1
|
Alimohammadi M, Rahimzadeh P, Khorrami R, Bonyadi M, Daneshi S, Nabavi N, Raesi R, Farani MR, Dehkhoda F, Taheriazam A, Hashemi M. A comprehensive review of the PTEN/PI3K/Akt axis in multiple myeloma: From molecular interactions to potential therapeutic targets. Pathol Res Pract 2024; 260:155401. [PMID: 38936094 DOI: 10.1016/j.prp.2024.155401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/02/2024] [Accepted: 06/09/2024] [Indexed: 06/29/2024]
Abstract
Phosphatase and tensin homolog (PTEN), phosphatidylinositol 3-kinase (PI3K), and protein kinase B (Akt) signaling pathways contribute to the development of several cancers, including multiple myeloma (MM). PTEN is a tumor suppressor that influences the PI3K/Akt/mTOR pathway, which in turn impacts vital cellular processes like growth, survival, and treatment resistance. The current study aims to present the role of PTEN and PI3K/Akt/mTOR signaling in the development of MM and its response to treatment. In addition, the molecular interactions in MM that underpin the PI3K/Akt/mTOR pathway and address potential implications for the development of successful treatment plans are also discussed in detail. We investigate their relationship to both upstream and downstream regulators, highlighting new developments in combined therapies that target the PTEN/PI3K/Akt axis to overcome drug resistance, including the use of PI3K and mitogen-activated protein kinase (MAPK) inhibitors. We also emphasize that PTEN/PI3K/Akt pathway elements may be used in MM diagnosis, prognosis, and therapeutic targets.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Islamic Republic of Iran
| | - Mojtaba Bonyadi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Islamic Republic of Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Islamic Republic of Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran; Department of Nursing, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Farshid Dehkhoda
- Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran.
| |
Collapse
|
2
|
Pu J, Liu T, Wang X, Sharma A, Schmidt-Wolf IGH, Jiang L, Hou J. Exploring the role of histone deacetylase and histone deacetylase inhibitors in the context of multiple myeloma: mechanisms, therapeutic implications, and future perspectives. Exp Hematol Oncol 2024; 13:45. [PMID: 38654286 DOI: 10.1186/s40164-024-00507-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Histone deacetylase inhibitors (HDACis) are a significant category of pharmaceuticals that have developed in the past two decades to treat multiple myeloma. Four drugs in this category have received approval from the U.S. Food and Drug Administration (FDA) for use: Panobinonstat (though canceled by the FDA in 2022), Vorinostat, Belinostat and Romidepsin. The efficacy of this group of drugs is attributed to the disruption of many processes involved in tumor growth through the inhibition of histone deacetylase, and this mode of action leads to significant anti-multiple myeloma (MM) activity. In MM, inhibition of histone deacetylase has many downstream consequences, including suppression of NF-κB signaling and HSP90, upregulation of cell cycle regulators (p21, p53), and downregulation of antiapoptotic proteins including Bcl-2. Furthermore, HDACis have a variety of direct and indirect oxidative effects on cellular DNA. HDAC inhibitors enhance normal immune function, thereby decreasing the proliferation of malignant plasma cells and promoting autophagy. The various biological effects of inhibiting histone deacetylase have a combined or additional impact when used alongside other chemotherapeutic and targeted drugs for multiple myeloma. This helps to decrease resistance to treatment. Combination treatment regimens that include HDACis have become an essential part of the therapy for multiple myeloma. These regimens incorporate drugs from other important classes of anti-myeloma agents, such as immunomodulatory drugs (IMiDs), conventional chemotherapy, monoclonal antibodies, and proteasome inhibitors. This review provides a comprehensive evaluation of the clinical efficacy and safety data pertaining to the currently approved histone deacetylase inhibitors, as well as an explanation of the crucial function of histone deacetylase in multiple myeloma and the characteristics of the different histone deacetylase inhibitors. Moreover, it provides a concise overview of the most recent developments in the use of histone deacetylase inhibitors for treating multiple myeloma, as well as potential future uses in treatment.
Collapse
Affiliation(s)
- Jingjing Pu
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Ting Liu
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, NRW, Germany
| | - Xuzhen Wang
- Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, Jiangsu, China
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127, Bonn, NRW, Germany
| | - Liping Jiang
- Wuxi Maternity and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Wuxi, 214002, Jiangsu, China.
| | - Jian Hou
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
3
|
Poorghobadi S, Hosseini SY, Sadat SM, Abdoli A, Irani S, Baesi K. The Combinatorial Effect of Ad-IL-24 and Ad-HSV-tk/GCV on Tumor Size, Autophagy, and UPR Mechanisms in Multiple Myeloma Mouse Model. Biochem Genet 2024:10.1007/s10528-024-10671-2. [PMID: 38436816 DOI: 10.1007/s10528-024-10671-2] [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: 08/26/2023] [Accepted: 01/02/2024] [Indexed: 03/05/2024]
Abstract
Multiple myeloma is a type of malignant neoplasia whose treatment has changed over the past decade. This study aimed to investigate the effects of combination of Adenovector-carrying interleukin-24 and herpes simplex virus 1 thymidine kinase/ganciclovir on tumor growth, autophagy, and unfolded protein response mechanisms in mouse model of multiple myeloma. Six groups of mice, including Ad-HSV-tk/GCV, Ad-IL-24, Ad-HSV-tk/IL-24, Ad-GFP, and positive and negative controls, were investigated, and each group was injected every 72 h. The tumor size was measured several times. The expression of LC3B evaluated through western blotting and ASK-1, CHOP, Caspase-3, and ATF-6 genes in the UPR and apoptosis pathways were also analyzed by the quantitative polymerase chain reaction (qPCR) method. The present results showed that the injection of Ad-HSV-tk/GCV, Ad-HSV-tk/IL-24, and metformin reduced the tumor size. The expression of LC3B was significantly higher in the treatment groups and positive control groups compared to the negative control group. The expression of CHOP, caspase-3, and ATF-6 genes was significantly higher in the Ad-IL-24 group compared to the other treatment groups. Besides, the ASK-1 expression was significantly lower in the Ad-IL-24 group as compared to the other groups. Overall, the results indicated that the presence of the HSV-tk gene in the adenovectors reduced the size of tumors and induced autophagy by triggering the expression of LC3B protein. The presence of the IL-24 might affect tumor growth but not as much the therapeutic effect of HSV-tk. Furthermore, the results indicated that co-administration of IL-24 and HSV-tk had no synergistic effect on tumor size control.
Collapse
Affiliation(s)
- Shima Poorghobadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, P.O. Box 14115-331, Tehran, Iran
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, P.O. Box 14115-331, Tehran, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Kazem Baesi
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, P.O. Box 14115-331, Tehran, Iran.
| |
Collapse
|
4
|
Yeşilaltay A, Muz D, Erdal B. Oncolytic Myxoma virus Increases Autophagy in Multiple Myeloma. Turk J Haematol 2024; 41:16-25. [PMID: 38258554 PMCID: PMC10918390 DOI: 10.4274/tjh.galenos.2024.2023.0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/23/2024] [Indexed: 01/24/2024] Open
Abstract
Objective Multiple myeloma, which affects plasma cells, is the second most common hematological malignancy. Despite the development of new drugs and treatment protocols, patient survival has not reached the desired level. In this study, we investigated the effects of Myxoma virus (MYXV), an oncolytic virus, on autophagy in myeloma cells. Materials and Methods We analyzed protein expressions of ATG-5, p62, Beclin-1, LC3B, and the apoptosis marker Bcl-2 as autophagy markers in human U-266 and mouse MOPC-315 myeloma cell lines subjected to different doses of MYXV. In addition, autophagic images of myeloma cells were investigated using transmission electron microscopy (TEM). Results In the first 24 h, which is the early stage of autophagy, ATG-5 and Beclin-1 expression levels were increased in the U-266 and MOPC-315 cell lines in the groups that had received MYXV at a multiplicity of infection of 15. At 48 h, a significant increase was detected in the expression of LC3B, which is a late indicator. Autophagosomes were observed in myeloma cells by TEM. Conclusion MYXV shows an antimyeloma effect by increasing autophagy in myeloma cells.
Collapse
Affiliation(s)
- Alpay Yeşilaltay
- Başkent University İstanbul Hospital, Department of Hematology, İstanbul, Türkiye
| | - Dilek Muz
- Tekirdağ Namık Kemal University Faculty of Veterinary Medicine, Department of Virology, Tekirdağ, Türkiye
| | - Berna Erdal
- Tekirdağ Namık Kemal University Faculty of Medicine, Department of Microbiology, Tekirdağ, Türkiye
| |
Collapse
|
5
|
Pakjoo M, Ahmadi SE, Zahedi M, Jaafari N, Khademi R, Amini A, Safa M. Interplay between proteasome inhibitors and NF-κB pathway in leukemia and lymphoma: a comprehensive review on challenges ahead of proteasome inhibitors. Cell Commun Signal 2024; 22:105. [PMID: 38331801 PMCID: PMC10851565 DOI: 10.1186/s12964-023-01433-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/11/2023] [Indexed: 02/10/2024] Open
Abstract
The current scientific literature has extensively explored the potential role of proteasome inhibitors (PIs) in the NF-κB pathway of leukemia and lymphoma. The ubiquitin-proteasome system (UPS) is a critical component in regulating protein degradation in eukaryotic cells. PIs, such as BTZ, are used to target the 26S proteasome in hematologic malignancies, resulting in the prevention of the degradation of tumor suppressor proteins, the activation of intrinsic mitochondrial-dependent cell death, and the inhibition of the NF-κB signaling pathway. NF-κB is a transcription factor that plays a critical role in the regulation of apoptosis, cell proliferation, differentiation, inflammation, angiogenesis, and tumor migration. Despite the successful use of PIs in various hematologic malignancies, there are limitations such as resistant to these inhibitors. Some reports suggest that PIs can induce NF-κB activation, which increases the survival of malignant cells. This article discusses the various aspects of PIs' effects on the NF-κB pathway and their limitations. Video Abstract.
Collapse
Affiliation(s)
- Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- ATMP department, Breast cancer research center, Motamed cancer institute, ACECR, P.O. BOX:15179/64311, Tehran, Iran
| | - Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Jaafari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reyhane Khademi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Amini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Zhu Y, Jian X, Chen S, An G, Jiang D, Yang Q, Zhang J, Hu J, Qiu Y, Feng X, Guo J, Chen X, Li Z, Zhou R, Hu C, He N, Shi F, Huang S, Liu H, Li X, Xie L, Zhu Y, Zhao L, Jiang Y, Li J, Wang J, Qiu L, Chen X, Jia W, He Y, Zhou W. Targeting gut microbial nitrogen recycling and cellular uptake of ammonium to improve bortezomib resistance in multiple myeloma. Cell Metab 2024; 36:159-175.e8. [PMID: 38113887 DOI: 10.1016/j.cmet.2023.11.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 10/17/2023] [Accepted: 11/30/2023] [Indexed: 12/21/2023]
Abstract
The gut microbiome has been found to play a crucial role in the treatment of multiple myeloma (MM), which is still considered incurable due to drug resistance. In previous studies, we demonstrated that intestinal nitrogen-recycling bacteria are enriched in patients with MM. However, their role in MM relapse remains unclear. This study highlights the specific enrichment of Citrobacter freundii (C. freundii) in patients with relapsed MM. Through fecal microbial transplantation experiments, we demonstrate that C. freundii plays a critical role in inducing drug resistance in MM by increasing levels of circulating ammonium. The ammonium enters MM cells through the transmembrane channel protein SLC12A2, promoting chromosomal instability and drug resistance by stabilizing the NEK2 protein. We show that furosemide sodium, a loop diuretic, downregulates SLC12A2, thereby inhibiting ammonium uptake by MM cells and improving progression-free survival and curative effect scores. These findings provide new therapeutic targets and strategies for the intervention of MM progression and drug resistance.
