1
|
Geng Q, Jiang Z, Byun WS, Donovan KA, Zhuang Z, Jiang F, Jones HM, Razumkov H, Tang MT, Sarott RC, Fischer ES, Corsello SM, Hinshaw SM, Gray NS. Development of Potent and Selective CK1α Molecular Glue Degraders. J Med Chem 2025; 68:3180-3196. [PMID: 39873536 DOI: 10.1021/acs.jmedchem.4c02415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Molecular glue degraders (MGDs) are small molecules that facilitate proximity between a target protein and an E3 ubiquitin ligase, thereby inducing target protein degradation. Glutarimide-containing compounds are MGDs that bind cereblon (CRBN) and recruit neosubstrates. Through explorative synthesis of a glutarimide-based library, we discovered a series of molecules that induce casein kinase 1 alpha (CK1α) degradation. By scaffold hopping and rational modification of the chemical scaffold, we identified an imidazo[1,2-a]pyrimidine compound that induces potent and selective CK1α degradation. A structure-activity relationship study of the lead compound, QXG-6442, identified the chemical features that contribute to degradation potency and selectivity compared to other frequently observed neosubstrates. The glutarimide library screening and structure-activity relationship medicinal chemistry approach we employed is generally useful for developing new molecular glue degraders toward new targets of interest.
Collapse
Affiliation(s)
- Qixiang Geng
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
| | - Zixuan Jiang
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
- Department of Chemistry, Stanford School of Humanities and Sciences and Department of Chemical and Systems Biology, ChEM-H, Stanford School of Medicine, Stanford University, Stanford, California 94305-6104, United States
| | - Woong Sub Byun
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
| | - Katherine A Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215 United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Zhe Zhuang
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
| | - Fen Jiang
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
| | - Hannah M Jones
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
| | - Hlib Razumkov
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
- Department of Chemistry, Stanford School of Humanities and Sciences and Department of Chemical and Systems Biology, ChEM-H, Stanford School of Medicine, Stanford University, Stanford, California 94305-6104, United States
| | - Michelle T Tang
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
| | - Roman C Sarott
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
| | - Eric S Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215 United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Steven M Corsello
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Department of Medicine, Stanford School of Medicine, Stanford, California 94305-6104, United States
| | - Stephen M Hinshaw
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
| | - Nathanael S Gray
- Department of Chemical and Systems Biology, Stanford Cancer Institute, School of Medicine Stanford University, Stanford, California 94305-6104, United States
- Sarafan ChEM-H, Stanford, California 94305-6104, United States
| |
Collapse
|
2
|
Tkachenko A, Havranek O. Erythronecroptosis: an overview of necroptosis or programmed necrosis in red blood cells. Mol Cell Biochem 2024; 479:3273-3291. [PMID: 38427167 DOI: 10.1007/s11010-024-04948-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/20/2024] [Indexed: 03/02/2024]
Abstract
Necroptosis is considered a programmed necrosis that requires receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3), and pore-forming mixed lineage kinase domain-like protein (MLKL) to trigger a regulated cell membrane lysis. Membrane rupture in necroptosis has been shown to fuel innate immune response due to release of damage-associated molecular patterns (DAMPs). Recently published studies indicate that mature erythrocytes can undergo necroptosis as well. In this review, we provide an outline of multiple cell death modes occurring in erythrocytes, discuss possible immunological aspects of diverse erythrocyte cell deaths, summarize available evidence related to the ability of erythrocytes to undergo necroptosis, outline key involved molecular mechanisms, and discuss the potential implication of erythrocyte necroptosis in the physiology and pathophysiology. Furthermore, we aim to highlight the interplay between necroptosis and eryptosis signaling in erythrocytes, emphasizing specific characteristics of these pathways distinct from their counterparts in nucleated cells. Thus, our review provides a comprehensive summary of the current knowledge of necroptosis in erythrocytes. To reflect critical differences between necroptosis of nucleated cells and necroptosis of erythrocytes, we suggest a term erythronecroptosis for necroptosis of enucleated cells.
