023; 102:151301. [PMID: 36907023 DOI: 10.1016/j.ejcb.2023.151301] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/07/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The actin cytoskeleton impacts practically every function of a eukaryotic cell. Historically, the best-characterized cytoskeletal activities are in cell morphogenesis, motility, and division. The structural and dynamic properties of the actin cytoskeleton are also crucial for establishing, maintaining, and changing the organization of membrane-bound organelles and other intracellular structures. Such activities are important in nearly all animal cells and tissues, although distinct anatomical regions and physiological systems rely on different regulatory factors. Recent work indicates that the Arp2/3 complex, a broadly expressed actin nucleator, drives actin assembly during several intracellular stress response pathways. These newly described Arp2/3-mediated cytoskeletal rearrangements are coordinated by members of the Wiskott-Aldrich Syndrome Protein (WASP) family of actin nucleation-promoting factors. Thus, the Arp2/3 complex and WASP-family proteins are emerging as crucial players in cytoplasmic and nuclear activities including autophagy, apoptosis, chromatin dynamics, and DNA repair. Characterizations of the functions of the actin assembly machinery in such stress response mechanisms are advancing our understanding of both normal and pathogenic processes, and hold great promise for providing insights into organismal development and interventions for disease.
Collapse
Affiliation(s)
- Kenneth G Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA.
| | - Nadine M Lebek
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA
| | - Virginia L King
- Department of Molecular and Cell Biology, Institute for Systems Genomics; University of Connecticut; Storrs, CT, USA
| |
Collapse
|
6
|
Kundu M, Das S, Das CK, Kulkarni G, Das S, Dhara D, Mandal M. Magnolol induces cytotoxic autophagy in glioma by inhibiting PI3K/AKT/mTOR signaling. Exp Cell Res 2023; 424:113488. [PMID: 36736226 DOI: 10.1016/j.yexcr.2023.113488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 12/18/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023]
Abstract
Glioma is difficult-to-treat because of its infiltrative nature and the presence of the blood-brain barrier. Temozolomide is the only FDA-approved drug for its management. Therefore, finding a novel chemotherapeutic agent for glioma is of utmost importance. Magnolol, a neolignan, has been known for its apoptotic role in glioma. In this work, we have explored a novel anti-glioma mechanism of Magnolol associated with its role in autophagy modulation. We found increased expression levels of Beclin-1, Atg5-Atg12, and LC3-II and lower p62 expression in Magnolol-treated glioma cells. PI3K/AKT/mTOR pathway proteins were also downregulated in Magnolol-treated glioma cells. Next, we treated the glioma cells with Insulin, a stimulator of PI3K/AKT/mTOR signaling, to confirm that Magnolol induced autophagy by inhibiting this pathway. Insulin reversed the effect on Magnolol-mediated autophagy induction. We also established the same in in vivo glioma model where Magnolol showed an anti-glioma effect by inducing autophagy. To confirm the cytotoxic effect of Magnolol-induced autophagy, we used Chloroquine, a late-stage autophagy inhibitor. Chloroquine efficiently reversed the anti-glioma effects of Magnolol both in vitro and in vivo. Our study revealed the cytotoxic effect of Magnolol-induced autophagy in glioma, which was not previously reported. Additionally, Magnolol showed no toxicity in non-cancerous cell lines as well as rat organs. Thus, we concluded that Magnolol is an excellent candidate for developing new therapeutic strategies for glioma management.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Chandan Kanta Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Gaurav Kulkarni
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Soumen Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Dibakar Dhara
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
7
|
Udupa P, Kumar A, Parit R, Ghosh DK. Acyl-CoA binding protein regulates nutrient-dependent autophagy. Metabolism 2022:155338. [PMID: 36280213 DOI: 10.1016/j.metabol.2022.155338] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Homeostasis of autophagy under normal conditions and nutrient stress is maintained by adaptive activation of regulatory proteins. However, the protein-lipid crosstalk that modulates the switch from suppression to activation of autophagy initiation is largely unknown. RESULTS Here, we show that human diazepam-binding inhibitor (DBI), also known as acyl-CoA binding protein (ACBP), binds to phosphatidylethanolamine of the phagophore membrane under nutrient-rich growth conditions, leading to inhibition of LC3 lipidation and suppression of autophagy initiation. Specific residues, including the conserved tyrosine residues of DBI, interact with phosphatidylethanolamine to stabilize the later molecule in the acyl-CoA binding cavity of the protein. Under starvation, phosphorylation of serine-21 of DBI mediated by the AMP-activated protein kinase results in a drastic reduction in the affinity of the protein for phosphatidylethanolamine. The release of serine-21 phosphorylated DBI from the phagophore upon nutrient starvation restores the high LC3 lipidation flux and maturation of the phagophore to autophagosome. CONCLUSION DBI acts as a strategic barrier against overactivation of phagophore maturation under nutrient-rich conditions, while triggering autophagy under nutrient-deficient conditions.
Collapse
Affiliation(s)
- Prajna Udupa
- Departmentof Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Abhishek Kumar
- School of Medicine, University of California, San Francisco, United States of America
| | - Rahul Parit
- Departmentof Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Debasish Kumar Ghosh
- Departmentof Medical Genetics, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
8
|
The effect of lactoferrin on ULK1 and ATG13 genes expression in breast cancer cell line MCF7 and bioinformatics studies of protein interaction between lactoferrin and the autophagy initiation complex. Cell Biochem Biophys 2022; 80:795-806. [PMID: 36169801 DOI: 10.1007/s12013-022-01097-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 09/12/2022] [Indexed: 11/03/2022]
Abstract
Recently, the study of autophagy and its mechanism on the cancer cell growth process has received much attention. lactoferrin (Lf) is a glycoprotein with various biological activities, including antibacterial, antiviral, anti-cancer, etc. In the present study, the effect of different concentrations of lactoferrin on the expression of ULK1 and ATG13 genes was evaluated in breast cancer cell line MCF7 using real-time PCR technique as well as the molecular mechanism of these two genes and their proteins in the autophagy pathway and the relationship between lactoferrin and these proteins were investigated by bioinformatics studies. The result showed that the expression of the ULK1 gene at a concentration of 500 μg/ml of lactoferrin was significantly (P < 0.007) increased compared to the control and two other concentrations. Also, the expression of the ATG13 gene at all three concentrations was not significantly different from each other and compared to the control (P = 0.635). In the immunoblot of ULK1 protein at a concentration of 500 µg, more protein expression was observed. The binding mode of lactoferrin with ULK1, ATG13, and ATG101 proteins was obtained using docking. According to docking results, the N-lobe region of lactoferrin interacts with the PS domain of the ULK1 protein, and the N-lobe region of lactoferrin interacts with the horma domain of the ATG 13 and ATG101 proteins. The results show that lactoferrin, in addition to acting on the gene, interacts with ULK1, ATG13, and ATG101 proteins. Since all three proteins are components of the autophagy initiation complex, lactoferrin can induce autophagy in this way.
Collapse
|
9
|
Berth SH, Rich DJ, Lloyd TE. The role of autophagic kinases in regulation of axonal function. Front Cell Neurosci 2022; 16:996593. [PMID: 36226074 PMCID: PMC9548526 DOI: 10.3389/fncel.2022.996593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
Autophagy is an essential process for maintaining cellular homeostasis. Highlighting the importance of proper functioning of autophagy in neurons, disruption of autophagy is a common finding in neurodegenerative diseases. In recent years, evidence has emerged for the role of autophagy in regulating critical axonal functions. In this review, we discuss kinase regulation of autophagy in neurons, and provide an overview of how autophagic kinases regulate axonal processes, including axonal transport and axonal degeneration and regeneration. We also examine mechanisms for disruption of this process leading to neurodegeneration, focusing on the role of TBK1 in pathogenesis of Amyotrophic Lateral Sclerosis.
Collapse
|
10
|
Function and regulation of ULK1: From physiology to pathology. Gene 2022; 840:146772. [PMID: 35905845 DOI: 10.1016/j.gene.2022.146772] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/03/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022]
Abstract
The expression of ULK1, a core protein of autophagy, is closely related to autophagic activity. Numerous studies have shown that pathological abnormal expression of ULK1 is associated with various human diseases such as neurological disorders, infections, cardiovascular diseases, liver diseases and cancers. In addition, new advances in the regulation of ULK1 have been identified. Furthermore, targeting ULK1 as a therapeutic strategy for diseases is gaining attention as new corresponding activators or inhibitors are being developed. In this review, we describe the structure and regulation of ULK1 as well as the current targeted activators and inhibitors. Moreover, we highlight the pathological disorders of ULK1 expression and its critical role in human diseases.
Collapse
|
11
|
Ki YS, Chung KS, Lee HW, Choi JH, Tapondjou LA, Jang E, Lee KT. Pennogenin-3-O-α-L-Rhamnopyranosyl-(1→2)-[α-L-Rhamnopyranosyl-(1→3)]-β-D-Glucopyranoside (Spiroconazol A) Isolated from Dioscorea bulbifera L. var. sativa Induces Autophagic Cell Death by p38 MAPK Activation in NSCLC Cells. Pharmaceuticals (Basel) 2022; 15:ph15070893. [PMID: 35890190 PMCID: PMC9319756 DOI: 10.3390/ph15070893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 02/01/2023] Open
Abstract
In our previous study, we reported the isolation of pennogenin-3-O-α-L-rhamnopyranosyl-(1→2)-[α-L-rhamnopyranosyl-(1→3)]-β-D-glucopyranoside (spiroconazol A), a steroidal saponin, from the flowers of Dioscorea bulbifera L. var. sativa. In the present study, we aimed to investigate the effects of spiroconazol A on autophagy and its underlying mechanisms in A549 and NCI-H358 human non-small cell lung cancer (NSCLC) cells. Spiroconazol A inhibited the proliferation of NSCLC cells in a concentration- and time-dependent manner. To determine the type of programmed cell death induced by spiroconazol A, we performed a characterization of apoptosis in spiroconazol A-treated A549 cells. Our results showed that spiroconazol A significantly suppressed A549 cell viability but did not influence cell apoptosis because phosphatidylserine and caspase activation were not detected. Furthermore, spiroconazol A treatment upregulated the expression of LC3-II and autophagy-related Beclin-1 protein, suggesting that spiroconazol A induces autophagy in A549 cells. Moreover, spiroconazol A activated the phosphorylation of p38 mitogen-activated protein kinase (MAPK) but did not affect the phosphorylation of Janus kinase or ERK1/2. Notably, SB203580, a p38 MAPK inhibitor, had a significant inhibitory effect on spiroconazol A-induced autophagic cell death in A549 cells. Our results indicated that spiroconazol A-induced autophagy is dependent on p38 MAPK signaling and has potential as a therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Yo Sook Ki
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Seoul 02447, Korea; (Y.S.K.); (K.-S.C.); (H.-W.L.)
| | - Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Seoul 02447, Korea; (Y.S.K.); (K.-S.C.); (H.-W.L.)
| | - Heon-Woo Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Seoul 02447, Korea; (Y.S.K.); (K.-S.C.); (H.-W.L.)
| | - Jung-Hye Choi
- Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Seoul 02447, Korea;
| | - Léon Azefack Tapondjou
- Department of Chemistry, Faculty of Science, University of Dschang, Dschang P.O. Box 183, Cameroon;
| | - Eungyeong Jang
- Department of Internal Medicine, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Seoul 02447, Korea;
- Department of Internal Medicine, Kyung Hee University Korean Medicine Hospital, 23, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Seoul 02447, Korea; (Y.S.K.); (K.-S.C.); (H.-W.L.)