Collapse
Affiliation(s)
- Yinghong Zhu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Xingxing Jian
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuping Chen
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Duanfeng Jiang
- Department of Hematology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qin Yang
- Department of Hematology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingyu Zhang
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Jian Hu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Qiu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangling Feng
- Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Jiaojiao Guo
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Xun Chen
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Zhengjiang Li
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Ruiqi Zhou
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Cong Hu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Nihan He
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Fangming Shi
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Siqing Huang
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Li
- Department of Hematology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lu Xie
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Zhu
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lia Zhao
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yichuan Jiang
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Li
- Peking Union Medical College Hospital, Chinese Academy Medical Society & Peking Union Medical College, Beijing, China
| | - Jinuo Wang
- Peking Union Medical College Hospital, Chinese Academy Medical Society & Peking Union Medical College, Beijing, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Wei Jia
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Yanjuan He
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Wen Zhou
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Experimental Hematology, Bioinformatics Center, National Clinical Research Center for Geriatric Disorders, Key Laboratory for Carcinogenesis and Invasion, Chinese Ministry of Education, Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Furong Laboratory, Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China; Xiangya School of Public Health, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Khalil MI, Ali MM, Holail J, Houssein M. Growth or death? Control of cell destiny by mTOR and autophagy pathways. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 185:39-55. [PMID: 37944568 DOI: 10.1016/j.pbiomolbio.2023.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/08/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
One of the central regulators of cell growth, proliferation, and metabolism is the mammalian target of rapamycin, mTOR, which exists in two structurally and functionally different complexes: mTORC1 and mTORC2; unlike m TORC2, mTORC1 is activated in response to the sufficiency of nutrients and is inhibited by rapamycin. mTOR complexes have critical roles not only in protein synthesis, gene transcription regulation, proliferation, tumor metabolism, but also in the regulation of the programmed cell death mechanisms such as autophagy and apoptosis. Autophagy is a conserved catabolic mechanism in which damaged molecules are recycled in response to nutrient starvation. Emerging evidence indicates that the mTOR signaling pathway is frequently activated in tumors. In addition, dysregulation of autophagy was associated with the development of a variety of human diseases, such as cancer and aging. Since mTOR can inhibit the induction of the autophagic process from the early stages of autophagosome formation to the late stage of lysosome degradation, the use of mTOR inhibitors to regulate autophagy could be considered a potential therapeutic option. The present review sheds light on the mTOR and autophagy signaling pathways and the mechanisms of regulation of mTOR-autophagy.
Collapse
Affiliation(s)
- Mahmoud I Khalil
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, 11072809, Lebanon; Molecular Biology Unit, Department of Zoology, Faculty of Science, Alexandria University, Alexandria, 21511, Egypt.
| | - Mohamad M Ali
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden.
| | - Jasmine Holail
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom.
| | - Marwa Houssein
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, 11072809, Lebanon.
| |
Collapse
|
8
|
Zhao N, Qu C, Yang Y, Li H, Li Y, Zhu H, Long Z. Identification of a cholesterol metabolism-related prognostic signature for multiple myeloma. Sci Rep 2023; 13:19395. [PMID: 37938654 PMCID: PMC10632470 DOI: 10.1038/s41598-023-46426-z] [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: 04/14/2023] [Accepted: 10/31/2023] [Indexed: 11/09/2023] Open
Abstract
Multiple myeloma (MM) is a prevalent hematological malignancy that poses significant challenges for treatment. Dysregulated cholesterol metabolism has been linked to tumorigenesis, disease progression, and therapy resistance. However, the correlation between cholesterol metabolism-related genes (CMGs) and the prognosis of MM remains unclear. Univariate Cox regression analysis and LASSO Cox regression analysis were applied to construct an overall survival-related signature based on the Gene Expression Omnibus database. The signature was validated using three external datasets. Enrichment analysis and immune analysis were performed between two risk groups. Furthermore, an optimal nomogram was established for clinical application, and its performance was assessed by the calibration curve and C-index. A total of 6 CMGs were selected to establish the prognostic signature, including ANXA2, CHKA, NSDHL, PMVK, SCAP and SQLE. The prognostic signature demonstrated good prognostic performance and correlated with several important clinical parameters, including number of transplants, International Staging System, albumin, beta2-Microglobulin and lactate dehydrogenase levels. The function analysis and immune analysis revealed that the metabolic pathways and immunologic status were associated with risk score. The nomogram incorporating the signature along with other clinical characteristics was constructed and the discrimination was verified by the calibration curve and C-index. Our findings indicated the potential prognostic connotation of cholesterol metabolism in MM. The development and validation of the prognostic signature is expected to aid in predicting prognosis and guiding precision treatment for MM.
Collapse
Affiliation(s)
- Na Zhao
- Department of Hematology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Chunxia Qu
- Department of Hematology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Yan Yang
- Department of Hematology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Huihui Li
- Department of Hematology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Yueyue Li
- Department of Hematology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China
| | - Hongbo Zhu
- Department of Pathology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| | - Zhiguo Long
- Department of Hematology, Shanghai Pudong Hospital, Fudan University, Shanghai, 201399, China.
| |
Collapse
|
9
|
Chen J, Cao W, Huang X, Chen Q, Ye S, Qu J, Liu Y, Guo X, Yao S, Zhang E, He J, Li A, Yang L, Cai Z. TRIM21 enhances bortezomib sensitivity in multiple myeloma by halting prosurvival autophagy. Blood Adv 2023; 7:5752-5770. [PMID: 37083684 PMCID: PMC10561007 DOI: 10.1182/bloodadvances.2022008241] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 03/22/2023] [Accepted: 04/09/2023] [Indexed: 04/22/2023] Open
Abstract
Bortezomib (bort) is an effective therapeutic agent for patients with multiple myeloma (MM); however, most patients develop drug resistance. Autophagy, a highly conserved process that recycles cytosol or entire organelles via lysosomal activity, is essential for the survival, homeostasis, and drug resistance in MM. Growing evidence has highlighted that E3 ligase tripartite motif-containing protein 21 (TRIM21) not only interacts with multiple autophagy regulators but also participates in drug resistance in various cancers. However, to date, the direct substrates and additional roles of TRIM21 in MM remain unexplored. In this study, we demonstrated that low TRIM21 expression is a factor for relapse in MM. TRIM21 knockdown (KD) made MM cells more resistant to bort, whereas TRIM21 overexpression (OE) resulted in increased MM sensitivity to bort. Proteomic and phosphoproteomic studies of TRIM21 KD MM cells showed that bort resistance was associated with increased oxidative stress and elevated prosurvival autophagy. Our results showed that TRIM21 KD MM cell lines induced prosurvival autophagy after bort treatment, suppressing autophagy by 3-methyladenine treatment or by the short hairpin RNA of autophagy-related gene 5 (ATG5)-restored-bort sensitivity. Indeed, ATG5 expression was increased and decreased by TRIM21 KD and OE, respectively. TRIM21 affected autophagy by ubiquitinating ATG5 through K48 for proteasomal degradation. Importantly, we confirmed that TRIM21 could potentiate the antimyeloma effect of bort through in vitro and in vivo experiments. Overall, our findings define the key role of TRIM21 in MM bort resistance and provide a foundation for a novel targeted therapeutic approach.
Collapse
Affiliation(s)
- Jing Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wen Cao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xi Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qingxiao Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuting Ye
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianwei Qu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xing Guo
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shunnan Yao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anqi Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Yang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Bisht K, Fukao T, Chiron M, Richardson P, Atanackovic D, Chini E, Chng WJ, Van De Velde H, Malavasi F. Immunomodulatory properties of CD38 antibodies and their effect on anticancer efficacy in multiple myeloma. Cancer Med 2023; 12:20332-20352. [PMID: 37840445 PMCID: PMC10652336 DOI: 10.1002/cam4.6619] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND CD38 has been established as an important therapeutic target for multiple myeloma (MM), for which two CD38 antibodies are currently approved-daratumumab and isatuximab. CD38 is an ectoenzyme that degrades NAD and its precursors and is involved in the production of adenosine and other metabolites. AIM Among the various mechanisms by which CD38 antibodies can induce MM cell death is immunomodulation, including multiple pathways for CD38-mediated T-cell activation. Patients who respond to anti-CD38 targeting treatment experience more marked changes in T-cell expansion, activity, and clonality than nonresponders. IMPLICATIONS Resistance mechanisms that undermine the immunomodulatory effects of CD38-targeting therapies can be tumor intrinsic, such as the downregulation of CD38 surface expression and expression of complement inhibitor proteins, and immune microenvironment-related, such as changes to the natural killer (NK) cell numbers and function in the bone marrow niche. There are numerous strategies to overcome this resistance, which include identifying and targeting other therapeutic targets involved in, for example, adenosine production, the activation of NK cells or monocytes through immunomodulatory drugs and their combination with elotuzumab, or with bispecific T-cell engagers.
Collapse
Affiliation(s)
| | - Taro Fukao
- Sanofi OncologyCambridgeMassachusettsUSA
| | | | - Paul Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma CenterDana Farber Cancer Institute, Harvard Medical SchoolBostonMassachusettsUSA
| | - Djordje Atanackovic
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer CenterBaltimoreMarylandUSA
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Eduardo Chini
- Department of Anesthesiology and Perioperative MedicineMayo ClinicJacksonvilleFloridaUSA
| | - Wee Joo Chng
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | | | - Fabio Malavasi
- Department of Medical SciencesUniversity of TurinTorinoItaly
- Fondazione Ricerca MolinetteTorinoItaly
| |
Collapse
|
11
|
Clavero E, Sanchez-Maldonado JM, Macauda A, Ter Horst R, Sampaio-Marques B, Jurczyszyn A, Clay-Gilmour A, Stein A, Hildebrandt MAT, Weinhold N, Buda G, García-Sanz R, Tomczak W, Vogel U, Jerez A, Zawirska D, Wątek M, Hofmann JN, Landi S, Spinelli JJ, Butrym A, Kumar A, Martínez-López J, Galimberti S, Sarasquete ME, Subocz E, Iskierka-Jażdżewska E, Giles GG, Rybicka-Ramos M, Kruszewski M, Abildgaard N, Verdejo FG, Sánchez Rovira P, da Silva Filho MI, Kadar K, Razny M, Cozen W, Pelosini M, Jurado M, Bhatti P, Dudzinski M, Druzd-Sitek A, Orciuolo E, Li Y, Norman AD, Zaucha JM, Reis RM, Markiewicz M, Rodríguez Sevilla JJ, Andersen V, Jamroziak K, Hemminki K, Berndt SI, Rajkumar V, Mazur G, Kumar SK, Ludovico P, Nagler A, Chanock SJ, Dumontet C, Machiela MJ, Varkonyi J, Camp NJ, Ziv E, Vangsted AJ, Brown EE, Campa D, Vachon CM, Netea MG, Canzian F, Försti A, Sainz J. Polymorphisms within Autophagy-Related Genes as Susceptibility Biomarkers for Multiple Myeloma: A Meta-Analysis of Three Large Cohorts and Functional Characterization. Int J Mol Sci 2023; 24:ijms24108500. [PMID: 37239846 DOI: 10.3390/ijms24108500] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/10/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Multiple myeloma (MM) arises following malignant proliferation of plasma cells in the bone marrow, that secrete high amounts of specific monoclonal immunoglobulins or light chains, resulting in the massive production of unfolded or misfolded proteins. Autophagy can have a dual role in tumorigenesis, by eliminating these abnormal proteins to avoid cancer development, but also ensuring MM cell survival and promoting resistance to treatments. To date no studies have determined the impact of genetic variation in autophagy-related genes on MM risk. We performed meta-analysis of germline genetic data on 234 autophagy-related genes from three independent study populations including 13,387 subjects of European ancestry (6863 MM patients and 6524 controls) and examined correlations of statistically significant single nucleotide polymorphisms (SNPs; p < 1 × 10-9) with immune responses in whole blood, peripheral blood mononuclear cells (PBMCs), and monocyte-derived macrophages (MDM) from a large population of healthy donors from the Human Functional Genomic Project (HFGP). We identified SNPs in six loci, CD46, IKBKE, PARK2, ULK4, ATG5, and CDKN2A associated with MM risk (p = 4.47 × 10-4-5.79 × 10-14). Mechanistically, we found that the ULK4rs6599175 SNP correlated with circulating concentrations of vitamin D3 (p = 4.0 × 10-4), whereas the IKBKErs17433804 SNP correlated with the number of transitional CD24+CD38+ B cells (p = 4.8 × 10-4) and circulating serum concentrations of Monocyte Chemoattractant Protein (MCP)-2 (p = 3.6 × 10-4). We also found that the CD46rs1142469 SNP correlated with numbers of CD19+ B cells, CD19+CD3- B cells, CD5+IgD- cells, IgM- cells, IgD-IgM- cells, and CD4-CD8- PBMCs (p = 4.9 × 10-4-8.6 × 10-4) and circulating concentrations of interleukin (IL)-20 (p = 0.00082). Finally, we observed that the CDKN2Ars2811710 SNP correlated with levels of CD4+EMCD45RO+CD27- cells (p = 9.3 × 10-4). These results suggest that genetic variants within these six loci influence MM risk through the modulation of specific subsets of immune cells, as well as vitamin D3-, MCP-2-, and IL20-dependent pathways.
