1
|
Tong Y, Wang X, Li R, Xu X, Dai M, Wang N, Fan B, Feng S, Ma T. LSD1 is a promising target to treat cancers by modulating cell stemness. Biochem Pharmacol 2024; 229:116549. [PMID: 39304105 DOI: 10.1016/j.bcp.2024.116549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
As the first discovered histone demethylase, LSD1 plays a vital role in maintaining pathological processes such as cancer, infection, and immune diseases. Based on previous researches, LSD1 is highly expressed in sorts of tumor cells such as acute myeloid leukemia, non-small cell lung cancer, prostate cancer, breast cancer and gastric cancer, etc. Therefore, targeting LSD1 is a prospective strategy for tumor treatment. Cancer stem cells could preserve self-renewal, cell proliferation, cell migration and malignant phenotype. So, the reduction of tumor cell stemness can effectively inhibit the growth of tumor cells, which may be a new strategy for the treatment of cancers. Up to now, there exist many researches confirming the significant role of LSD1 in regulating the stemness characteristics such as embryonic stem cells differentiation. Many reports show that inhibition of LSD1 effectively decreases the property of cancer cell stemness. However, there lacks a detailed review about the relationship between LSD1 and cancer cell stemness. Herein, in this review, we summarized the mechanisms how LSD1 regulates cell stemness comprehensively. In addition, some related inhibitors targeting LSD1 to reduce the proliferation characteristics of cancer stem cells are also described.
Collapse
Affiliation(s)
- Yaoyuan Tong
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Xiaoru Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ruonan Li
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Xiangyu Xu
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Mengge Dai
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Nan Wang
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China
| | - Boyi Fan
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Siqi Feng
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China.
| | - Ting Ma
- School of Pharmaceutical Sciences, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
2
|
Ghannad-Zadeh K, Ivanova A, Wu M, Wilson TM, Lau A, Flick R, Munoz DG, Das S. One-carbon-mediated purine synthesis underlies temozolomide resistance in glioblastoma. Cell Death Dis 2024; 15:774. [PMID: 39455562 PMCID: PMC11511812 DOI: 10.1038/s41419-024-07170-y] [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: 04/29/2024] [Revised: 09/30/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Glioblastoma accounts for nearly half of all primary malignant brain tumors in adults, and despite an aggressive standard of care, including excisional surgery and adjuvant chemoradiation, recurrence remains universal, with an overall median survival of 14.6 months. Recent work has revealed the importance of passenger mutations as critical mediators of metabolic adaptation in cancer progression. In our previous work, we identified a role for the epigenetic modifier ID-1 in temozolomide resistance in glioblastoma. Here, we show that ID-1-mediated glioblastoma tumourigenesis is accompanied by upregulation of one-carbon (1-C) mediated de novo purine synthesis. ID-1 knockout results in a significant reduction in the expression of 1-C metabolism and purine synthesis enzymes. Analysis of glioblastoma surgical specimens at initial presentation and recurrence reveals that 1-C purine synthesis metabolic enzymes are enriched in recurrent glioblastoma and that their expression correlates with a shorter time to tumor recurrence. Further, we show that the 1-C metabolic phenotype underlies proliferative capacity and temozolomide resistance in glioblastoma cells. Supplementation with exogenous purines restores proliferation in ID-1-deficient cells, while inhibition of purine synthesis with AICAR sensitizes temozolomide-resistant glioblastoma cells to temozolomide chemotherapy. Our data suggest that the metabolic phenotype observed in treatment-resistant glioma cells is a potential therapeutic target in glioblastoma.
Collapse
Affiliation(s)
- Kimia Ghannad-Zadeh
- The Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Alyona Ivanova
- The Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Megan Wu
- The Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, ON, Canada
| | - Taylor M Wilson
- The Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Alyssa Lau
- The Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, ON, Canada
| | - Robert Flick
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - David G Munoz
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sunit Das
- The Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, ON, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
- Neurosurgery Research Department, St. Michael's Hospital, Toronto Unity Health, Toronto, ON, Canada.
- Division of Neurosurgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Liu Y, Qu HQ, Chang X, Mentch FD, Qiu H, Nguyen K, Ostberg K, Wang T, Glessner J, Hakonarson H. Deciphering protective genomic factors of tumor development in pediatric down syndrome via deep learning approach to whole genome and RNA sequencing. Cancer Commun (Lond) 2024. [PMID: 39387321 DOI: 10.1002/cac2.12612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Affiliation(s)
- Yichuan Liu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hui-Qi Qu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Xiao Chang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Frank D Mentch
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Haijun Qiu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kenny Nguyen
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kayleigh Ostberg
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Tiancheng Wang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joseph Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
4
|
La HM, Chan AL, Hutchinson AM, Su BYM, Rossello FJ, Schittenhelm RB, Hobbs RM. Functionally redundant roles of ID family proteins in spermatogonial stem cells. Stem Cell Reports 2024; 19:1379-1388. [PMID: 39332405 DOI: 10.1016/j.stemcr.2024.08.011] [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: 02/13/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/29/2024] Open
Abstract
Spermatogonial stem cells (SSCs) are essential for sustained sperm production, but SSC regulatory mechanisms and markers remain poorly defined. Studies have suggested that the Id family transcriptional regulator Id4 is expressed in SSCs and involved in SSC maintenance. Here, we used reporter and knockout models to define the expression and function of Id4 in the adult male germline. Within the spermatogonial pool, Id4 reporter expression and inhibitor of DNA-binding 4 (ID4) protein are found throughout the GFRα1+ fraction, comprising the self-renewing population. However, Id4 deletion is tolerated by adult SSCs while revealing roles in meiotic spermatocytes. Cultures of undifferentiated spermatogonia could be established following Id4 deletion. Importantly, ID4 loss in undifferentiated spermatogonia triggers ID3 upregulation, and both ID proteins associate with transcription factor partner TCF3 in wild-type cells. Combined inhibition of IDs in cultured spermatogonia disrupts the stem cell state and blocks proliferation. Our data therefore demonstrate critical but functionally redundant roles of IDs in SSC function.
Collapse
Affiliation(s)
- Hue M La
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Melbourne, VIC 3800, Australia; University of Melbourne Centre for Cancer Research, University of Melbourne, Melbourne, VIC 3000, Australia; Department of Clinical Pathology, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Ai-Leen Chan
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Ashlee M Hutchinson
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Bianka Y M Su
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Fernando J Rossello
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC 3000, Australia; Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia; Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia; Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics & Metabolomics Platform, Monash Biomedicine Discovery Institute & Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Robin M Hobbs
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia; Department of Molecular and Translational Sciences, Monash University, Melbourne, VIC 3800, Australia.
| |
Collapse
|
5
|
Jha PK, Valekunja UK, Reddy AB. An integrative analysis of cell-specific transcriptomics and nuclear proteomics of sleep-deprived mouse cerebral cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.611806. [PMID: 39386443 PMCID: PMC11463534 DOI: 10.1101/2024.09.24.611806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Sleep regulation follows a homeostatic pattern. The mammalian cerebral cortex is the repository of homeostatic sleep drive and neurons and astrocytes of the cortex are principal responders of sleep need. The molecular mechanisms by which these two cell types respond to sleep loss are not yet clearly understood. By combining cell-type specific transcriptomics and nuclear proteomics we investigated how sleep loss affects the cellular composition and molecular profiles of these two cell types in a focused approach. The results indicate that sleep deprivation regulates gene expression and nuclear protein abundance in a cell-type-specific manner. Our integrated multi-omics analysis suggests that this distinction arises because neurons and astrocytes employ different gene regulatory strategies under accumulated sleep pressure. These findings provide a comprehensive view of the effects of sleep deprivation on gene regulation in neurons and astrocytes.
Collapse
Affiliation(s)
- Pawan K. Jha
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Utham K. Valekunja
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Akhilesh B. Reddy
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Chronobiology and Sleep Institute (CSI), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
6
|
Foley KG, Adli M, Kim JJ. Single-nuclei sequencing of uterine serous carcinoma reveals racial differences in immune signaling. Proc Natl Acad Sci U S A 2024; 121:e2402998121. [PMID: 39133838 PMCID: PMC11348309 DOI: 10.1073/pnas.2402998121] [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: 02/15/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Significant racial disparities exist between Black and White patients with uterine serous carcinoma (USC). While the reasons for these disparities are unclear, several studies have demonstrated significantly different rates of driver mutations between racial groups, including TP53. However, limited research has investigated the transcriptional differences of tumors or the composition of the tumor microenvironment (TME) between these groups. Here, we report the single-nuclei RNA-sequencing profiles of primary USC tumors from diverse racial backgrounds. We find that there are significant differences between the tumors of Black and White patients. Tumors from Black patients exhibited higher expression of specific genes associated with aggressiveness, such as PAX8, and axon guidance and synaptic signaling pathways. We also demonstrated that T cell populations are reduced in the tumor tissue compared to matched benign, while anti-inflammatory macrophage populations are retained within the TME. Furthermore, we investigated the connection between PAX8 overexpression and immunosuppression in USC through regulation of several cytokines and chemokines. Notably, we show that PAX8 activity can influence macrophage gene expression and protein secretion. These studies provide a detailed understanding of the USC transcriptome and TME, and identify differences in tumor biology from patients of different racial backgrounds.
Collapse
Affiliation(s)
- K. Grace Foley
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL60611
| | - Mazhar Adli
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL60611
| | - J. Julie Kim
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL60611
| |
Collapse
|
7
|
Phadte P, Bishnu A, Dey P, M M, Mehrotra M, Singh P, Chakrabarty S, Majumdar R, Rekhi B, Patra M, De A, Ray P. Autophagy-mediated ID1 turnover dictates chemo-resistant fate in ovarian cancer stem cells. J Exp Clin Cancer Res 2024; 43:222. [PMID: 39123206 PMCID: PMC11316295 DOI: 10.1186/s13046-024-03147-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The mechanisms enabling dynamic shifts between drug-resistant and drug-sensitive states in cancer cells are still underexplored. This study investigated the role of targeted autophagic protein degradation in regulating ovarian cancer stem cell (CSC) fate decisions and chemo-resistance. METHODS Autophagy levels were compared between CSC-enriched side population (SP) and non-SP cells (NSP) in multiple ovarian cancer cell lines using immunoblotting, immunofluorescence, and transmission electron microscopy. The impact of autophagy modulation on CSC markers and differentiation was assessed by flow cytometry, immunoblotting and qRT-PCR. In silico modeling and co-immunoprecipitation identified ID1 interacting proteins. Pharmacological and genetic approaches along with Annexin-PI assay, ChIP assay, western blotting, qRT-PCR and ICP-MS were used to evaluate effects on cisplatin sensitivity, apoptosis, SLC31A1 expression, promoter binding, and intracellular platinum accumulation in ID1 depleted backdrop. Patient-derived tumor spheroids were analyzed for autophagy and SLC31A1 levels. RESULTS Ovarian CSCs exhibited increased basal autophagy compared to non-CSCs. Further autophagy stimulation by serum-starvation and chemical modes triggered proteolysis of the stemness regulator ID1, driving the differentiation of chemo-resistant CSCs into chemo-sensitive non-CSCs. In silico modeling predicted TCF12 as a potent ID1 interactor, which was validated by co-immunoprecipitation. ID1 depletion freed TCF12 to transactivate the cisplatin influx transporter SLC31A1, increasing intracellular cisplatin levels and cytotoxicity. Patient-derived tumor spheroids exhibited a functional association between autophagy, ID1, SLC31A1, and platinum sensitivity. CONCLUSIONS This study reveals a novel autophagy-ID1-TCF12-SLC31A1 axis where targeted autophagic degradation of ID1 enables rapid remodeling of CSCs to reverse chemo-resistance. Modulating this pathway could counter drug resistance in ovarian cancer.
Collapse
Affiliation(s)
- Pratham Phadte
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Aniketh Bishnu
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Pranay Dey
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Manikandan M
- Laboratory of Medicinal Chemistry and Cell Biology, Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, 400005, India
| | - Megha Mehrotra
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Prerna Singh
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Shritama Chakrabarty
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
- Indian Institute of Science Education and Research, Bhopal, 462066, India
| | - Rounak Majumdar
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
- Indian Institute of Science Education and Research, Kolkata, 741246, India
| | - Bharat Rekhi
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
- Department of Pathology, Tata Memorial Hospital, Mumbai, 400012, India
| | - Malay Patra
- Laboratory of Medicinal Chemistry and Cell Biology, Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, 400005, India
| | - Abhijit De
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Pritha Ray
- Imaging Cell Signalling & Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, 410210, India.