Collapse
Affiliation(s)
- Anton Tkachenko
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250, Vestec, Czech Republic.
| | - Ondrej Havranek
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 25250, Vestec, Czech Republic
- First Department of Internal Medicine-Hematology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
3
|
Huang L, Chen L, Chen L, Peng B, Zhou L, Sun Y, Shi T, Lu J, Lin W, Liu Y, Cao L, Li L, Han Q, Chen X, Yang P, Zhang S, Wang Z, Yang J, Guo Z, Jiang B, Lu W. Development of Oral, Potent, and Selective CK1α Degraders for AML Therapy. JACS AU 2024; 4:4423-4434. [PMID: 39610741 PMCID: PMC11600170 DOI: 10.1021/jacsau.4c00762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024]
Abstract
Molecular glue degraders (MGDs) are proximity-inducing agents that mediate the cooperative interaction between a target protein and an E3 ligase, introducing an additional layer of specificity beyond that afforded by traditional small molecules. Historically, molecular glues that stabilize protein-protein interactions were often discovered serendipitously. In this study, we leveraged the reprogramming potential of cereblon (CRBN)-based ligands and conducted a CRBN-dependent proliferation screen to identify CRBN-based MGDs capable of inducing the degradation of proteins essential for cell viability. Through our screening and subsequent medicinal chemistry optimization, we identified dCK1α-1 as a potent and selective CK1α-targeting molecular glue degrader. Furthermore, we synthesized an orally active derivative, dCK1α-2, with enhanced pharmacokinetic properties, which exhibited pronounced degradation activity and demonstrated efficacy in mouse models following oral gavage. These findings indicate that phenotypic drug discovery campaigns, in combination with chemically distinct CRBN ligand libraries, can accelerate the development of therapeutically relevant MGDs. Furthermore, the development of dCK1α-1 and dCK1α-2 provides new therapeutic options for cancers with functional p53 signaling and offers valuable chemical tools for future investigations into the role of CK1α.
Collapse
Affiliation(s)
- Lu Huang
- Department
of Cardiovascular Surgery, The First Affiliated
Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230022, China
- Lingang
Laboratory, Shanghai 200031, China
| | - Lu Chen
- Department
of Radiation and Medical Oncology, Medical Research Institute, Frontier
Science Center of Immunology and Metabolism, Zhongnan Hospital of
Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Lu Chen
- Lingang
Laboratory, Shanghai 200031, China
| | - Bo Peng
- Lingang
Laboratory, Shanghai 200031, China
| | - Lixin Zhou
- Department
of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell
Research, Shanghai Key Laboratory of Signaling and Disease Research,
School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yanli Sun
- Lingang
Laboratory, Shanghai 200031, China
| | - Taiting Shi
- Lingang
Laboratory, Shanghai 200031, China
- Department
of Neurosurgery, The First Affiliated Hospital
of Wenzhou Medical University, Wenzhou 325000, China
| | - Jiayi Lu
- College
of
Science, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Weiye Lin
- Lingang
Laboratory, Shanghai 200031, China
- School
of Life Science and Technology, ShanghaiTech
University, 393 Middle
Huaxia Road, Shanghai 201210, China
| | - Yuhang Liu
- Lingang
Laboratory, Shanghai 200031, China
- School
of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation
(Yantai University), Ministry of Education; Collaborative Innovation
Center of Advanced Drug Delivery System and Biotech Drugs in Universities
of Shandong, Yantai University, Yantai 264005, China
| | - Linhui Cao
- Lingang
Laboratory, Shanghai 200031, China
- School
of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation
(Yantai University), Ministry of Education; Collaborative Innovation
Center of Advanced Drug Delivery System and Biotech Drugs in Universities
of Shandong, Yantai University, Yantai 264005, China
| | - Lanlan Li
- Lingang
Laboratory, Shanghai 200031, China
- Institute
of Entomology, The Provincial Special Key Laboratory for Development
and Utilization of Insect Resources, Guizhou
University, Guiyang 550025, China
| | | | - Xi Chen
- SpecAlly
Life Technology Co., Ltd., Wuhan 430075, China
| | - Ping Yang
- Department
of Cardiovascular Surgery, The First Affiliated
Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230022, China
| | - Shuo Zhang
- Department
of Cardiovascular Surgery, The First Affiliated
Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230022, China
| | - Zhe Wang
- Department
of Cardiovascular Surgery, The First Affiliated
Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230022, China
| | - Jing Yang
- Department
of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell
Research, Shanghai Key Laboratory of Signaling and Disease Research,
School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Zhixiang Guo
- Department
of Cardiovascular Surgery, The First Affiliated
Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230022, China
| | - Baishan Jiang
- Department
of Radiation and Medical Oncology, Medical Research Institute, Frontier
Science Center of Immunology and Metabolism, Zhongnan Hospital of
Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Wenchao Lu
- Lingang
Laboratory, Shanghai 200031, China
| |
Collapse
|
4
|
Zhong G, Chang X, Xie W, Zhou X. Targeted protein degradation: advances in drug discovery and clinical practice. Signal Transduct Target Ther 2024; 9:308. [PMID: 39500878 PMCID: PMC11539257 DOI: 10.1038/s41392-024-02004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/19/2024] [Accepted: 09/28/2024] [Indexed: 11/08/2024] Open
Abstract
Targeted protein degradation (TPD) represents a revolutionary therapeutic strategy in disease management, providing a stark contrast to traditional therapeutic approaches like small molecule inhibitors that primarily focus on inhibiting protein function. This advanced technology capitalizes on the cell's intrinsic proteolytic systems, including the proteasome and lysosomal pathways, to selectively eliminate disease-causing proteins. TPD not only enhances the efficacy of treatments but also expands the scope of protein degradation applications. Despite its considerable potential, TPD faces challenges related to the properties of the drugs and their rational design. This review thoroughly explores the mechanisms and clinical advancements of TPD, from its initial conceptualization to practical implementation, with a particular focus on proteolysis-targeting chimeras and molecular glues. In addition, the review delves into emerging technologies and methodologies aimed at addressing these challenges and enhancing therapeutic efficacy. We also discuss the significant clinical trials and highlight the promising therapeutic outcomes associated with TPD drugs, illustrating their potential to transform the treatment landscape. Furthermore, the review considers the benefits of combining TPD with other therapies to enhance overall treatment effectiveness and overcome drug resistance. The future directions of TPD applications are also explored, presenting an optimistic perspective on further innovations. By offering a comprehensive overview of the current innovations and the challenges faced, this review assesses the transformative potential of TPD in revolutionizing drug development and disease management, setting the stage for a new era in medical therapy.