- Correspondence: ; Tel.: +82-2-961-0860
| |
Collapse
|
12
|
Lahiri V, Metur SP, Hu Z, Song X, Mari M, Hawkins WD, Bhattarai J, Delorme-Axford E, Reggiori F, Tang D, Dengjel J, Klionsky DJ. Post-transcriptional regulation of ATG1 is a critical node that modulates autophagy during distinct nutrient stresses. Autophagy 2022; 18:1694-1714. [PMID: 34836487 PMCID: PMC9298455 DOI: 10.1080/15548627.2021.1997305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy/autophagy is a highly conserved nutrient-recycling pathway that eukaryotes utilize to combat diverse stresses including nutrient depletion. Dysregulation of autophagy disrupts cellular homeostasis leading to starvation susceptibility in yeast and disease development in humans. In yeast, the robust autophagy response to starvation is controlled by the upregulation of ATG genes, via regulatory processes involving multiple levels of gene expression. Despite the identification of several regulators through genetic studies, the predominant mechanism of regulation modulating the autophagy response to subtle differences in nutrient status remains undefined. Here, we report the unexpected finding that subtle changes in nutrient availability can cause large differences in autophagy flux, governed by hitherto unknown post-transcriptional regulatory mechanisms affecting the expression of the key autophagyinducing kinase Atg1 (ULK1/ULK2 in mammals). We have identified two novel post-transcriptional regulators of ATG1 expression, the kinase Rad53 and the RNA-binding protein Ded1 (DDX3 in mammals). Furthermore, we show that DDX3 regulates ULK1 expression post-transcriptionally, establishing mechanistic conservation and highlighting the power of yeast biology in uncovering regulatory mechanisms that can inform therapeutic approaches.
Collapse
Affiliation(s)
- Vikramjit Lahiri
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Shree Padma Metur
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Zehan Hu
- Department of Biology, University of Fribourg, FribourgSwitzerland
| | - Xinxin Song
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Muriel Mari
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, GroningenThe Netherlands
| | - Wayne D. Hawkins
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Janakraj Bhattarai
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | | | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, GroningenThe Netherlands
| | - Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joern Dengjel
- Department of Biology, University of Fribourg, FribourgSwitzerland
| | - Daniel J. Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Chemosensitization Effect of Seabuckthorn ( Hippophae rhamnoides L.) Pulp Oil via Autophagy and Senescence in NSCLC Cells. Foods 2022; 11:foods11101517. [PMID: 35627086 PMCID: PMC9140501 DOI: 10.3390/foods11101517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/03/2022] Open
Abstract
The research has demonstrated a synergistic anticancer effect of Seabuckthorn pulp oil (SBO) and the standard chemotherapy regimen Docetaxel (DTX) against two non-small cell lung cancer (NSCLC) cell lines: A549 and H23. The synergizing effect of an SBO and DTX combination was detected utilizing SRB assay and combination index (CI) approaches. Flow cytometry was carried out using fluorescent probes to measure cell cycle analysis by DNA content and reactive oxygen species (ROS) generation. Further, we demonstrated that the synergistic anticancer activity of SBO merged with DTX was achieved by caspase-independent autophagy and senescence induction. These changes were concomitant with increased generation of ROS production and microtubule-associated protein 1 light chain 3 (LC3) protein expression, G1-phase arrest, and enhanced senescence-associated β-galactosidase staining activity. Our data also demonstrated that SBO or DTX treatment groups solely upregulated the phosphorylation of ERK, which coincided with the induction of autophagy vacuoles and was functionally associated with ROS activation. Moreover, endogenous LC3 puncta staining was performed and monitored by confocal microscopy. Overall, these results suggest new mechanisms for the antitumor activity of SBO co-treated with DTX through triggering autophagic cell death and senescence against cancer cells as a result of sustained ERK phosphorylation and intracellular ROS production in NSCLC. In addition, we also highlight SBO as an alternative therapeutic option or adjunct therapeutic strategy in combination with chemotherapeutic agents in lung cancer therapy management.
Collapse
|
14
|
Li H, Liu L, Chen HY, Yan X, Li RL, Lan J, Xue KY, Li X, Zhuo CL, Lin L, Li LY, Wu Z, Zhang D, Wang XM, Huang WJ, Wang Y, Jiang W, Zhou L. Mogrol suppresses lung cancer cell growth by activating AMPK-dependent autophagic death and inducing p53-dependent cell cycle arrest and apoptosis. Toxicol Appl Pharmacol 2022; 444:116037. [PMID: 35489526 DOI: 10.1016/j.taap.2022.116037] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/23/2022] [Accepted: 04/22/2022] [Indexed: 02/08/2023]
Abstract
Lung carcinoma is the leading cause of cancer-related death worldwide. Chemotherapy remains the cornerstone of lung cancer treatment. Unfortunately, most types of cancer will develop resistance to chemotherapies over the time. One of the efforts to prevent the chemotherapy resistance is to find alternative chemotherapy drugs. Mogrol has been found to have antitumor activity. However, little is known about the pharmacological mechanisms underlying the suppression of mogrol on lung cancers. In this study, we observed that mogrol exposure significantly reduced the tumor volume and weight in tumor-bearing nude mice without obvious effect on body weight and cardiac function. Mogrol also significantly inhibited the proliferation and migration of lung cancer cells, including non-small-cell lung carcinoma cells, A549, H1299, H1975 and SK-MES-1 cells, with no obvious effect on control human bronchial epithelial cells (HBE). Further studies revealed that mogrol stirred excessive autophagy and autophagic flux, and finally, autophagic cell death, in lung cancer cells, which could be attenuated by autophagy inhibitors, 3-MA and chloroquine. Furthermore, mogrol significantly activated AMPK to induce autophagy and autophagic cell death, which could be abrogated by Compound C, an AMPK inhibitor. In addition, mogrol induced a significant increase in p53 activity in lung cancer cells, accompanied with cell cycle arrest and apoptosis, which could be weakened by p53 silence. Our results indicated that mogrol effectively suppressed lung cancer cells in vivo and in vitro by inducing the excessive autophagy and autophagic cell death via activating AMPK signaling pathway, as well as cell cycle arrest and apoptosis via activating p53 pathway.
Collapse
Affiliation(s)
- He Li
- School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, PR China; Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Linling Liu
- School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, PR China; Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Hong-Ying Chen
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xin Yan
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Ru-Li Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Jie Lan
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Kun-Yue Xue
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xue Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Cai-Li Zhuo
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Lan Lin
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Ling-Yu Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Zhuang Wu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Die Zhang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Xue-Mei Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Wen-Jing Huang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Yingling Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China
| | - Wei Jiang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, PR China.
| | - Liming Zhou
- School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
15
|
Eshraghi M, Ahmadi M, Afshar S, Lorzadeh S, Adlimoghaddam A, Rezvani Jalal N, West R, Dastghaib S, Igder S, Torshizi SRN, Mahmoodzadeh A, Mokarram P, Madrakian T, Albensi BC, Łos MJ, Ghavami S, Pecic S. Enhancing autophagy in Alzheimer's disease through drug repositioning. Pharmacol Ther 2022; 237:108171. [PMID: 35304223 DOI: 10.1016/j.pharmthera.2022.108171] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is one of the biggest human health threats due to increases in aging of the global population. Unfortunately, drugs for treating AD have been largely ineffective. Interestingly, downregulation of macroautophagy (autophagy) plays an essential role in AD pathogenesis. Therefore, targeting autophagy has drawn considerable attention as a therapeutic approach for the treatment of AD. However, developing new therapeutics is time-consuming and requires huge investments. One of the strategies currently under consideration for many diseases is "drug repositioning" or "drug repurposing". In this comprehensive review, we have provided an overview of the impact of autophagy on AD pathophysiology, reviewed the therapeutics that upregulate autophagy and are currently used in the treatment of other diseases, including cancers, and evaluated their repurposing as a possible treatment option for AD. In addition, we discussed the potential of applying nano-drug delivery to neurodegenerative diseases, such as AD, to overcome the challenge of crossing the blood brain barrier and specifically target molecules/pathways of interest with minimal side effects.
Collapse
Affiliation(s)
- Mehdi Eshraghi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Mazaher Ahmadi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Aida Adlimoghaddam
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada
| | | | - Ryan West
- Department of Chemistry and Biochemistry, California State University, Fullerton, United States of America
| | - Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tayyebeh Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benedict C Albensi
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada; Nova Southeastern Univ. College of Pharmacy, Davie, FL, United States of America; University of Manitoba, College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University, Fullerton, United States of America.
| |
Collapse
|
16
|
Mázala-de-Oliveira T, de Figueiredo CS, de Rezende Corrêa G, da Silva MS, Miranda RL, de Azevedo MA, Cossenza M, Dos Santos AA, Giestal-de-Araujo E. Ouabain-Na +/K +-ATPase Signaling Regulates Retinal Neuroinflammation and ROS Production Preventing Neuronal Death by an Autophagy-Dependent Mechanism Following Optic Nerve Axotomy In Vitro. Neurochem Res 2022; 47:723-738. [PMID: 34783975 DOI: 10.1007/s11064-021-03481-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/25/2022]
Abstract
Ouabain is a classic Na+K+ATPase ligand and it has been described to have neuroprotective effects on neurons and glial cells at nanomolar concentrations. In the present work, the neuroprotective and immunomodulatory potential of ouabain was evaluated in neonatal rat retinal cells using an optic nerve axotomy model in vitro. After axotomy, cultured retinal cells were treated with ouabain (3 nM) at different periods. The levels of important inflammatory receptors in the retina such as TNFR1/2, TLR4, and CD14 were analyzed. We observed that TNFR1, TLR4, and CD14 were decreased in all tested periods (15 min, 45 min, 24 h, and 48 h). On the other hand, TNFR2 was increased after 24 h, suggesting an anti-inflammatory potential for ouabain. Moreover, we showed that ouabain also decreased Iba-1 (microglial marker) density. Subsequently, analyses of retrograde labeling of retinal ganglion cells (RGC) were performed after 48 h and showed that ouabain-induced RGC survival depends on autophagy. Using an autophagy inhibitor (3-methyladenine), we observed a complete blockage of the ouabain effect. Western blot analyses showed that ouabain increases the levels of autophagy proteins (LC3 and Beclin-1) coupled to p-CREB transcription factor and leads to autophagosome formation. Additionally, we found that the ratio of cleaved/pro-caspase-3 did not change after ouabain treatment; however, p-JNK density was enhanced. Also, ouabain decreased reactive oxygen species production immediately after axotomy. Taken together, our results suggest that ouabain controls neuroinflammation in the retina following optic nerve axotomy and promotes RGC neuroprotection through activation of the autophagy pathway.
Collapse
Affiliation(s)
- Thalita Mázala-de-Oliveira
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil
- Souza Marques School of Medicine, Souza Marques Technical-Educational Foundation, Rio de Janeiro, 21310-310, Brazil
| | - Camila Saggioro de Figueiredo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil
| | - Gustavo de Rezende Corrêa
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil
- Souza Marques School of Medicine, Souza Marques Technical-Educational Foundation, Rio de Janeiro, 21310-310, Brazil
| | - Mayra Santos da Silva
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil
- Souza Marques School of Medicine, Souza Marques Technical-Educational Foundation, Rio de Janeiro, 21310-310, Brazil
| | - Renan Lyra Miranda
- Department of Physiology and Pharmacology and Program of Neurosciences, Laboratory of Neurochemical I`nteractions & Laboratory of Molecular Pharmacology, Biomedical Institute, Federal Fluminense University, Niterói, 24020-141, Brazil
| | - Mariana Almeida de Azevedo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil
| | - Marcelo Cossenza
- Department of Physiology and Pharmacology and Program of Neurosciences, Laboratory of Neurochemical I`nteractions & Laboratory of Molecular Pharmacology, Biomedical Institute, Federal Fluminense University, Niterói, 24020-141, Brazil
| | - Aline Araujo Dos Santos
- Department of Physiology and Pharmacology and Program of Neurosciences, Laboratory of Neurochemical I`nteractions & Laboratory of Molecular Pharmacology, Biomedical Institute, Federal Fluminense University, Niterói, 24020-141, Brazil
| | - Elizabeth Giestal-de-Araujo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil.