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India.
| |
Collapse
|
8
|
Lima AS, Bezerra MF, Moreira-Aguiar A, Weinhäuser I, Santos BL, Falcão RM, Salustiano-Bandeira ML, Franca-Neto PL, Lima MM, Saldanha-Araujo F, Coelho-Silva JL, Pereira-Martins DA, Bezerra MA, Lucena-Araujo AR. Prognostic implications of the ID1 expression in acute myeloid leukemia patients treated in a resource-constrained setting. Hematol Transfus Cell Ther 2024; 46:250-255. [PMID: 37393163 PMCID: PMC11221249 DOI: 10.1016/j.htct.2023.04.005] [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: 07/29/2022] [Revised: 11/03/2022] [Accepted: 04/17/2023] [Indexed: 07/03/2023] Open
Abstract
INTRODUCTION The aberrant expression of the inhibitor of DNA binding (ID1) gene has been frequently associated with the leukemogenesis and prognostication acute myeloid leukemia (AML), although its clinical importance has never been investigated in patients treated outside well-controlled clinical trials. METHODS Using quantitative real-time polymerase chain reaction, we investigated the role of the ID1 expression in the clinical outcomes of non-selected patients with acute myeloid leukemia treated in a real-life setting. RESULTS Overall, 128 patients were enrolled. Patients with high ID1 expression had a lower 3-year overall survival (OS) rate of 9%, with the 95% confidence interval (95%CI) at 3 to 20%, compared to patients with a low ID1 expression (22%, 95%CI: 11 - 34%) (p = 0.037), although these findings did not retain significance after adjustment (hazard ratio (HR): 1.5, 95%CI: 0.98 - 2.28; p = 0.057). The ID1 expression had no impact on post-induction outcomes (disease-free survival, p = 0.648; cumulative incidence of relapse, p = 0.584). CONCLUSIONS Although we are aware thar our data are confronted with many variables that cannot be fully controlled, including drug unavailability, risk-adapted treatment, comorbidities and the time from diagnosis to treatment initiation, we are firm believers that such an initiative can provide more realistic data on understudied populations, in particular those from low- and middle-income countries.
Collapse
Affiliation(s)
- Aleide S Lima
- Universidade Federal de Pernambuco (UFPE), Recife, PE, Brazil
| | | | | | - Isabel Weinhäuser
- Cancer Research Centre Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bianca L Santos
- Universidade Federal de Pernambuco (UFPE), Recife, PE, Brazil
| | - Raul M Falcão
- Bioinformatics Multidisciplinary Environment (BioME), Metrópole Digital Institute, Universidade Federal do Rio Grande do Norte (UFRN), Natal, RN, Brazil
| | | | | | - Marinus M Lima
- Universidade Federal de Pernambuco (UFPE), Recife, PE, Brazil
| | - Felipe Saldanha-Araujo
- Laboratório de Hematologia e Células-Tronco, Universidade de Brasília (UnB) Brasília, DF, Brazil
| | - Juan L Coelho-Silva
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Diego A Pereira-Martins
- Cancer Research Centre Groningen, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | |
Collapse
|
9
|
Verdura S, Encinar JA, Gratchev A, Llop-Hernández À, López J, Serrano-Hervás E, Teixidor E, López-Bonet E, Martin-Castillo B, Micol V, Bosch-Barrera J, Cuyàs E, Menendez JA. Silibinin is a suppressor of the metastasis-promoting transcription factor ID3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155493. [PMID: 38484626 DOI: 10.1016/j.phymed.2024.155493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND ID3 (inhibitor of DNA binding/differentiation-3) is a transcription factor that enables metastasis by promoting stem cell-like properties in endothelial and tumor cells. The milk thistle flavonolignan silibinin is a phytochemical with anti-metastatic potential through largely unknown mechanisms. HYPOTHESIS/PURPOSE We have mechanistically investigated the ability of silibinin to inhibit the aberrant activation of ID3 in brain endothelium and non-small cell lung cancer (NSCLC) models. METHODS Bioinformatic analyses were performed to investigate the co-expression correlation between ID3 and bone morphogenic protein (BMP) ligands/BMP receptors (BMPRs) genes in NSCLC patient datasets. ID3 expression was assessed by immunoblotting and qRT-PCR. Luciferase reporter assays were used to evaluate the gene sequences targeted by silibinin to regulate ID3 transcription. In silico computational modeling and LanthaScreen TR-FRET kinase assays were used to characterize and validate the BMPR inhibitory activity of silibinin. Tumor tissues from NSCLC xenograft models treated with oral silibinin were used to evaluate the in vivo anti-ID3 effects of silibinin. RESULTS Analysis of lung cancer patient datasets revealed a top-ranked positive association of ID3 with the BMP9 endothelial receptor ACVRL1/ALK1 and the BMP ligand BMP6. Silibinin treatment blocked the BMP9-induced activation of the ALK1-phospho-SMAD1/5-ID3 axis in brain endothelial cells. Constitutive, acquired, and adaptive expression of ID3 in NSCLC cells were all significantly downregulated in response to silibinin. Silibinin blocked ID3 transcription via BMP-responsive elements in ID3 gene enhancers. Silibinin inhibited the kinase activities of BMPRs in the micromolar range, with the lower IC50 values occurring against ACVRL1/ALK1 and BMPR2. In an in vivo NSCLC xenograft model, tumoral overexpression of ID3 was completely suppressed by systematically achievable oral doses of silibinin. CONCLUSIONS ID3 is a largely undruggable metastasis-promoting transcription factor. Silibinin is a novel suppressor of ID3 that may be explored as a novel therapeutic approach to interfere with the metastatic dissemination capacity of NSCLC.
Collapse
Affiliation(s)
- Sara Verdura
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - José Antonio Encinar
- Institute of Research, Development and Innovation in Health Biotechnology of Elche (IDiBE), Universitas Miguel Hernández (UMH), Elche 03202, Spain
| | - Alexei Gratchev
- Laboratory for Tumor Stromal Cell Biology, Institute of Carcinogenesis, Nikolaj Nikolajevich (N.N.) Blokhin National Medical Research Center of Oncology, Moscow 115478, Russia
| | - Àngela Llop-Hernández
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Júlia López
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Eila Serrano-Hervás
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Eduard Teixidor
- Precision Oncology Group (OncoGir-Pro), Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Medical Oncology, Catalan Institute of Oncology, Girona, 17007, Spain
| | - Eugeni López-Bonet
- Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Department of Anatomical Pathology, Dr. Josep Trueta Hospital of Girona, Girona 17007, Spain
| | - Begoña Martin-Castillo
- Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Unit of Clinical Research, Catalan Institute of Oncology, Girona, 17007, Spain
| | - Vicente Micol
- Institute of Research, Development and Innovation in Health Biotechnology of Elche (IDiBE), Universitas Miguel Hernández (UMH), Elche 03202, Spain; CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, 28029, Spain
| | - Joaquim Bosch-Barrera
- Precision Oncology Group (OncoGir-Pro), Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain; Medical Oncology, Catalan Institute of Oncology, Girona, 17007, Spain; Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | - Elisabet Cuyàs
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain
| | - Javier A Menendez
- Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology, Girona, 17007, Spain; Metabolism and Cancer Group, Girona Biomedical Research Institute (IDIBGI), Girona 17190, Spain.
| |
Collapse
|
10
|
Nagasaka M, Miyajima C, Inoue Y, Hashiguchi S, Suzuki Y, Morishita D, Aoki H, Toriuchi K, Katayama R, Aoyama M, Hayashi H. ID3 is a novel target gene of p53 and modulates lung cancer cell metastasis. Biochem Biophys Res Commun 2024; 708:149789. [PMID: 38513475 DOI: 10.1016/j.bbrc.2024.149789] [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: 02/28/2024] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
The tumor suppressor p53 prevents cancer development by regulating dozens of target genes with diverse biological functions. Although numerous p53 target genes have been identified to date, the dynamics and function of the regulatory network centered on p53 have not yet been fully elucidated. We herein identified inhibitor of DNA-binding/differentiation-3 (ID3) as a direct p53 target gene. p53 bound the distal promoter of ID3 and positively regulated its transcription. ID3 expression was significantly decreased in clinical lung cancer tissues, and was closely associated with overall survival outcomes in these patients. Functionally, ID3 deficiency promoted the metastatic ability of lung cancer cells through its effects on the transcriptional regulation of CDH1. Furthermore, the ectopic expression of ID3 in p53-knockdown cells restored E-cadherin expression. Collectively, the present results demonstrate that ID3 plays a tumor-suppressive role as a downstream effector of p53 and impedes lung cancer cell metastasis by regulating E-cadherin expression.
Collapse
Affiliation(s)
- Mai Nagasaka
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Chiharu Miyajima
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Yasumichi Inoue
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| | - Sakura Hashiguchi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Yuya Suzuki
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Daisuke Morishita
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Hiromasa Aoki
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Kohki Toriuchi
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Mineyoshi Aoyama
- Department of Pathobiology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan
| | - Hidetoshi Hayashi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, 467-8603, Japan.
| |
Collapse
|
11
|
Kaise T, Kageyama R. Transcriptional control of neural stem cell activity. Biochem Soc Trans 2024; 52:617-626. [PMID: 38477464 DOI: 10.1042/bst20230439] [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: 12/20/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024]
Abstract
In the adult brain, neural stem cells (NSCs) are under the control of various molecular mechanisms to produce an appropriate number of neurons that are essential for specific brain functions. Usually, the majority of adult NSCs stay in a non-proliferative and undifferentiated state known as quiescence, occasionally transitioning to an active state to produce newborn neurons. This transition between the quiescent and active states is crucial for the activity of NSCs. Another significant state of adult NSCs is senescence, in which quiescent cells become more dormant and less reactive, ceasing the production of newborn neurons. Although many genes involved in the regulation of NSCs have been identified using genetic manipulation and omics analyses, the entire regulatory network is complicated and ambiguous. In this review, we focus on transcription factors, whose importance has been elucidated in NSCs by knockout or overexpression studies. We mainly discuss the transcription factors with roles in the active, quiescent, and rejuvenation states of adult NSCs.
Collapse
Affiliation(s)
- Takashi Kaise
- RIKEN Center for Brain Science, Wako 351-0198, Japan
| | | |
Collapse
|
12
|
Puyalto A, Rodríguez-Remírez M, López I, Macaya I, Guruceaga E, Olmedo M, Vilalta-Lacarra A, Welch C, Sandiego S, Vicent S, Valencia K, Calvo A, Pio R, Raez LE, Rolfo C, Ajona D, Gil-Bazo I. Trametinib sensitizes KRAS-mutant lung adenocarcinoma tumors to PD-1/PD-L1 axis blockade via Id1 downregulation. Mol Cancer 2024; 23:78. [PMID: 38643157 PMCID: PMC11031964 DOI: 10.1186/s12943-024-01991-3] [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: 11/15/2023] [Accepted: 04/02/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND The identification of novel therapeutic strategies to overcome resistance to the MEK inhibitor trametinib in mutant KRAS lung adenocarcinoma (LUAD) is a challenge. This study analyzes the effects of trametinib on Id1 protein, a key factor involved in the KRAS oncogenic pathway, and investigates the role of Id1 in the acquired resistance to trametinib as well as the synergistic anticancer effect of trametinib combined with immunotherapy in KRAS-mutant LUAD. METHODS We evaluated the effects of trametinib on KRAS-mutant LUAD by Western blot, RNA-seq and different syngeneic mouse models. Genetic modulation of Id1 expression was performed in KRAS-mutant LUAD cells by lentiviral or retroviral transductions of specific vectors. Cell viability was assessed by cell proliferation and colony formation assays. PD-L1 expression and apoptosis were measured by flow cytometry. The anti-tumor efficacy of the combined treatment with trametinib and PD-1 blockade was investigated in KRAS-mutant LUAD mouse models, and the effects on the tumor immune infiltrate were analyzed by flow cytometry and immunohistochemistry. RESULTS We found that trametinib activates the proteasome-ubiquitin system to downregulate Id1 in KRAS-mutant LUAD tumors. Moreover, we found that Id1 plays a major role in the acquired resistance to trametinib treatment in KRAS-mutant LUAD cells. Using two preclinical syngeneic KRAS-mutant LUAD mouse models, we found that trametinib synergizes with PD-1/PD-L1 blockade to hamper lung cancer progression and increase survival. This anti-tumor activity depended on trametinib-mediated Id1 reduction and was associated with a less immunosuppressive tumor microenvironment and increased PD-L1 expression on tumor cells. CONCLUSIONS Our data demonstrate that Id1 expression is involved in the resistance to trametinib and in the synergistic effect of trametinib with anti-PD-1 therapy in KRAS-mutant LUAD tumors. These findings suggest a potential therapeutic approach for immunotherapy-refractory KRAS-mutant lung cancers.
Collapse
Affiliation(s)
- Ander Puyalto
- Department of Medical Oncology, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - María Rodríguez-Remírez
- Department of Medical Oncology, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Inés López
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Irati Macaya
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - María Olmedo
- Department of Medical Oncology, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
| | - Anna Vilalta-Lacarra
- Department of Medical Oncology, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
| | - Connor Welch
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Sergio Sandiego
- Department of Oncology, Fundación Instituto Valenciano de Oncología (FIVO), C/Beltrán Báguena 8. 46009, Valencia, Spain
| | - Silvestre Vicent
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Karmele Valencia
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
| | - Alfonso Calvo
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ruben Pio
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain
| | - Luis E Raez
- Memorial Cancer Institute, Memorial Healthcare System, Florida Atlantic University (FAU), Pembroke Pines, FL, USA
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Health System, New York, USA
| | - Daniel Ajona
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain.
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- Department of Biochemistry and Genetics, School of Sciences, Universidad de Navarra, Pamplona, Spain.
| | - Ignacio Gil-Bazo
- Department of Medical Oncology, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain.