Collapse
Affiliation(s)
- Esther Clavero
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain
| | - José Manuel Sanchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
| | - Angelica Macauda
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rob Ter Horst
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
| | - Artur Jurczyszyn
- Plasma Cell Dyscrasias Center, Department of Hematology, Jagiellonian University Medical College, 31-066 Kraków, Poland
| | - Alyssa Clay-Gilmour
- Department of Biostatistics and Epidemiology, Arnold School of Public Health, University of South Carolina, Greenville, SC 29208, USA
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
| | - Angelika Stein
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Michelle A T Hildebrandt
- Department of Lymphoma-Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Niels Weinhold
- Myeloma Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
| | - Gabriele Buda
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy
| | - Ramón García-Sanz
- Diagnostic Laboratory Unit in Hematology, University Hospital of Salamanca, IBSAL, CIBERONC, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Waldemar Tomczak
- Department of Hematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-059 Lublin, Poland
| | - Ulla Vogel
- National Research Centre for the Working Environment, DK-2100 Copenhagen, Denmark
| | - Andrés Jerez
- Department of Hematology, Experimental Hematology Unit, Vall d'Hebron Institute of Oncology (VHIO), University Hospital Vall d'Hebron, 08035 Barcelona, Spain
| | - Daria Zawirska
- Department of Hematology, University Hospital, 30-688 Kraków, Poland
| | - Marzena Wątek
- Holycross Medical Oncology Center, 25-735 Kielce, Poland
- Institute of Hematology and Transfusion Medicine, 00-791 Warsaw, Poland
| | - Jonathan N Hofmann
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Stefano Landi
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - John J Spinelli
- Division of Population Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Aleksandra Butrym
- Department of Cancer Prevention and Therapy, Wroclaw Medical University, 50-367 Wroclaw, Poland
- Alfred Sokolowski Specialist Hospital in Walbrzych Oncology Support Centre for Clinical Trials, 58-309 Walbrzych, Poland
| | - Abhishek Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
- Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | | | - Sara Galimberti
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy
| | - María Eugenia Sarasquete
- Diagnostic Laboratory Unit in Hematology, University Hospital of Salamanca, IBSAL, CIBERONC, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Edyta Subocz
- Department of Hematology, Military Institute of Medicine, 04-141 Warsaw, Poland
| | | | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC 3004, Australia
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, VIC 3010, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia
| | - Malwina Rybicka-Ramos
- Department of Hematology, Specialist Hospital No. 1 in Bytom, Academy of Silesia, Faculty of Medicine, 40-055 Katowice, Poland
| | - Marcin Kruszewski
- Department of Hematology, University Hospital No. 2, 85-168 Bydgoszcz, Poland
| | - Niels Abildgaard
- Department of Hematology, Odense University Hospital, DK-5000 Odense, Denmark
| | | | - Pedro Sánchez Rovira
- Department of Medical Oncology, Complejo Hospitalario de Jaén, 23007 Jaén, Spain
| | - Miguel Inacio da Silva Filho
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany
| | | | - Małgorzata Razny
- Department of Hematology, Rydygier Hospital, 31-826 Cracow, Poland
| | - Wendy Cozen
- Division of Hematology/Oncology, Department of Medicine, School of Medicine, Department of Pathology, School of Medicine, Susan and Henry Samueli College of Health Sciences, Chao Family Comprehensive Cancer Center, University of California at Irvine, Irvine, CA 92697, USA
| | - Matteo Pelosini
- U.O. Dipartimento di Ematologia, Azienda USL Toscana Nord Ovest, 57124 Livorno, Italy
| | - Manuel Jurado
- Hematology Department, Virgen de las Nieves University Hospital, 18012 Granada, Spain
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
- Department of Medicine, University of Granada, 18012 Granada, Spain
| | - Parveen Bhatti
- Cancer Control Research, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- Program in Epidemiology, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Marek Dudzinski
- Department of Hematology, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
| | - Agnieszka Druzd-Sitek
- Department of Lymphoproliferative Diseases, Maria Skłodowska Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Enrico Orciuolo
- Haematology Unit, Department of Clinical and Experimental Medicine, University of Pisa/AOUP, 56126 Pisa, Italy
| | - Yang Li
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Aaron D Norman
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
- Genetic Epidemiology and Risk Assessment Program, Mayo Clinic Comprehensive Cancer Center, Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
| | - Jan Maciej Zaucha
- Department of Hematology and Transplantology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Rui Manuel Reis
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal and ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil
| | - Miroslaw Markiewicz
- Department of Hematology, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
| | | | - Vibeke Andersen
- Molecular Diagnostics and Clinical Research Unit, Institute of Regional Health Research, University Hospital of Southern Denmark, DK-6200 Aabenraa, Denmark
| | - Krzysztof Jamroziak
- Department of Hematology, Transplantology and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Kari Hemminki
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, 30605 Pilsen, Czech Republic
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vicent Rajkumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Grzegorz Mazur
- Department of Internal Diseases, Occupational Medicine, Hypertension and Clinical Oncology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Shaji K Kumar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
| | - Arnon Nagler
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer 52621, Israel
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Charles Dumontet
- UMR INSERM 1052/CNRS 5286, University of Lyon, Hospices Civils de Lyon, 69008 Lyon, France
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Nicola J Camp
- Division of Hematology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Elad Ziv
- Department of Medicine, University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94143, USA
| | - Annette Juul Vangsted
- Department of Hematology, Rigshospitalet, Copenhagen University, DK-2100 Copenhagen, Denmark
| | - Elizabeth E Brown
- Department of Pathology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniele Campa
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Celine M Vachon
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55902, USA
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Asta Försti
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Hopp Children's Cancer Center (KiTZ), 69120 Heidelberg, Germany
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain
- Instituto de Investigación Biosanataria IBs, Granada, 18014 Granada, Spain
- Department of Biochemistry and Molecular Biology I, University of Granada, 18071 Granada, Spain
| |
Collapse
|
12
|
Bashiri H, Tabatabaeian H. Autophagy: A Potential Therapeutic Target to Tackle Drug Resistance in Multiple Myeloma. Int J Mol Sci 2023; 24:ijms24076019. [PMID: 37046991 PMCID: PMC10094562 DOI: 10.3390/ijms24076019] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
Multiple myeloma (MM) is the second most prevalent hematologic malignancy. In the past few years, the survival of MM patients has increased due to the emergence of novel drugs and combination therapies. Nevertheless, one of the significant obstacles in treating most MM patients is drug resistance, especially for individuals who have experienced relapses or developed resistance to such cutting-edge treatments. One of the critical processes in developing drug resistance in MM is autophagic activity, an intracellular self-digestive process. Several possible strategies of autophagy involvement in the induction of MM-drug resistance have been demonstrated thus far. In multiple myeloma, it has been shown that High mobility group box protein 1 (HMGB1)-dependent autophagy can contribute to drug resistance. Moreover, activation of autophagy via proteasome suppression induces drug resistance. Additionally, the effectiveness of clarithromycin as a supplemental drug in treating MM has been reported recently, in which autophagy blockage is proposed as one of the potential action mechanisms of CAM. Thus, a promising therapeutic approach that targets autophagy to trigger the death of MM cells and improve drug susceptibility could be considered. In this review, autophagy has been addressed as a survival strategy crucial for drug resistance in MM.
Collapse
Affiliation(s)
- Hamed Bashiri
- Institute of Molecular and Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore 138673, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | | |
Collapse
|
13
|
De Novellis D, Fontana R, Giudice V, Serio B, Selleri C. Innovative Anti-CD38 and Anti-BCMA Targeted Therapies in Multiple Myeloma: Mechanisms of Action and Resistance. Int J Mol Sci 2022; 24:645. [PMID: 36614086 PMCID: PMC9820921 DOI: 10.3390/ijms24010645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
CD38 and B-cell maturation antigens (BCMAs) are prevalently expressed on neoplastic plasma cells in multiple myeloma (MM), making them ideal therapeutic targets. Anti-CD38 monoclonal antibodies, such as approved daratumumab and isatuximab, are currently the milestone in MM treatment because they induce plasma cell apoptosis and kill through several mechanisms, including antibody-dependent cellular cytotoxicity or phagocytosis. BCMA is considered an excellent target in MM, and three different therapeutic strategies are either already available in clinical practice or under investigation: antibody-drug conjugates, such as belantamab-mafodotin; bispecific T cell engagers; and chimeric antigen receptor-modified T cell therapies. Despite the impressive clinical efficacy of these new strategies in the treatment of newly diagnosed or multi-refractory MM patients, several mechanisms of resistance have already been described, including antigen downregulation, the impairment of antibody-dependent cell cytotoxicity and phagocytosis, T- and natural killer cell senescence, and exhaustion. In this review, we summarize the current knowledge on the mechanisms of action and resistance of anti-CD38 and anti-BCMA agents and their clinical efficacy and safety.
Collapse
Affiliation(s)
- Danilo De Novellis
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Raffaele Fontana
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Bianca Serio
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
14
|
Chen P, Wu S, Dong X, Zhou M, Xu P, Chen B. Formosanin C induces autophagy-mediated apoptosis in multiple myeloma cells through the PI3K/AKT/mTOR signaling pathway. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2022; 27:977-986. [PMID: 36053135 DOI: 10.1080/16078454.2022.2117126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Multiple myeloma (MM) is an incurable plasma cell malignancy associated with poor survival. Novel therapeutic drugs are urgently needed to improve MM therapy and patient outcomes. This study aimed to investigate the effect of formosanin C (FC), a Chinese medicine monomer, on MM in vitro and disclose the underlying molecular mechanism. METHODS The effect of FC on the viability, proliferation, apoptosis, and autophagy of MM cell lines (NCI-H929 and ARP1) was studied through CCK-8, colony formation, flow cytometry, GFP-LC3, and western blotting assays, respectively. A pharmacological approach and network pharmacology technology were implemented to explore the potential mechanisms of the action of FC on MM cells. RESULTS FC efficiently suppressed the viability and colony-forming capacity, but promoted the number of autophagic vacuoles with GFP-LC3 localization and the percentage of apoptotic cells in MM cells. Additionally, FC significantly increased the levels of the autophagy-related proteins LC3-Ⅱ and Beclin 1, as well as the apoptosis-related proteins Bax and cleaved caspase-3, but blocked the expression of the proapoptotic protein Bcl-2 in the cells; these effects were reversed by an inhibitor of autophagy, 3-methyladenine. What's more, we found that the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway was involved in the FC-mediated inhibition of MM. Pharmacological inhibition of this pathway dramatically relieved FC-triggered excessive expression of autophagy-related proteins and rescued MM cells from FC-induced apoptosis. CONCLUSION Our findings indicate that FC exhibits an anti-MM effect by activating cell autophagy through the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Ping Chen
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China.,Department of Hematology, Nanjing Hospital of Chinese Medicine, Nanjing, People's Republic of China
| | - Sungui Wu
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Xiaoqing Dong
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Min Zhou
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Peipei Xu
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| |
Collapse
|
15
|
Zalpoor H, Bakhtiyari M, Akbari A, Aziziyan F, Shapourian H, Liaghat M, Zare-Badie Z, Yahyazadeh S, Tarhriz V, Ganjalikhani-Hakemi M. Potential role of autophagy induced by FLT3-ITD and acid ceramidase in acute myeloid leukemia chemo-resistance: new insights. Cell Commun Signal 2022; 20:172. [PMCID: PMC9620650 DOI: 10.1186/s12964-022-00956-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/09/2022] [Indexed: 11/10/2022] Open
Abstract
Acute myeloid leukemia (AML) is a type of leukemia with a poor prognosis and survival characterized by abnormal cell proliferation and differentiation. Despite advances in treatment, AML still has a low complete remission rate, particularly in elderly patients, and recurrences are frequently seen even after complete remissions. The major challenge in treating AML is the resistance of leukemia cells to chemotherapy drugs. Thus, to overcome this issue, it can be crucial to conduct new investigations to explore the mechanisms of chemo-resistance in AML and target them. In this review, the potential role of autophagy induced by FLT3-ITD and acid ceramidase in chemo-resistance in AML patients are analyzed. With regard to the high prevalence of FLT3-ITD mutation (about 25% of AML cases) and high level of acid ceramidase in these patients, we hypothesized that both of these factors could lead to chemo-resistance by inducing autophagy. Therefore, pharmacological targeting of autophagy, FLT3-ITD, and acid ceramidase production could be a promising therapeutic approach for such AML patients to overcome chemo-resistance.