- National Institute of Science and Technology on Neuroimmunomodulation - INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, 21040-360, Brazil.
| |
Collapse
|
17
|
Kritschil R, Scott M, Sowa G, Vo N. Role of autophagy in intervertebral disc degeneration. J Cell Physiol 2022; 237:1266-1284. [PMID: 34787318 PMCID: PMC8866220 DOI: 10.1002/jcp.30631] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/13/2021] [Accepted: 11/01/2021] [Indexed: 02/03/2023]
Abstract
Intervertebral disc degeneration (IDD) is a leading contributor to low back pain. The intervertebral disc (IVD) is composed of three tissue types: the central gelatinous nucleus pulposus (NP) tissue, the surrounding annulus fibrosus (AF) tissue, and the inferior and superior cartilage endplates. The IVD microenvironment is hypoxic, acidic, hyperosmotic, and low in nutrients because it is mostly avascular. The cellular processes that underlie IDD initiation and progression are still poorly understood. Specifically, a lack of understanding regarding NP cell metabolism and physiology hinders the development of effective therapeutics to treat IDD patients. Autophagy is a vital intracellular degradation process that removes damaged organelles, misfolded proteins, and intracellular pathogens and recycles the degraded components for cellular energy and function. NP cells have adapted to survive within their harsh tissue microenvironment using processes that are largely unknown, and we postulate autophagy is one of these undiscovered mechanisms. In this review, we describe unique features of the IVD tissue, review how physiological stressors impact autophagy in NP cells in vitro, survey the current understanding of autophagy regulation in the IVD, and assess the relationship between autophagy and IDD. Published studies confirm autophagy markers are present in IVD tissue, and IVD cells can regulate autophagy in response to cellular stressors in vitro. However, data are still lacking to determine the exact mechanisms regulating autophagy in IVD cells. More in-depth research is needed to establish whether autophagy is necessary to maintain IVD cell health and validate autophagy as a relevant therapeutic target for treating IDD.
Collapse
Affiliation(s)
- Rebecca Kritschil
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Melanie Scott
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA,Pittsburgh Trauma Research Center, Pittsburgh, PA
| | - Gwendolyn Sowa
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA,Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
| | - Nam Vo
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
18
|
SARS-CoV-2 spike promotes inflammation and apoptosis through autophagy by ROS-suppressed PI3K/AKT/mTOR signaling. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166260. [PMID: 34461258 PMCID: PMC8390448 DOI: 10.1016/j.bbadis.2021.166260] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022]
Abstract
Background Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection-induced inflammatory responses are largely responsible for the death of novel coronavirus disease 2019 (COVID-19) patients. However, the mechanism by which SARS-CoV-2 triggers inflammatory responses remains unclear. Here, we aimed to explore the regulatory role of SARS-CoV-2 spike protein in infected cells and attempted to elucidate the molecular mechanism of SARS-CoV-2-induced inflammation. Methods SARS-CoV-2 spike pseudovirions (SCV-2-S) were generated using the spike-expressing virus packaging system. Western blot, mCherry-GFP-LC3 labeling, immunofluorescence, and RNA-seq were performed to examine the regulatory mechanism of SCV-2-S in autophagic response. The effects of SCV-2-S on apoptosis were evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), Western blot, and flow cytometry analysis. Enzyme-linked immunosorbent assay (ELISA) was carried out to examine the mechanism of SCV-2-S in inflammatory responses. Results Angiotensin-converting enzyme 2 (ACE2)-mediated SCV-2-S infection induced autophagy and apoptosis in human bronchial epithelial and microvascular endothelial cells. Mechanistically, SCV-2-S inhibited the PI3K/AKT/mTOR pathway by upregulating intracellular reactive oxygen species (ROS) levels, thus promoting the autophagic response. Ultimately, SCV-2-S-induced autophagy triggered inflammatory responses and apoptosis in infected cells. These findings not only improve our understanding of the mechanism underlying SARS-CoV-2 infection-induced pathogenic inflammation but also have important implications for developing anti-inflammatory therapies, such as ROS and autophagy inhibitors, for COVID-19 patients.
Collapse
|
19
|
Chen X, Chen X, Huang Y, Lin J, Wu Y, Chen Y. TCP1 increases drug resistance in acute myeloid leukemia by suppressing autophagy via activating AKT/mTOR signaling. Cell Death Dis 2021; 12:1058. [PMID: 34750375 PMCID: PMC8575913 DOI: 10.1038/s41419-021-04336-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 09/29/2021] [Accepted: 10/18/2021] [Indexed: 12/17/2022]
Abstract
T-complex protein 1 (TCP1) is one of the subunits of chaperonin-containing T complex (CCT), which is involved in protein folding, cell proliferation, apoptosis, cell cycle regulation, and drug resistance. Investigations have demonstrated that TCP1 is a factor being responsible for drug resistance in breast and ovarian cancer. However, the TCP1 role in acute myeloid leukemia (AML) remains elusive. In the present study, we discovered that the TCP1 expression was elevated in AML patients and high TCP1 expression was associated with low complete response rate along with poor overall survival. TCP1 showed higher expression in the adriamycin-resistant leukemia cell line HL60/A and K562/A, comparing to their respective parent cells HL60 and K562 cells. TCP1 inhibition suppressed drug resistance in HL60/A and K562/A cells, whereas TCP1 overexpression in HL60 cells incremented drug resistance, both in vitro and in vivo. Mechanistic investigations revealed that TCP1 inhibited autophagy and adriamycin-induced cell apoptosis, and TCP1-mediated autophagy inhibition conferred resistance to adriamycin-induced cell apoptosis. Furthermore, TCP1 interacted with AKT and mTOR to activate AKT/mTOR signaling, which negatively regulates apoptosis and autophagy. Pharmacological inhibition of AKT/mTOR signal particularly activated autophagy and resensitized TCP1-overexpressing HL60 cells to adriamycin. These findings identify a novel role of TCP1 regarding drug resistance in AML, which advise a new strategy for overcoming drug resistance in AML through targeting TCP1/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiaofang Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.,Department of Infectious Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Xianling Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yiping Huang
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Jia Lin
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yong Wu
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| | - Yuanzhong Chen
- Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
20
|
Mao C, Xu X, Ding Y, Xu N. Optimization of BCG Therapy Targeting Neutrophil Extracellular Traps, Autophagy, and miRNAs in Bladder Cancer: Implications for Personalized Medicine. Front Med (Lausanne) 2021; 8:735590. [PMID: 34660642 PMCID: PMC8514698 DOI: 10.3389/fmed.2021.735590] [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: 07/02/2021] [Accepted: 08/05/2021] [Indexed: 01/07/2023] Open
Abstract
Bladder cancer (BC) is the ninth most common cancer and the thirteenth most common cause of mortality worldwide. Bacillus Calmette Guerin (BCG) instillation is a common treatment option for BC. BCG therapy is associated with the less adversary effects, compared to chemotherapy, radiotherapy, and other conventional treatments. BCG could inhibit the progression and recurrence of BC by triggering apoptosis pathways, arrest cell cycle, autophagy, and neutrophil extracellular traps (NETs) formation. However, BCG therapy is not efficient for metastatic cancer. NETs and autophagy were induced by BCG and help to suppress the growth of tumor cells especially in the primary stages of BC. Activated neutrophils can stimulate autophagy pathway and release NETs in the presence of microbial pathogenesis, inflammatory agents, and tumor cells. Autophagy can also regulate NETs formation and induce production of reactive oxygen species (ROS) and NETs. Moreover, miRNAs are important regulator of gene expression. These small non-coding RNAs are also considered as an essential factor to control the levels of tumor development. However, the interaction between BCG and miRNAs has not been well-understood yet. Therefore, the present study discusses the roles of miRNAs in regulations of autophagy and NETs formation in BCG therapy in the treatment of BC. The roles of autophagy and NETs formation in BC treatment and efficiency of BCG are also discussed.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of Medical Oncology Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xin Xu
- Department of Medical Oncology Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yongfeng Ding
- Department of Medical Oncology Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Nong Xu
- Department of Medical Oncology Cancer Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
21
|
Lan CN, Cai WJ, Shi J, Yi ZJ. MAPK inhibitors protect against early‑stage osteoarthritis by activating autophagy. Mol Med Rep 2021; 24:829. [PMID: 34590154 PMCID: PMC8503737 DOI: 10.3892/mmr.2021.12469] [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: 12/04/2020] [Accepted: 09/01/2021] [Indexed: 12/26/2022] Open
Abstract
Osteoarthritis (OA) is a chronic, age-related osteoarthropathy that causes a considerable decline in quality of life, as well as economic losses due to its high incidence and poor prognosis. Mitogen-activated protein kinases (MAPKs) regulate multiple cellular processes, including proliferation, differentiation and apoptosis, in certain diseases, such as cancer, diabetes and Alzheimer's disease. The present study aimed to investigate the regulatory role of the MAPK signaling pathway in early-stage OA. A rabbit model of early-stage OA was induced by treatment with the enzyme papain. U0126 [an extracellular signal-regulated kinase (ERK) inhibitor], SP600125 [a Jun NH2-terminal kinase (JNK) inhibitor] and SB203580 (a p38 inhibitor) were administered to the rabbits via intra-articular injection. The severity of OA was assessed by histological examination using H&E, toluidine blue and safranin-O/fast green staining, as well by analyzing the glycosaminoglycan (GAG) content and determining the OA Research Society International (OARSI) score. Western blotting was used to detect the protein expression levels of matrix metalloproteinase-3 (MMP3), ERK, phosphorylated (p)-ERK, p38, p-p38, JNK, p-JNK, Beclin1, UNC-51-like kinase 1 (ULK1) and microtubule-associated protein 1 light chain 3 (LC3)II/I. U0126, SP600125 or SB203580 treatment significantly decreased the OARSI scores and significantly increased the GAG levels in the cartilaginous tissues of OA model rabbits. These results indicated that the MAPK inhibitors reduced the severity of OA-induced injury at the early stage. Western blotting results demonstrated that MAPK inhibition significantly decreased the protein expression levels of MMP3 in OA cartilage. The protective effect of MAPK inhibitors in OA was mediated via the activation of autophagy, as demonstrated by the increased protein expression levels of LC3II/I, ULK1 and Beclin1. Overall, the data indicated that MAPK inhibitors may exert a protective effect against OA by restoring compromised autophagy. Furthermore, the present study suggested that MAPK inhibitors may represent a potential pharmacological strategy for treating OA in the future.
Collapse
Affiliation(s)
- Chun-Na Lan
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Wei-Jun Cai
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jie Shi
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhong-Jie Yi
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
22
|
Yang DL, Zhang YJ, He LJ, Hu CS, Gao LX, Huang JH, Tang Y, Luo J, Tang DY, Chen ZZ. Demethylzeylasteral (T-96) initiates extrinsic apoptosis against prostate cancer cells by inducing ROS-mediated ER stress and suppressing autophagic flux. Biol Res 2021; 54:27. [PMID: 34488902 PMCID: PMC8420005 DOI: 10.1186/s40659-021-00350-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/22/2021] [Indexed: 02/05/2023] Open
Abstract
Background Demethylzeylasteral (T-96) is a pharmacologically active triterpenoid monomer extracted from Tripterygium wilfordii Hook F (TWHF) that has been reported to exhibit anti-neoplastic effects against several types of cancer cells. However, the potential anti-tumour effects of T-96 against human Prostate cancer (CaP) cells and the possible underlying mechanisms have not been well studied. Results In the current study, T-96 exerted significant cytotoxicity to CaP cells in vitro and induced cell cycle arrest at S-phase in a dose-dependent manner. Mechanistically, T-96 promoted the initiation of autophagy but inhibited autophagic flux by inducing ROS-mediated endoplasmic reticulum (ER) stress which subsequently activated the extrinsic apoptosis pathway in CaP cells. These findings implied that T-96-induced ER stress activated the caspase-dependent apoptosis pathway to inhibit proliferation of CaP cells. Moreover, we observed that T-96 enhances the sensitivity of CaP cells to the chemotherapeutic drug, cisplatin. Conclusions Taken together, our data demonstrated that T-96 is a novel modulator of ER stress and autophagy, and has potential therapeutic applications against CaP in the clinic. Supplementary Information The online version contains supplementary material available at 10.1186/s40659-021-00350-6.