- Program in Solid Tumors, Cancer Division, Cima Universidad de Navarra, CCUN, Av. Pio XII, 55, 31008, Pamplona, Spain.
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- Department of Oncology, Fundación Instituto Valenciano de Oncología (FIVO), C/Beltrán Báguena 8. 46009, Valencia, Spain.
| |
Collapse
|
13
|
Li KQ, Bai X, Ke AT, Ding SQ, Zhang CD, Dai DQ. Ubiquitin-specific proteases: From biological functions to potential therapeutic applications in gastric cancer. Biomed Pharmacother 2024; 173:116323. [PMID: 38401523 DOI: 10.1016/j.biopha.2024.116323] [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: 12/04/2023] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024] Open
Abstract
Deubiquitination, a post-translational modification regulated by deubiquitinases, is essential for cancer initiation and progression. Ubiquitin-specific proteases (USPs) are essential elements of the deubiquitinase family, and are overexpressed in gastric cancer (GC). Through the regulation of several signaling pathways, such as Wnt/β-Catenin and nuclear factor-κB signaling, and the promotion of the expression of deubiquitination- and stabilization-associated proteins, USPs promote the proliferation, metastasis, invasion, and epithelial-mesenchymal transition of GC. In addition, the expression of USPs is closely related to clinicopathological features, patient prognosis, and chemotherapy resistance. USPs therefore could be used as prognostic biomarkers. USP targeting small molecule inhibitors have demonstrated strong anticancer activity. However, they have not yet been tested in the clinic. This article provides an overview of the latest fundamental research on USPs in GC, aiming to enhance the understanding of how USPs contribute to GC progression, and identifying possible targets for GC treatment to improve patient survival.
Collapse
Affiliation(s)
- Kai-Qiang Li
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Xiao Bai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Ang-Ting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Si-Qi Ding
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Chun-Dong Zhang
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Dong-Qiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China; Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China.
| |
Collapse
|
14
|
Li Y, Han M, Wei H, Huang W, Chen Z, Zhang T, Qian M, Jing L, Nan G, Sun X, Dai S, Wang K, Jiang J, Zhu P, Chen L. Id2 epigenetically controls CD8 + T-cell exhaustion by disrupting the assembly of the Tcf3-LSD1 complex. Cell Mol Immunol 2024; 21:292-308. [PMID: 38287103 PMCID: PMC10902300 DOI: 10.1038/s41423-023-01118-6] [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: 07/31/2022] [Accepted: 12/01/2023] [Indexed: 01/31/2024] Open
Abstract
CD8+ T-cell exhaustion is a state of dysfunction that promotes tumor progression and is marked by the generation of Slamf6+ progenitor exhausted (Texprog) and Tim-3+ terminally exhausted (Texterm) subpopulations. Inhibitor of DNA binding protein 2 (Id2) has been shown to play important roles in T-cell development and CD8+ T-cell immunity. However, the role of Id2 in CD8+ T-cell exhaustion is unclear. Here, we found that Id2 transcriptionally and epigenetically regulates the generation of Texprog cells and their conversion to Texterm cells. Genetic deletion of Id2 dampens CD8+ T-cell-mediated immune responses and the maintenance of stem-like CD8+ T-cell subpopulations, suppresses PD-1 blockade and increases tumor susceptibility. Mechanistically, through its HLH domain, Id2 binds and disrupts the assembly of the Tcf3-Tal1 transcriptional regulatory complex, and thus modulates chromatin accessibility at the Slamf6 promoter by preventing the interaction of Tcf3 with the histone lysine demethylase LSD1. Therefore, Id2 increases the abundance of the permissive H3K4me2 mark on the Tcf3-occupied E-boxes in the Slamf6 promoter, modulates chromatin accessibility at the Slamf6 promoter and epigenetically regulates the generation of Slamf6+ Texprog cells. An LSD1 inhibitor GSK2879552 can rescue the Id2 knockout phenotype in tumor-bearing mice. Inhibition of LSD1 increases the abundance of Slamf6+Tim-3- Texprog cells in tumors and the expression level of Tcf1 in Id2-deleted CD8+ T cells. This study demonstrates that Id2-mediated transcriptional and epigenetic modification drives hierarchical CD8+ T-cell exhaustion, and the mechanistic insights gained may have implications for therapeutic intervention with tumor immune evasion.
Collapse
Affiliation(s)
- Yiming Li
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Mingwei Han
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Haolin Wei
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Wan Huang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Zhinan Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Tianjiao Zhang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Meirui Qian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Lin Jing
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Gang Nan
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Xiuxuan Sun
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Shuhui Dai
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Kun Wang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China
| | - Jianli Jiang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China.
| | - Ping Zhu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China.
| | - Liang Chen
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Ganzhou, Jiangxi, 341000, Xi'an, Shaanxi, 710032, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
15
|
Yeh CY, Cai HY, Kuo HH, Lin YY, He ZJ, Cheng HC, Yang CJ, Huang CYF, Chang YC. ALDOA coordinates PDE3A through the β-catenin/ID3 axis to stimulate cancer metastasis and M2 polarization in lung cancer with EGFR mutations. Biochem Biophys Res Commun 2024; 696:149489. [PMID: 38244313 DOI: 10.1016/j.bbrc.2024.149489] [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] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/22/2024]
Abstract
Lung cancer has a high incidence rate and requires more effective treatment strategies and drug options for clinical patients. EGFR is a common genetic alteration event in lung cancer that affects patient survival and drug strategy. Our study discovered aberrant aldolase A (ALDOA) expression and dysfunction in lung cancer patients with EGFR mutations. In addition to investigating relevant metabolic processes like glucose uptake, lactate production, and ATPase activity, we examined multi-omics profiles (transcriptomics, proteomics, and pull-down assays). It was observed that phosphodiesterase 3A (PDE3A) enzyme and ALDOA exhibit correlation, and furthermore, they impact M2 macrophage polarization through β-catenin and downstream ID3. In addition to demonstrating the aforementioned mechanism of action, our experiments discovered that the PDE3 inhibitor trequinsin has a substantial impact on lung cancer cell lines with EGFR mutants. The trequinsin medication was found to decrease the M2 macrophage polarization status and several cancer phenotypes, in addition to transduction. These findings have potential prognostic and therapeutic applications for clinical patients with EGFR mutation and lung cancer.
Collapse
Affiliation(s)
- Chia-Ying Yeh
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Huei Yu Cai
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Han-His Kuo
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - You-Yu Lin
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Zhao-Jing He
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Chen Cheng
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Jen Yang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
16
|
Shalev N, Kendall M, Kumar N, Tiwari S, Anil SM, Hauschner H, Swamy SG, Doron-Faingenboim A, Belausov E, Kendall BE, Koltai H. Integrated transcriptome and cell phenotype analysis suggest involvement of PARP1 cleavage, Hippo/Wnt, TGF-β and MAPK signaling pathways in ovarian cancer cells response to cannabis and PARP1 inhibitor treatment. Front Genet 2024; 15:1333964. [PMID: 38322025 PMCID: PMC10844430 DOI: 10.3389/fgene.2024.1333964] [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] [Received: 11/06/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Introduction: Cannabis sativa is utilized mainly for palliative care worldwide. Ovarian cancer (OC) is a lethal gynecologic cancer. A particular cannabis extract fraction ('F7') and the Poly(ADP-Ribose) Polymerase 1 (PARP1) inhibitor niraparib act synergistically to promote OC cell apoptosis. Here we identified genetic pathways that are altered by the synergistic treatment in OC cell lines Caov3 and OVCAR3. Materials and methods: Gene expression profiles were determined by RNA sequencing and quantitative PCR. Microscopy was used to determine actin arrangement, a scratch assay to determine cell migration and flow cytometry to determine apoptosis, cell cycle and aldehyde dehydrogenase (ALDH) activity. Western blotting was used to determine protein levels. Results: Gene expression results suggested variations in gene expression between the two cell lines examined. Multiple genetic pathways, including Hippo/Wnt, TGF-β/Activin and MAPK were enriched with genes differentially expressed by niraparib and/or F7 treatments in both cell lines. Niraparib + F7 treatment led to cell cycle arrest and endoplasmic reticulum (ER) stress, inhibited cell migration, reduced the % of ALDH positive cells in the population and enhanced PARP1 cleavage. Conclusion: The synergistic effect of the niraparib + F7 may result from the treatment affecting multiple genetic pathways involving cell death and reducing mesenchymal characteristics.
Collapse
Affiliation(s)
- Nurit Shalev
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- Volcani Center, Agriculture Research Organization, Institute of Plant Science, Rishon LeZion, Israel
| | | | - Navin Kumar
- Volcani Center, Agriculture Research Organization, Institute of Plant Science, Rishon LeZion, Israel
| | - Sudeep Tiwari
- Volcani Center, Agriculture Research Organization, Institute of Plant Science, Rishon LeZion, Israel
| | - Seegehalli M. Anil
- Volcani Center, Agriculture Research Organization, Institute of Plant Science, Rishon LeZion, Israel
| | - Hagit Hauschner
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Savvemala G. Swamy
- Volcani Center, Agriculture Research Organization, Institute of Plant Science, Rishon LeZion, Israel
| | - Adi Doron-Faingenboim
- Volcani Center, Agriculture Research Organization, Institute of Plant Science, Rishon LeZion, Israel
| | - Eduard Belausov
- Volcani Center, Agriculture Research Organization, Institute of Plant Science, Rishon LeZion, Israel
| | | | - Hinanit Koltai
- Volcani Center, Agriculture Research Organization, Institute of Plant Science, Rishon LeZion, Israel
| |
Collapse
|
17
|
Romo A, Rodríguez TM, Yu G, Dewey RA. Chimeric TβRII-SE/Fc overexpression by a lentiviral vector exerts strong antitumoral activity on colorectal cancer-derived cell lines in vitro and on xenografts. Cancer Gene Ther 2024; 31:174-185. [PMID: 37993543 DOI: 10.1038/s41417-023-00694-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/16/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023]
Abstract
The TGF signaling pathway is a key regulator of cancer progression. In this work, we report for the first time the antitumor activity of TβRII-SE/Fc, a novel peptibody whose targeting domain is comprised of the soluble endogenous isoform of the human TGF-β type II receptor (TβRII-SE). Overexpression of TβRIISE/Fc reduces in vitro cell proliferation and migration while inducing cell cycle arrest and apoptosis in human colorectal cancer-derived cell lines. Moreover, TβRII-SE/Fc overexpression reduces tumorigenicity in BALB/c nude athymic mice. Our results revealed that TRII-SE/Fc-expressing tumors were significantly reduced in size or were even incapable of developing. We also demonstrated that the novel peptibody has the ability to inhibit the canonical TGF-β and BMP signaling pathways while identifying SMAD-dependent and independent proteins involved in tumor progression that are modulated by TβRII-SE/Fc. These findings provide insights into the underlying mechanism responsible for the antitumor activity of TβRII-SE/Fc. Although more studies are required to demonstrate the effectiveness and safety of the novel peptibody as a new therapeutic for the treatment of cancer, our initial in vitro and in vivo results in human colorectal tumor-derived cell lines are highly encouraging. Our results may serve as the foundation for further research and development of a novel biopharmaceutical for oncology.
Collapse
Affiliation(s)
- Ana Romo
- Laboratorio de Terapia Génica y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM), Chascomús, Argentina
- Escuela de Bio y Nanotecnologías, Universidad Nacional de San Martín (UNSAM), Buenos Aires, Argentina
- RADBIO S.A.S., Sunchales, Argentina
| | - Tania Melina Rodríguez
- Laboratorio de Terapia Génica y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM), Chascomús, Argentina
- Escuela de Bio y Nanotecnologías, Universidad Nacional de San Martín (UNSAM), Buenos Aires, Argentina
| | - Guo Yu
- Bio X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Ricardo Alfredo Dewey
- Laboratorio de Terapia Génica y Células Madre, Instituto Tecnológico de Chascomús (INTECH), CONICET-UNSAM), Chascomús, Argentina.
- Escuela de Bio y Nanotecnologías, Universidad Nacional de San Martín (UNSAM), Buenos Aires, Argentina.
- RADBIO S.A.S., Sunchales, Argentina.
- Centro de Medicina Traslacional (CEMET), Hospital de Alta Complejidad en Red "El Cruce" Nestor Carlos Kirchner, Florencio Varela, Argentina.
| |
Collapse
|
18
|
Shang S, Yang C, Chen F, Xiang RS, Zhang H, Dai SY, Liu J, Lv XX, Zhang C, Liu XT, Zhang Q, Lu SB, Song JW, Yu JJ, Zhou JC, Zhang XW, Cui B, Li PP, Zhu ST, Zhang HZ, Hua F. ID1 expressing macrophages support cancer cell stemness and limit CD8 + T cell infiltration in colorectal cancer. Nat Commun 2023; 14:7661. [PMID: 37996458 PMCID: PMC10667515 DOI: 10.1038/s41467-023-43548-w] [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: 03/13/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
Elimination of cancer stem cells (CSCs) and reinvigoration of antitumor immunity remain unmet challenges for cancer therapy. Tumor-associated macrophages (TAMs) constitute the prominant population of immune cells in tumor tissues, contributing to the formation of CSC niches and a suppressive immune microenvironment. Here, we report that high expression of inhibitor of differentiation 1 (ID1) in TAMs correlates with poor outcome in patients with colorectal cancer (CRC). ID1 expressing macrophages maintain cancer stemness and impede CD8+ T cell infiltration. Mechanistically, ID1 interacts with STAT1 to induce its cytoplasmic distribution and inhibits STAT1-mediated SerpinB2 and CCL4 transcription, two secretory factors responsible for cancer stemness inhibition and CD8+ T cell recruitment. Reducing ID1 expression ameliorates CRC progression and enhances tumor sensitivity to immunotherapy and chemotherapy. Collectively, our study highlights the pivotal role of ID1 in controlling the protumor phenotype of TAMs and paves the way for therapeutic targeting of ID1 in CRC.