Collapse
Affiliation(s)
- Guangcai Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaoyu Chang
- School of Pharmaceutical Sciences, Pingyuan Laboratory, Zhengzhou University, Zhengzhou, 450001, China
| | - Weilin Xie
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
5
|
Cunningham JL, Frankovich J, Dubin RA, Pedrosa E, Baykara RN, Schlenk NC, Maqbool SB, Dolstra H, Marino J, Edinger J, Shea JM, Laje G, Swagemakers SMA, Sinnadurai S, Zhang ZD, Lin JR, van der Spek PJ, Lachman HM. Ultrarare Variants in DNA Damage Repair Genes in Pediatric Acute-Onset Neuropsychiatric Syndrome or Acute Behavioral Regression in Neurodevelopmental Disorders. Dev Neurosci 2024:1-20. [PMID: 39396515 DOI: 10.1159/000541908] [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: 05/21/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
INTRODUCTION Acute onset of severe psychiatric symptoms or regression may occur in children with premorbid neurodevelopmental disorders, although typically developing children can also be affected. Infections or other stressors are likely triggers. The underlying causes are unclear, but a current hypothesis suggests the convergence of genes that influence neuronal and immunological function. We previously identified 11 genes in pediatric acute-onset neuropsychiatric syndrome (PANS), in which two classes of genes related to either synaptic function or the immune system were found. Among the latter, three affect the DNA damage response (DDR): PPM1D, CHK2, and RAG1. We now report an additional 17 cases with mutations in PPM1D and other DDR genes in patients with acute onset of psychiatric symptoms and/or regression that their clinicians classified as PANS or another inflammatory brain condition. METHODS We analyzed genetic findings obtained from parents and carried out whole-exome sequencing on a total of 17 cases, which included 3 sibling pairs and a family with 4 affected children. RESULTS The DDR genes include clusters affecting p53 DNA repair (PPM1D, ATM, ATR, 53BP1, and RMRP), and the Fanconi Anemia Complex (FANCE, SLX4/FANCP, FANCA, FANCI, and FANCC). We hypothesize that defects in DNA repair genes, in the context of infection or other stressors, could contribute to decompensated states through an increase in genomic instability with a concomitant accumulation of cytosolic DNA in immune cells triggering DNA sensors, such as cGAS-STING and AIM2 inflammasomes, as well as central deficits on neuroplasticity. In addition, increased senescence and defective apoptosis affecting immunological responses could be playing a role. CONCLUSION These compelling preliminary findings motivate further genetic and functional characterization as the downstream impact of DDR deficits may point to novel treatment strategies.