Collapse
Affiliation(s)
- Dong-Lin Yang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China. .,College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, 400715, China.
| | - Ya-Jun Zhang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China
| | - Liu-Jun He
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China
| | - Chun-Sheng Hu
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China
| | - Li-Xia Gao
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China
| | - Jiu-Hong Huang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China.,College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, 400715, China
| | - Yan Tang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China
| | - Jie Luo
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China
| | - Dian-Yong Tang
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China.
| | - Zhong-Zhu Chen
- College of Pharmacy, National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, Chongqing University of Arts and Sciences, Chongqing, 402160, China.
| |
Collapse
|
23
|
Chen SJ, Hseu YC, Gowrisankar YV, Chung YT, Zhang YZ, Way TD, Yang HL. The anti-melanogenic effects of 3-O-ethyl ascorbic acid via Nrf2-mediated α-MSH inhibition in UVA-irradiated keratinocytes and autophagy induction in melanocytes. Free Radic Biol Med 2021; 173:151-169. [PMID: 34314818 DOI: 10.1016/j.freeradbiomed.2021.07.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/07/2021] [Accepted: 07/23/2021] [Indexed: 02/07/2023]
Abstract
3-O-ethyl ascorbic acid (EAA) is an ether-derivative of ascorbic acid, known to inhibit tyrosinase activity, and is widely used in skincare formulations. Nevertheless, the molecular mechanisms underlying the EAA's effects are poorly understood. Here, the anti-melanogenic activity of EAA was demonstrated through Nrf2-mediated α-MSH inhibition in UVA-irradiated keratinocytes (HaCaT) and autophagy induction and inhibition of α-MSH-stimulated melanogenesis in melanocytes (B16F10). EAA pretreatment increased the HaCaT cell viability but suppressed ROS-mediated p53/POMC/α-MSH pathways in UVA-irradiated cells. Further, the conditioned medium from EAA-pretreated and UVA-irradiated HaCaT cells suppressed the MITF-CREB-tyrosinase pathways leading to the inhibition of melanin synthesis in B16F10 cells. EAA treatment increased nuclear Nrf2 translocation via the p38, PKC, and ROS pathways leading to HO-1, γ-GCLC, and NQO-1 antioxidant expression in HaCaT cells. However, Nrf2 silencing reduced the EAA-mediated anti-melanogenic activity, evidenced by impaired antioxidant gene expression and uncontrolled ROS (H202) generation following UVA irradiation. In B16F10 cells, EAA-induced autophagy was shown by enhanced LC3-II levels, AVO formation, Beclin-1 upregulation, and activation of p62/SQSTM1. Further, EAA-induced anti-melanogenic activity was substantially decreased in autophagy inhibitor (3-MA) pretreated or LC3 knockdown B16F10 cells. Notably, transmission electron microscopy data showed increased melanosome-engulfing autophagosomes in EAA-treated B16F10 cells. Moreover, EAA also down-regulated MC1R, TRP-1/-2, tyrosinase expressions, and melanin synthesis by suppressing the cAMP-CREB-mediated MITF expression in B16F10 cells stimulated with α-MSH. In vivo studies on the zebrafish model further confirmed that EAA inhibited tyrosinase expression/activity and endogenous pigmentation. In conclusion, 3-O-ethyl ascorbic acid is an effective skin-whitening agent and could be used as a topical agent for cosmetic purposes.
Collapse
Affiliation(s)
- Siang-Jyun Chen
- Institute of Nutrition, College of Health Care, China Medical University, Taichung, 406040, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung, 406040, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, 413005, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, 406040, Taiwan; Research Center of Chinese Herbal Medicine, China Medical University, Taichung, 406040, Taiwan.
| | | | - Yi-Ting Chung
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung, 406040, Taiwan
| | - Yan-Zhen Zhang
- Department of Life Sciences, China Medical University, Taichung, 406040, Taiwan
| | - Tzong-Der Way
- Department of Life Sciences, China Medical University, Taichung, 406040, Taiwan
| | - Hsin-Ling Yang
- Institute of Nutrition, College of Health Care, China Medical University, Taichung, 406040, Taiwan.
| |
Collapse
|
24
|
Ristic B, Harhaji-Trajkovic L, Bosnjak M, Dakic I, Mijatovic S, Trajkovic V. Modulation of Cancer Cell Autophagic Responses by Graphene-Based Nanomaterials: Molecular Mechanisms and Therapeutic Implications. Cancers (Basel) 2021; 13:cancers13164145. [PMID: 34439299 PMCID: PMC8392723 DOI: 10.3390/cancers13164145] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Graphene-based nanomaterials (GNM) are one-to-several carbon atom-thick flakes of graphite with at least one lateral dimension <100 nm. The unique electronic structure, high surface-to-volume ratio, and relatively low toxicity make GNM potentially useful in cancer treatment. GNM such as graphene, graphene oxide, graphene quantum dots, and graphene nanofibers are able to induce autophagy in cancer cells. During autophagy the cell digests its own components in organelles called lysosomes, which can either kill cancer cells or promote their survival, as well as influence the immune response against the tumor. However, a deeper understanding of GNM-autophagy interaction at the mechanistic and functional level is needed before these findings could be exploited to increase GNM effectiveness as cancer therapeutics and drug delivery systems. In this review, we analyze molecular mechanisms of GNM-mediated autophagy modulation and its possible implications for the use of GNM in cancer therapy. Abstract Graphene-based nanomaterials (GNM) are plausible candidates for cancer therapeutics and drug delivery systems. Pure graphene and graphene oxide nanoparticles, as well as graphene quantum dots and graphene nanofibers, were all able to trigger autophagy in cancer cells through both transcriptional and post-transcriptional mechanisms involving oxidative/endoplasmic reticulum stress, AMP-activated protein kinase, mechanistic target of rapamycin, mitogen-activated protein kinase, and Toll-like receptor signaling. This was often coupled with lysosomal dysfunction and subsequent blockade of autophagic flux, which additionally increased the accumulation of autophagy mediators that participated in apoptotic, necrotic, or necroptotic death of cancer cells and influenced the immune response against the tumor. In this review, we analyze molecular mechanisms and structure–activity relationships of GNM-mediated autophagy modulation, its consequences for cancer cell survival/death and anti-tumor immune response, and the possible implications for the use of GNM in cancer therapy.
Collapse
Affiliation(s)
- Biljana Ristic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
| | - Ljubica Harhaji-Trajkovic
- Department of Neurophysiology, Institute for Biological Research “Sinisa Stankovic”, National Institute of Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia;
| | - Mihajlo Bosnjak
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Ivana Dakic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
| | - Srdjan Mijatovic
- Clinic for Emergency Surgery, Clinical Centre of Serbia, 11000 Belgrade, Serbia;
| | - Vladimir Trajkovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (B.R.); (I.D.)
- Correspondence:
| |
Collapse
|
25
|
Grisan F, Iannucci LF, Surdo NC, Gerbino A, Zanin S, Di Benedetto G, Pozzan T, Lefkimmiatis K. PKA compartmentalization links cAMP signaling and autophagy. Cell Death Differ 2021; 28:2436-2449. [PMID: 33742135 PMCID: PMC8328970 DOI: 10.1038/s41418-021-00761-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 01/31/2023] Open
Abstract
Autophagy is a highly regulated degradative process crucial for maintaining cell homeostasis. This important catabolic mechanism can be nonspecific, but usually occurs with fine spatial selectivity (compartmentalization), engaging only specific subcellular sites. While the molecular machines driving autophagy are well understood, the involvement of localized signaling events in this process is not well defined. Among the pathways that regulate autophagy, the cyclic AMP (cAMP)/protein kinase A (PKA) cascade can be compartmentalized in distinct functional units called microdomains. However, while it is well established that, depending on the cell type, cAMP can inhibit or promote autophagy, the role of cAMP/PKA microdomains has not been tested. Here we show not only that the effects on autophagy of the same cAMP elevation differ in different cell types, but that they depend on a highly complex sub-compartmentalization of the signaling cascade. We show in addition that, in HT-29 cells, in which autophagy is modulated by cAMP rising treatments, PKA activity is strictly regulated in space and time by phosphatases, which largely prevent the phosphorylation of soluble substrates, while membrane-bound targets are less sensitive to the action of these enzymes. Interestingly, we also found that the subcellular distribution of PKA type-II regulatory PKA subunits hinders the effect of PKA on autophagy, while displacement of type-I regulatory PKA subunits has no effect. Our data demonstrate that local PKA activity can occur independently of local cAMP concentrations and provide strong evidence for a link between localized PKA signaling events and autophagy.
Collapse
Affiliation(s)
- Francesca Grisan
- Foundation for Advanced Biomedical Research, Veneto Institute of Molecular Medicine, Padua, Italy
- Department of Biology, University of Padua, Padua, Italy
| | - Liliana F Iannucci
- Foundation for Advanced Biomedical Research, Veneto Institute of Molecular Medicine, Padua, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Nicoletta C Surdo
- Foundation for Advanced Biomedical Research, Veneto Institute of Molecular Medicine, Padua, Italy
- Department of Biology, University of Padua, Padua, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Bari, Italy
| | - Sofia Zanin
- Foundation for Advanced Biomedical Research, Veneto Institute of Molecular Medicine, Padua, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Giulietta Di Benedetto
- Foundation for Advanced Biomedical Research, Veneto Institute of Molecular Medicine, Padua, Italy
- Neuroscience Institute, National Research Council, Padua, Italy
| | - Tullio Pozzan
- Neuroscience Institute, National Research Council, Padua, Italy
| | - Konstantinos Lefkimmiatis
- Foundation for Advanced Biomedical Research, Veneto Institute of Molecular Medicine, Padua, Italy.
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
26
|
Kocak M, Ezazi Erdi S, Jorba G, Maestro I, Farrés J, Kirkin V, Martinez A, Pless O. Targeting autophagy in disease: established and new strategies. Autophagy 2021; 18:473-495. [PMID: 34241570 PMCID: PMC9037468 DOI: 10.1080/15548627.2021.1936359] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionarily conserved pathway responsible for clearing cytosolic aggregated proteins, damaged organelles or invading microorganisms. Dysfunctional autophagy leads to pathological accumulation of the cargo, which has been linked to a range of human diseases, including neurodegenerative diseases, infectious and autoimmune diseases and various forms of cancer. Cumulative work in animal models, application of genetic tools and pharmacologically active compounds, has suggested the potential therapeutic value of autophagy modulation in disease, as diverse as Huntington, Salmonella infection, or pancreatic cancer. Autophagy activation versus inhibition strategies are being explored, while the role of autophagy in pathophysiology is being studied in parallel. However, the progress of preclinical and clinical development of autophagy modulators has been greatly hampered by the paucity of selective pharmacological agents and biomarkers to dissect their precise impact on various forms of autophagy and cellular responses. Here, we summarize established and new strategies in autophagy-related drug discovery and indicate a path toward establishing a more efficient discovery of autophagy-selective pharmacological agents. With this knowledge at hand, modern concepts for therapeutic exploitation of autophagy might become more plausible. Abbreviations: ALS: amyotrophic lateral sclerosis; AMPK: AMP-activated protein kinase; ATG: autophagy-related gene; AUTAC: autophagy-targeting chimera; CNS: central nervous system; CQ: chloroquine; GABARAP: gamma-aminobutyric acid type A receptor-associated protein; HCQ: hydroxychloroquine; LYTAC: lysosome targeting chimera; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; NDD: neurodegenerative disease; PDAC: pancreatic ductal adenocarcinoma; PE: phosphatidylethanolamine; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol 3-phosphate; PROTAC: proteolysis-targeting chimera; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2; SQSTM1/p62: sequestosome 1; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Muhammed Kocak
- Cancer Research UK, Cancer Therapeutics Unit, the Institute of Cancer Research London, Sutton, UK
| | | | | | - Inés Maestro
- Centro De Investigaciones Biologicas "Margarita Salas"-CSIC, Madrid, Spain
| | | | - Vladimir Kirkin
- Cancer Research UK, Cancer Therapeutics Unit, the Institute of Cancer Research London, Sutton, UK
| | - Ana Martinez
- Centro De Investigaciones Biologicas "Margarita Salas"-CSIC, Madrid, Spain.,Centro De Investigación Biomédica En Red En Enfermedades Neurodegenerativas (CIBERNED), Instituto De Salud Carlos III, Madrid, Spain
| | - Ole Pless
- Fraunhofer ITMP ScreeningPort, Hamburg, Germany
| |
Collapse
|
27
|
Zia A, Pourbagher-Shahri AM, Farkhondeh T, Samarghandian S. Molecular and cellular pathways contributing to brain aging. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2021; 17:6. [PMID: 34118939 PMCID: PMC8199306 DOI: 10.1186/s12993-021-00179-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
Aging is the leading risk factor for several age-associated diseases such as neurodegenerative diseases. Understanding the biology of aging mechanisms is essential to the pursuit of brain health. In this regard, brain aging is defined by a gradual decrease in neurophysiological functions, impaired adaptive neuroplasticity, dysregulation of neuronal Ca2+ homeostasis, neuroinflammation, and oxidatively modified molecules and organelles. Numerous pathways lead to brain aging, including increased oxidative stress, inflammation, disturbances in energy metabolism such as deregulated autophagy, mitochondrial dysfunction, and IGF-1, mTOR, ROS, AMPK, SIRTs, and p53 as central modulators of the metabolic control, connecting aging to the pathways, which lead to neurodegenerative disorders. Also, calorie restriction (CR), physical exercise, and mental activities can extend lifespan and increase nervous system resistance to age-associated neurodegenerative diseases. The neuroprotective effect of CR involves increased protection against ROS generation, maintenance of cellular Ca2+ homeostasis, and inhibition of apoptosis. The recent evidence about the modem molecular and cellular methods in neurobiology to brain aging is exhibiting a significant potential in brain cells for adaptation to aging and resistance to neurodegenerative disorders.