Collapse
Affiliation(s)
- Shuang Shang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Chen Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Fei Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Ren-Shen Xiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
| | - Huan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Shu-Yuan Dai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Jing Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Xiao-Xi Lv
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Cheng Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Department of Pharmacy, China-Japan Friendship Hospital, 100029, Beijing, P. R. China
| | - Xiao-Tong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Qi Zhang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
| | - Shuai-Bing Lu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China
| | - Jia-Wei Song
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Jiao-Jiao Yu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Ji-Chao Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Xiao-Wei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Bing Cui
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Ping-Ping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China
| | - Sheng-Tao Zhu
- Beijing Digestive Diseases Center, Beijing Friendship Hospital, 100050, Beijing, P. R. China
- Beijing Key Laboratory for Precancerous Lesion of Digestive Diseases, Beijing Friendship Hospital, 100050, Beijing, P. R. China
| | - Hai-Zeng Zhang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China.
- State Key Lab of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, P. R. China.
| | - Fang Hua
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China.
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study (BZ0150), Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China.
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, Beijing, P. R. China.
| |
Collapse
|
19
|
Doke M, Álvarez-Cubela S, Klein D, Altilio I, Schulz J, Mateus Gonçalves L, Almaça J, Fraker CA, Pugliese A, Ricordi C, Qadir MMF, Pastori RL, Domínguez-Bendala J. Dynamic scRNA-seq of live human pancreatic slices reveals functional endocrine cell neogenesis through an intermediate ducto-acinar stage. Cell Metab 2023; 35:1944-1960.e7. [PMID: 37898119 DOI: 10.1016/j.cmet.2023.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/23/2023] [Accepted: 10/03/2023] [Indexed: 10/30/2023]
Abstract
Human pancreatic plasticity is implied from multiple single-cell RNA sequencing (scRNA-seq) studies. However, these have been invariably based on static datasets from which fate trajectories can only be inferred using pseudotemporal estimations. Furthermore, the analysis of isolated islets has resulted in a drastic underrepresentation of other cell types, hindering our ability to interrogate exocrine-endocrine interactions. The long-term culture of human pancreatic slices (HPSs) has presented the field with an opportunity to dynamically track tissue plasticity at the single-cell level. Combining datasets from same-donor HPSs at different time points, with or without a known regenerative stimulus (BMP signaling), led to integrated single-cell datasets storing true temporal or treatment-dependent information. This integration revealed population shifts consistent with ductal progenitor activation, blurring of ductal/acinar boundaries, formation of ducto-acinar-endocrine differentiation axes, and detection of transitional insulin-producing cells. This study provides the first longitudinal scRNA-seq analysis of whole human pancreatic tissue, confirming its plasticity in a dynamic fashion.
Collapse
Affiliation(s)
- Mayur Doke
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dagmar Klein
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Isabella Altilio
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joseph Schulz
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Luciana Mateus Gonçalves
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Christopher A Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alberto Pugliese
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mirza M F Qadir
- Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ricardo L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Juan Domínguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
20
|
Kim K, Lee SB. Regulation of CMGC kinases by hypoxia. BMB Rep 2023; 56:584-593. [PMID: 37915135 PMCID: PMC10689084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/25/2023] [Accepted: 10/12/2023] [Indexed: 11/03/2023] Open
Abstract
Hypoxia, a widespread occurrence observed in various malignant tumors, results from rapid tumor growth that outpaces the oxygen supply. Tumor hypoxia precipitates several effects on tumor biology; these include activating angiogenesis, intensifying invasiveness, enhancing the survival of tumor cells, suppressing anti-tumor immunity, and fostering resistance to therapy. Aligned with the findings that correlate CMGC kinases with the regulation of Hypoxia-Inducible Factor (HIF), a pivotal modulator, reports also indicate that hypoxia governs the activity of CMGC kinases, including DYRK1 kinases. Prolyl hydroxylation of DYRK1 kinases by PHD1 constitutes a novel mechanism of kinase maturation and activation. This modification "primes" DYRK1 kinases for subsequent tyrosine autophosphorylation, a vital step in their activation cascade. This mechanism adds a layer of intricacy to comprehending the regulation of CMGC kinases, and underscores the complex interplay between distinct post-translational modifications in harmonizing precise kinase activity. Overall, hypoxia assumes a substantial role in cancer progression, influencing diverse aspects of tumor biology that include angiogenesis, invasiveness, cell survival, and resistance to treatment. CMGC kinases are deeply entwined in its regulation. To fathom the molecular mechanisms underpinning hypoxia's impact on cancer cells, comprehending how hypoxia and prolyl hydroxylation govern the activity of CMGC kinases, including DYRK1 kinases, becomes imperative. This insight may pave the way for pioneering therapeutic approaches that target the hypoxic tumor microenvironment and its associated challenges. [BMB Reports 2023; 56(11): 584-593].
Collapse
Affiliation(s)
- KyeongJin Kim
- Department of Biomedical Sciences, Program in Biomedical Science & Engineering and Research Center for Controlling Intercellular Communication (RCIC), Inha University College of Medicine, Incheon 22212, Korea
| | - Sang Bae Lee
- Division of Life Sciences, Jeonbuk National University, Jeonju 54896, Korea
| |
Collapse
|
21
|
Wu Y, Liang H, Luo A, Li Y, Liu Z, Li X, Li W, Liang K, Li J, Liu Z, Du Y. Gelatin-based 3D biomimetic scaffolds platform potentiates culture of cancer stem cells in esophageal squamous cell carcinoma. Biomaterials 2023; 302:122323. [PMID: 37717405 DOI: 10.1016/j.biomaterials.2023.122323] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/02/2023] [Accepted: 09/09/2023] [Indexed: 09/19/2023]
Abstract
Cancer stem cells (CSCs) are crucial for tumorigenesis, metastasis, and therapy resistance in esophageal squamous cell carcinoma (ESCC). To further elucidate the mechanism underlying characteristics of CSCs and develop CSCs-targeted therapy, an efficient culture system that could expand and maintain CSCs is needed. CSCs reside in a complex tumor microenvironment, and three-dimensional (3D) culture systems of biomimetic scaffolds are expected to better support the growth of CSCs by recapitulating the biophysical properties of the extracellular matrix (ECM). Here, we established gelatin-based 3D biomimetic scaffolds mimicking the stiffness and collagen content of ESCC, which could enrich ESCC CSCs efficiently. Biological changes of ESCC cells laden in scaffolds with three different viscoelasticity emulating physiological stiffness of esophageal tissues were thoroughly investigated in varied aspects such as cell morphology, viability, cell phenotype markers, and transcriptomic profiling. The results demonstrated the priming effects of viscoelasticity on the stemness of ESCC. The highly viscous scaffolds (G': 6-403 Pa; G'': 2-75 Pa) better supported the enrichment of ESCC CSCs, and the TGF-beta signaling pathway might be involved in regulating the stemness of ESCC cells. Compared to two-dimensional (2D) cultures, highly viscous scaffolds significantly promoted the clonal expansion of ESCC cells in vitro and tumor formation ability in vivo. Our findings highlight the crucial role of biomaterials' viscoelasticity for the 3D culture of ESCC CSCs in vitro, and this newly-established culture system represents a valuable platform to support their growth, which could facilitate the CSCs-targeted therapy in the future.
Collapse
Affiliation(s)
- Yenan Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Haiwei Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Aiping Luo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiqiang Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xin Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenxin Li
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Junyang Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
22
|
Puyalto A, Rodríguez-Remírez M, López I, Iribarren F, Simón JA, Ecay M, Collantes M, Vilalta-Lacarra A, Francisco-Cruz A, Solórzano JL, Sandiego S, Peñuelas I, Calvo A, Ajona D, Gil-Bazo I. A novel [ 89Zr]-anti-PD-1-PET-CT to assess response to PD-1/PD-L1 blockade in lung cancer. Front Immunol 2023; 14:1272570. [PMID: 37841258 PMCID: PMC10569300 DOI: 10.3389/fimmu.2023.1272570] [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: 08/04/2023] [Accepted: 09/12/2023] [Indexed: 10/17/2023] Open
Abstract
Background Harnessing the anti-tumor immune system response by targeting the program cell death protein (PD-1) and program cell death ligand protein (PD-L1) axis has been a major breakthrough in non-small cell lung cancer (NSCLC) therapy. Nonetheless, conventional imaging tools cannot accurately assess response in immunotherapy-treated patients. Using a lung cancer syngeneic mouse model responder to immunotherapy, we aimed to demonstrate that [89Zr]-anti-PD-1 immuno-PET is a safe and feasible imaging modality to assess the response to PD-1/PD-L1 blockade in NSCLC. Materials and methods A syngeneic mouse model responder to anti-PD-1 therapy was used. Tumor growth and response to PD-1 blockade were monitored by conventional 2-deoxy-2-[18F]fluoro-D-glucose ([18F]-FDG) PET scans. Additionally, tumor lymphocyte infiltration was analyzed by the use of an [89Zr]-labeled anti-PD-1 antibody and measured as 89Zr tumor uptake. Results Conventional [18F]-FDG-PET scans failed to detect the antitumor activity exerted by anti-PD-1 therapy. However, [89Zr]-anti-PD-1 uptake was substantially higher in mice that responded to PD-1 blockade. The analysis of tumor-infiltrating immune cell populations and interleukins demonstrated an increased anti-tumor effect elicited by activation of effector immune cells in PD-1-responder mice. Interestingly, a positive correlation between [89Zr]-anti-PD-1 uptake and the proportion of tumor-infiltrating lymphocytes (TILs) was found (Cor = 0.8; p = 0.001). Conclusion Our data may support the clinical implementation of immuno-PET as a promising novel imaging tool to predict and assess the response of PD-1/PD-L1 inhibitors in patients with NSCLC.
Collapse
Affiliation(s)
- Ander Puyalto
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - María Rodríguez-Remírez
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Inés López
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
| | - Fabiola Iribarren
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
| | - Jon Ander Simón
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
- Department of Nuclear Medicine, Clínica Universidad de Navarra, Pamplona, Spain
- Translational Molecular Imaging Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Marga Ecay
- Department of Nuclear Medicine, Clínica Universidad de Navarra, Pamplona, Spain
- Translational Molecular Imaging Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - María Collantes
- Department of Nuclear Medicine, Clínica Universidad de Navarra, Pamplona, Spain
- Translational Molecular Imaging Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Anna Vilalta-Lacarra
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
| | | | - Jose Luis Solórzano
- Departamento de Anatomía Patológica y Diagnóstico Molecular, Md Anderson Cancer Center, Madrid, Spain
- Unidad de Investigación Clínica de Cáncer de Pulmón Hospital Universitario 12 de octubre- Centro Nacional de Investigaciones Oncologicas (H12O-CNIO), Madrid, Spain
| | - Sergio Sandiego
- Department of Oncology, Fundación Instituto Valenciano de Oncología (FIVO), Valencia, Spain
| | - Iván Peñuelas
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Department of Nuclear Medicine, Clínica Universidad de Navarra, Pamplona, Spain
- Translational Molecular Imaging Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Alfonso Calvo
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red - Cáncer (CIBERONC), Madrid, Spain
| | - Daniel Ajona
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red - Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Gil-Bazo
- Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
- University of Navarra, Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain
- Department of Oncology, Fundación Instituto Valenciano de Oncología (FIVO), Valencia, Spain
- Centro de Investigación Biomédica en Red - Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
23
|
Fei MY, Wang Y, Chang BH, Xue K, Dong F, Huang D, Li XY, Li ZJ, Hu CL, Liu P, Wu JC, Yu PC, Hong MH, Chen SB, Xu CH, Chen BY, Jiang YL, Liu N, Zhao C, Jin JC, Hou D, Chen XC, Ren YY, Deng CH, Zhang JY, Zong LJ, Wang RJ, Gao FF, Liu H, Zhang QL, Wu LY, Yan J, Shen S, Chang CK, Sun XJ, Wang L. The non-cell-autonomous function of ID1 promotes AML progression via ANGPTL7 from the microenvironment. Blood 2023; 142:903-917. [PMID: 37319434 PMCID: PMC10644073 DOI: 10.1182/blood.2022019537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/04/2023] [Accepted: 05/24/2023] [Indexed: 06/17/2023] Open
Abstract
The bone marrow microenvironment (BMM) can regulate leukemia stem cells (LSCs) via secreted factors. Increasing evidence suggests that dissecting the mechanisms by which the BMM maintains LSCs may lead to the development of effective therapies for the eradication of leukemia. Inhibitor of DNA binding 1 (ID1), a key transcriptional regulator in LSCs, previously identified by us, controls cytokine production in the BMM, but the role of ID1 in acute myeloid leukemia (AML) BMM remains obscure. Here, we report that ID1 is highly expressed in the BMM of patients with AML, especially in BM mesenchymal stem cells, and that the high expression of ID1 in the AML BMM is induced by BMP6, secreted from AML cells. Knocking out ID1 in mesenchymal cells significantly suppresses the proliferation of cocultured AML cells. Loss of Id1 in the BMM results in impaired AML progression in AML mouse models. Mechanistically, we found that Id1 deficiency significantly reduces SP1 protein levels in mesenchymal cells cocultured with AML cells. Using ID1-interactome analysis, we found that ID1 interacts with RNF4, an E3 ubiquitin ligase, and causes a decrease in SP1 ubiquitination. Disrupting the ID1-RNF4 interaction via truncation in mesenchymal cells significantly reduces SP1 protein levels and delays AML cell proliferation. We identify that the target of Sp1, Angptl7, is the primary differentially expression protein factor in Id1-deficient BM supernatant fluid to regulate AML progression in mice. Our study highlights the critical role of ID1 in the AML BMM and aids the development of therapeutic strategies for AML.