Video abstract
Collapse
Affiliation(s)
- Hamidreza Zalpoor
- grid.412571.40000 0000 8819 4698Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Maryam Bakhtiyari
- grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran ,grid.412606.70000 0004 0405 433XDepartment of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Abdullatif Akbari
- grid.412571.40000 0000 8819 4698Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Fatemeh Aziziyan
- grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran ,grid.412266.50000 0001 1781 3962Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hooriyeh Shapourian
- grid.411036.10000 0001 1498 685XDepartment of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Liaghat
- grid.510410.10000 0004 8010 4431Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran ,grid.472315.60000 0004 0494 0825Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Zahra Zare-Badie
- grid.412571.40000 0000 8819 4698Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sheida Yahyazadeh
- grid.411036.10000 0001 1498 685XDepartment of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahideh Tarhriz
- grid.412888.f0000 0001 2174 8913Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mazdak Ganjalikhani-Hakemi
- grid.411036.10000 0001 1498 685XDepartment of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
16
|
Wang G, Fan F, Sun C, Hu Y. Looking into Endoplasmic Reticulum Stress: The Key to Drug-Resistance of Multiple Myeloma? Cancers (Basel) 2022; 14:5340. [PMID: 36358759 PMCID: PMC9654020 DOI: 10.3390/cancers14215340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 09/22/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy, resulting from the clonal proliferation of malignant plasma cells within the bone marrow. Despite significant advances that have been made with novel drugs over the past two decades, MM patients often develop therapy resistance, especially to bortezomib, the first-in-class proteasome inhibitor that was approved for treatment of MM. As highly secretory monoclonal protein-producing cells, MM cells are characterized by uploaded endoplasmic reticulum stress (ERS), and rely heavily on the ERS response for survival. Great efforts have been made to illustrate how MM cells adapt to therapeutic stresses through modulating the ERS response. In this review, we summarize current knowledge on the mechanisms by which ERS response pathways influence MM cell fate and response to treatment. Moreover, based on promising results obtained in preclinical studies, we discuss the prospect of applying ERS modulators to overcome drug resistance in MM.
Collapse
Affiliation(s)
- Guangqi Wang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
| | - Fengjuan Fan
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
| | - Chunyan Sun
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
17
|
Blasiak J, Kaarniranta K. Secretory autophagy: a turn key for understanding AMD pathology and developing new therapeutic targets? Expert Opin Ther Targets 2022; 26:883-895. [PMID: 36529978 DOI: 10.1080/14728222.2022.2157260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Age-related macular degeneration (AMD) is an eye disease leading to vision loss with poorly known pathogenesis and limited therapeutic options. Degradative autophagy (DA) is impaired in AMD, but emerging evidence points to secretary autophagy (SA) as a key element in AMD pathogenesis. AREAS COVERED SA may cause the release of proteins and protein aggregates, lipofuscin, beta amyloid, faulty mitochondria, pro-inflammatory and pro-angiogenic factors from the retinal pigment epithelium (RPE) that may contribute to drusen formation and choroidal neovascularization. SA may replace DA, when formation of autolysosome is impaired, and then a harmful cargo, instead of being degraded, is extruded from the RPE contributing to drusen and/or angiogenic environment. Therefore, the interplay between DA and SA may be critical for drusen formation and choroidal neovascularization, so it can be a turn key to understand AMD pathogenesis. EXPERT OPINION Although SA fulfills some beneficial functions, it is detrimental for the retina in many cases. Therefore, inhibiting SA may be a therapeutic strategy in AMD, but it is challenged by the development of selective SA inhibitors that would not affect DA. The TRIM16, SEC22B and RAB8A proteins, specific for secretory autophagosome, may be primary candidates as therapeutic targets, but their action is not limited to autophagy and therefore requires further studies.
Collapse
Affiliation(s)
- Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, Kuopio, Finland.,Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
18
|
Ayna Duran G, Benderli Cihan Y. Autophagy-related genes affect the survival of multiple myeloma patients depending on chromosomal abnormality. ASIAN BIOMED 2022; 16:249-264. [PMID: 37551318 PMCID: PMC10321186 DOI: 10.2478/abm-2022-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Background Targeting autophagy at gene level may be promising in multiple myeloma (MM) treatment depending on chromosomal abnormality (ABN) status. Objectives We aimed to investigate the role of ABN on survival of MM patients and to identify prognosis related autophagy-related genes (ARGs) for patients with or without ABN. Methods Gene intensity values of 222 ARG for 548 MM patients were obtained from the Affymetrix Human Genome U133 Plus 2.0 Array (GPL570) platform containing 54,675 probes (GSE24080). A dataset containing data from 1576 MM patients with 1q21 amplification (GSE4204, GSE4452, GSE4581, and GSE2658) was used for validation. Survival analysis of the patients was analyzed using univariate and multivariate Cox regression method with the help of R3.53 programming language and Kaplan-Meier graphics were created. The Gene Ontology enRIchmentanaLysis and visuaLizAtion (GOrilla) tool was used to define the related biological processes and pathways. Results The overall survival (OS) and event-free survival (EFS) in all MM patients were strongly influenced by ABN. In the group of patients with ABN, 41 ARGs were found to be important in prognosis, whereas in the group of patients without ABN, 13 ARGs were found to be important in prognosis. CDKN1A, FKBP1B, FOXO3, and NCKAP1 ARGs were commonly significant in both groups and found to be survival triggering. Conclusions The classification of MM patients according to the absence or presence of ABN is important in the determination of survival status. Detection of survival related ARGs in patients with chromosomal anomalies may be a new therapeutic target in treatment.
Collapse
Affiliation(s)
- Gizem Ayna Duran
- Department of Biomedical Engineering, Faculty of Engineering, Izmir University of Economics, Balçova, İzmir35330, Turkey
| | - Yasemin Benderli Cihan
- Department of Radiation Oncology, Kayseri City Education and Research Hospital, Kocasinan, Kayseri38080, Turkey
| |
Collapse
|
19
|
Sharma A, Nair R, Achreja A, Mittal A, Gupta P, Balakrishnan K, Edgar CL, Animasahun O, Dwivedi B, Barwick BG, Gupta VA, Matulis SM, Bhasin M, Lonial S, Nooka AK, Wiita AP, Boise LH, Nagrath D, Shanmugam M. Therapeutic implications of mitochondrial stress-induced proteasome inhibitor resistance in multiple myeloma. SCIENCE ADVANCES 2022; 8:eabq5575. [PMID: 36170375 PMCID: PMC9519052 DOI: 10.1126/sciadv.abq5575] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The connections between metabolic state and therapy resistance in multiple myeloma (MM) are poorly understood. We previously reported that electron transport chain (ETC) suppression promotes sensitivity to the BCL-2 antagonist venetoclax. Here, we show that ETC suppression promotes resistance to proteasome inhibitors (PIs). Interrogation of ETC-suppressed MM reveals integrated stress response-dependent suppression of protein translation and ubiquitination, leading to PI resistance. ETC and protein translation gene expression signatures from the CoMMpass trial are down-regulated in patients with poor outcome and relapse, corroborating our in vitro findings. ETC-suppressed MM exhibits up-regulation of the cystine-glutamate antiporter SLC7A11, and analysis of patient single-cell RNA-seq shows that clusters with low ETC gene expression correlate with higher SLC7A11 expression. Furthermore, erastin or venetoclax treatment diminishes mitochondrial stress-induced PI resistance. In sum, our work demonstrates that mitochondrial stress promotes PI resistance and underscores the need for implementing combinatorial regimens in MM cognizant of mitochondrial metabolic state.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Remya Nair
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Abhinav Achreja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Anjali Mittal
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Pulkit Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Kamakshi Balakrishnan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Claudia L. Edgar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Olamide Animasahun
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Bhakti Dwivedi
- Department of Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Vikas A. Gupta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Shannon M. Matulis
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Manoj Bhasin
- Department of Biostatistics and Bioinformatics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Ajay K. Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Arun P. Wiita
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Lawrence H. Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Deepak Nagrath
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
- Corresponding author.
| |
Collapse
|
20
|
Zhang C, Kang T, Wang X, Wang J, Liu L, Zhang J, Liu X, Li R, Wang J, Zhang J. LINC-PINT suppresses cisplatin resistance in gastric cancer by inhibiting autophagy activation via epigenetic silencing of ATG5 by EZH2. Front Pharmacol 2022; 13:968223. [PMID: 36091809 PMCID: PMC9452659 DOI: 10.3389/fphar.2022.968223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/11/2022] [Indexed: 12/24/2022] Open
Abstract
Resistance to cisplatin (DDP) is a major obstacle in the clinical treatment of advanced gastric cancer (GC). Long noncoding RNA (lncRNA) play a significant regulatory role in the development and drug resistance of GC. In this study, we reported that the lncRNA LINC-PINT was downregulated in DDP-resistant GC cells. Functional studies showed that LINC-PINT inhibited proliferation and migration of DDP-resistant GC cells in vitro, and overexpression of LINC-PINT could enhance the sensitivity of DDP-resistant GC cells to DDP. Further investigation revealed that LINC-PINT recruited enhancer of zeste homolog 2 (EZH2) to the promotor of ATG5 to inhibit its transcription, leading to the suppression of autophagy and DDP resensitization. Collectively, our results revealed how the LINC-PINT/EZH2/ATG5 axis regulates autophagy and DDP resistance in GC. These data suggest that LINC-PINT may be a potential therapeutic target in GC.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Tong Kang
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xinyi Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jizhao Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lin Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jiawei Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Rong Li
- Department of Radiotherapy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jiansheng Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Jiansheng Wang, ; Jia Zhang,
| | - Jia Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Jiansheng Wang, ; Jia Zhang,
| |
Collapse
|
21
|
Development of Olaparib-Resistance Prostate Cancer Cell Lines to Identify Mechanisms Associated with Acquired Resistance. Cancers (Basel) 2022; 14:cancers14163877. [PMID: 36010871 PMCID: PMC9405809 DOI: 10.3390/cancers14163877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary PARP inhibitors (PARPi; olaparib) are presently in clinical trials for advanced prostate cancer (PC). Resistance mechanisms are not fully understood in PC compared to ovarian and breast cancers. Our study aimed to identify new molecular mechanisms that affect acquired olaparib-resistance. We developed new resistant PC cell line models derived from original PC cell lines. We identified that DNA repair, autophagy, and the Rho-associated coiled-coil containing protein kinase 2 (ROCK2) could be potential targets to reverse the acquired olaparib-resistance. Abstract Background: Poly (ADP-ribose) polymerase inhibitors (PARPi) were initially deployed to target breast and ovarian tumors with mutations in DNA damage response genes. Recently, PARPi have been shown to be beneficial in the treatment of prostate cancer (PC) patients having exhausted conventional therapeutics. Despite demonstrating promising response rates, all patients treated with PARPi eventually develop resistance. However, PARPi resistance in PC is not well understood, and further studies are required to understand PARPi resistance in PC to propose strategies to circumvent resistance. Methods: Starting from well-established olaparib-sensitive PC cell lines (LNCaP, C4-2B and DU145), we derived olaparib-resistant (OR) PC cell lines and performed a microarray analysis. Results: The olaparib IC50 values of OR cell lines increased significantly as compared to the parental cell lines. Gene expression analyses revealed that different pathways, including DNA repair, cell cycle regulation and autophagy, were affected by acquired resistance. A total of 195 and 87 genes were significantly upregulated and downregulated, respectively, in all three OR cell lines compared to their parental counterparts. Among these genes, we selected BRCC3, ROCK2 and ATG2B for validation. We showed that ROCK2 expression, basal autophagy and homologous recombination (HR) efficiency were increased in all OR cell lines. Conclusions: Our study provides a new in vitro model to study PARPi resistance in PC and suggests new possible targets to reverse resistance and prolong the benefits of PARPi treatment.
Collapse
|
22
|
Majeed ST, Majeed R, Andrabi KI. Expanding the view of the molecular mechanisms of autophagy pathway. J Cell Physiol 2022; 237:3257-3277. [DOI: 10.1002/jcp.30819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 01/18/2023]
Affiliation(s)
- Sheikh Tahir Majeed
- Department of Biotechnology Central University of Kashmir Ganderbal Jammu and Kashmir India
- Growth Factor Signaling Laboratory, Department of Biotechnology University of Kashmir Srinagar Jammu and Kashmir India
| | - Rabiya Majeed
- Growth Factor Signaling Laboratory, Department of Biotechnology University of Kashmir Srinagar Jammu and Kashmir India
- Department of Biochemistry University of Kashmir Srinagar Jammu and Kashmir India
| | - Khurshid I. Andrabi
- Growth Factor Signaling Laboratory, Department of Biotechnology University of Kashmir Srinagar Jammu and Kashmir India
| |
Collapse
|
23
|
Schwestermann J, Besse A, Driessen C, Besse L. Contribution of the Tumor Microenvironment to Metabolic Changes Triggering Resistance of Multiple Myeloma to Proteasome Inhibitors. Front Oncol 2022; 12:899272. [PMID: 35692781 PMCID: PMC9178120 DOI: 10.3389/fonc.2022.899272] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Virtually all patients with multiple myeloma become unresponsive to treatment with proteasome inhibitors over time. Relapsed/refractory multiple myeloma is accompanied by the clonal evolution of myeloma cells with heterogeneous genomic aberrations, diverse proteomic and metabolic alterations, and profound changes of the bone marrow microenvironment. However, the molecular mechanisms that drive resistance to proteasome inhibitors within the context of the bone marrow microenvironment remain elusive. In this review article, we summarize the latest knowledge about the complex interaction of malignant plasma cells with its surrounding microenvironment. We discuss the pivotal role of metabolic reprograming of malignant plasma cells within the tumor microenvironment with a subsequent focus on metabolic rewiring in plasma cells upon treatment with proteasome inhibitors, driving multiple ways of adaptation to the treatment. At the same time, mutual interaction of plasma cells with the surrounding tumor microenvironment drives multiple metabolic alterations in the bone marrow. This provides a tumor-promoting environment, but at the same time may offer novel therapeutic options for the treatment of relapsed/refractory myeloma patients.