Collapse
Affiliation(s)
- Aliabbas Zia
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ali Mohammad Pourbagher-Shahri
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), 9717853577 Birjand, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
28
|
Subbiah R, Tiwari RR. The herbicide paraquat-induced molecular mechanisms in the development of acute lung injury and lung fibrosis. Crit Rev Toxicol 2021; 51:36-64. [PMID: 33528289 DOI: 10.1080/10408444.2020.1864721] [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] [Indexed: 02/08/2023]
Abstract
The herbicide paraquat (PQ; 1,1'-dimethyl-4,4'-bipyridylium dichloride) is a highly toxic organic heterocyclic herbicide that has been widely used in agricultural settings. Since its commercial introduction in the early 1960s, numerous cases of fatal PQ poisonings attributed to accidental and/or intentional ingestion of PQ concentrated formulations have been reported. The clinical manifestations of the respiratory system during the acute phase of PQ poisoning mainly include acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), followed by pulmonary fibrosis in a later phase. The focus of this review is to summarize the most recent publications related to PQ-induced lung toxicity as well as the underlying molecular mechanisms for PQ-mediated pathologic processes. Growing sets of data from in vitro and in vivo models have demonstrated the involvement of the PQ in regulating lung oxidative stress, inflammatory response, epigenetics, apoptosis, autophagy, and the progression of lung fibrosis. The article also summarizes novel therapeutic avenues based on a literature review, which can be explored as potential means to combat PQ-induced lung toxicity. Finally, we also presented clinical studies on the association of PQ exposure with the incidence of lung injury and pulmonary fibrosis.
Collapse
Affiliation(s)
- Rajasekaran Subbiah
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Rajnarayan R Tiwari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| |
Collapse
|
29
|
Pal A, Verma P, Paul S, Majumder I, Kundu R. Two species of Ulva inhibits the progression of cervical cancer cells SiHa by means of autophagic cell death induction. 3 Biotech 2021; 11:52. [PMID: 33489671 PMCID: PMC7801572 DOI: 10.1007/s13205-020-02576-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022] Open
Abstract
Edible green algal seaweeds, namely Ulva intestinalis and Ulva lactuca constitute a significant repository of popular herbal medicines in the Traditional Chinese Medicinal system. The present study aimed to assess the anticancer potential of these algal members and its mode of action in cervical cancer cells SiHa. The algal samples primarily extracted in methanol was fractionated into hexane, chloroform, and aqueous algal fractions, which inhibited the proliferation of SiHa cells in a dose-dependent manner, with the algal chloroform fractions harbouring the lowest IC50 dose of 141.38 µg/ml in U. intestinalis and 445.278 µg/ml in U. lactuca. These algal chloroform fractions when studied for their in-depth mode of action, were found to damage and pulverise the nuclei, resulting in a concomitant increase in subG0-phase of SiHa cells, studied by flow cytometry. The algal treatment also caused an increase in the number of acidic vesicles and enhanced the expression of LC3BII, p62 and atg12 proteins, which together pointed out autophagy as the induced mode of cell death. Upregulated Bax and p53 expression along with decreased Bcl2 expression also correlated to autophagic cell death. Decreased expression of E6 viral oncogene was noted as a significant response to algal fractions. Lastly, these potent algal fractions when characterised pharmacologically through GC-MS analysis were found to be rich in unsaturated fatty acids, majorly palmitic acid. Hence, this study concludes that the two species of Ulva successfully decreased the proliferation of SiHa cervical cancer cells through autophagy, hinting at palmitic acid being the major responsible bioactive compound in both.
Collapse
Affiliation(s)
- Asmita Pal
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
| | - Preeti Verma
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
| | - Subhabrata Paul
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
- Present Address: Bioprospecting Laboratory, School of Biotechnology, Presidency University, Kolkata, West Bengal India
| | - Indira Majumder
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
| | - Rita Kundu
- Department of Botany, Centre of Advanced Studies, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 West Bengal India
| |
Collapse
|
30
|
Wang Y, Wang X, Wang L, Cheng G, Zhang M, Xing Y, Zhao X, Liu Y, Liu J. Mitophagy Induced by Mitochondrial Function Damage in Chicken Kidney Exposed to Cr(VI). Biol Trace Elem Res 2021; 199:703-711. [PMID: 32440992 DOI: 10.1007/s12011-020-02176-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 04/24/2020] [Indexed: 01/08/2023]
Abstract
Cr(VI) is a heavy metal environmental pollutant and carcinogen. Excessive Cr(VI) exposure injures kidneys. This study aimed to investigate mitophagy induced by mitochondrial function damage in chicken kidney exposed to Cr(VI). To explore the mechanism involved, we randomly divided 40 one-day-old Hy-line Brown cockerels into four groups, with each group exposed to different concentrations of Cr(VI), i.e., 0, 10, 30 and 50 mg kg-1, which were orally administered daily for 45 days. Excessive Cr(VI) increased tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and chemokine (C-X-C motif) ligand 1(CXCL1) expression and decreased Ca2+-adenosine triphosphatase (Ca2+-ATPase), Mg2+-ATPase and Na+/k+-ATPase activities in chicken kidney. Furthermore, Cr(VI) significantly increased reactive oxygen species (ROS) production and induced mitochondrial membrane potential (MMP) collapse and typical autophagosome formation. With the increase of Cr(VI) concentration, the Parkin translocation, value of LC3-II increased and decreased the content of p62/SQSTM1 and the translocase of outer mitochondrial membrane 20 (TOMM20). In summary, our findings explicated that mitochondrial function damage and mitophagy-related indicators were related to Cr(VI) concentration in chicken kidney.
Collapse
Affiliation(s)
- Yue Wang
- College of Veterinary Medicine, Shandong Agricultural University, Taiàn, 271018, Shandong, China
| | - Xiaozhou Wang
- College of Veterinary Medicine, Shandong Agricultural University, Taiàn, 271018, Shandong, China
| | - Lumei Wang
- College of Veterinary Medicine, Shandong Agricultural University, Taiàn, 271018, Shandong, China
| | - Guodong Cheng
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Taiàn, 271018, Shandong, China
| | - Meihua Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Taiàn, 271018, Shandong, China
| | - Yuxiao Xing
- College of Veterinary Medicine, Shandong Agricultural University, Taiàn, 271018, Shandong, China
| | - Xiaona Zhao
- College of Veterinary Medicine, Shandong Agricultural University, Taiàn, 271018, Shandong, China
| | - Yongxia Liu
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Taiàn, 271018, Shandong, China
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Taiàn, 271018, Shandong, China.
| |
Collapse
|
31
|
Jin KT, Tao XH, Fan YB, Wang SB. Crosstalk between oncolytic viruses and autophagy in cancer therapy. Biomed Pharmacother 2020; 134:110932. [PMID: 33370632 DOI: 10.1016/j.biopha.2020.110932] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/15/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Oncolytic viruses have attracted attention as a promising strategy in cancer therapy owing to their ability to selectively infect and kill tumor cells, without affecting healthy cells. They also exert their anti-tumor effects by releasing immunostimulatory molecules from dying cancer cells. Several regulatory mechanisms, such as autophagy, contribute to the anti-tumor properties of oncolytic viruses. Autophagy is a conserved catabolic process in responses to various stresses, such as nutrient deprivation, hypoxia, and infection that produces energy by lysosomal degradation of intracellular contents. Autophagy can support infectivity and replication of the oncolytic virus and enhance their anti-tumor effects via mediating oncolysis, autophagic cell death, and immunogenic cell death. On the other hand, autophagy can reduce the cytotoxicity of oncolytic viruses by providing survival nutrients for tumor cells. In his review, we summarize various types of oncolytic viruses in clinical trials, their mechanism of action, and autophagy machinery. Furthermore, we precisely discuss the interaction between oncolytic viruses and autophagy in cancer therapy and their combinational effects on tumor cells.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang Province, PR China
| | - Xiao-Hua Tao
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, PR China
| | - Yi-Bin Fan
- Department of Dermatology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, PR China.
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, PR China.