Collapse
Affiliation(s)
- Ming-Yue Fei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bin-He Chang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kai Xue
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyi Dong
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xi-Ya Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zi-Juan Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Cheng-Long Hu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ping Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ji-Chuan Wu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peng-Cheng Yu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ming-Hua Hong
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Shu-Bei Chen
- Department of Life Sciences and Biotechnology, Shanghai Jiao Tong University School of Life Sciences and Biotechnology, Shanghai, China
| | - Chun-Hui Xu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bing-Yi Chen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi-Lun Jiang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Na Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chong Zhao
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Cheng Jin
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Dan Hou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xin-Chi Chen
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi-Yi Ren
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chu-Han Deng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jia-Ying Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Li-juan Zong
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rou-Jia Wang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Fei-Fei Gao
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Eighth People’s Hospital, Shanghai, China
| | - Hui Liu
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology and Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Qun-Ling Zhang
- Department of Lymphoma, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ling-Yun Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jinsong Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Dalian Key Laboratory of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Diamond Bay Institute of Hematology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Shuhong Shen
- Department of Hematology/Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Key Laboratory of Pediatric Hematology and Oncology of China Ministry of Health, and National Children's Medical Center, Shanghai, China
| | - Chun-Kang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiao-Jian Sun
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Life Sciences and Biotechnology, Shanghai Jiao Tong University School of Life Sciences and Biotechnology, Shanghai, China
| | - Lan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
24
|
Nguyen A, Sung Y, Lee SH, Martin CE, Srikanth S, Chen W, Kang MK, Kim RH, Park NH, Gwack Y, Kim Y, Shin KH. Orai3 Calcium Channel Contributes to Oral/Oropharyngeal Cancer Stemness through the Elevation of ID1 Expression. Cells 2023; 12:2225. [PMID: 37759448 PMCID: PMC10527097 DOI: 10.3390/cells12182225] [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: 07/27/2023] [Revised: 08/24/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Emerging evidence indicates that intracellular calcium (Ca2+) levels and their regulatory proteins play essential roles in normal stem cell proliferation and differentiation. Cancer stem-like cells (CSCs) are subpopulations of cancer cells that retain characteristics similar to stem cells and play an essential role in cancer progression. Recent studies have reported that the Orai3 calcium channel plays an oncogenic role in human cancer. However, its role in CSCs remains underexplored. In this study, we explored the effects of Orai3 in the progression and stemness of oral/oropharyngeal squamous cell carcinoma (OSCC). During the course of OSCC progression, the expression of Orai3 exhibited a stepwise augmentation. Notably, Orai3 was highly enriched in CSC populations of OSCC. Ectopic Orai3 expression in non-tumorigenic immortalized oral epithelial cells increased the intracellular Ca2+ levels, acquiring malignant growth and CSC properties. Conversely, silencing of the endogenous Orai3 in OSCC cells suppressed the CSC phenotype, indicating a pivotal role of Orai3 in CSC regulation. Moreover, Orai3 markedly increased the expression of inhibitor of DNA binding 1 (ID1), a stemness transcription factor. Orai3 and ID1 exhibited elevated expression within CSCs compared to their non-CSC counterparts, implying the functional importance of the Orai3/ID1 axis in CSC regulation. Furthermore, suppression of ID1 abrogated the CSC phenotype in the cell with ectopic Orai3 overexpression and OSCC. Our study reveals that Orai3 is a novel functional CSC regulator in OSCC and further suggests that Orai3 plays an oncogenic role in OSCC by promoting cancer stemness via ID1 upregulation.
Collapse
Affiliation(s)
- Anthony Nguyen
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Youngjae Sung
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Sung Hee Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Charlotte Ellen Martin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Wei Chen
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
| | - Mo K. Kang
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - Reuben H. Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Yong Kim
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
- Laboratory of Stem Cell and Cancer Epigenetics, UCLA School of Dentistry, Los Angeles, CA 90095, USA
- UCLA Broad Stem Cell Research Center, Los Angeles, CA 90095, USA
| | - Ki-Hyuk Shin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, Los Angeles, CA 90095, USA; (A.N.)
- UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA 90095, USA
| |
Collapse
|
25
|
Chowdhary S, Deka R, Panda K, Kumar R, Solomon AD, Das J, Kanoujiya S, Gupta AK, Sinha S, Ruokolainen J, Kesari KK, Gupta PK. Recent Updates on Viral Oncogenesis: Available Preventive and Therapeutic Entities. Mol Pharm 2023; 20:3698-3740. [PMID: 37486263 PMCID: PMC10410670 DOI: 10.1021/acs.molpharmaceut.2c01080] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/25/2023]
Abstract
Human viral oncogenesis is a complex phenomenon and a major contributor to the global cancer burden. Several recent findings revealed cellular and molecular pathways that promote the development and initiation of malignancy when viruses cause an infection. Even, antiviral treatment has become an approach to eliminate the viral infections and prevent the activation of oncogenesis. Therefore, for a better understanding, the molecular pathogenesis of various oncogenic viruses like, hepatitis virus, human immunodeficiency viral (HIV), human papillomavirus (HPV), herpes simplex virus (HSV), and Epstein-Barr virus (EBV), could be explored, especially, to expand many potent antivirals that may escalate the apoptosis of infected malignant cells while sparing normal and healthy ones. Moreover, contemporary therapies, such as engineered antibodies antiviral agents targeting signaling pathways and cell biomarkers, could inhibit viral oncogenesis. This review elaborates the recent advancements in both natural and synthetic antivirals to control viral oncogenesis. The study also highlights the challenges and future perspectives of using antivirals in viral oncogenesis.
Collapse
Affiliation(s)
- Shivam Chowdhary
- Department
of Industrial Microbiology, Sam Higginbottom
University of Agriculture, Technology and Sciences, Prayagraj 211007, Uttar Pradesh India
| | - Rahul Deka
- Department
of Bioengineering and Biotechnology, Birla
Institute of Technology, Mesra, Ranchi 835215, Jharkhand, India
| | - Kingshuk Panda
- Department
of Applied Microbiology, Vellore Institute
of Technology, Vellore 632014, Tamil Nadu, India
| | - Rohit Kumar
- Department
of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, India
| | - Abhishikt David Solomon
- Department
of Molecular & Cellular Engineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, Uttar Pradesh, India
| | - Jimli Das
- Centre
for
Biotechnology and Bioinformatics, Dibrugarh
University, Assam 786004, India
| | - Supriya Kanoujiya
- School
of
Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Ashish Kumar Gupta
- Department
of Biophysics, All India Institute of Medical
Sciences, New Delhi 110029, India
| | - Somya Sinha
- Department
of Biotechnology, Graphic Era Deemed to
Be University, Dehradun 248002, Uttarakhand, India
| | - Janne Ruokolainen
- Department
of Applied Physics, School of Science, Aalto
University, 02150 Espoo, Finland
| | - Kavindra Kumar Kesari
- Department
of Applied Physics, School of Science, Aalto
University, 02150 Espoo, Finland
- Division
of Research and Development, Lovely Professional
University, Phagwara 144411, Punjab, India
| | - Piyush Kumar Gupta
- Department
of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- Department
of Biotechnology, Graphic Era Deemed to
Be University, Dehradun 248002, Uttarakhand, India
- Faculty
of Health and Life Sciences, INTI International
University, Nilai 71800, Malaysia
| |
Collapse
|
26
|
Chang YT, Chiu I, Wang Q, Bustamante J, Jiang W, Rycaj K, Yi S, Li J, Kowalski-Muegge J, Matsui W. Loss of p53 enhances the tumor-initiating potential and drug resistance of clonogenic multiple myeloma cells. Blood Adv 2023; 7:3551-3560. [PMID: 37042949 PMCID: PMC10368840 DOI: 10.1182/bloodadvances.2022009387] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/05/2023] [Accepted: 04/05/2023] [Indexed: 04/13/2023] Open
Abstract
Tumor relapse and drug resistance are major factors that limit the curability of multiple myeloma (MM). New regimens have improved overall MM survival rates, but patients with high-risk features continue to have inferior outcomes. Chromosome 17p13 deletion (del17p) that includes the loss of the TP53 gene is a high-risk cytogenetic abnormality and is associated with poor clinical outcomes owing to relatively short remissions and the development of pan-drug resistant disease. Increased relapse rates suggest that del17p enhances clonogenic growth, and we found that the loss of p53 increased both the frequency and drug resistance of tumor-initiating MM cells (TICs). Subsequent RNA sequencing (RNA-seq) studies demonstrated significant activation of the Notch signaling pathway and upregulation of inhibitor of DNA binding (ID1/ID2) genes in p53-knock out (p53-KO) cells. We found that the loss of ID1 or HES-1 expression or treatment with a gamma-secretase inhibitor (GSI) significantly decreased the clonogenic growth of p53-KO but not p53 wild-type cells. GSI treatment in a small set of MM specimens also reduced the clonogenic growth in del17p samples but not in non-del17p samples. This effect was specific as overexpression of the Notch intracellular domain (NICD) rescued the effects of GSI treatment. Our study demonstrates that the Notch signaling and ID1 expression are required for TIC expansion in p53-KO MM cells. These findings also suggest that GSI may be specifically active in patients with p53 mutant MM.
Collapse
Affiliation(s)
- Yu-Tai Chang
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Ian Chiu
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
- College of Natural Sciences, The University of Texas at Austin, Austin, TX
| | - Qiuju Wang
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Jorge Bustamante
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Wenxuan Jiang
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Kiera Rycaj
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Song Yi
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Joey Li
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jeanne Kowalski-Muegge
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
| | - William Matsui
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX
| |
Collapse
|
27
|
Yan C, Li Y, Liu H, Chen D, Wu J. Antitumor mechanism of cannabidiol hidden behind cancer hallmarks. Biochim Biophys Acta Rev Cancer 2023; 1878:188905. [PMID: 37164234 DOI: 10.1016/j.bbcan.2023.188905] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
Cannabinoids have been utilized for recreational and therapeutic purposes for over 4,000 years. As the primary ingredient in exogenous cannabinoids, Cannabidiol (CBD) has drawn a lot of interest from researchers due to its negligible psychotropic side effects and potential tumor-suppressing properties. However, the obscure mechanisms that underlie them remain a mystery. Complex biological mechanisms are involved in the progression of cancer, and malignancies have a variety of acquired biological capabilities, including sustained proliferation, death evasion, neovascularization, tissue invasion and metastasis, immune escape, metabolic reprogramming, induction of tumor-associated inflammation, cancerous stemness and genomic instability. Nowadays, the role of CBD hidden in these hallmarks is gradually revealed. Nevertheless, flaws or inconsistencies in the recent studies addressing the anti-cancer effects of CBD still exist. The purpose of this review is to evaluate the potential mechanisms underlying the role of CBD in a range of tumor-acquired biological capabilities. We propose potential drugs that may have a synergistic effect with CBD and provide optional directions for future research.