Collapse
Affiliation(s)
- Janet L Cunningham
- Department of Medical Sciences, Psychiatry, Uppsala University, Uppsala, Sweden
| | - Jennifer Frankovich
- Department of Pediatrics, Division of Pediatric Allergy, Immunology, Rheumatology and Immune Behavioral Health Program, Stanford Children's Health and Stanford University School of Medicine, Palo Alto, California, USA
| | - Robert A Dubin
- Center for Epigenomics, Computational Genomics Core, Albert Einstein College of Medicine, New York, New York, USA
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York, USA
| | - Refia Nur Baykara
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York, USA
| | - Noelle Cathleen Schlenk
- Stanford Children's Health, PANS Clinic and Research Program, Stanford University School of Medicine, Stanford, California, USA
| | - Shahina B Maqbool
- Department of Genetics Epigenetics Shared Facility, Albert Einstein College of Medicine, New York, New York, USA
| | - Hedwig Dolstra
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York, USA
| | - Jacqueline Marino
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York, USA
| | - Jacob Edinger
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York, USA
| | - Julia M Shea
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, New York, New York, USA
| | - Gonzalo Laje
- Department of Psychiatry, Permian Basin, Texas Tech University Health Sciences Center School of Medicine, Lubbock, Texas, USA
| | - Sigrid M A Swagemakers
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, Rotterdam, The Netherlands
| | - Siamala Sinnadurai
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, Rotterdam, The Netherlands
- Department of Epidemiology and Health Promotion at the School of Public Health Medical Center for Postgraduate Education, Warsaw, Poland
| | - Zhengdong D Zhang
- Department of Genetics, Albert Einstein College of Medicine, New York, New York, USA
| | - Jhih-Rong Lin
- Department of Genetics, Albert Einstein College of Medicine, New York, New York, USA
| | - Peter J van der Spek
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, Rotterdam, The Netherlands
| | - Herbert M Lachman
- Department of Psychiatry and Behavioral Sciences, Department of Medicine, Department of Genetics, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
6
|
Ayoubi R, Alende C, Fotouhi M, Ruíz Moleón V, González Bolívar S, Southern K, Laflamme C, NeuroSGC/YCharOS/EDDU collaborative group, ABIF consortium. A guide to selecting high-performing antibodies for CSNK1A1 (UniProt ID: P48729) for use in western blot, immunoprecipitation, and immunofluorescence. F1000Res 2024; 13:1055. [PMID: 40028451 PMCID: PMC11868743 DOI: 10.12688/f1000research.155928.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 03/05/2025] Open
Abstract
CSNK1A1 is a key regulator of various signalling pathways, including the Wnt/β-catenin pathway. Playing a central role in cellular function and disease pathology, CSNK1A1 has emerged as an attractive protein target for therapeutic development. In this study we characterize ten CSNK1A1 commercial antibodies for western blot, immunoprecipitation, and immunofluorescence using a standardized experimental protocol based on comparing read-outs in knockout cell lines and isogenic parental controls. This study is part of a larger, collaborative initiative seeking to address antibody reproducibility issues by characterizing commercially available antibodies for human proteins and publishing the results openly as a resource for the scientific community. While the use of antibodies and protocols vary between laboratories, we encourage readers to use this report as a guide to select the most appropriate antibodies for their specific needs.
Collapse
Affiliation(s)
- Riham Ayoubi
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Charles Alende
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Maryam Fotouhi
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Vera Ruíz Moleón
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Sara González Bolívar
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Kathleen Southern
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Carl Laflamme
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | | | - ABIF consortium
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| |
Collapse
|
7
|
Abstract
Eryptosis is a regulated cell death (RCD) of mature erythrocytes initially described as a counterpart of apoptosis for enucleated cells. However, over the recent years, a growing number of studies have emphasized certain differences between both cell death modalities. In this review paper, we underline the hallmarks of eryptosis and apoptosis and highlight resemblances and dissimilarities between both RCDs. We summarize and critically discuss differences in the impact of caspase-3, Ca2+ signaling, ROS signaling pathways, opposing roles of casein kinase 1α, protein kinase C, Janus kinase 3, cyclin-dependent kinase 4, and AMP-activated protein kinase to highlight a certain degree of divergence between apoptosis and eryptosis. This review emphasizes the crucial importance of further studies that focus on deepening our knowledge of cell death machinery and identifying novel differences between cell death of nucleated and enucleated cells. This might provide evidence that erythrocytes can be defined as viable entities capable of programmed cell destruction. Additionally, the revealed cell type-specific patterns in cell death can facilitate the development of cell death-modulating therapeutic agents.
Collapse
Affiliation(s)
- Anton Tkachenko
- 1st Faculty of Medicine, BIOCEV, Charles University, Průmyslová 595, 25250, Vestec, Czech Republic.