| |
Collapse
|
32
|
Fattahi S, Amjadi-Moheb F, Tabaripour R, Ashrafi GH, Akhavan-Niaki H. PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci 2020; 262:118513. [PMID: 33011222 DOI: 10.1016/j.lfs.2020.118513] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
PI3K/AKT/mTOR pathway is one of the most important signaling pathways involved in normal cellular processes. Its aberrant activation modulates autophagy, epithelial-mesenchymal transition, apoptosis, chemoresistance, and metastasis in many human cancers. Emerging evidence demonstrates that some infections as well as epigenetic regulatory mechanisms can control PI3K/AKT/mTOR signaling pathway. In this review, we focused on the role of this pathway in gastric cancer development, prognosis, and metastasis, with an emphasis on epigenetic alterations including DNA methylation, histone modifications, and post-transcriptional modulations through non-coding RNAs fluctuations as well as H. pylori and Epstein-Barr virus infections. Finally, we reviewed different molecular targets and therapeutic agents in clinical trials as a potential strategy for gastric cancer treatment through the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Sadegh Fattahi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; North Research Center, Pasteur Institute, Amol, Iran
| | - Fatemeh Amjadi-Moheb
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Reza Tabaripour
- Department of Cellular and Molecular Biology, Islamic Azad University Babol-Branch, Iran
| | - Gholam Hossein Ashrafi
- Kingston University London, Cancer theme, School of Life Science, Pharmacy and Chemistry, SEC Faculty, Kingston upon Thames, KT12EE, London, UK
| | - Haleh Akhavan-Niaki
- Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
33
|
Yang L, Zhang C, Chen J, Zhang S, Pan G, Xin Y, Lin L, You Z. Shenmai injection suppresses multidrug resistance in MCF-7/ADR cells through the MAPK/NF-κB signalling pathway. PHARMACEUTICAL BIOLOGY 2020; 58:276-285. [PMID: 32251615 PMCID: PMC7170370 DOI: 10.1080/13880209.2020.1742167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Context: Shenmai Injection (SMI) is usually used to treat atherosclerotic coronary heart disease and viral myocarditis in China. However, the effect of SMI on multidrug resistance has not been reported.Objective: To investigate the reversal effect of SMI in adriamycin (ADR) resistant breast cancer cell line (MCF-7/ADR) and explore the related molecular mechanisms.Materials and methods: The effect of SMI (0.25, 0.5, 1 mg/mL) to reverse chemoresistance in MCF-7/ADR cells was elucidated by MTT, HPLC-FLD, DAPI staining, flow cytometric analysis, western blotting. At the same time, in vivo test was conducted to probe into the effect of SMI on reversing ADR resistance, and verapamil (10 μM) was used as a positive control.Results: The results showed that the toxicity of ADR to MCF-7/ADR cells was strengthened significantly after treated with SMI (0.25, 0.5, 1 mg/mL), the IC50 of ADR was decreased 54.4-fold. The intracellular concentrations of ADR were increased 2.2-fold (p < 0.05) and ADR accumulation was enhanced in the nuclei (p < 0.05). SMI could strongly enhance the ADR-induced apoptosis and increase intracellular rhodamine 123 accumulation in MCF-7/ADR cells. Additionally, a combination of ADR and SMI (5 mg/kg) could dramatically reduce the weight and volume of tumour (p < 0.05). Furthermore, the results revealed that SMI might reverse MDR via inhibiting ADR-induced activation of the mitogen-activated protein kinase/nuclear factor (NF)-κB pathway to down-regulated the expression of P-glycoprotein (P-gp).Discussion and conclusions: SMI could potentially be used to treat ADR-resistance. This suggests possibilities for future clinical research.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Animals
- Antineoplastic Combined Chemotherapy Protocols/metabolism
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Down-Regulation/drug effects
- Doxorubicin/metabolism
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Combinations
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Female
- Humans
- MAP Kinase Signaling System/drug effects
- MCF-7 Cells
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NF-kappa B/metabolism
- Rhodamine 123/metabolism
- Signal Transduction/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lin Yang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Chengda Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Jiaoting Chen
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Sheng Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Guixuan Pan
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yanfei Xin
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
- Yanfei Xin
| | - Lin Lin
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Lin Lin Zhejiang Academy of Medical Sciences, 182 Tianmushan Road, Hangzhou310013, China
| | - Zhenqiang You
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- CONTACT Zhenqiang You
| |
Collapse
|
34
|
Cardioprotective Effects of Taurisolo® in Cardiomyoblast H9c2 Cells under High-Glucose and Trimethylamine N-Oxide Treatment via De Novo Sphingolipid Synthesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2961406. [PMID: 33273998 PMCID: PMC7683148 DOI: 10.1155/2020/2961406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/21/2020] [Accepted: 10/24/2020] [Indexed: 12/27/2022]
Abstract
In addition to high plasma glucose, increased levels of trimethylamine N-oxide (TMAO) have been found in obese subjects, where are considered as a novel risk factor for cardiovascular diseases. The present study aimed to investigate the effect of a novel nutraceutical formulation based on grape polyphenols (registered as Taurisolo®) in counteracting TMAO- and high glucose (HG)-induced cytotoxicity in cardiomyoblast H9c2 cells. Cell damage was induced with HG (HG-H9c2) and HG+TMAO (THG-H9c2); both experimental cell models were, thus, incubated for 72 h in the presence or absence of Taurisolo®. It was observed that Taurisolo® significantly increased the cell viability and reduced lactate dehydrogenase and aspartate transaminase release in both HG- and THG-H9c2 cells. Additionally, through its antioxidant activity, Taurisolo® modulated cell proliferation via ERK activation in THG-H9c2. Furthermore, Taurisolo® was able to induce autophagic process via increasing the expression of LC3II, a protein marker involved in formation of autophagosome and ex novo synthesis of sphingomyelin, ceramides, and their metabolites both in HG- and THG-H9c2 cells. Finally, Taurisolo® reduced hypertrophy and induced differentiation of HG-H9C2 cells into cardiomyocyte-like cells. These data suggest that Taurisolo® counteracts the toxicity induced by TMAO and HG concentrations increasing autophagic process and activating de novo sphingolipid synthesis, resulting in a morphological cell remodeling. In conclusion, our results allow speculating that Taurisolo®, combined with energy restriction, may represent a useful nutraceutical approach for prevention of cardiomyopathy in obese subjects.
Collapse
|
35
|
Nakatani K, Matsuo H, Harata Y, Higashitani M, Koyama A, Noura M, Nishinaka-Arai Y, Kamikubo Y, Adachi S. Inhibition of CDK4/6 and autophagy synergistically induces apoptosis in t(8;21) acute myeloid leukemia cells. Int J Hematol 2020; 113:243-253. [PMID: 33068248 DOI: 10.1007/s12185-020-03015-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/10/2020] [Accepted: 09/18/2020] [Indexed: 12/19/2022]
Abstract
The t(8;21) translocation is the most common cytogenetic abnormality in acute myeloid leukemia (AML). Although t(8;21) AML patients have a relatively favorable prognosis, relapse is a frequent occurrence, underscoring the need to develop novel therapeutic approaches. Here, we showed that t(8;21) AML is characterized by frequent mutation and overexpression of CCND2. Analysis of 19 AML cell lines showed that t(8;21) AML cells had lower IC50 values for the selective CDK4/6 inhibitors palbociclib and abemaciclib than non-t(8;21) AML cells. CDK4/6 inhibitors caused cell cycle arrest at G1 phase and impaired cell proliferation in t(8;21) AML cells. CDK4/6 inhibition decreased MAP-ERK and PI3K-AKT-mTOR signaling pathway activity, induced LC3B-I to LC3B-II conversion, and enhanced autophagosome formation, suggesting autophagy induction. Treatment of t(8;21) AML cells with the autophagy inhibitors chloroquine (CQ) or LY294002 in combination with the CDK4/6 inhibitor abemaciclib significantly increased the percentage of apoptotic (Annexin V positive) cells, whereas CQ or LY294002 single treatment had no significant effects. The effectiveness of co-inhibiting CDK4/6 and autophagy was confirmed in primary t(8;21) AML cells. The results suggest that the combination of CDK4/6 and autophagy inhibitors had a synergistic effect on inducing apoptosis, suggesting a novel therapeutic approach for the treatment of t(8;21) AML.
Collapse
Affiliation(s)
- Kana Nakatani
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Hidemasa Matsuo
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Yutarou Harata
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Moe Higashitani
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Asami Koyama
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Mina Noura
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Yoko Nishinaka-Arai
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan.,Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yasuhiko Kamikubo
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan
| | - Souichi Adachi
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyoku, Kyoto, 606-8507, Japan.
| |
Collapse
|
36
|
Soto-Avellaneda A, Morrison BE. Signaling and other functions of lipids in autophagy: a review. Lipids Health Dis 2020; 19:214. [PMID: 32998777 PMCID: PMC7525950 DOI: 10.1186/s12944-020-01389-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 09/23/2020] [Indexed: 12/19/2022] Open
Abstract
The process of autophagy is integral to cellular function. In this process, proteins, organelles, and metabolites are engulfed in a lipid vesicle and trafficked to a lysosome for degradation. Its central role in protein and organelle homeostasis has piqued interest for autophagy dysfunction as a driver of pathology for a number of diseases including cancer, muscular disorders, neurological disorders, and non-alcoholic fatty liver disease. For much of its history, the study of autophagy has centered around proteins, however, due to advances in mass spectrometry and refined methodologies, the role of lipids in this essential cellular process has become more apparent. This review discusses the diverse endogenous lipid compounds shown to mediate autophagy. Downstream lipid signaling pathways are also reviewed in the context of autophagy regulation. Specific focus is placed upon the Mammalian Target of Rapamycin (mTOR) and Peroxisome Proliferator-Activated Receptor (PPAR) signaling pathways as integration hubs for lipid regulation of autophagy.
Collapse
Affiliation(s)
| | - Brad E Morrison
- Biomolecular Sciences Graduate programs, Boise State University, Boise, ID, 83725, USA.
- Department of Biological Sciences, Boise State University, Boise, ID, 83725, USA.
| |
Collapse
|
37
|
Ding H, Ge G, Tseng Y, Ma Y, Zhang J, Liu J. Hepatic autophagy fluctuates during the development of non-alcoholic fatty liver disease. Ann Hepatol 2020; 19:516-522. [PMID: 32553647 DOI: 10.1016/j.aohep.2020.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Autophagy has emerged as a critical regulatory pathway in non-alcoholic fatty liver disease (NAFLD). However, the variability of hepatic autophagy during NAFLD development remains controversial. This study aimed to elucidate the dynamics of hepatic autophagy and its underlying mechanism during NAFLD development both in vivo and in vitro. MATERIALS AND METHODS Autophagy markers were evaluated in the livers of mice fed a high fat diet or a methionine-choline-deficient diet and in HepG2 cells treated with palmitic acid (PA) by western blotting. Intrahepatic and intracellular triacylglycerol levels were assessed using biochemical quantification and lipid staining. Autophagic flux was monitored using an LC3 turnover assay and tandem mRFP-GFP-LC3 fluorescence analysis. RESULTS Hepatic autophagy was enhanced in early stages but blocked at later stages of NAFLD development both in vivo and in vitro. Analysis of autophagic flux revealed that both autophagic synthesis and degradation were initially activated and progressively inhibited afterwards. The activation of mammalian target of rapamycin complex 1 (mTORC1), a central regulator of autophagy, was found to be negatively correlated with autophagic synthesis; moreover, pharmacological inhibition of mTORC1 by rapamycin alleviated hepatic steatosis through recovery of autophagic flux in hepatocytes with prolonged PA treatment. CONCLUSIONS Hepatic autophagy fluctuates during the development of NAFLD in which mTORC1 signalling plays a critical regulatory role, suggesting a therapeutic potential of autophagy modulation by targeting the mTORC1 signalling pathway in NAFLD.
Collapse
Affiliation(s)
- Hao Ding
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Ge Ge
- Department of Dermatology, Air Force Medical Center, Beijing, China; China Medical University, Shenyang, China
| | - Yujen Tseng
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanyun Ma
- Human Phenome Institute, Fudan University, Shanghai, 201203, China; Six-sector Industrial Research Institute, Fudan University, Shanghai, 200433, China; Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Jun Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
38
|
Regulation of Autophagy by Protein Kinase C-ε in Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21124247. [PMID: 32549199 PMCID: PMC7352677 DOI: 10.3390/ijms21124247] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 11/24/2022] Open
Abstract
Protein kinase C-ε (PKCε), an anti-apoptotic protein, plays critical roles in breast cancer development and progression. Although autophagy is an important survival mechanism, it is not known if PKCε regulates autophagy in breast cancer cells. We have shown that silencing of PKCε by siRNA inhibited basal and starvation-induced autophagy in T47D breast cancer cells as determined by the decrease in LC3-II, increase in p62, and decrease in autophagy puncta both in the presence and absence of bafilomycin A1. The mechanistic target of rapamycin (mTOR) associates with Raptor or Rictor to form complex-1 (mTORC1) or complex-2 (mTORC2), respectively. Knockdown of PKCε attenuated an increase in autophagy caused by the depletion of Raptor and Rictor. Overexpression of PKCε in MCF-7 cells caused activation of mTORC1 and an increase in LC3-I, LC3-II, and p62. The mTORC1 inhibitor rapamycin abolished the increase in LC3-I and p62. Knockdown of mTOR and Rictor or starvation enhanced autophagy in PKCε overexpressing cells. While overexpression of PKCε in MCF-7 cells inhibited apoptosis, it induced autophagy in response to tumor necrosis factor-α. However, inhibition of autophagy by Atg5 knockdown restored apoptosis in PKCε-overexpressing cells. Thus, PKCε promotes breast cancer cell survival not only by inhibiting apoptosis but also by inducing autophagy.
Collapse
|
39
|
Sun W, Qian K, Guo K, Chen L, Xiang J, Li D, Wu Y, Ji Q, Sun T, Wang Z. LHPP inhibits cell growth and migration and triggers autophagy in papillary thyroid cancer by regulating the AKT/AMPK/mTOR signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2020; 52:382-389. [PMID: 32227107 DOI: 10.1093/abbs/gmaa015] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/20/2019] [Accepted: 02/24/2020] [Indexed: 12/24/2022] Open
Abstract
In recent decades, the incidence rate of papillary thyroid carcinoma (PTC) has been rapidly increasing. However, the molecular mechanism of the physiological and pathological processes of PTC is still largely unknown. Phospholysine phosphohistidine inorganic pyrophosphate phosphatase (LHPP) is a tumor suppressor and exerts anti-tumor effect in several human cancers, while the role and underlying mechanism of LHPP in PTC remain vague. In this study, we firstly evaluated the roles of LHPP in PTC and explored its underlying mechanism. Using clinical tissue samples, we detected the level of LHPP in PTC tissues and in matched adjacent normal tissues. Lower level of LHPP was found in PTC tissues than in matched adjacent normal tissues. Similar LHPP expression pattern was found in PTC cell lines when compared with that in normal human thyroid follicular epithelial cells. Then, we over-expressed LHPP in three PTC cell lines and results showed that ectopic LHPP expression consistently reduced cell viability, proliferation, and migration and triggered cell autophagy. Furthermore, over-expression of LHPP inhibited the activation of the AKT/mTOR pathway and promoted the AMPK signaling pathway. In addition, the activation of AKT/mTOR and inhibition of AMPK signaling pathways restored the role of ectopic LHPP expression in PTC cell lines, indicating that LHPP exerts its anti-tumor activity through regulating the AKT/AMPK/mTOR pathway. Ultimately, we illustrated that ectopic LHPP expression inhibited PTC tumor growth in vivo. In conclusion, we revealed that LHPP has an anti-tumor effect in PTC and indicated that LHPP might serve as an effective diagnostic and therapeutic target for PTC.