Collapse
Affiliation(s)
- Chaobiao Yan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, China; NHC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences(2019RU019), China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou 310003, Zhejiang Province, China.
| | - Yu Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, China; NHC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences(2019RU019), China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou 310003, Zhejiang Province, China.
| | - Hanqing Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, China; NHC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences(2019RU019), China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou 310003, Zhejiang Province, China.
| | - Diyu Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, China; NHC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences(2019RU019), China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou 310003, Zhejiang Province, China.
| | - Jian Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, China; NHC Key Laboratory of Combined Multi-organ Transplantation, China; Key Laboratory of the diagnosis and treatment of organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences(2019RU019), China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Hangzhou 310003, Zhejiang Province, China.
| |
Collapse
|
28
|
Emri E, Cappa O, Kelly C, Kortvely E, SanGiovanni JP, McKay BS, Bergen AA, Simpson DA, Lengyel I. Zinc Supplementation Induced Transcriptional Changes in Primary Human Retinal Pigment Epithelium: A Single-Cell RNA Sequencing Study to Understand Age-Related Macular Degeneration. Cells 2023; 12:773. [PMID: 36899910 PMCID: PMC10000409 DOI: 10.3390/cells12050773] [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: 02/05/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Zinc supplementation has been shown to be beneficial to slow the progression of age-related macular degeneration (AMD). However, the molecular mechanism underpinning this benefit is not well understood. This study used single-cell RNA sequencing to identify transcriptomic changes induced by zinc supplementation. Human primary retinal pigment epithelial (RPE) cells could mature for up to 19 weeks. After 1 or 18 weeks in culture, we supplemented the culture medium with 125 µM added zinc for one week. RPE cells developed high transepithelial electrical resistance, extensive, but variable pigmentation, and deposited sub-RPE material similar to the hallmark lesions of AMD. Unsupervised cluster analysis of the combined transcriptome of the cells isolated after 2, 9, and 19 weeks in culture showed considerable heterogeneity. Clustering based on 234 pre-selected RPE-specific genes divided the cells into two distinct clusters, we defined as more and less differentiated cells. The proportion of more differentiated cells increased with time in culture, but appreciable numbers of cells remained less differentiated even at 19 weeks. Pseudotemporal ordering identified 537 genes that could be implicated in the dynamics of RPE cell differentiation (FDR < 0.05). Zinc treatment resulted in the differential expression of 281 of these genes (FDR < 0.05). These genes were associated with several biological pathways with modulation of ID1/ID3 transcriptional regulation. Overall, zinc had a multitude of effects on the RPE transcriptome, including several genes involved in pigmentation, complement regulation, mineralization, and cholesterol metabolism processes associated with AMD.
Collapse
Affiliation(s)
- Eszter Emri
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
- Section Ophthalmogenetics, Department of Human Genetics, Queen Emma Centre for Precision Medicine, Amsterdam UMC, Location AMC, 1105AZ Amsterdam, The Netherlands
| | - Oisin Cappa
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Caoimhe Kelly
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Elod Kortvely
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - John Paul SanGiovanni
- Biosciences Research Laboratories, BIO5 Institute, University of Arizona, 1230 North Cherry Avenue, Tucson, AZ 85724, USA
| | - Brian S. McKay
- Department of Ophthalmology and Vision Science, University of Arizona, 1656 E. Mabel Street, Tucson, AZ 85724, USA
| | - Arthur A. Bergen
- Section Ophthalmogenetics, Department of Human Genetics, Queen Emma Centre for Precision Medicine, Amsterdam UMC, Location AMC, 1105AZ Amsterdam, The Netherlands
- The Netherlands Institute for Neuroscience (NIN-KNAW), 1105AZ Amsterdam, The Netherlands
| | - David A. Simpson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| | - Imre Lengyel
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT97BL, UK
| |
Collapse
|
29
|
Murugesan P, Begum H, Tangutur AD. Inhibitor of DNA binding/differentiation proteins as IDs for pancreatic cancer: Role in pancreatic cancer initiation, development and prognosis. Gene 2023; 853:147092. [PMID: 36464175 DOI: 10.1016/j.gene.2022.147092] [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: 08/01/2022] [Revised: 11/11/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
A family of inhibitors of cell differentiation or DNA-binding proteins, known as ID proteins (ID1-4), function as mighty transcription factors in various cellular processes, such as inhibiting differentiation, promoting cell-cycle progression, senescence, angiogenesis, tumorigenesis, and metastasis in cancer. Pancreatic cancer represents the deadliest cancer with the lowest survival rate of 10% due to the diagnosis at an advanced fatal stage and therapeutic resistance. Modestly, the only curative option for this lethal cancer is surgery but is done in less than 15-20% of patients because of the locally aggressive and early metastatic nature. Finding the earliest biomarkers and targeting the various hallmarks of pancreatic cancer can improve the treatment and survival of pancreatic cancer patients. Therefore, herein in this review, we explore in depth the potential roles of ID proteins function in hallmarks of pancreatic cancer, signaling pathways, and its oncogenic and tumor-suppressive effects. Hence, understanding the roles of dysregulated ID proteins would provide new insights into its function in pancreatic cancer tumorigenesis.
Collapse
Affiliation(s)
- Periyasamy Murugesan
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Habeebunnisa Begum
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
30
|
Meligova AK, Siakouli D, Stasinopoulou S, Xenopoulou DS, Zoumpouli M, Ganou V, Gkotsi EF, Chatziioannou A, Papadodima O, Pilalis E, Alexis MN, Mitsiou DJ. ERβ1 Sensitizes and ERβ2 Desensitizes ERα-Positive Breast Cancer Cells to the Inhibitory Effects of Tamoxifen, Fulvestrant and Their Combination with All-Trans Retinoic Acid. Int J Mol Sci 2023; 24:ijms24043747. [PMID: 36835157 PMCID: PMC9959521 DOI: 10.3390/ijms24043747] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
Adjuvant endocrine therapy (AET) is the treatment of choice for early-stage estrogen receptor alpha (ERα)-positive breast cancer (BC). However, almost 40% of tamoxifen-treated cases display no response or a partial response to AET, thus increasing the need for new treatment options and strong predictors of the therapeutic response of patients at high risk of relapse. In addition to ERα, BC research has focused on ERβ1 and ERβ2 (isoforms of ERβ), the second ER isotype. At present, the impact of ERβ isoforms on ERα-positive BC prognosis and treatment remains elusive. In the present study, we established clones of MCF7 cells constitutively expressing human ERβ1 or ERβ2 and investigated their role in the response of MCF7 cells to antiestrogens [4-hydroxytamoxifen (OHΤ) and fulvestrant (ICI182,780)] and retinoids [all-trans retinoic acid (ATRA)]. We show that, compared to MCF7 cells, MCF7-ERβ1 and MCF7-ERβ2 cells were sensitized and desensitized, respectively, to the antiproliferative effect of the antiestrogens, ATRA and their combination and to the cytocidal effect of the combination of OHT and ATRA. Analysis of the global transcriptional changes upon OHT-ATRA combinatorial treatment revealed uniquely regulated genes associated with anticancer effects in MCF7-ERβ1 cells and cancer-promoting effects in MCF7-ERβ2 cells. Our data are favorable to ERβ1 being a marker of responsiveness and ERβ2 being a marker of resistance of MCF7 cells to antiestrogens alone and in combination with ATRA.
Collapse
Affiliation(s)
- Aggeliki K. Meligova
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Dimitra Siakouli
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Sotiria Stasinopoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Despoina S. Xenopoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Zoumpouli
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Vassiliki Ganou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Eleni-Fani Gkotsi
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Aristotelis Chatziioannou
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Olga Papadodima
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | | | - Michael N. Alexis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
- Correspondence: (M.N.A.); (D.J.M.)
| | - Dimitra J. Mitsiou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
- Correspondence: (M.N.A.); (D.J.M.)
| |
Collapse
|
31
|
Cancer Stem Cell Relationship with Pro-Tumoral Inflammatory Microenvironment. Biomedicines 2023; 11:biomedicines11010189. [PMID: 36672697 PMCID: PMC9855358 DOI: 10.3390/biomedicines11010189] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Inflammatory processes and cancer stem cells (CSCs) are increasingly recognized as factors in the development of tumors. Emerging evidence indicates that CSCs are associated with cancer properties such as metastasis, treatment resistance, and disease recurrence. However, the precise interaction between CSCs and the immune microenvironment remains unexplored. Although evasion of the immune system by CSCs has been extensively studied, new research demonstrates that CSCs can also control and even profit from the immune response. This review provides an overview of the reciprocal interplay between CSCs and tumor-infiltrating immune cells, collecting pertinent data about how CSCs stimulate leukocyte reprogramming, resulting in pro-tumor immune cells that promote metastasis, chemoresistance, tumorigenicity, and even a rise in the number of CSCs. Tumor-associated macrophages, neutrophils, Th17 and regulatory T cells, mesenchymal stem cells, and cancer-associated fibroblasts, as well as the signaling pathways involved in these pro-tumor activities, are among the immune cells studied. Although cytotoxic leukocytes have the potential to eliminate CSCs, immune evasion mechanisms in CSCs and their clinical implications are also known. We intended to compile experimental findings that provide direct evidence of interactions between CSCs and the immune system and CSCs and the inflammatory milieu. In addition, we aimed to summarize key concepts in order to comprehend the cross-talk between CSCs and the tumor microenvironment as a crucial process for the effective design of anti-CSC therapies.
Collapse
|
32
|
Zhao Q, Wang Y, Yu D, Leng JY, Zhao Y, Chu M, Xu Z, Ding H, Zhou J, Zhang T. Comprehensive analysis of ID genes reveals the clinical and prognostic value of ID3 expression in acute myeloid leukemia using bioinformatics identification and experimental validation. BMC Cancer 2022; 22:1229. [PMID: 36443709 PMCID: PMC9707109 DOI: 10.1186/s12885-022-10352-6] [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: 08/03/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Dysregulation of inhibitor of differentiation/DNA binding (ID) genes is linked to cancer growth, angiogenesis, invasiveness, metastasis and patient survival. Nevertheless, few investigations have systematically determined the expression and prognostic value of ID genes in acute myeloid leukemia (AML). METHODS The expression and clinical prognostic value of ID genes in AML were first identified by public databases and further validated by our research cohort. RESULTS Using public data, the expression of ID1/ID3 was markedly downregulated in AML, and the expression of ID2 was greatly upregulated in AML, whereas ID4 showed no significant difference. Among the ID genes, only ID3 expression may be the most valuable prognostic biomarker in both total AML and cytogenetically normal AML (CN-AML) and especially in CN-AML. Clinically, reduced ID3 expression was greatly associated with higher white blood cell counts, peripheral blood/bone marrow blasts, normal karyotypes and intermediate cytogenetic risk. In addition, low ID3 expression was markedly related to FLT3 and NPM1 mutations as well as wild-type TP53. Despite these associations, multivariate Cox regression analysis revealed that ID3 expression was an independent risk factor affecting overall survival (OS) and disease free survival (DFS) in CN-AML patients. Biologically, a total of 839 mRNAs/lncRNAs and 72 microRNAs were found to be associated with ID3 expression in AML. Importantly, the expression of ID3 with discriminative value in AML was further confirmed in our research cohort. CONCLUSION The bioinformatics analysis and experimental verification demonstrate that low ID3 expression independently affects OS and DFS in patients with CN-AML, which might be seen as a potential prognostic indicator in CN-AML.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China.,Zhenjiang Clinical Research Center of Hematology, 212002, Zhenjiang, Jiangsu, P. R. China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Jiangsu, 212002, Zhenjiang, P. R. China.,Laboratory Center, Affiliated People's Hospital of Jiangsu University, 212002, Zhenjiang, Jiangsu, P. R. China.,Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China
| | - Yun Wang
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China.,Zhenjiang Clinical Research Center of Hematology, 212002, Zhenjiang, Jiangsu, P. R. China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Jiangsu, 212002, Zhenjiang, P. R. China
| | - Di Yu
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China.,Zhenjiang Clinical Research Center of Hematology, 212002, Zhenjiang, Jiangsu, P. R. China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Jiangsu, 212002, Zhenjiang, P. R. China
| | - Jia-Yan Leng
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China.,Zhenjiang Clinical Research Center of Hematology, 212002, Zhenjiang, Jiangsu, P. R. China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Jiangsu, 212002, Zhenjiang, P. R. China
| | - Yangjing Zhao
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, 212013, Zhenjiang, Jiangsu, P. R. China
| | - Mingqiang Chu
- Zhenjiang Clinical Research Center of Hematology, 212002, Zhenjiang, Jiangsu, P. R. China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Jiangsu, 212002, Zhenjiang, P. R. China.,Laboratory Center, Affiliated People's Hospital of Jiangsu University, 212002, Zhenjiang, Jiangsu, P. R. China.,Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China
| | - Zijun Xu
- Zhenjiang Clinical Research Center of Hematology, 212002, Zhenjiang, Jiangsu, P. R. China.,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Jiangsu, 212002, Zhenjiang, P. R. China.,Laboratory Center, Affiliated People's Hospital of Jiangsu University, 212002, Zhenjiang, Jiangsu, P. R. China
| | - Hao Ding
- Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China.
| | - Jingdong Zhou
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China. .,Zhenjiang Clinical Research Center of Hematology, 212002, Zhenjiang, Jiangsu, P. R. China. .,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Jiangsu, 212002, Zhenjiang, P. R. China.
| | - Tingjuan Zhang
- Zhenjiang Clinical Research Center of Hematology, 212002, Zhenjiang, Jiangsu, P. R. China. .,The Key Lab of Precision Diagnosis and Treatment of Zhenjiang City, Jiangsu, 212002, Zhenjiang, P. R. China. .,Department of Oncology, Affiliated People's Hospital of Jiangsu University, 8 Dianli Rd, 212002, Zhenjiang, Jiangsu, P. R. China.