| |
Collapse
|
8
|
Gao Y, Wu Z, Chen Y, Shang G, Zeng Y, Gao Y. A global bibliometric and visualized analysis of the links between the autophagy and acute myeloid leukemia. Front Pharmacol 2024; 14:1291195. [PMID: 38322702 PMCID: PMC10844427 DOI: 10.3389/fphar.2023.1291195] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/31/2023] [Indexed: 02/08/2024] Open
Abstract
Background and objectives: Autophagy is a cellular process where damaged organelles or unwanted proteins are packaged into a double-membrane structure and transported to lysosomes for degradation. Autophagy plays a regulatory role in various hematologic malignancies, including acute myeloid leukemia (AML). However, there are few bibliometric studies on the role of autophagy in AML. The purpose of this study is to clarify the role of autophagy in acute myeloid leukemia through bibliometric analysis. Methods: The literature on autophagy and AML research from 2003 to 2023 was searched in Web of Science Core Collection, and bibliometric tools such as VOSviewer 1.6.18, Cite Space (6.1.R3), RStudio (R package bibliometrix), and Scimago Graphica were used to understand the current status and hotspots of autophagy and AML research. The study conducted an analysis of various dimensions including the quantity of publications, countries, institutions, journals, authors, co-references, keywords, and to predict future development trends in this field by drawing relevant visualization maps. Results: A total of 343 articles were obtained, published in 169 journals, written by 2,323 authors from 295 institutions in 43 countries. The journals with the most publications were Blood and Oncotarget. China had the most publications, and Chongqing Medical University and Sun Yat-sen University had the most publications. The author with the highest number of publications was Tschan, Mario P. The main types of research included clinical research, in vitro experiments, in vivo experiments, public database information, and reviews, and the forms of therapeutic effects mainly focused on genetic regulation, traditional Chinese medicine combination, autophagy inhibitors, and drug targets. The research hotspots of autophagy and AML in the past 17 years have focused on genetic regulation, autophagy inhibition, and targeted drugs. Chemotherapy resistance and mitochondrial autophagy will be the forefront of research. Conclusion: The gradual increase in the literature on autophagy and AML research and the decline after 2022 could be a result of authors focusing more on the type of research and the quality of the literature. The current research hotspots are mainly genetic regulation, autophagy inhibition, and autophagy-related targeted drugs. In future, autophagy will remain the focus of the AML field, with research trends likely to focus more on AML chemotherapy resistance and mitochondrial autophagy.
Collapse
Affiliation(s)
- Yao Gao
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Zhenhui Wu
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, China
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yingfan Chen
- Department of Traditional Chinese Medicine, Sixth Medical Center, PLA General Hospital, Beijing, China
| | - Guangbin Shang
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yingjian Zeng
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, China
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yue Gao
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, China
- Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
9
|
Nishiguchi G, Mascibroda LG, Young SM, Caine EA, Abdelhamed S, Kooijman JJ, Miller DJ, Das S, McGowan K, Mayasundari A, Shi Z, Barajas JM, Hiltenbrand R, Aggarwal A, Chang Y, Mishra V, Narina S, Thomas M, Loughran AJ, Kalathur R, Yu K, Zhou S, Wang X, High AA, Peng J, Pruett-Miller SM, Daniels DL, Urh M, Shelat AA, Mullighan CG, Riching KM, Zaman GJR, Fischer M, Klco JM, Rankovic Z. Selective CK1α degraders exert antiproliferative activity against a broad range of human cancer cell lines. Nat Commun 2024; 15:482. [PMID: 38228616 PMCID: PMC10791743 DOI: 10.1038/s41467-024-44698-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024] Open
Abstract
Molecular-glue degraders are small molecules that induce a specific interaction between an E3 ligase and a target protein, resulting in the target proteolysis. The discovery of molecular glue degraders currently relies mostly on screening approaches. Here, we describe screening of a library of cereblon (CRBN) ligands against a panel of patient-derived cancer cell lines, leading to the discovery of SJ7095, a potent degrader of CK1α, IKZF1 and IKZF3 proteins. Through a structure-informed exploration of structure activity relationship (SAR) around this small molecule we develop SJ3149, a selective and potent degrader of CK1α protein in vitro and in vivo. The structure of SJ3149 co-crystalized in complex with CK1α + CRBN + DDB1 provides a rationale for the improved degradation properties of this compound. In a panel of 115 cancer cell lines SJ3149 displays a broad antiproliferative activity profile, which shows statistically significant correlation with MDM2 inhibitor Nutlin-3a. These findings suggest potential utility of selective CK1α degraders for treatment of hematological cancers and solid tumors.