Collapse
Affiliation(s)
- Wenyu Sun
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Kai Qian
- Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Kai Guo
- Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Lili Chen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jun Xiang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Duanshu Li
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yi Wu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Tuanqi Sun
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Zhuoying Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College of Fudan University, Shanghai 200032, China
- Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| |
Collapse
|
40
|
Shi Q, Pei F, Silverman GA, Pak SC, Perlmutter DH, Liu B, Bahar I. Mechanisms of Action of Autophagy Modulators Dissected by Quantitative Systems Pharmacology Analysis. Int J Mol Sci 2020; 21:ijms21082855. [PMID: 32325894 PMCID: PMC7215584 DOI: 10.3390/ijms21082855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy plays an essential role in cell survival/death and functioning. Modulation of autophagy has been recognized as a promising therapeutic strategy against diseases/disorders associated with uncontrolled growth or accumulation of biomolecular aggregates, organelles, or cells including those caused by cancer, aging, neurodegeneration, and liver diseases such as α1-antitrypsin deficiency. Numerous pharmacological agents that enhance or suppress autophagy have been discovered. However, their molecular mechanisms of action are far from clear. Here, we collected a set of 225 autophagy modulators and carried out a comprehensive quantitative systems pharmacology (QSP) analysis of their targets using both existing databases and predictions made by our machine learning algorithm. Autophagy modulators include several highly promiscuous drugs (e.g., artenimol and olanzapine acting as activators, fostamatinib as an inhibitor, or melatonin as a dual-modulator) as well as selected drugs that uniquely target specific proteins (~30% of modulators). They are mediated by three layers of regulation: (i) pathways involving core autophagy-related (ATG) proteins such as mTOR, AKT, and AMPK; (ii) upstream signaling events that regulate the activity of ATG pathways such as calcium-, cAMP-, and MAPK-signaling pathways; and (iii) transcription factors regulating the expression of ATG proteins such as TFEB, TFE3, HIF-1, FoxO, and NF-κB. Our results suggest that PKA serves as a linker, bridging various signal transduction events and autophagy. These new insights contribute to a better assessment of the mechanism of action of autophagy modulators as well as their side effects, development of novel polypharmacological strategies, and identification of drug repurposing opportunities.
Collapse
Affiliation(s)
- Qingya Shi
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Fen Pei
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
| | - Gary A. Silverman
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - Stephen C. Pak
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - David H. Perlmutter
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - Bing Liu
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- Correspondence: (B.L.); (I.B.)
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- Correspondence: (B.L.); (I.B.)
| |
Collapse
|
41
|
Gugliandolo A, Pollastro F, Bramanti P, Mazzon E. Cannabidiol exerts protective effects in an in vitro model of Parkinson's disease activating AKT/mTOR pathway. Fitoterapia 2020; 143:104553. [PMID: 32184097 DOI: 10.1016/j.fitote.2020.104553] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 01/12/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of the nigrostriatal dopaminergic pathway with loss of substantia nigra pars compacta neurons and dopamine depletion. Various natural compounds showed protective actions against PD. In this work, the protective effects of cannabidiol (CBD), obtained from Cannabis sativa, were evaluated in retinoic acid differentiated SH-SY5Y cells exposed to 1-methyl-4-phenylpyridinium (MPP+), an in vitro PD model. In order to evaluate which receptor is involved in CBD actions CB1, CB2 and TRPV1 receptor antagonists were used. CBD counteracted the loss of cell viability caused by MPP+, reducing apoptosis as demonstrated by the reduction of Bax and caspase 3. Moreover, CBD reduced the nuclear levels of PARP-1. The protective effects of CBD seem to be mediated by the activation of ERK and AKT/mTOR pathways. The treatment with AKT1/2 inhibitor and the mTOR inhibitor rapamycin abolished CBD protective effects. The CBD-induced ERK activation may be mediated by CBD interaction with CB2 and TRPV1. We also investigated the protein levels of the autophagic proteins LC3 and beclin 1. CBD reduced the MPP+-induced increase of LC3 by CB2 and TRPV1 receptors. These data suggested the involvement of ERK in the modulation of autophagy. However, beclin 1 levels were not modified neither by MPP+ nor by CBD. These results indicated that CBD may exert preventive and protective actions in PD.
Collapse
Affiliation(s)
- Agnese Gugliandolo
- IRCCS Centro Neurolesi "Bonino Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Federica Pollastro
- Dipartimento di Scienze del Farmaco, Università del Piemonte Orientale, Largo Donegani 2, 28100 Novara, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
42
|
Clarithromycin inhibits autophagy in colorectal cancer by regulating the hERG1 potassium channel interaction with PI3K. Cell Death Dis 2020; 11:161. [PMID: 32123164 PMCID: PMC7052256 DOI: 10.1038/s41419-020-2349-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
We have studied how the macrolide antibiotic Clarithromycin (Cla) regulates autophagy, which sustains cell survival and resistance to chemotherapy in cancer. We found Cla to inhibit the growth of human colorectal cancer (CRC) cells, by modulating the autophagic flux and triggering apoptosis. The accumulation of cytosolic autophagosomes accompanied by the modulation of autophagic markers LC3-II and p62/SQSTM1, points to autophagy exhaustion. Because Cla is known to bind human Ether-à-go-go Related Gene 1 (hERG1) K+ channels, we studied if its effects depended on hERG1 and its conformational states. By availing of hERG1 mutants with different gating properties, we found that fluorescently labelled Cla preferentially bound to the closed channels. Furthermore, by sequestering the channel in the closed conformation, Cla inhibited the formation of a macromolecular complex between hERG1 and the p85 subunit of PI3K. This strongly reduced Akt phosphorylation, and stimulated the p53-dependent cell apoptosis, as witnessed by late caspase activation. Finally, Cla enhanced the cytotoxic effect of 5-fluorouracil (5-FU), the main chemotherapeutic agent in CRC, in vitro and in a xenograft CRC model. We conclude that Cla affects the autophagic flux by impairing the signaling pathway linking hERG1 and PI3K. Combining Cla with 5-FU might be a novel therapeutic option in CRC.
Collapse
|
43
|
Kiruthiga C, Devi KP, Nabavi SM, Bishayee A. Autophagy: A Potential Therapeutic Target of Polyphenols in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12030562. [PMID: 32121322 PMCID: PMC7139730 DOI: 10.3390/cancers12030562] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a conserved biological phenomenon that maintains cellular homeostasis through the clearing of damaged cellular components under cellular stress and offers the cell building blocks for cellular survival. Aberrations in autophagy subsidize to various human pathologies, such as dementia, cardiovascular diseases, leishmaniosis, influenza, hepatic diseases, and cancer, including hepatocellular carcinoma (HCC). HCC is the fifth common mortal type of liver cancer globally, with an inhomogeneous topographical distribution and highest incidence tripled in men than women. Existing treatment procedures with liver cancer patients result in variable success rates and poor prognosis due to their drug resistance and toxicity. One of the pathophysiological mechanisms that are targeted during the development of anti-liver cancer drugs is autophagy. Generally, overactivated autophagy may lead to a non-apoptotic form of programmed cell death (PCD) or autophagic cell death or type II PCD. Emerging evidence suggests that manipulation of autophagy could induce type II PCD in cancer cells, acting as a potential tumor suppressor. Hence, altering autophagic signaling offers new hope for the development of novel drugs for the therapy of resistant cancer cells. Natural polyphenolic compounds, including flavonoids and non-flavonoids, execute their anticarcinogenic mechanism through upregulating tumor suppressors and autophagy by modulating canonical (Beclin-1-dependent) and non-canonical (Beclin-1-independent) signaling pathways. Additionally, there is evidence signifying that plant polyphenols target angiogenesis and metastasis in HCC via interference with multiple intracellular signals and decrease the risk against HCC. The current review offers a comprehensive understanding of how natural polyphenolic compounds exhibit their anti-HCC effects through regulation of autophagy, the non-apoptotic mode of cell death.
Collapse
Affiliation(s)
- Chandramohan Kiruthiga
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India;
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University (Science Campus), Karaikudi 630 003, Tamil Nadu, India;
- Correspondence: (K.P.D.); or (A.B.); Tel.: +91-4565223325 (K.P.D.); +1-941-782-5950 (A.B.)
| | - Seyed M. Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran;
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
- Correspondence: (K.P.D.); or (A.B.); Tel.: +91-4565223325 (K.P.D.); +1-941-782-5950 (A.B.)
| |
Collapse
|
44
|
Qian S, Tong S, Wu J, Tian L, Qi Z, Chen B, Zhu D, Zhang Y. Paris saponin VII extracted from Trillium tschonoskii induces autophagy and apoptosis in NSCLC cells. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112304. [PMID: 31626908 DOI: 10.1016/j.jep.2019.112304] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/26/2019] [Accepted: 10/12/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Trillium tschonoskii Maxim, a perennial herb of the Trilliaceae, has been widely used to treat inflammation, hypertension and cancer. We investigated Paris saponin VII's (PS VII), isolated from Trillium tschonoskii Maxim, function in mediating autophagy and apoptosis in NSCLC cells. MATERIALS AND METHODS We treated various NSCLC cells with different concentrations of PS Ⅶ and then measure the cell apoptosis by using flow cytometry assays and western blot. Autophagy were investigated by using western blot, transmission electron microscopy and immunofluorescence analysis. We also use a xenograft model of nude mice to measure the effect of PS Ⅶ in vivo. RESULTS Treatment with PS Ⅶ significantly inhibit NSCLC cell growth, especially for A549 (IC50 = 1.53 μM). Moreover, PS VII induces caspase-dependent apoptosis and autophagy through AMPK-ULK1 pathway. After blocking autophagy by 3-methyladenine (3-MA), PS VII induced cell death was significantly increased. In vivo, the co-treatment with PS VII and 3-MA dramatically inhibited A549 tumor growth in immune deficient mice and has similar inhibition rates as cisplatin group. CONCLUSION Our results suggest that a combination of PS VII and autophagy inhibitor may be a potential anticancer strategy in the NSCLC therapy.