| |
Collapse
|
33
|
Wang T, Xu C, Zhang Z, Wu H, Li X, Zhang Y, Deng N, Dang N, Tang G, Yang X, Shi B, Li Z, Li L, Ye K. Cellular heterogeneity and transcriptomic profiles during intrahepatic cholangiocarcinoma initiation and progression. Hepatology 2022; 76:1302-1317. [PMID: 35340039 PMCID: PMC9790314 DOI: 10.1002/hep.32483] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIMS Intrahepatic cholangiocarcinoma (ICC) is not fully investigated, and how stromal cells contribute to ICC formation is poorly understood. We aimed to uncover ICC origin, cellular heterogeneity, and critical modulators during ICC initiation/progression, and to decipher how fibroblast and endothelial cells in the stromal compartment favor ICC progression. APPROACH AND RESULTS We performed single-cell RNA sequencing (scRNA-seq) using AKT/Notch intracellular domain-induced mouse ICC tissues at early, middle, and late stages. We analyzed the transcriptomic landscape, cellular classification and evolution, and intercellular communication during ICC initiation/progression. We confirmed the findings using quantitative real-time PCR, western blotting, immunohistochemistry or immunofluorescence, and gene knockout/knockdown analysis. We identified stress-responding and proliferating subpopulations in late-stage mouse ICC tissues and validated them using human scRNA-seq data sets. By integrating weighted correlation network analysis and protein-protein interaction through least absolute shrinkage and selection operator regression, we identified zinc finger, MIZ-type containing 1 (Zmiz1) and Y box protein 1 (Ybx1) as core transcription factors required by stress-responding and proliferating ICC cells, respectively. Knockout of either one led to the blockade of ICC initiation/progression. Using two other ICC mouse models (YAP/AKT, KRAS/p19) and human ICC scRNA-seq data sets, we confirmed the orchestrating roles of Zmiz1 and Ybx1 in ICC occurrence and development. In addition, hes family bHLH transcription factor 1, cofilin 1, and inhibitor of DNA binding 1 were identified as driver genes for ICC. Moreover, periportal liver sinusoidal endothelial cells could differentiate into tip endothelial cells to promote ICC development, and this was Dll4-Notch4-Efnb2 signaling-dependent. CONCLUSIONS Stress-responding and ICC proliferating subtypes were identified, and Zmiz1 and Ybx1 were revealed as core transcription factors in these subtypes. Fibroblast-endothelial cell interaction promotes ICC development.
Collapse
Affiliation(s)
- Tingjie Wang
- School of Automation Science and EngineeringFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityXi’anShaanxiChina
| | - Chuanrui Xu
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhijing Zhang
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hua Wu
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiujuan Li
- School of Automation Science and EngineeringFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityXi’anShaanxiChina
| | - Yu Zhang
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Nan Deng
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ningxin Dang
- Genome Institutethe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiChina
| | - Guangbo Tang
- School of Life Science and TechnologyXi’an Jiaotong UniversityXi’anShaanxiChina
| | - Xiaofei Yang
- School of Computer Science and TechnologyFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityXi’anShaanxiChina,Genome Institutethe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiChina,MOE Key Lab for Intelligent Networks & Networks SecurityFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityXi’anShaanxiChina
| | - Bingyin Shi
- Department of Endocrinologythe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiChina
| | - Zihang Li
- School of Life Science and TechnologyXi’an Jiaotong UniversityXi’anShaanxiChina
| | - Lei Li
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Kai Ye
- School of Automation Science and EngineeringFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityXi’anShaanxiChina,Genome Institutethe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anShaanxiChina,School of Life Science and TechnologyXi’an Jiaotong UniversityXi’anShaanxiChina,Faculty of ScienceLeiden UniversityLeidenthe Netherlands,MOE Key Lab for Intelligent Networks & Networks SecurityFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityXi’anShaanxiChina
| |
Collapse
|
34
|
Kim M, Singh M, Lee BK, Hibbs M, Richardson K, Ellies L, Wintle L, Stuart LM, Wang JY, Voon DC, Blancafort P, Wang J, Kim J, Leedman PJ, Woo AJ. A MYC-ZNF148-ID1/3 regulatory axis modulating cancer stem cell traits in aggressive breast cancer. Oncogenesis 2022; 11:60. [PMID: 36207293 PMCID: PMC9546828 DOI: 10.1038/s41389-022-00435-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
The MYC proto-oncogene (MYC) is one of the most frequently overexpressed genes in breast cancer that drives cancer stem cell-like traits, resulting in aggressive disease progression and poor prognosis. In this study, we identified zinc finger transcription factor 148 (ZNF148, also called Zfp148 and ZBP-89) as a direct target of MYC. ZNF148 suppressed cell proliferation and migration and was transcriptionally repressed by MYC in breast cancer. Depletion of ZNF148 by short hairpin RNA (shRNA) and CRISPR/Cas9 increased triple-negative breast cancer (TNBC) cell proliferation and migration. Global transcriptome and chromatin occupancy analyses of ZNF148 revealed a central role in inhibiting cancer cell de-differentiation and migration. Mechanistically, we identified the Inhibitor of DNA binding 1 and 3 (ID1, ID3), drivers of cancer stemness and plasticity, as previously uncharacterized targets of transcriptional repression by ZNF148. Silencing of ZNF148 increased the stemness and tumorigenicity in TNBC cells. These findings uncover a previously unknown tumor suppressor role for ZNF148, and a transcriptional regulatory circuitry encompassing MYC, ZNF148, and ID1/3 in driving cancer stem cell traits in aggressive breast cancer.
Collapse
Affiliation(s)
- Mijeong Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Manjot Singh
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, 6027, Australia
| | - Bum-Kyu Lee
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY, 12144, USA
| | - Moira Hibbs
- RPH Research Centre, Royal Perth Hospital, Perth, WA, 6000, Australia
| | - Kirsty Richardson
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
| | - Lesley Ellies
- Division of Pharmacology and Toxicology, School of Biomedical Sciences, The University of Western Australia, Perth, WA, 6000, Australia
| | - Larissa Wintle
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
| | - Lisa M Stuart
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
| | - Jenny Y Wang
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Dominic C Voon
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, 920-1192, Japan
- Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Pilar Blancafort
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia
- School of Human Sciences, The University of Western Australia, Perth, WA, 6000, Australia
- The Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Jianlong Wang
- Department of Medicine, Columbia Center for Human Development, Columbia Stem Cell Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Peter J Leedman
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia.
| | - Andrew J Woo
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, WA, 6000, Australia.
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, 6027, Australia.
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, 6000, Australia.
| |
Collapse
|
35
|
Anti-Cancer Activity of Cannabis sativa Phytocannabinoids: Molecular Mechanisms and Potential in the Fight against Ovarian Cancer and Stem Cells. Cancers (Basel) 2022; 14:cancers14174299. [PMID: 36077833 PMCID: PMC9454933 DOI: 10.3390/cancers14174299] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy, with about 70% of cases diagnosed only at an advanced stage. Cannabis sativa, which produces more than 150 phytocannabinoids, is used worldwide to alleviate numerous symptoms associated with various medical conditions. Recently, studies across a range of cancer types have demonstrated that the phytocannabinoids Δ9-trans-tetrahydrocannabinol (THC) and cannabidiol (CBD) have anti-cancer activity in vitro and in vivo, but also the potential to increase other drugs’ adverse effects. THC and CBD act via several different biological and signaling pathways, including receptor-dependent and receptor-independent pathways. However, very few studies have examined the effectiveness of cannabis compounds against OC. Moreover, little is known about the effectiveness of cannabis compounds against cancer stem cells (CSCs) in general and OC stem cells (OCSCs) in particular. CSCs have been implicated in tumor initiation, progression, and invasion, as well as tumor recurrence, metastasis, and drug resistance. Several hallmarks and concepts describe CSCs. OCSCs, too, are characterized by several markers and specific drug-resistance mechanisms. While there is no peer-reviewed information regarding the effect of cannabis and cannabis compounds on OCSC viability or development, cannabis compounds have been shown to affect genetic pathways and biological processes related to CSCs and OCSCs. Based on evidence from other cancer-type studies, the use of phytocannabinoid-based treatments to disrupt CSC homeostasis is suggested as a potential intervention to prevent chemotherapy resistance. The potential benefits of the combination of chemotherapy with phytocannabinoid treatment should be examined in ovarian cancer patients.
Collapse
|
36
|
Current Opportunities for Targeting Dysregulated Neurodevelopmental Signaling Pathways in Glioblastoma. Cells 2022; 11:cells11162530. [PMID: 36010607 PMCID: PMC9406959 DOI: 10.3390/cells11162530] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most common and highly lethal type of brain tumor, with poor survival despite advances in understanding its complexity. After current standard therapeutic treatment, including tumor resection, radiotherapy and concomitant chemotherapy with temozolomide, the median overall survival of patients with this type of tumor is less than 15 months. Thus, there is an urgent need for new insights into GBM molecular characteristics and progress in targeted therapy in order to improve clinical outcomes. The literature data revealed that a number of different signaling pathways are dysregulated in GBM. In this review, we intended to summarize and discuss current literature data and therapeutic modalities focused on targeting dysregulated signaling pathways in GBM. A better understanding of opportunities for targeting signaling pathways that influences malignant behavior of GBM cells might open the way for the development of novel GBM-targeted therapies.
Collapse
|
37
|
Shi X, Zhang Q, Wang J, Zhang Y, Yan Y, Liu Y, Yang N, Wang Q, Xu X. Differential expression analysis of mRNAs, lncRNAs, and miRNAs expression profiles and construction of ceRNA networks in PEDV infection. BMC Genomics 2022; 23:586. [PMID: 35964002 PMCID: PMC9375197 DOI: 10.1186/s12864-022-08805-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 07/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Porcine Epidemic Diarrhea Virus (PEDV) is a coronavirus that seriously affects the swine industry. MicroRNAs and long noncoding RNAs are two relevant non-coding RNAs (ncRNAs) class and play crucial roles in a variety of physiological processes. Increased evidence indicates a complex interaction between mRNA and ncRNA. However, our understanding of the function of ncRNA involved in host-PEDV interaction is limited. RESULTS A total of 1,197 mRNA transcripts, 539 lncRNA transcripts, and 208 miRNA transcripts were differentially regulated at 24 h and 48 h post-infection. Gene ontology (GO) and KEGG pathway enrichment analysis showed that DE mRNAs and DE lncRNAs were mainly involved in biosynthesis, innate immunity, and lipid metabolism. Moreover, we constructed a miRNA-mRNA-pathway network using bioinformatics, including 12 DE mRNAs, 120 DE miRNAs, and 11 pathways. Finally, the target genes of DE miRNAs were screened by bioinformatics, and we constructed immune-related lncRNA-miRNA-mRNA ceRNA networks. Then, the selected DE genes were validated by qRT-PCR, which were consistent with the results from RNA-Seq data. CONCLUSIONS This study provides the comprehensive analysis of the expression profiles of mRNAs, lncRNAs, and miRNAs during PEDV infection. We characterize the ceRNA networks which can provide new insights into the pathogenesis of PEDV.
Collapse
Affiliation(s)
- Xiaojie Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Qi Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Jingjing Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Yuting Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Yuchao Yan
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Yi Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Naling Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Quanqiong Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China
| | - Xingang Xu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Xianyang, 712100, Shaanxi, China.
| |
Collapse
|
38
|
ID1 marks the tumorigenesis of pancreatic ductal adenocarcinoma in mouse and human. Sci Rep 2022; 12:13555. [PMID: 35941362 PMCID: PMC9359991 DOI: 10.1038/s41598-022-17827-3] [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: 04/28/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a deadly disease that has an increasing death rate but no effective treatment to now. Although biological and immunological hallmarks of PDAC have been frequently reported recently, early detection and the particularly aggressive biological features are the major challenges remaining unclear. In the current study, we retrieved multiple scRNA-seq datasets and illustrated the genetic programs of PDAC development in genetically modified mouse models. Notably, the transcription levels of Id1 were elevated specifically along with the PDAC development. Pseudotime trajectory analysis revealed that Id1 was closely correlated with the malignancy of PDAC. The gene expression patterns of human PDAC cells were determined by the comparative analysis of the scRNA-seq data on human PDAC and normal pancreas tissues. ID1 levels in human PDAC cancer cells were dramatically increased compared to normal epithelial cells. ID1 deficiency in vitro significantly blunt the invasive tumor-formation related phenotypes. IPA analysis on the differentially expressed genes suggested that EIF2 signaling was the core pathway regulating the development of PDAC. Blocking EFI2 signaling remarkably decreased the expression of ID1 and attenuated the tumor-formation related phenotypes. These observations confirmed that ID1 was regulated by EIF2 signaling and was the critical determinator of PDAC development and progression. This study suggests that ID1 is a potential malignant biomarker of PDAC in both mouse models and human and detecting and targeting ID1 may be a promising strategy to treat or even rescue PDAC.
Collapse
|
39
|
Singh S, Sarkar T, Jakubison B, Gadomski S, Spradlin A, Gudmundsson KO, Keller JR. Inhibitor of DNA binding proteins revealed as orchestrators of steady state, stress and malignant hematopoiesis. Front Immunol 2022; 13:934624. [PMID: 35990659 PMCID: PMC9389078 DOI: 10.3389/fimmu.2022.934624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
Adult mammalian hematopoiesis is a dynamic cellular process that provides a continuous supply of myeloid, lymphoid, erythroid/megakaryocyte cells for host survival. This process is sustained by regulating hematopoietic stem cells (HSCs) quiescence, proliferation and activation under homeostasis and stress, and regulating the proliferation and differentiation of downstream multipotent progenitor (MPP) and more committed progenitor cells. Inhibitor of DNA binding (ID) proteins are small helix-loop-helix (HLH) proteins that lack a basic (b) DNA binding domain present in other family members, and function as dominant-negative regulators of other bHLH proteins (E proteins) by inhibiting their transcriptional activity. ID proteins are required for normal T cell, B cell, NK and innate lymphoid cells, dendritic cell, and myeloid cell differentiation and development. However, recent evidence suggests that ID proteins are important regulators of normal and leukemic hematopoietic stem and progenitor cells (HSPCs). This chapter will review our current understanding of the function of ID proteins in HSPC development and highlight future areas of scientific investigation.