Collapse
Affiliation(s)
- Gisele Nishiguchi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Lauren G Mascibroda
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sarah M Young
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Elizabeth A Caine
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Sherif Abdelhamed
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | | | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sourav Das
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kevin McGowan
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anand Mayasundari
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Zhe Shi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Juan M Barajas
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Ryan Hiltenbrand
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anup Aggarwal
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Yunchao Chang
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Vibhor Mishra
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Shilpa Narina
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Melvin Thomas
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Allister J Loughran
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Ravi Kalathur
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kaiwen Yu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Suiping Zhou
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anthony A High
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Memphis, TN, 38105, USA
| | - Danette L Daniels
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Marjeta Urh
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Anang A Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kristin M Riching
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Guido J R Zaman
- Oncolines B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Marcus Fischer
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
10
|
Huang Z, Zhou J, Jiang Y, Han Y, Wang X, Li F, Jiang S, Yu K, Zhang S. Combined inhibition of XIAP and autophagy induces apoptosis and differentiation in acute myeloid leukaemia. J Cell Mol Med 2023; 27:1682-1696. [PMID: 37154878 PMCID: PMC10273072 DOI: 10.1111/jcmm.17765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023] Open
Abstract
Perturbations in autophagy, apoptosis and differentiation have greatly affected the progression and therapy of acute myeloid leukaemia (AML). The role of X-linked inhibitor of apoptosis (XIAP)-related autophagy remains unclear in AML therapeutics. Here, we found that XIAP was highly expressed and associated with poor overall survival in patients with AML. Furthermore, pharmacologic inhibition of XIAP using birinapant or XIAP knockdown via siRNA impaired the proliferation and clonogenic capacity by inducing autophagy and apoptosis in AML cells. Intriguingly, birinapant-induced cell death was aggravated in combination with ATG5 siRNA or an autophagy inhibitor spautin-1, suggesting that autophagy may be a pro-survival signalling. Spautin-1 further enhanced the ROS level and myeloid differentiation in THP-1 cells treated with birinapant. The mechanism analysis showed that XIAP interacted with MDM2 and p53, and XIAP inhibition notably downregulated p53, substantially increased the AMPKα1 phosphorylation and downregulated the mTOR phosphorylation. Combined treatment using birinapant and chloroquine significantly retarded AML progression in both a subcutaneous xenograft model injected with HEL cells and an orthotopic xenograft model injected intravenously with C1498 cells. Collectively, our data suggested that XIAP inhibition can induce autophagy, apoptosis and differentiation, and combined inhibition of XIAP and autophagy may be a promising therapeutic strategy for AML.
Collapse
Affiliation(s)
- Ziyang Huang
- Department of HematologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
| | - Jifan Zhou
- Department of HematologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
| | - Yinyan Jiang
- Department of HematologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
| | - Yixiang Han
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
- Central LaboratoryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Xiaofang Wang
- Department of HematologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
| | - Fanfan Li
- Department of HematologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
| | - Songfu Jiang
- Department of HematologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
| | - Kang Yu
- Department of HematologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
| | - Shenghui Zhang
- Department of HematologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Institute of HematologyWenzhou Medical UniversityWenzhouZhejiangChina
- Wenzhou Key Laboratory of HematologyWenzhouZhejiangChina
- Laboratory Animal CenterThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
11
|
Han MH, Baek JM, Min KW, Cheong JH, Ryu JI, Won YD, Kwon MJ, Koh SH. DKK3 expression is associated with immunosuppression and poor prognosis in glioblastoma, in contrast to lower-grade gliomas. BMC Neurol 2023; 23:183. [PMID: 37149563 PMCID: PMC10163766 DOI: 10.1186/s12883-023-03236-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023] Open
Abstract
PURPOSE We previously reported that expression of dickkopf-3 (DKK3), which is involved in the Wnt/β-catenin pathway, is significantly associated with prognosis in patients with glioblastoma multiforme (GBM). The aim of this study was to compare the association of DKK3 with other Wnt/β-catenin pathway-related genes and immune responses between lower grade glioma (LGG) and GBM. METHODS We obtained the clinicopathological data of 515 patients with LGG (World Health Organization [WHO] grade II and III glioma) and 525 patients with GBM from the Cancer Genome Atlas (TCGA) database. We performed Pearson's correlation analysis to investigate the relationships between Wnt/β-catenin-related gene expression in LGG and GBM. Linear regression analysis was performed to identify the association between DKK3 expression and immune cell fractions in all grade II to IV gliomas. RESULTS A total of 1,040 patients with WHO grade II to IV gliomas were included in the study. As the grade of glioma increased, DKK3 showed a tendency to be more strongly positively correlated with the expression of other Wnt/β-catenin pathway-related genes. DKK3 was not associated with immunosuppression in LGG but was associated with downregulation of immune responses in GBM. We hypothesized that the role of DKK3 in the Wnt/β-catenin pathway might be different between LGG and GBM. CONCLUSION According to our findings, DKK3 expression had a weak effect on LGG but a significant effect on immunosuppression and poor prognosis in GBM. Therefore, DKK3 expression seems to play different roles, through the Wnt/β-catenin pathway, between LGG and GBM.