Collapse
Affiliation(s)
- Shijing Qian
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, PR China.
| | - Shanshan Tong
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, PR China.
| | - Juan Wu
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, PR China.
| | - Lulu Tian
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, PR China.
| | - Zhan Qi
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, PR China.
| | - Beilei Chen
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, PR China.
| | - Deqiu Zhu
- Department of oncology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, PR China.
| | - Yan Zhang
- Department of oncology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, PR China.
| |
Collapse
|
45
|
Arora N, Raj A, Anjum F, Kaur R, Rawat SS, Kumar R, Tripathi S, Singh G, Prasad A. Unveiling Taenia solium kinome profile and its potential for new therapeutic targets. Expert Rev Proteomics 2020; 17:85-94. [DOI: 10.1080/14789450.2020.1719835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Naina Arora
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Anand Raj
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
- Department of Biotechnology, Motilal Nehru Institute of Technology, Allahabad, India
| | - Farhan Anjum
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Rimanpreet Kaur
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Suraj Singh Rawat
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Rajiv Kumar
- Department of Biotechnology, CSIR-Institute of Himalayan Bioresource Technology, Palampur, India
| | - Shweta Tripathi
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Gagandeep Singh
- Department of Neurology, Dayanand Medical College, Ludhiana, India
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| |
Collapse
|
46
|
Hanyu X, Lanyue L, Miao D, Wentao F, Cangran C, Hui S. Effect of Ganoderma applanatum polysaccharides on MAPK/ERK pathway affecting autophagy in breast cancer MCF-7 cells. Int J Biol Macromol 2020; 146:353-362. [PMID: 31911173 DOI: 10.1016/j.ijbiomac.2020.01.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/21/2019] [Accepted: 01/02/2020] [Indexed: 12/14/2022]
Abstract
Fungal polysaccharides have become hotspots in the field of health foods due to their antitumor activity in recent years. In this experiment, antitumor effect of the medicinal fungus Ganoderma applanatum polysaccharide (GAP) was investigated in human breast cancer MCF-7 cells, as well as the molecular mechanism of its effect on autophagy. Results showed that GAP contains three polysaccharides with molecular weights of 6.36 × 105 Da, 4.25 × 105 Da, and 2.53 × 105 Da and which composed of rhamnose, glucose, arabinose, fucose in the molar ratio of 1:22:16.1:3.2. GAP inhibited the proliferation and migration of MCF-7 cells in a time-dose-dependent manner, the maximum inhibition rate reached 50.2% at 500 μg/mL in 48 h. Flow cytometry analysis showed that GAP could induce apoptosis, treatment of cells with GAP could result in up-regulation of gene and protein levels of autophagy-associated markers LC3 and Beclin-1; addition of autocrine late inhibitor CQ significantly raised the protein expression level of LC3II. The mitogen-activated protein kinases (MAPK) signaling pathway was not only related to the apoptotic pathway but also to the autophagy pathway; Western blot analysis showed that MAPK signaling pathway is involved in GAP-induced autophagy in MCF-7 cells. Detection of the relevant signaling pathway protein showed that the expression of p-ERK1/2 protein was down-regulated, however the expression of p-p38 and p-JNK protein was up-regulated. These results indicate that GAP could induce early autophagy in MCF-7 cells via the MAPK/ERK pathway. In conclusion, GAP showed strong antitumor activity by inducing apoptosis and autophagy through MAPK signaling pathway in MCF-7 cells, suggesting the molecular mechanism of fungal polysaccharide on its antitumor activity.
Collapse
Affiliation(s)
- Xu Hanyu
- School of Life Science, Jilin Agricultural University, Changchun, Jilin, China
| | - Liu Lanyue
- School of Life Science, Jilin Agricultural University, Changchun, Jilin, China
| | - Ding Miao
- School of Life Science, Jilin Agricultural University, Changchun, Jilin, China
| | - Fan Wentao
- School of Life Science, Jilin Agricultural University, Changchun, Jilin, China
| | - Chen Cangran
- School of Life Science, Jilin Agricultural University, Changchun, Jilin, China
| | - Song Hui
- School of Life Science, Jilin Agricultural University, Changchun, Jilin, China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Changchun, China.
| |
Collapse
|
47
|
Wang XS, Yue J, Hu LN, Tian Z, Zhang K, Yang L, Zhang HN, Guo YY, Feng B, Liu HY, Wu YM, Zhao MG, Liu SB. Activation of G protein-coupled receptor 30 protects neurons by regulating autophagy in astrocytes. Glia 2020; 68:27-43. [PMID: 31429156 DOI: 10.1002/glia.23697] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 12/24/2022]
Abstract
Ischemic stroke leads to neuronal damage induced by excitotoxicity, inflammation, and oxidative stress. Astrocytes play diverse roles in stroke and ischemia-induced inflammation, and autophagy is critical for maintaining astrocytic functions. Our previous studies showed that the activation of G protein-coupled receptor 30 (GPR30), an estrogen membrane receptor, protected neurons from excitotoxicity. However, the role of astrocytic GPR30 in maintaining autophagy and neuroprotection remained unclear. In this study, we found that the neuroprotection induced by G1 (GPR30 agonist) in wild-type mice after a middle cerebral artery occlusion was completely blocked in GPR30 conventional knockout (KO) mice but partially attenuated in astrocytic or neuronal GPR30 KO mice. In cultured primary astrocytes, glutamate exposure induced astrocyte proliferation and decreased astrocyte autophagy by activating mammalian target of rapamycin (mTOR) and c-Jun N-terminal kinase (JNK) and inhibiting p38 mitogen-activated protein kinase (MAPK) pathway. G1 treatment restored autophagy to its basal level by regulating the p38 pathway but not the mTOR and JNK signaling pathways. Our findings revealed a key role of GPR30 in neuroprotection via the regulation of astrocyte autophagy and support astrocytic GPR30 as a potential drug target against ischemic brain damage.
Collapse
Affiliation(s)
- Xin-Shang Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiao Yue
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Li-Ning Hu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen Tian
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.,Department of Pharmacy, The 154th Central Hospital of PLA, Xinyang, China
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Le Yang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui-Nan Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan-Yan Guo
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Bin Feng
- State Key Laboratory of Military Stomatology, Department of pharmacy, School of Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Fourth Military Medical University, Xi'an, China
| | - Hai-Yan Liu
- State Key Laboratory of Military Stomatology, Department of pharmacy, School of Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Fourth Military Medical University, Xi'an, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ming-Gao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China.,Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
48
|
Zambrano JN, Eblen ST, Abt M, Rhett JM, Muise-Helmericks R, Yeh ES. HUNK Phosphorylates Rubicon to Support Autophagy. Int J Mol Sci 2019; 20:E5813. [PMID: 31752345 PMCID: PMC6888122 DOI: 10.3390/ijms20225813] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/01/2019] [Accepted: 11/15/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Autophagy is a catabolic cellular recycling pathway that is essential for maintaining intracellular homeostasis. Autophagosome formation is achieved via the coordination of the Beclin-1 protein complex. Rubicon is a Beclin-1 associated protein that suppresses autophagy by impairing the activity of the class III PI3K, Vps34. However, very little is known about the molecular mechanisms that regulate Rubicon function. METHODS In this study, co-immunoprecipitation and kinase assays were used to investigate the ability of Hormonally Upregulated Neu-associated Kinase (HUNK) to bind to and phosphorylate Rubicon. LC3B was monitored by immunofluorescence and immunoblotting to determine whether phosphorylation of Rubicon by HUNK controls the autophagy suppressive function of Rubicon. RESULTS Findings from this study identify Rubicon as a novel substrate of HUNK and show that phosphorylation of Rubicon inhibits its function, promoting autophagy.
Collapse
Affiliation(s)
- Joelle N. Zambrano
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA; (J.N.Z.); (S.T.E.); (J.M.R.)
| | - Scott T. Eblen
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA; (J.N.Z.); (S.T.E.); (J.M.R.)
| | - Melissa Abt
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Simon Cancer Center, Indianapolis, IN 46202, USA;
| | - J. Matthew Rhett
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA; (J.N.Z.); (S.T.E.); (J.M.R.)
| | - Robin Muise-Helmericks
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Elizabeth S. Yeh
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Simon Cancer Center, Indianapolis, IN 46202, USA;
| |
Collapse
|
49
|
Jung M, Lee JH, Lee C, Park JH, Park YR, Moon KC. Prognostic Implication of pAMPK Immunohistochemical Staining by Subcellular Location and Its Association with SMAD Protein Expression in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2019; 11:cancers11101602. [PMID: 31640193 PMCID: PMC6826619 DOI: 10.3390/cancers11101602] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/17/2019] [Accepted: 10/19/2019] [Indexed: 11/18/2022] Open
Abstract
Although cytoplasmic AMP-activated protein kinase (AMPK) has been known as a tumor-suppressor protein, nuclear AMPK is suggested to support clear cell renal cell carcinoma (ccRCC). In addition, pAMPK interacts with TGF-β/SMAD, which is one of the frequently altered pathways in ccRCC. In this study, we investigated the prognostic significance of pAMPK with respect to subcellular location and investigated its interaction with TGF-β/SMAD in ccRCC. Immunohistochemical staining for pAMPK, pSMAD2 and SMAD4 was conducted on tissue microarray of 987 ccRCC specimens. Moreover, the levels of pSMAD2 were measured in Caki-1 cells treated with 5-aminoimidazole-4-carboxamide ribonucleotide. The relationship between AMPK/pAMPK and TGFB1 expression was determined using the TCGA database. As a result, pAMPK positivity, either in the cytoplasm or nuclei, was independently associated with improved ccRCC prognosis, after adjusting for TNM stage and WHO grade. Furthermore, pAMPK-positive ccRCC displayed increased pSMAD2 and SMAD4 expression, while activation of pAMPK increased pSMAD2 in Caki-1 cells. However, AMPK/pAMPK expression was inversely correlated with TGFB1 expression in the TCGA database. Therefore, pAMPK immunostaining, both in the cytoplasm and nuclei, is a useful prognostic biomarker for ccRCC. pAMPK targets TGF-β-independent phosphorylation of SMAD2 and activates pSMAD2/SMAD4, representing a novel anti-tumoral mechanism of pAMPK in ccRCC.
Collapse
Affiliation(s)
- Minsun Jung
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.
| | - Jeong Hoon Lee
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Cheol Lee
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.
| | - Jeong Hwan Park
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul 07061, Korea.
| | - Yu Rang Park
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea.
- Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
50
|
Zou Y, Sarem M, Xiang S, Hu H, Xu W, Shastri VP. Autophagy inhibition enhances Matrine derivative MASM induced apoptosis in cancer cells via a mechanism involving reactive oxygen species-mediated PI3K/Akt/mTOR and Erk/p38 signaling. BMC Cancer 2019; 19:949. [PMID: 31615459 PMCID: PMC6794878 DOI: 10.1186/s12885-019-6199-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 09/24/2019] [Indexed: 12/26/2022] Open
Abstract
Background In the quest for new anti-cancer drugs, the drug discovery process has shifted to screening of active ingredients in traditional eastern medicine. Matrine is an active alkaloid isolated from plants of the Sophora genus used in traditional Chinese herbal medicine that exhibits a wide spectrum of biological properties and has a potential as an anti-proliferative agent. In this study, we investigated the anticancer property of MASM, ([(6aS, 10S, 11aR, 11bR, 11cS)210-Methylamino-dodecahydro-3a, 7a-diaza-benzo (de)anthracene-8-thione]), a potent derivative of matrine. Methods Four epithelial cancer cell lines representing the dominant cancers, namely: A549 (non-small-cell lung cancer cell line), MCF-7 and MDA-MB-231 (breast cancer cell lines), and Hela (cervical cancer cell line) were employed, and the mechanistic underpinning of MASM-induced apoptosis was investigated using flow cytometry, western blot and immunofluorescence. Results MASM, induced apoptosis via caspase 3 dependent and independent pathways, and autophagy in all the four cancer cell lines, but post-EMT (epithelial mesenchymal transition) cells showed greater sensitivity to MASM. Scavenging reactive oxygen species using N-acetylcysteine rescued all cancer cell lines from apoptosis and autophagy. Mechanistic analysis revealed that MASM induced autophagy involves inhibition of Akt signaling and the activation of Erk and p38 signaling, and inhibition of autophagy further enhanced the apoptosis induced by MASM. Conclusions These results indicate that MASM possesses potency against cancer cells and modulating autophagy during MASM administration could be used to further enhance its therapeutic effects.
Collapse
Affiliation(s)
- Yuming Zou
- Institute for Macromolecular Chemistry, University of Freiburg, 79104, Freiburg, Germany.,Department of Orthopaedics, Department of Orthopaedics, People's Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China.,Department of Orthopaedics, Changhai hospital, Second Military Medical University, Shanghai, 200433, People's Republic of China.,Department of Orthopaedics, the 904th Hospital of Joint Logistic Support Force, Chinese People's Liberation Army, Wuxi, Jiangsu Province, People's Republic of China
| | - Melika Sarem
- Institute for Macromolecular Chemistry, University of Freiburg, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Shengnan Xiang
- Institute for Macromolecular Chemistry, University of Freiburg, 79104, Freiburg, Germany
| | - Honggang Hu
- Department of Organic Chemistry, School of Pharmacy, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Weidong Xu
- Department of Orthopaedics, Changhai hospital, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - V Prasad Shastri
- Institute for Macromolecular Chemistry, University of Freiburg, 79104, Freiburg, Germany. .,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|