Collapse
Affiliation(s)
| | | | | | - Lenka Besse
- Laboratory of Experimental Oncology, Clinics for Medical Hematology and Oncology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
24
|
Han Q, Bai H, Xu Y, Zhou M, Zhou H, Dong X, Chen B. Solamargine induces autophagy-mediated apoptosis and enhances bortezomib activity in multiple myeloma. Clin Exp Pharmacol Physiol 2022; 49:674-685. [PMID: 35294057 PMCID: PMC9310729 DOI: 10.1111/1440-1681.13643] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/04/2022] [Accepted: 03/09/2022] [Indexed: 11/29/2022]
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy with a poor survival rate. Conventional chemotherapeutic agent‐induced adverse events, including toxicity, neuropathy or drug resistance, significantly decrease the patients' quality of life and can even lead to interruption of treatment. Therefore, novel therapeutic drugs and strategies are urgently needed to improve MM therapy and patient outcomes. Here, we show that solamargine (SM), a steroidal alkaloid glycoside isolated from a Chinese herb Solanum nigrum L., exhibits promising anti‐MM activity. In particular, SM suppressed the viability of MM cell lines (ARP‐1 and NCI‐H929) in a concentration‐ and time‐dependent manner, inducing apoptosis in these cells. RNA‐seq analysis showed that treatment with SM led to the upregulation of genes associated with cell death and autophagy in H929 cells. Further, we found that treatment with SM activated autophagy in the MM cells, as incubation with 3‐Methyladenine, an inhibitor of autophagy, significantly alleviated SM‐triggered apoptosis and inhibition of viability in MM cells. Interestingly, we also observed a synergistic effect between SM and bortezomib (BTZ), a common chemotherapeutic agent for MM, in both MM cells and human bone marrow CD138+ primary myeloma cells. We also confirmed the single‐agent efficacy of SM and the synergistic effects between SM and BTZ in an MM xenograft mouse model. Collectively, these findings indicate that SM exerts an anti‐MM effect, at least in part, by activating cell autophagy and reveal that SM alone or in combination with BTZ is a potential therapeutic strategy for treating MM.
Collapse
Affiliation(s)
- Qiaoyan Han
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.,Department of Hematology, Jingjiang People's Hospital, Jingjiang, Jiangsu, China
| | - Hua Bai
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yong Xu
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Min Zhou
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - He Zhou
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaoqing Dong
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Bing Chen
- Department of Hematology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
25
|
Arora M, Bogenberger JM, Abdelrahman AM, Yonkus J, Alva-Ruiz R, Leiting JL, Chen X, Serrano Uson Junior PL, Dumbauld CR, Baker AT, Gamb SI, Egan JB, Zhou Y, Nagalo BM, Meurice N, Eskelinen EL, Salomao MA, Kosiorek HE, Braggio E, Barrett MT, Buetow KH, Sonbol MB, Mansfield AS, Roberts LR, Bekaii-Saab TS, Ahn DH, Truty MJ, Borad MJ. Synergistic combination of cytotoxic chemotherapy and cyclin-dependent kinase 4/6 inhibitors in biliary tract cancers. Hepatology 2022; 75:43-58. [PMID: 34407567 DOI: 10.1002/hep.32102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Biliary tract cancers (BTCs) are uncommon, but highly lethal, gastrointestinal malignancies. Gemcitabine/cisplatin is a standard-of-care systemic therapy, but has a modest impact on survival and harbors toxicities, including myelosuppression, nephropathy, neuropathy, and ototoxicity. Whereas BTCs are characterized by aberrations activating the cyclinD1/cyclin-dependent kinase (CDK)4/6/CDK inhibitor 2a/retinoblastoma pathway, clinical use of CDK4/6 inhibitors as monotherapy is limited by lack of validated biomarkers, diffident preclinical efficacy, and development of acquired drug resistance. Emerging studies have explored therapeutic strategies to enhance the antitumor efficacy of CDK4/6 inhibitors by the combination with chemotherapy regimens, but their mechanism of action remains elusive. APPROACH AND RESULTS Here, we report in vitro and in vivo synergy in BTC models, showing enhanced efficacy, reduced toxicity, and better survival with a combination comprising gemcitabine/cisplatin and CDK4/6 inhibitors. Furthermore, we demonstrated that abemaciclib monotherapy had only modest efficacy attributable to autophagy-induced resistance. Notably, triplet therapy was able to potentiate efficacy through elimination of the autophagic flux. Correspondingly, abemaciclib potentiated ribonucleotide reductase catalytic subunit M1 reduction, resulting in sensitization to gemcitabine. CONCLUSIONS As such, these data provide robust preclinical mechanistic evidence of synergy between gemcitabine/cisplatin and CDK4/6 inhibitors and delineate a path forward for translation of these findings to preliminary clinical studies in advanced BTC patients.
Collapse
Affiliation(s)
- Mansi Arora
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | - James M Bogenberger
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | | | - Jennifer Yonkus
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Xianfeng Chen
- Department of Informatics, Mayo Clinic, Scottsdale, Arizona, USA
| | | | - Chelsae R Dumbauld
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Alexander T Baker
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Scott I Gamb
- Microscopy and Cell Analysis Core, Mayo Clinic, Rochester, Minnesota, USA
| | - Jan B Egan
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yumei Zhou
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Bolni Marius Nagalo
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Nathalie Meurice
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | | | - Marcela A Salomao
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona, USA
| | - Heidi E Kosiorek
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, Arizona, USA
| | - Esteban Braggio
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Michael T Barrett
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | - Kenneth H Buetow
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Mohamad B Sonbol
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Aaron S Mansfield
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lewis R Roberts
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Tanios S Bekaii-Saab
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | - Daniel H Ahn
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA
| | - Mark J Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Mitesh J Borad
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, Arizona, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
26
|
Chen Q, Hao M, Wang L, Li L, Chen Y, Shao X, Tian Z, Pfuetzner RA, Zhong Q, Brunger AT, Guan JL, Diao J. Prefused lysosomes cluster on autophagosomes regulated by VAMP8. Cell Death Dis 2021; 12:939. [PMID: 34645799 PMCID: PMC8514493 DOI: 10.1038/s41419-021-04243-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/13/2021] [Accepted: 10/04/2021] [Indexed: 11/21/2022]
Abstract
Lysosome–autophagosome fusion is critical to autophagosome maturation. Although several proteins that regulate this fusion process have been identified, the prefusion architecture and its regulation remain unclear. Herein, we show that upon stimulation, multiple lysosomes form clusters around individual autophagosomes, setting the stage for membrane fusion. The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein on lysosomes—vesicle-associated membrane protein 8 (VAMP8)—plays an important role in forming this prefusion state of lysosomal clusters. To study the potential role of phosphorylation on spontaneous fusion, we investigated the effect of phosphorylation of C-terminal residues of VAMP8. Using a phosphorylation mimic, we observed a decrease of fusion in an ensemble lipid mixing assay and an increase of unfused lysosomes associated with autophagosomes. These results suggest that phosphorylation not only reduces spontaneous fusion for minimizing autophagic flux under normal conditions, but also preassembles multiple lysosomes to increase the fusion probability for resuming autophagy upon stimulation. VAMP8 phosphorylation may thus play an important role in chemotherapy drug resistance by influencing autophagosome maturation.
Collapse
Affiliation(s)
- Qixin Chen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Mingang Hao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Lei Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Linsen Li
- State Key Lab of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Yang Chen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Xintian Shao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Zhiqi Tian
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Richard A Pfuetzner
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, 94305, CA, USA.,Howard Hughes Medical Institute, Stanford University, Stanford, 94305, CA, USA
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Axel T Brunger
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, 94305, CA, USA.,Howard Hughes Medical Institute, Stanford University, Stanford, 94305, CA, USA
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
27
|
Siri M, Behrouj H, Dastghaib S, Zamani M, Likus W, Rezaie S, Hudecki J, Khazayel S, Łos MJ, Mokarram P, Ghavami S. Casein Kinase-1-Alpha Inhibitor (D4476) Sensitizes Microsatellite Instable Colorectal Cancer Cells to 5-Fluorouracil via Authophagy Flux Inhibition. Arch Immunol Ther Exp (Warsz) 2021; 69:26. [PMID: 34536148 PMCID: PMC8449776 DOI: 10.1007/s00005-021-00629-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023]
Abstract
Adjuvant chemotherapy with 5-fluorouracil (5-FU) does not improve survival of patients suffering from a form of colorectal cancer (CRC) characterized by high level of microsatellite instability (MSI-H). Given the importance of autophagy and multi-drug-resistant (MDR) proteins in chemotherapy resistance, as well as the role of casein kinase 1-alpha (CK1α) in the regulation of autophagy, we tested the combined effect of 5-FU and CK1α inhibitor (D4476) on HCT116 cells as a model of MSI-H colorectal cancer. To achieve this goal, the gene expression of Beclin1 and MDR genes, ABCG2 and ABCC3 were analyzed using quantitative real-time polymerase chain reaction. We used immunoblotting to measure autophagy flux (LC3, p62) and flow cytometry to detect apoptosis. Our findings showed that combination treatment with 5-FU and D4476 inhibited autophagy flux. Moreover, 5-FU and D4476 combination therapy induced G2, S and G1 phase arrests and it depleted mRNA of both cell proliferation-related genes and MDR-related genes (ABCG2, cyclin D1 and c-myc). Hence, our data indicates that targeting of CK1α may increase the sensitivity of HCT116 cells to 5-FU. To our knowledge, this is the first description of sensitization of CRC cells to 5-FU chemotherapy by CK1α inhibitor.
Collapse
Affiliation(s)
- Morvarid Siri
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran
| | - Hamid Behrouj
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran
| | - Sanaz Dastghaib
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhdeh Zamani
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Wirginia Likus
- Department of Anatomy, School of Health Science in Katowice, Medical University of Silesia, ul. Medyków 18, 40-762, Katowice, Poland
| | - Sedigheh Rezaie
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran
| | - Jacek Hudecki
- Laryngology Department, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Saeed Khazayel
- Department of Research and Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marek J Łos
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pathology, Unii Lubelskiej 1, Pomeranian Medical University, 71-344, Szczecin, Poland.
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, P.O Box: 1167, Shiraz, Iran.
| | - Saeid Ghavami
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada.
- Faculty of Medicine, Katowice School of Technology, Katowice, Poland.
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
28
|
Hosseini A, Hamblin MR, Mirzaei H, Mirzaei HR. Role of the bone marrow microenvironment in drug resistance of hematological malignances. Curr Med Chem 2021; 29:2290-2305. [PMID: 34514979 DOI: 10.2174/0929867328666210910124319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
The unique features of the tumor microenvironment (TME) govern the biological properties of many cancers, including hematological malignancies. TME factors can trigger invasion, and protect against drug cytotoxicity by inhibiting apoptosis and activating specific signaling pathways (e.g. NF-ΚB). TME remodeling is facilitated due to the high self-renewal ability of the bone marrow. Progressing tumor cells can alter some extracellular matrix (ECM) components which act as a barrier to drug penetration in the TME. The initial progression of the cell cycle is controlled by the MAPK pathway (Raf/MEK/ERK) and Hippo pathway, while the final phase is regulated by the PI3K/Akt /mTOR and WNT pathways. In this review we summarize the main signaling pathways involved in drug resistance (DR) and some mechanisms by which DR can occur in the bone marrow. The relationship between autophagy, endoplasmic reticulum stress, and cellular signaling pathways in DR and apoptosis are covered in relation to the TME.
Collapse
Affiliation(s)
- Alireza Hosseini
- Laboratory Hematology and Blood Banking, Tehran University of Medical Sciences, Tehran. Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028. South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan. Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran. Iran
| |
Collapse
|
29
|
Zhang N, Xue M, Sun T, Yang J, Pei Z, Qin K. Fucoidan as an Autophagy Regulator: Mechanisms and Therapeutic Potentials for Cancer and Other Diseases. Nutr Cancer 2021; 74:1568-1579. [PMID: 34477470 DOI: 10.1080/01635581.2021.1973045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Fucoidan, a natural polysaccharide with a variety of classical bioactivities mainly sourced from brown algae, has been extensively studied owing to its favorable pharmacological effects, including anti-inflammatory, anti-tumor, anticoagulant and liver protection. Recently it has been found to play a regulatory role in the processes of autophagy. Autophagy is an important cellular process that effectively protects cells and organisms from stimulating factors such as nutrient deficiency, low cellular ATP levels, metabolic stress, growth factor deprivation and hypoxic conditions. In recent years, many studies have shown that fucoidan can treat human diseases by regulating autophagy process though cell signaling pathways. In this review, we summarize the latest progress in the discovery of natural autophagy regulator of fucoidan for the therapeutic application in cardiac diseases, cancers and liver diseases, aiming to provide the new pharmacological application that fucoidan may treat human diseases by regulating autophagy. Furthermore, we look forward to seeing more diseases that would be treated by autophagy modulator of fucoidan and the discovery of more elaborate autophagy regulation mechanism.