Collapse
Affiliation(s)
- Myung-Hoon Han
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Jeong Min Baek
- Department of Translational Medicine, Graduate School of Biomedical Science & Engineering, Hanyang University, Hanyang University, Seoul, South Korea
| | - Kyueng-Whan Min
- Department of Pathology Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Gyeonggi-do, Republic of Korea.
| | - Jin Hwan Cheong
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Je Il Ryu
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Yu Deok Won
- Department of Neurosurgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Gyeonggi-do, Republic of Korea
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Gyeonggi-do, Republic of Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, 11923 Gyeongchun-ro, Guri-Si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
12
|
Abstract
Eryptosis is a coordinated non-lytic cell death of erythrocytes characterized by cell shrinkage, cell membrane scrambling, Ca2+ influx, ceramide accumulation, oxidative stress, activation of calpain and caspases. Physiologically, it aims at removing damaged or aged erythrocytes from circulation. A plethora of diseases are associated with enhanced eryptosis, including metabolic diseases, cardiovascular pathology, renal and hepatic diseases, hematological disorders, systemic autoimmune pathology, and cancer. This makes eryptosis and eryptosis-regulating signaling pathways a target for therapeutic interventions. This review highlights the eryptotic signaling machinery containing several protein kinases and its small molecular inhibitors with a special emphasis on casein kinase 1α (CK1α), a serine/threonine protein kinase with a broad spectrum of activity. In this review article, we provide a critical analysis of the regulatory role of CK1α in eryptosis, highlight triggers of CK1α-mediated suicidal death of red blood cells, cover the knowledge gaps in understanding CK1α-driven eryptosis and discover the opportunity of CK1α-targeted pharmacological modulation of eryptosis. Moreover, we discuss the directions of future research focusing on uncovering crosstalks between CK1α and other eryptosis-regulating kinases and pathways.
Collapse
Affiliation(s)
- Anton Tkachenko
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, 4 Nauky ave, 61022, Kharkiv, Ukraine.
| | - Anatolii Onishchenko
- Research Institute of Experimental and Clinical Medicine, Kharkiv National Medical University, 4 Nauky ave, 61022, Kharkiv, Ukraine
| |
Collapse
|
13
|
Behrouj H, Mokarram P. BAMLET (Bovine α-lactalbumin made lethal to tumor cells) inhibits autophagy flux and induces apoptosis via down-regulation of protein kinase CK1α and attenuation of the AKT/p-ß-catenin (S552) pathway in RAS-mutated human colorectal HCT 116 cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:1212-1219. [PMID: 37736507 PMCID: PMC10510486 DOI: 10.22038/ijbms.2023.69343.15114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 06/06/2023] [Indexed: 09/23/2023]
Abstract
Objectives Oncogenic RAS mutations occur in nearly 50% of colorectal cancer cases and are usually dependent on the autophagy mechanism to maintain tumorigenesis. We have recently demonstrated that CK1α controls autophagy machinery possibly through the AKT/p-ß-catenin (S552) signaling in colorectal cancer cells harboring RAS mutation. It has been found that a lipid-protein complex comprising oleic acid binds to human α-lactalbumin, known as HAMLET (human α -lactalbumin made lethal to tumor cells), targets a broad range of kinases including CK1α. Therefore, this study was designed to investigate the effects of BAMLET (bovine α -lactalbumin made lethal to tumor cells, the bovine counterpart of HAMLET) on CK1α expression, AKT/Phospho-ß-catenin (S552) pathway, and autophagy flux in RAS-mutated human colorectal HCT 116 cells. Materials and Methods For this purpose, HCT116 cells were treated with BAMLET and casein kinase 1 inhibitor (D4476), and quantitative real-time polymerase chain reaction (RT-qPCR) and western blot analysis were used to measure the proteins and genes of the AKT/Phospho-ß-catenin (S552) pathway and autophagy. Apoptosis was measured by flow-cytometry. Results We found that BAMLET significantly reduced cell viability and decreased the expression of CK1α. Additionally, BAMLET inhibited autophagy flux and enhanced the ability of CK1α inhibitor D4476 to impair autophagy flux, which was accompanied by an increase in the apoptosis percentage. We also observed that BAMLET empowered D4476 to down-regulate the AKT/Phospho-ß-catenin (S552) axis. Conclusion BAMLET hampers autophagy flux and leads to apoptosis induction, possibly, by reducing the expression of CK1α and attenuation of the AKT/Phospho-ß-catenin (S552) axis.