Collapse
Affiliation(s)
- Nan Zhang
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Meilan Xue
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Ting Sun
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Jia Yang
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Zhongqian Pei
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| | - Kunpeng Qin
- Basic Medical College, Qingdao University of Medicine, Qingdao, PR China
| |
Collapse
|
30
|
Zheng YH, Shen HY, Chen X, Feng J, Gao GX. Prognostic model and immune-infiltrating cell landscape based on differentially expressed autophagy-related genes in TP53-mutated multiple myeloma. Arch Med Sci 2021; 20:1619-1630. [PMID: 39649276 PMCID: PMC11623155 DOI: 10.5114/aoms/140293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 07/18/2021] [Indexed: 12/10/2024] Open
Abstract
Introduction Autophagy functions as a prosurvival mechanism in multiple myeloma (MM). The objective of this research was to establish an autophagy-related gene (ARG) signature for predicting the survival outcomes of MM patients with TP53 mutations. Material and methods Information about MM patients with TP53 mutations was downloaded from the Gene Expression Omnibus (GEO) database. Cox proportional hazard regression was employed to determine the independent prognostic ARG and construct a risk signature. Time-dependent receiver-operating characteristic (tROC) curve analysis was used to explore the predictive accuracy of the prognostic model. A nomogram was constructed to give a more precise prediction of the probability of 5-year, 8-year and 10-year overall survival (OS). In addition, we used the CIBERSORT algorithm to explore the distribution difference of 22 immune-infiltrating cells. Results Three differentially expressed ARGs (CASP8, MAPK8, RB1CC1) were finally incorporated to construct the risk model. Area under the curve (AUC) values of the corresponding tROC curve for 5-year, 8-year and 10-year OS were 0.735, 0.686 and 0.662, respectively. Multiple myeloma patients were categorized into high and low-risk groups in accordance with the median threshold value (-1.724549). An ARG-based risk score model was an independent prognostic element correlated with OS, giving an hazard ratio (HR) of 3.29 (95% CI 2.35-4.60, p < 0.001). Thirteen immune infiltrating cells were found to have distribution differences between the two groups. Conclusions We established a three-ARG risk signature which manifested an independent prognostic factor. The nomogram was testified to perform well in forecasting the long-term survival of TP53-mutated MM patients.
Collapse
Affiliation(s)
- Yan-Hua Zheng
- Department of Hematology, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
- National Clinical Research Center for Hematological Diseases of China, Shaanxi Branch Center, Clinical Research Center for Hematologic Disease of Shaanxi Province, China
| | - Hong-Yuan Shen
- Department of Hematology, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Xiang Chen
- Department of Pharmacy, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Juan Feng
- Department of Hematology, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| | - Guang-Xun Gao
- Department of Hematology, Xijing Hospital, Fourth Military Medical University (Air Force Medical University), Xi’an, Shaanxi, China
| |
Collapse
|
31
|
Immunological Prognostic Factors in Multiple Myeloma. Int J Mol Sci 2021; 22:ijms22073587. [PMID: 33808304 PMCID: PMC8036885 DOI: 10.3390/ijms22073587] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm characterized by an abnormal proliferation of clonal, terminally differentiated B lymphocytes. Current approaches for the treatment of MM focus on developing new diagnostic techniques; however, the search for prognostic markers is also crucial. This enables the classification of patients into risk groups and, thus, the selection of the most optimal treatment method. Particular attention should be paid to the possible use of immune factors, as the immune system plays a key role in the formation and course of MM. In this review, we focus on characterizing the components of the immune system that are of prognostic value in MM patients, in order to facilitate the development of new diagnostic and therapeutic directions.
Collapse
|
32
|
Dai H, Ma B, Dai X, Pang J, Wang J, Zhao Y, Wang M, Zhang H, Gao H, Qian S, Tian F, Sun X. Shengma Biejia Decoction Inhibits Cell Growth in Multiple Myeloma by Inducing Autophagy-Mediated Apoptosis Through the ERK/mTOR Pathway. Front Pharmacol 2021; 12:585286. [PMID: 33854428 PMCID: PMC8039907 DOI: 10.3389/fphar.2021.585286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
Shengma Biejia decoction (SMBJD), a traditional Chinese formula recorded in the Golden Chamber, has been widely used for the treatment of malignant tumors. However, its underlying molecular targets and mechanisms are still unclear. This study showed that SMBJD inhibited tumor growth and stimulated hemogram recovery significantly in a multiple myeloma xenograft model. Western blot and immunohistochemistry assays of tumor tissues showed that SMBJD reduced the ratio of autophagy-related proteins LC3-II/LC3-I, while P62 and apoptosis-related proteins cleaved caspase-3/caspase-3 and Bax/Bcl-2 were upregulated. In vitro experiments demonstrated the time-dependent and dose-dependent cytotoxicity of SMBJD on multiple myeloma cell lines H929 and U266 through MTT assays. The LC3-II/LC3-I ratio and number of GFP-LC3 puncta showed that SMBJD inhibited the autophagy process of H929 and U266 cells. Moreover, both SMBJD and 3-methyladenine (3-MA) caused a decrease in LC3-II/LC3-I, and SMBJD could not reverse the upregulation of LC3-II/LC3-I caused by bafilomycin A1 (Baf-A1). Furthermore, the results of annexin V-FITC and propidium iodide double staining demonstrated that SMBJD treatment induced the apoptosis of H929 and U266 cells. These data prove that SMBJD inhibits autophagy and promotes apoptosis in H929 and U266 cells. The results also show that rapamycin could reduce the rate of SMBJD-induced apoptosis in H929 and U266 cells, at a concentration which had no effect on apoptosis but activated autophagy. In addition, analysis of the mechanism indicated that levels of phosphorylated ERK and phosphorylated mTOR were increased by treatment with SMBJD in vivo and in vitro. These results indicate that SMBJD, an old and effective herbal compound, could inhibit the viability of H929 and U266 cells and induce autophagy-mediated apoptosis through the ERK/mTOR pathway. Thus, it represents a potential therapy strategy for multiple myeloma.
Collapse
Affiliation(s)
- Huibo Dai
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Bangyun Ma
- Department of Hematology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xingbin Dai
- Department of Hematology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Pang
- Department of Hematology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingyu Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yandong Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Mengya Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Haoran Gao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shushu Qian
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Fang Tian
- Research Center of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuemei Sun
- Department of Hematology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
33
|
Chien CH, Hsueh WT, Chuang JY, Chang KY. Dissecting the mechanism of temozolomide resistance and its association with the regulatory roles of intracellular reactive oxygen species in glioblastoma. J Biomed Sci 2021; 28:18. [PMID: 33685470 PMCID: PMC7938520 DOI: 10.1186/s12929-021-00717-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is the most common primary malignant brain tumor that is usually considered fatal even with treatment. This is often a result for tumor to develop resistance. Regarding the standard chemotherapy, the alkylating agent temozolomide is effective in disease control but the recurrence will still occur eventually. The mechanism of the resistance is various, and differs in terms of innate or acquired. To date, aberrations in O6-methylguanine-DNA methyltransferase are the clear factor that determines drug susceptibility. Alterations of the other DNA damage repair genes such as DNA mismatch repair genes are also known to affect the drug effect. Together these genes have roles in the innate resistance, but are not sufficient for explaining the mechanism leading to acquired resistance. Recent identification of specific cellular subsets with features of stem-like cells may have role in this process. The glioma stem-like cells are known for its superior ability in withstanding the drug-induced cytotoxicity, and giving the chance to repopulate the tumor. The mechanism is complicated to administrate cellular protection, such as the enhancing ability against reactive oxygen species and altering energy metabolism, the important steps to survive. In this review, we discuss the possible mechanism for these specific cellular subsets to evade cancer treatment, and the possible impact to the following treatment courses. In addition, we also discuss the possibility that can overcome this obstacle.
Collapse
Affiliation(s)
- Chia-Hung Chien
- National Institute of Cancer Research, National Health Research Institutes, 367 Sheng-Li Road, Tainan, 70456, Taiwan
| | - Wei-Ting Hsueh
- Department of Oncology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Jian-Ying Chuang
- Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan.,The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kwang-Yu Chang
- National Institute of Cancer Research, National Health Research Institutes, 367 Sheng-Li Road, Tainan, 70456, Taiwan. .,Department of Oncology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
34
|
Indirubin-3'-monoxime induces paraptosis in MDA-MB-231 breast cancer cells by transmitting Ca 2+ from endoplasmic reticulum to mitochondria. Arch Biochem Biophys 2020; 698:108723. [PMID: 33321111 DOI: 10.1016/j.abb.2020.108723] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Indirubin-3'-monoxime (I3M) induces cell death in many cancer cells; however, whether I3M regulates paraptosis is unclear. The present study aimed to investigate I3M-induced paraptosis. METHODS We treated various cancer cells with I3M, and measured vacuole formation (a paraptosis marker) and the regulating signaling pathway such as endoplasmic reticulum (ER) stress, reactive oxygen species, and proteasomal dysfunction. RESULTS We found that I3M induced small vacuole formation in MDA-MB-231 breast cancer cells and transient knockdown of eIF2α and CHOP significantly downregulated vacuolation in the ER and mitochondria, as well as cell death in response to I3M, indicating that I3M-meditaed paraptosis was upregulated by ER stress. Moreover, I3M accumulated ubiquitinylated proteins via proteasome dysfunction, which stimulated ER stress-mediated Ca2+ release. A Ca2+ chelator significantly downregulated vacuolation in the ER and mitochondria as well as cell death, suggesting that Ca2+ was a key regulator in I3M-induced paraptosis. Our results also revealed that Ca2+ finally transited in mitochondria through mitochondrial Ca2+ uniporter (MCU), causing I3M-mediated paraptosis; however, the paraptosis was completely inhibited by, ruthenium red, an MCU inhibitor. CONCLUSION I3M induced proteasomal dysfunction-mediated ER stress and subsequently promoted Ca2+ release, which was accumulated in the mitochondria via MCU, thus causing paraptosis in MDA-MB-231 breast cancer cells.
Collapse
|
35
|
Zeng J, Shirihai OS, Grinstaff MW. Modulating lysosomal pH: a molecular and nanoscale materials design perspective. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2020; 2:25-37. [PMID: 33403369 PMCID: PMC7781074 DOI: 10.36069/jols/20201204] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lysosomes, membrane-bound organelles, play important roles in cellular processes including endocytosis, phagocytosis, and autophagy. Lysosomes maintain cellular homeostasis by generating a highly acidic environment of pH 4.5 - 5.0 and by housing hydrolytic enzymes that degrade engulfed biomolecules. Impairment of lysosomal function, especially in its acidification, is a driving force in the pathogenesis of diseases including neurodegeneration, cancer, metabolic disorders, and infectious diseases. Therefore, lysosomal pH is an attractive and targetable site for therapeutic intervention. Currently, there is a dearth of strategies or materials available to specifically modulate lysosomal acidification. This review focuses on the key aspects of how lysosomal pH is implicated in various diseases and discusses design strategies and molecular or nanoscale agents for lysosomal pH modulation, with the ultimate goal of developing novel therapeutic solutions.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
- Department of Neurology, School of Medicine, Yale University, New Haven, CT 06511
| | - Orian S Shirihai
- Division of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90045
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118
- Department of Chemistry, Boston University, Boston, MA 02215
| |
Collapse
|
36
|
Molecular Insights into the Multifunctional Role of Natural Compounds: Autophagy Modulation and Cancer Prevention. Biomedicines 2020; 8:biomedicines8110517. [PMID: 33228222 PMCID: PMC7699596 DOI: 10.3390/biomedicines8110517] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
Autophagy is a vacuolar, lysosomal degradation pathway for injured and damaged protein molecules and organelles in eukaryotic cells, which is controlled by nutrients and stress responses. Dysregulation of cellular autophagy may lead to various diseases such as neurodegenerative disease, obesity, cardiovascular disease, diabetes, and malignancies. Recently, natural compounds have come to attention for being able to modulate the autophagy pathway in cancer prevention, although the prospective role of autophagy in cancer treatment is very complex and not yet clearly elucidated. Numerous synthetic chemicals have been identified that modulate autophagy and are favorable candidates for cancer treatment, but they have adverse side effects. Therefore, different phytochemicals, which include natural compounds and their derivatives, have attracted significant attention for use as autophagy modulators in cancer treatment with minimal side effects. In the current review, we discuss the promising role of natural compounds in modulating the autophagy pathway to control and prevent cancer, and provide possible therapeutic options.
Collapse
|
37
|
Gunda V, Pathania AS, Chava S, Prathipati P, Chaturvedi NK, Coulter DW, Pandey MK, Durden DL, Challagundla KB. Amino Acids Regulate Cisplatin Insensitivity in Neuroblastoma. Cancers (Basel) 2020; 12:cancers12092576. [PMID: 32927667 PMCID: PMC7563727 DOI: 10.3390/cancers12092576] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Neuroblastomas mostly show poor response to the Cisplatin therapy. Amino acids serve as building blocks for proteins, which are acquired either through diet or protein breakdown. Our study reveals high amino acid pools and dependence of Cisplatin-tolerant neuroblastomas cells on amino acids for their survival, especially, in drug treated conditions. Our study also demonstrates that response of neuroblastomas to Cisplatin can be improved by decreasing cellular amino acid levels either by reducing amino acid supplements or by applying autophagy inhibitor, Hydroxychloroquine. Thus, our findings establish that neuroblastomas can be sensitized to Cisplatin by targeting amino acid metabolism. Abstract Neuroblastoma are pediatric, extracranial malignancies showing alarming survival prognosis outcomes due to their resilience to current aggressive treatment regimens, including chemotherapies with cisplatin (CDDP) provided in the first line of therapy regimens. Metabolic deregulation supports tumor cell survival in drug-treated conditions. However, metabolic pathways underlying cisplatin-resistance are least studied in neuroblastoma. Our metabolomics analysis revealed that cisplatin-insensitive cells alter their metabolism; especially, the metabolism of amino acids was upregulated in cisplatin-insensitive cells compared to the cisplatin-sensitive neuroblastoma cell line. A significant increase in amino acid levels in cisplatin-insensitive cells led us to hypothesize that the mechanisms upregulating intracellular amino acid pools facilitate insensitivity in neuroblastoma. We hereby report that amino acid depletion reduces cell survival and cisplatin-insensitivity in neuroblastoma cells. Since cells regulate their amino acids levels through processes, such as autophagy, we evaluated the effects of hydroxychloroquine (HCQ), a terminal autophagy inhibitor, on the survival and amino acid metabolism of cisplatin-insensitive neuroblastoma cells. Our results demonstrate that combining HCQ with CDDP abrogated the amino acid metabolism in cisplatin-insensitive cells and sensitized neuroblastoma cells to sub-lethal doses of cisplatin. Our results suggest that targeting of amino acid replenishing mechanisms could be considered as a potential approach in developing combination therapies for treating neuroblastomas.
Collapse
Affiliation(s)
- Venugopal Gunda
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.G.); (A.S.P.); (S.C.)
| | - Anup S. Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.G.); (A.S.P.); (S.C.)
| | - Srinivas Chava
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.G.); (A.S.P.); (S.C.)
| | - Philip Prathipati
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan;
| | - Nagendra K. Chaturvedi
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.C.); (D.W.C.)
| | - Don W. Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (N.K.C.); (D.W.C.)
| | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, 401 South Broadway, Camden, NJ 08103, USA;
| | - Donald L. Durden
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Moores Cancer Center, University of California, San Diego, 3855 Health Science Drive, MC-0815, La Jolla, CA 92093, USA;
- SignalRx Pharmaceuticals, Inc. 8330, Loveland Drive, Omaha, NE 68124, USA
| | - Kishore B. Challagundla
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (V.G.); (A.S.P.); (S.C.)
- The Children’s Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-559-9032
| |
Collapse
|
38
|
Ikeda S, Abe F, Matsuda Y, Kitadate A, Takahashi N, Tagawa H. Hypoxia-inducible hexokinase-2 enhances anti-apoptotic function via activating autophagy in multiple myeloma. Cancer Sci 2020; 111:4088-4101. [PMID: 32790954 PMCID: PMC7648043 DOI: 10.1111/cas.14614] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/30/2020] [Accepted: 08/09/2020] [Indexed: 12/21/2022] Open
Abstract
Multiple myeloma (MM) is an incurable hematopoietic neoplasm derived from plasma cells, and existing in the bone marrow. Recent developments in the field of myeloma onco-biology have enabled the use of proteasome inhibitors (PIs) as key drugs for MM. PIs can increase cell sensitivity to endoplasmic reticulum stress, leading to apoptosis of myeloma cells. PI cannot kill all myeloma cells, however; one reason of this might be activation of autophagy via hypoxic stress in the bone marrow microenvironment. Hypoxia-inducible gene(s) that regulate autophagy may be novel therapeutic target(s) for PI-resistant myeloma cells. Here, a hypoxia-inducible glycolytic enzyme hexokinase-2 (HK2) was demonstrated to contribute by autophagy activation to the acquisition of an anti-apoptotic phenotype in myeloma cells. We found that hypoxic stress led to autophagy activation accompanied by HK2 upregulation in myeloma cells. Under hypoxic conditions, HK2 knockdown inhibited glycolysis and impaired autophagy, inducing apoptosis. The cooperative effects of a PI (bortezomib) against immunodeficient mice inoculated with HK2-knocked down myeloma cells were examined and significant tumor reduction was observed. An HK2 inhibitor, 3-bromopyruvate (3-BrPA), also induced apoptosis under hypoxic rather than normoxic conditions. Further examination of the cooperative effects between 3-BrPA and bortezomib on myeloma cells revealed a significant increase in apoptotic myeloma cells. These results strongly suggested that HK2 regulates the activation of autophagy in hypoxic myeloma cells. Cooperative treatment using PI against a dominant fraction, and HK2 inhibitor against a minor fraction, adapted to the bone marrow microenvironment, may lead to deeper remission for refractory MM.
Collapse
Affiliation(s)
- Sho Ikeda
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Fumito Abe
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuka Matsuda
- Department of Life Science, Akita University Graduate School of Engineering Science, Akita, Japan
| | - Akihiro Kitadate
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroyuki Tagawa
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
39
|
Therachiyil L, Haroon J, Sahir F, Siveen KS, Uddin S, Kulinski M, Buddenkotte J, Steinhoff M, Krishnankutty R. Dysregulated Phosphorylation of p53, Autophagy and Stemness Attributes the Mutant p53 Harboring Colon Cancer Cells Impaired Sensitivity to Oxaliplatin. Front Oncol 2020; 10:1744. [PMID: 32984059 PMCID: PMC7485421 DOI: 10.3389/fonc.2020.01744] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/04/2020] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer (CRC) forms one of the highest ranked cancer types in the world with its increasing incidence and mortality rates despite the advancement in cancer therapeutics. About 50% of human CRCs are reported to have defective p53 expression resultant of TP53 gene mutation often contributing to drug resistance. The current study was aimed to investigate the response of wild-type TP53 harboring HCT 116 and mutant TP53 harboring HT 29 colon cancer cells to chemotherapeutic drug oxaliplatin (OX) and to elucidate the underlying molecular mechanisms of sensitivity/resistance in correlation to their p53 status. OX inhibited growth of wild-type p53-harboring colon cancer cells via p53/p21-Bax mediated apoptosis. Our study revealed that dysregulated phosphorylation of p53, autophagy as well as cancer stemness attributes the mutant p53-harboring colon cancer cells impaired sensitivity to OX.
Collapse
Affiliation(s)
- Lubna Therachiyil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha, Qatar
| | - Javeria Haroon
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Fairooz Sahir
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kodappully S. Siveen
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Michal Kulinski
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Joerg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
- Department of Medicine, Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, Doha, Qatar
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- College of Medicine, Qatar University, Doha, Qatar
| | - Roopesh Krishnankutty
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
40
|
Kim SM, Ha SE, Lee HJ, Rampogu S, Vetrivel P, Kim HH, Venkatarame Gowda Saralamma V, Lee KW, Kim GS. Sinensetin Induces Autophagic Cell Death through p53-Related AMPK/mTOR Signaling in Hepatocellular Carcinoma HepG2 Cells. Nutrients 2020; 12:nu12082462. [PMID: 32824273 PMCID: PMC7468969 DOI: 10.3390/nu12082462] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/19/2022] Open
Abstract
Sinensetin (SIN) has been reported to exhibit anti-inflammatory and anti-cancer activity. However, the cellular and molecular mechanism by which SIN promotes hepatocellular carcinoma (HCC) cell death remains unclear. In the present study, we investigated the induction of cell death by SIN and its underlying mechanism in HepG2 cells, an HCC cell line. We found that SIN significantly induced cell death in HepG2 cells, whereas the proliferation rate of Thle2, human liver epithelial cells, was unaffected by SIN. SIN-treated HepG2 cells were not affected by apoptotic cell death; instead, autophagic cell death was induced through the p53-mediated AMPK/mTOR signaling pathway. Inhibition of p53 degradation led to both autophagy and apoptosis in HepG2 cells. p53 translocation led to SIN-induced autophagy, whereas p53 translocation inhibited SIN-induced apoptosis. However, SIN showed apoptosis in the p53-mutant Hep3B cell line. Molecular docking simulation of the p53 core domain showed effective binding with SIN, which was found significant compared with the known p53 activator, RITA. Collectively, these data suggest that SIN may be a potential anti-cancer agent targeting autophagic cell death in human liver cancer.
Collapse
Affiliation(s)
- Seong Min Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
| | - Sang Eun Ha
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
| | - Ho Jeong Lee
- Biological Resources Research Group, Bioenvironmental Science & Toxicology Division, Gyeongnam Branch Institute, Korea Institute of Toxicology (KIT), 17 Jeigok-gil, Jinju 52834, Korea;
| | - Shailima Rampogu
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (K.W.L.)
| | - Preethi Vetrivel
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
| | - Hun Hwan Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
| | | | - Keun Woo Lee
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (K.W.L.)
| | - Gon Sup Kim
- Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; (S.M.K.); (S.E.H.); (P.V.); (H.H.K.)
- Correspondence: ; Tel.: +82-55-772-2346
| |
Collapse
|
41
|
Naz F, Wu Y, Zhang N, Yang Z, Yu C. Anticancer Attributes of Cantharidin: Involved Molecular Mechanisms and Pathways. Molecules 2020; 25:E3279. [PMID: 32707651 PMCID: PMC7397086 DOI: 10.3390/molecules25143279] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/15/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer is a preeminent threat to the human race, causing millions of deaths each year on the Earth. Traditionally, natural compounds are deemed promising agents for cancer treatment. Cantharidin (CTD)-a terpenoid isolated from blister beetles-has been used extensively in traditional Chinese medicines for healing various maladies and cancer. CTD has been proven to be protein phosphatase 2A (PP2A) and heat shock transcription factor 1 (HSF-1) inhibitor, which can be potential targets for its anticancer activity. Albeit, it harbors some toxicities, its immense anticancer potential cannot be overlooked, as the cancer-specific delivery of CTD could help to rescue its lethal effects. Furthermore, several derivatives have been designed to weaken its toxicity. In light of extensive research, the antitumor activity of CTD is evident in both in vitro as well as in vivo cancer models. CTD has also proven efficacious in combination with chemotherapy and radiotherapy and it can also target some drug-resistant cancer cells. This mini-review endeavors to interpret and summarize recent information about CTD anticancer potential and underlying molecular mechanisms. The pertinent anticancer strength of CTD could be employed to develop an effective anticarcinogenic drug.
Collapse
Affiliation(s)
| | | | | | - Zhao Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (F.N.); (Y.W.); (N.Z.)
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (F.N.); (Y.W.); (N.Z.)
| |
Collapse
|
42
|
Montemagno C, Pagès G. Resistance to Anti-angiogenic Therapies: A Mechanism Depending on the Time of Exposure to the Drugs. Front Cell Dev Biol 2020; 8:584. [PMID: 32775327 PMCID: PMC7381352 DOI: 10.3389/fcell.2020.00584] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels from preexisting one, represents a critical process for oxygen and nutrient supply to proliferating cells, therefore promoting tumor growth and metastasis. The Vascular Endothelial Growth Factor (VEGF) pathway is one of the key mediators of angiogenesis in cancer. Therefore, several therapies including monoclonal antibodies or tyrosine kinase inhibitors target this axis. Although preclinical studies demonstrated strong antitumor activity, clinical studies were disappointing. Antiangiogenic drugs, used to treat metastatic patients suffering of different types of cancers, prolonged survival to different extents but are not curative. In this review, we focused on different mechanisms involved in resistance to antiangiogenic therapies from early stage resistance involving mainly tumor cells to late stages related to the adaptation of the microenvironment.