Collapse
Affiliation(s)
- Hamid Behrouj
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Pooneh Mokarram
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
14
|
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
|
15
|
Lei H, Wang J, Hu J, Zhu Q, Wu Y. Deubiquitinases in hematological malignancies. Biomark Res 2021; 9:66. [PMID: 34454635 PMCID: PMC8401176 DOI: 10.1186/s40364-021-00320-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022] Open
Abstract
Deubiquitinases (DUBs) are enzymes that control the stability, interactions or localization of most cellular proteins by removing their ubiquitin modification. In recent years, some DUBs, such as USP7, USP9X and USP10, have been identified as promising therapeutic targets in hematological malignancies. Importantly, some potent inhibitors targeting the oncogenic DUBs have been developed, showing promising inhibitory efficacy in preclinical models, and some have even undergone clinical trials. Different DUBs perform distinct function in diverse hematological malignancies, such as oncogenic, tumor suppressor or context-dependent effects. Therefore, exploring the biological roles of DUBs and their downstream effectors will provide new insights and therapeutic targets for the occurrence and development of hematological malignancies. We summarize the DUBs involved in different categories of hematological malignancies including leukemia, multiple myeloma and lymphoma. We also present the recent development of DUB inhibitors and their applications in hematological malignancies. Together, we demonstrate DUBs as potential therapeutic drug targets in hematological malignancies.
Collapse
Affiliation(s)
- Hu Lei
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jiaqi Wang
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiacheng Hu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qian Zhu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingli Wu
- Department of Pathophysiology, International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
16
|
Spinello Z, Fregnani A, Quotti Tubi L, Trentin L, Piazza F, Manni S. Targeting Protein Kinases in Blood Cancer: Focusing on CK1α and CK2. Int J Mol Sci 2021; 22:ijms22073716. [PMID: 33918307 PMCID: PMC8038136 DOI: 10.3390/ijms22073716] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
Disturbance of protein kinase activity may result in dramatic consequences that often lead to cancer development and progression. In tumors of blood origin, both tyrosine kinases and serine/threonine kinases are altered by different types of mutations, critically regulating cancer hallmarks. CK1α and CK2 are highly conserved, ubiquitously expressed and constitutively active pleiotropic kinases, which participate in multiple biological processes. The involvement of these kinases in solid and blood cancers is well documented. CK1α and CK2 are overactive in multiple myeloma, leukemias and lymphomas. Intriguingly, they are not required to the same degree for the viability of normal cells, corroborating the idea of “druggable” kinases. Different to other kinases, mutations on the gene encoding CK1α and CK2 are rare or not reported. Actually, these two kinases are outside the paradigm of oncogene addiction, since cancer cells’ dependency on these proteins resembles the phenomenon of “non-oncogene” addiction. In this review, we will summarize the general features of CK1α and CK2 and the most relevant oncogenic and stress-related signaling nodes, regulated by kinase phosphorylation, that may lead to tumor progression. Finally, we will report the current data, which support the positioning of these two kinases in the therapeutic scene of hematological cancers.
Collapse
Affiliation(s)
- Zaira Spinello
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Anna Fregnani
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Laura Quotti Tubi
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Livio Trentin
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
- Correspondence: (F.P.); (S.M.); Tel.: +39-049-792-3263 (F.P. & S.M.); Fax: +39-049-792-3250 (F.P. & S.M.)
| | - Sabrina Manni
- Department of Medicine, Hematology Section, University of Padova, Via N. Giustiniani 2, 35128 Padova, Italy; (Z.S.); (A.F.); (L.Q.T.); (L.T.)
- Veneto Institute of Molecular Medicine, Via G. Orus 2, 35129 Padova, Italy
- Correspondence: (F.P.); (S.M.); Tel.: +39-049-792-3263 (F.P. & S.M.); Fax: +39-049-792-3250 (F.P. & S.M.)
| |
Collapse
|
17
|
Wang X, Xu Z, Cai Y, Zeng S, Peng B, Ren X, Yan Y, Gong Z. Rheostatic Balance of Circadian Rhythm and Autophagy in Metabolism and Disease. Front Cell Dev Biol 2020; 8:616434. [PMID: 33330516 PMCID: PMC7732583 DOI: 10.3389/fcell.2020.616434] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/04/2020] [Indexed: 02/05/2023] Open
Abstract
Circadian rhythms are physical, behavioral and environmental cycles that respond primarily to light and dark, with a period of time of approximately 24 h. The most essential physiological functions of mammals are manifested in circadian rhythm patterns, including the sleep-wake cycle and nutrient and energy metabolism. Autophagy is a conserved biological process contributing to nutrient and cellular homeostasis. The factors affecting autophagy are numerous, such as diet, drugs, and aging. Recent studies have indicated that autophagy is activated rhythmically in a clock-dependent manner whether the organism is healthy or has certain diseases. In addition, autophagy can affect circadian rhythm by degrading circadian proteins. This review discusses the interaction and mechanisms between autophagy and circadian rhythm. Moreover, we introduce the molecules influencing both autophagy and circadian rhythm. We then discuss the drugs affecting the circadian rhythm of autophagy. Finally, we present the role of rhythmic autophagy in nutrient and energy metabolism and its significance in physiology and metabolic disease.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cai
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Bi Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Xinxin Ren
- Key Laboratory of Molecular Radiation Oncology of Hunan Province, Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|