Collapse
Affiliation(s)
- Christopher Montemagno
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco.,CNRS UMR 7284, Institute for Research on Cancer and Aging of Nice, Université Côte d'Azur, Nice, France.,INSERM U1081, Centre Antoine Lacassagne, Nice, France
| | - Gilles Pagès
- Département de Biologie Médicale, Centre Scientifique de Monaco, Monaco, Monaco.,CNRS UMR 7284, Institute for Research on Cancer and Aging of Nice, Université Côte d'Azur, Nice, France.,INSERM U1081, Centre Antoine Lacassagne, Nice, France
| |
Collapse
|
43
|
Jia Y, Wen X, Gong Y, Wang X. Current scenario of indole derivatives with potential anti-drug-resistant cancer activity. Eur J Med Chem 2020; 200:112359. [PMID: 32531682 DOI: 10.1016/j.ejmech.2020.112359] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Abstract
Cancer chemotherapy is frequently hampered by drug resistance, so the resistance to anticancer agents represents one of the major obstacles for the effective cancer treatment. Indole derivatives have the potential to act on diverse targets in cancer cells and exhibit promising activity against drug-resistant cancers. Moreover, some indole-containing compounds such as Semaxanib, Sunitinib, Vinorelbine, and Vinblastine have already been applied in clinics for various kinds of cancer even drug-resistant cancer therapy. Thus, indole derivatives are one of significant resources for the development of novel anti-drug-resistant cancer agents. This review focuses on the recent development of indole derivatives with potential therapeutic application for drug-resistant cancers, and the mechanisms of action, the critical aspects of design as well as structure-activity relationships, covering articles published from 2010 to 2020.
Collapse
Affiliation(s)
- Yanshu Jia
- Chongqing Institute of Engineering, Chongqing, 400056, China
| | - Xiaoyue Wen
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang, Hubei, 443000, China
| | - Yufeng Gong
- The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Xuefeng Wang
- Department of Surgery, Zhuji Affiliated Hospital of Shaoxing University, Zhejiang Province, 311800, China.
| |
Collapse
|
44
|
Bortezomib Treatment Modulates Autophagy in Multiple Myeloma. J Clin Med 2020; 9:jcm9020552. [PMID: 32085480 PMCID: PMC7073518 DOI: 10.3390/jcm9020552] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
Although the introduction of bortezomib as a therapeutic strategy has improved the overall survival of multiple myeloma (MM) patients, 15–20% of high-risk patients do not respond to bortezomib over time or become resistant to treatment. Therefore, the development of new therapeutic strategies, such as combination therapies, is urgently needed. Methods: Given that bortezomib resistance may be mediated by activation of the autophagy pathway as an alternative mechanism of protein degradation, and that an enormous amounts of misfolded protein is generated in myeloma plasma cells (PCs), we investigated the effect of the simultaneous inhibition of proteasome by bortezomib and autophagy by hydroxychloroquine (HCQ) treatment on PCs and endothelial cells (ECs) isolated from patients with monoclonal gammopathy of undetermined significance (MGUS) and MM. Results: We found that bortezomib combined with HCQ induces synergistic cytotoxicity in myeloma PCs whereas this effect is lost on ECs. Levels of microtubule-associated protein light chain beta (LC3B) and p62 are differentially modulated in PCs and ECs, with effects on cell viability and proliferation. Conclusions: Our results suggest that treatment with bortezomib and HCQ should be associated with an anti-angiogenic drug to prevent the pro-angiogenic effect of bortezomib, the proliferation of a small residual tumor PC clone, and thus the relapse.
Collapse
|
45
|
Generation of endoplasmic reticulum stress and inhibition of autophagy by plitidepsin induces proteotoxic apoptosis in cancer cells. Biochem Pharmacol 2020; 172:113744. [DOI: 10.1016/j.bcp.2019.113744] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022]
|
46
|
Liu J, Zhang K, Cheng L, Zhu H, Xu T. Progress in Understanding the Molecular Mechanisms Underlying the Antitumour Effects of Ivermectin. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:285-296. [PMID: 32021111 PMCID: PMC6982461 DOI: 10.2147/dddt.s237393] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022]
Abstract
Ivermectin, a dihydro derivative of avermectin (AVM), was introduced into the veterinary, agricultural and aquaculture markets for animal health in 1981. Ivermectin was soon adopted in 1987 as a human medicine that was originally used for the treatment of onchocerciasis, a parasitic infection. Since then, ivermectin has also been used to control other human diseases and has exerted a significant effect on human health and welfare. In the past decade, many published studies have attempted to determine the role of ivermectin in cancer. In this review, we summarize the published studies to define the current progress in the characterization of ivermectin. Ivermectin causes cell death in cancer cell lines by inducing PAK1-mediated cytostatic autophagy, caspase-dependent apoptosis and immunogenic cell death (ICD) through the modulation of some pathways, including the WNT-T cell factor (TCF), Hippo and Akt/mTOR pathways. Ivermectin can affect the growth and proliferation of cancer cells and plays several different roles, such as its functions as an RNA helicase, a small-molecule mimetic of the surface-induced dissociation (SID) peptide, an activator of chloride channel receptors, and an inducer of mitochondrial dysfunction and oxidative stress. In addition, ivermectin induces the multidrug resistance protein (MDR), has potent anti-mitotic activity, targets angiogenesis and inhibits cancer stem-like cells (CSCs). Many studies have proven that ivermectin exerts antitumour effects and might thus benefit patients with cancer after sufficient clinical trials.
Collapse
Affiliation(s)
- Jian Liu
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| | - Kun Zhang
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| | - Lin Cheng
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| | - He Zhu
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| | - Tianmin Xu
- Department of Obstetrics and Gynecology, Jilin University Second Hospital, ChangChun 130041, People's Republic of China
| |
Collapse
|
47
|
Mechanisms of Resistance to Anti-CD38 Daratumumab in Multiple Myeloma. Cells 2020; 9:cells9010167. [PMID: 31936617 PMCID: PMC7017193 DOI: 10.3390/cells9010167] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023] Open
Abstract
Daratumumab (Dara) is the first-in-class human-specific anti-CD38 mAb approved for the treatment of multiple myeloma (MM). Although recent data have demonstrated very promising results in clinical practice and trials, some patients do not achieve a partial response, and ultimately all patients undergo progression. Dara exerts anti-MM activity via antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), complement-dependent cytotoxicity (CDC), and immunomodulatory effects. Deregulation of these pleiotropic mechanisms may cause development of Dara resistance. Knowledge of this resistance may improve the therapeutic management of MM patients.
Collapse
|
48
|
Qi Y, Qi W, Liu S, Sun L, Ding A, Yu G, Li H, Wang Y, Qiu W, Lv J. TSPAN9 suppresses the chemosensitivity of gastric cancer to 5-fluorouracil by promoting autophagy. Cancer Cell Int 2020; 20:4. [PMID: 31911756 PMCID: PMC6942356 DOI: 10.1186/s12935-019-1089-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 12/23/2019] [Indexed: 02/11/2023] Open
Abstract
Background The issue of drug resistance in gastric cancer has attracted global attention. TSPAN9, a 4-transmembrane protein that plays an important role in tumor progression and signal transduction, has been found to be closely related to tumor invasion, metastasis, and autophagy. Methods Immunoblotting was used to evaluate TSPAN9 expression in parental and drug-resistant gastric cancer cells. Functional assays, such as the CCK-8 assay, were used to detect the proliferation of gastric cancer cells and the response of TSPAN9 to 5-fluorouracil (5-FU). Western blotting was used to analyze the expression of constituents of the PI3K/AKT/mTOR-mediated autophagy pathway induced by TSPAN9. Coimmunoprecipitation was performed to assess the specific mechanism by which TSPAN9 affects the PI3K pathway. Results We demonstrated that TSPAN9 is overexpressed in 5-FU-resistant cells compared to parental cells. 5-FU-mediated inhibition of cell proliferation can be significantly restored by increasing TSPAN9 expression, and inhibiting this expression in drug-resistant cells can restore the sensitivity of the cells to 5-FU. In addition, TSPAN9 also significantly promoted autophagy in gastric cancer cells in vitro. Further studies indicated that TSPAN9 downregulates the expression of PI3K and proteins associated with PI3K-mediated autophagy. In addition, TSPAN9 interacts with PI3K and inhibits its catalytic activity. Conclusion The current study reveals the important role of TSPAN9 in drug resistance to 5-FU in gastric cancer. It also provides a new target to clinically address drug-resistant gastric cancer and will contribute to the treatment strategy of this disease.
Collapse
Affiliation(s)
- Yaoyue Qi
- 1Qingdao University, Qingdao, Shandong China
| | - Weiwei Qi
- 2Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China.,3Key Laboratory of Cancer Molecular and Translational Research, Qingdao University, Qingdao, Shandong China
| | - Shihai Liu
- 4Central Laboratory, Affiliated Hospital of Qingdao University, Qingdao, Shandong China
| | - Libin Sun
- 2Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China.,3Key Laboratory of Cancer Molecular and Translational Research, Qingdao University, Qingdao, Shandong China
| | - Aiping Ding
- 2Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China.,3Key Laboratory of Cancer Molecular and Translational Research, Qingdao University, Qingdao, Shandong China
| | - Guohong Yu
- 2Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China.,3Key Laboratory of Cancer Molecular and Translational Research, Qingdao University, Qingdao, Shandong China
| | - Hui Li
- 1Qingdao University, Qingdao, Shandong China
| | - Yixuan Wang
- 1Qingdao University, Qingdao, Shandong China
| | - Wensheng Qiu
- 2Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China.,3Key Laboratory of Cancer Molecular and Translational Research, Qingdao University, Qingdao, Shandong China
| | - Jing Lv
- 2Department of Oncology, Affiliated Hospital of Qingdao University, Qingdao, Shandong China.,3Key Laboratory of Cancer Molecular and Translational Research, Qingdao University, Qingdao, Shandong China
| |
Collapse
|
49
|
Saavedra-García P, Martini F, Auner HW. Proteasome inhibition in multiple myeloma: lessons for other cancers. Am J Physiol Cell Physiol 2019; 318:C451-C462. [PMID: 31875696 DOI: 10.1152/ajpcell.00286.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cellular protein homeostasis (proteostasis) depends on the controlled degradation of proteins that are damaged or no longer required by the ubiquitin-proteasome system (UPS). The 26S proteasome is the principal executer of substrate-specific proteolysis in eukaryotic cells and regulates a myriad of cellular functions. Proteasome inhibitors were initially developed as chemical tools to study proteasomal function but rapidly became widely used anticancer drugs that are now used at all stages of treatment for the bone marrow cancer multiple myeloma (MM). Here, we review the mechanisms of action of proteasome inhibitors that underlie their preferential toxicity to MM cells, focusing on endoplasmic reticulum stress, depletion of amino acids, and effects on glucose and lipid metabolism. We also discuss mechanisms of resistance to proteasome inhibition such as autophagy and metabolic rewiring and what lessons we may learn from the success and failure of proteasome inhibition in MM for treating other cancers with proteostasis-targeting drugs.
Collapse
Affiliation(s)
- Paula Saavedra-García
- Cancer Cell Metabolism Group, Hugh and Josseline Langmuir Centre for Myeloma Research, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Francesca Martini
- Department of Translational Research on New Technologies in Medicine and Surgery, Hematology Unit, Ospedale Santa Chiara, Pisa, Italy
| | - Holger W Auner
- Cancer Cell Metabolism Group, Hugh and Josseline Langmuir Centre for Myeloma Research, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
50
|
Li N, Zhan X. Mitochondrial Dysfunction Pathway Networks and Mitochondrial Dynamics in the Pathogenesis of Pituitary Adenomas. Front Endocrinol (Lausanne) 2019; 10:690. [PMID: 31649621 PMCID: PMC6794370 DOI: 10.3389/fendo.2019.00690] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 09/23/2019] [Indexed: 12/17/2022] Open
Abstract
Mitochondrion is a multi-functional organelle, which is associated with various signaling pathway networks, including energy metabolism, oxidative stress, cell apoptosis, cell cycles, autophagy, and immunity process. Mitochondrial proteins have been discovered to modulate these signaling pathway networks, and multiple biological behaviors to adapt to various internal environments or signaling events of human pathogenesis. Accordingly, mitochondrial dysfunction that alters the bioenergetic and biosynthetic state might contribute to multiple diseases, including cell transformation and tumor. Multiomics studies have revealed that mitochondrial dysfunction, oxidative stress, and cell cycle dysregulation signaling pathways operate in human pituitary adenomas, which suggest mitochondria play critical roles in pituitary adenomas. Some drugs targeting mitochondria are found as a therapeutic strategy for pituitary adenomas, including melatonin, melatonin inhibitors, temozolomide, pyrimethamine, 18 beta-glycyrrhetinic acid, gossypol acetate, Yougui pill, T-2 toxin, grifolic acid, cyclosporine A, dopamine agonists, and paeoniflorin. This article reviews the latest experimental evidence and potential biological roles of mitochondrial dysfunction and mitochondrial dynamics in pituitary adenoma progression, potential molecular mechanisms between mitochondria and pituitary adenoma progression, and current status and perspectives of mitochondria-based biomarkers and targeted drugs for effective management of pituitary adenomas.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha, China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|