Collapse
Affiliation(s)
- Shweta Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Tanmoy Sarkar
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Brad Jakubison
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Stephen Gadomski
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Andrew Spradlin
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
| | - Kristbjorn O. Gudmundsson
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Jonathan R. Keller
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI)- Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
- *Correspondence: Jonathan R. Keller,
| |
Collapse
|
40
|
Lu Z, Zhang Z, Yang M, Xiao M. Ubiquitin-specific protease 1 inhibition sensitizes hepatocellular carcinoma cells to doxorubicin by ubiquitinated proliferating cell nuclear antigen-mediated attenuation of stemness. Anticancer Drugs 2022; 33:622-631. [PMID: 35324534 DOI: 10.1097/cad.0000000000001311] [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: 12/24/2022]
Abstract
Currently, resistance to the chemotherapeutic agent doxorubicin (Dox) in hepatocellular carcinoma (HCC) cells is an obstacle in developing effective Dox-targeted clinical therapies. Ubiquitin-specific protease 1 (USP1) plays a crucial role in the progression of multiple cancers. In this study, the purpose was to investigate the effect of USP1 depletion with chemotherapeutant Dox on the HCC cells. Flow cytometry was used to detect the ratio of apoptosis. The expression levels of selected proteins were evaluated by western blotting. In addition, the expression of genes was quantitated by quantitative real-time PCR assay. Coimmunoprecipitation was performed to confirm the interaction between USP1 and proliferating cell nuclear antigen (PCNA). Sphere formation assay was carried out to investigate the cancer stemness. Subcutaneous xenograft and orthotopic liver tumor models were established to examine the growth of tumor. Knockdown of USP1 increased the rate of Dox-induced apoptosis in stem-like and nonstem-like HCC cells. The combination of Dox and the USP1 inhibitor SJB3-019A (SJB3) markedly enhanced apoptosis in the primary liver carcinoma/PRF/5 and MHCC-97H cell lines. Notably, Dox/SJB3-induced tumor inhibition was further determined in vivo using a xenograft and orthotopic liver tumor model. Mechanically, USP1 inhibition via SJB3 or short hairpin RNA significantly decreased cancer stemness, including sphere formation ability and the expression of Nanog, Sox2, and c-Myc. The sensitization of HCC to Dox by SJB3 is attributed to the upregulation of PCNA ubiquitylation. Thus, genetic or pharmacological inhibition of USP1 restored the sensitivity of HCC cells to Dox in vitro and in vivo , representing a new potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Zhe Lu
- Clinical Laboratory, Women and Children's Health Care Center of Hainan Province and Departments of
| | | | - Min Yang
- Medical Oncology, Hainan Cancer Hospital, Haikou, P.R. China
| | - Meifang Xiao
- Clinical Laboratory, Women and Children's Health Care Center of Hainan Province and Departments of
| |
Collapse
|
41
|
Jakubison BL, Sarkar T, Gudmundsson KO, Singh S, Sun L, Morris HM, Klarmann KD, Keller JR. ID2 and HIF-1α collaborate to protect quiescent hematopoietic stem cells from activation, differentiation, and exhaustion. J Clin Invest 2022; 132:152599. [PMID: 35775482 PMCID: PMC9246389 DOI: 10.1172/jci152599] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
Defining mechanism(s) that maintain tissue stem quiescence is important for improving tissue regeneration, cell therapies, aging, and cancer. We report here that genetic ablation of Id2 in adult hematopoietic stem cells (HSCs) promotes increased HSC activation and differentiation, which results in HSC exhaustion and bone marrow failure over time. Id2Δ/Δ HSCs showed increased cycling, ROS production, mitochondrial activation, ATP production, and DNA damage compared with Id2+/+ HSCs, supporting the conclusion that Id2Δ/Δ HSCs are less quiescent. Mechanistically, HIF-1α expression was decreased in Id2Δ/Δ HSCs, and stabilization of HIF-1α in Id2Δ/Δ HSCs restored HSC quiescence and rescued HSC exhaustion. Inhibitor of DNA binding 2 (ID2) promoted HIF-1α expression by binding to the von Hippel-Lindau (VHL) protein and interfering with proteasomal degradation of HIF-1α. HIF-1α promoted Id2 expression and enforced a positive feedback loop between ID2 and HIF-1α to maintain HSC quiescence. Thus, sustained ID2 expression could protect HSCs during stress and improve HSC expansion for gene editing and cell therapies.
Collapse
Affiliation(s)
- Brad L Jakubison
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.,Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI) - Frederick, NIH, Frederick, Maryland, USA
| | - Tanmoy Sarkar
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI) - Frederick, NIH, Frederick, Maryland, USA
| | - Kristbjorn O Gudmundsson
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.,Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI) - Frederick, NIH, Frederick, Maryland, USA
| | - Shweta Singh
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI) - Frederick, NIH, Frederick, Maryland, USA
| | - Lei Sun
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI) - Frederick, NIH, Frederick, Maryland, USA
| | - Holly M Morris
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI) - Frederick, NIH, Frederick, Maryland, USA
| | - Kimberly D Klarmann
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Jonathan R Keller
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA.,Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute (NCI) - Frederick, NIH, Frederick, Maryland, USA
| |
Collapse
|
42
|
Park CG, Choi SH, Lee SY, Eun K, Park MG, Jang J, Jeong HJ, Kim SJ, Jeong S, Lee K, Kim H. Cytoplasmic LMO2-LDB1 Complex Activates STAT3 Signaling through Interaction with gp130-JAK in Glioma Stem Cells. Cells 2022; 11:cells11132031. [PMID: 35805116 PMCID: PMC9265747 DOI: 10.3390/cells11132031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/19/2022] [Accepted: 06/24/2022] [Indexed: 12/10/2022] Open
Abstract
The oncogenic role of nuclear LIM domain only 2 (LMO2) as a transcriptional regulator is well established, but its function in the cytoplasm is largely unknown. Here, we identified LMO2 as a cytoplasmic activator for signal transducer and activator of transcription 3 (STAT3) signaling in glioma stem cells (GSCs) through biochemical and bioinformatics analyses. LMO2 increases STAT3 phosphorylation by interacting with glycoprotein 130 (gp130) and Janus kinases (JAKs). LMO2-driven activation of STAT3 signaling requires the LDB1 protein and leads to increased expression of an inhibitor of differentiation 1 (ID1), a master regulator of cancer stemness. Our findings indicate that the cytoplasmic LMO2-LDB1 complex plays a crucial role in the activation of the GSC signaling cascade via interaction with gp130 and JAK1/2. Thus, LMO2-LDB1 is a bona fide oncogenic protein complex that activates either the JAK-STAT signaling cascade in the cytoplasm or direct transcriptional regulation in the nucleus.
Collapse
Affiliation(s)
- Cheol Gyu Park
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Sang-Hun Choi
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Seon Yong Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Kiyoung Eun
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Korea
| | - Min Gi Park
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Junseok Jang
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Hyeon Ju Jeong
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Seong Jin Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Sohee Jeong
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Kanghun Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
| | - Hyunggee Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul 02841, Korea; (C.G.P.); (S.-H.C.); (S.Y.L.); (K.E.); (M.G.P.); (J.J.); (H.J.J.); (S.J.K.); (S.J.); (K.L.)
- Institute of Animal Molecular Biotechnology, Korea University, Seoul 02841, Korea
- Correspondence: ; Tel.: +82-2-3290-3059; Fax: +82-2-3290-3040
| |
Collapse
|
43
|
Combs LR, Spires LM, Alonso JD, Papp B, Toth Z. KSHV RTA Induces Degradation of the Host Transcription Repressor ID2 To Promote the Viral Lytic Cycle. J Virol 2022; 96:e0010122. [PMID: 35604218 PMCID: PMC9215225 DOI: 10.1128/jvi.00101-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/05/2022] [Indexed: 11/20/2022] Open
Abstract
The immediate early viral protein replication and transcription activator (RTA) of Kaposi's sarcoma-associated herpesvirus (KSHV) is essential for activating the lytic cycle of KSHV. RTA induces the KSHV lytic cycle by several mechanisms, acting as a viral transcription factor that directly induces viral and host genes and acting as a viral E3 ubiquitin ligase by degrading host proteins that block viral lytic replication. Recently, we have characterized the global gene expression changes in primary effusion lymphoma (PEL) upon lytic reactivation of KSHV, which also led to the identification of rapidly downregulated genes such as ID2, an inhibitor of basic helix-loop-helix transcription factors. Here, we demonstrate that ID2 overexpression in PEL ablates KSHV lytic reactivation, indicating that ID2 inhibits the KSHV lytic cycle. Furthermore, we show that while ID2 is highly expressed during latency, its protein level is rapidly reduced by 4 h postinduction during lytic reactivation. Our results indicate that RTA binds to ID2 and induces its degradation during the KSHV lytic cycle by N-terminal ubiquitination through the ubiquitin-proteasome pathway. Importantly, we found that not only KSHV RTA but also its Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68) homologs interact with ID2, and they can induce the degradation of all four members of the ID protein family, suggesting an evolutionarily conserved interplay between gammaherpesvirus RTAs and ID proteins. Taken together, we propose that ID2 acts as a repressor of the KSHV lytic cycle, which is counteracted by its RTA-mediated degradation. We also predict that ID proteins may act as restriction factors of the lytic phase of the other gammaherpesviruses as well. IMPORTANCE In addition to its transcription regulatory role, RTA is also known to have an E3 ubiquitin ligase activity, which RTA utilizes for inducing protein degradation. However, it is still largely unknown what host factors are downregulated during KSHV lytic reactivation by RTA-mediated protein degradation and what the biological significance of the degradation of these host factors is. In this study, we discovered that RTA employs N-terminal ubiquitination to induce degradation of ID2, a potent transcription repressor of host genes, via the ubiquitin-proteasome pathway to promote KSHV lytic reactivation in PEL cells. Furthermore, we found that not only KSHV RTA but also RTA of EBV and MHV68 gammaherpesviruses can induce the degradation of all four human ID proteins, indicating that the interplay between gammaherpesvirus RTAs and ID proteins is evolutionarily conserved.
Collapse
Affiliation(s)
- Lauren R. Combs
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Lauren McKenzie Spires
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Juan D. Alonso
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Bernadett Papp
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
- University of Florida Genetics Institute, Gainesville, Florida, USA
- University of Florida Health Cancer Center, Gainesville, Florida, USA
- University of Florida Center for Orphaned Autoimmune Disorders, Gainesville, Florida, USA
- University of Florida Informatics Institute, Gainesville, Florida, USA
| | - Zsolt Toth
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
- University of Florida Genetics Institute, Gainesville, Florida, USA
- University of Florida Health Cancer Center, Gainesville, Florida, USA
| |
Collapse
|
44
|
Wang T, Zhou J, Wang Y, Zheng Q, Lin Z, Li G, Mei J, Liu L. Clinicopathological characteristics and prognosis of resectable lung adenosquamous carcinoma: a population-based study of the SEER database. Jpn J Clin Oncol 2022; 52:1191-1200. [PMID: 35726160 DOI: 10.1093/jjco/hyac096] [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: 11/18/2021] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Adenosquamous carcinoma is a rare subtype of non-small cell lung cancer characterized by aggressive behavior, with combination of adenocarcinoma and squamous cell carcinoma components. The clinicopathological characteristics and prognosis of resectable adenosquamous carcinoma are incompletely understood and this study aimed to depict those in a large population. METHODS A total of 805 adenosquamous carcinoma, 7875 squamous cell carcinoma and 23 957 adenocarcinoma patients who underwent lobectomy or sublobectomy were queried from the Surveillance, Epidemiology, and End Results database (2010-17). Clinicopathological characteristics of adenosquamous carcinoma patients were compared with those of squamous cell carcinoma and adenocarcinoma patients. Prognostic factors were identified by univariable and multivariable Cox regression analyses. Propensity score matching was applied to reduce confounding effects. RESULTS Adenosquamous carcinoma was associated with higher pleural invasion incidence and poorer differentiation compared with squamous cell carcinoma or adenocarcinoma (P values < 0.001). The independent risk factors of cancer-specific survival of adenosquamous carcinoma patients were increasing age, male sex, invading through visceral pleura, poor differentiation and higher stage. Stage IB adenosquamous carcinoma patients whose tumor invaded through visceral pleura had significantly worse survival than those not (P = 0.003). Adenosquamous carcinoma patients had worse survival compared with squamous cell carcinoma (5-year-survival: 64.55 vs. 69.09%, P = 0.003) and adenocarcinoma (5-year-survival: 64.55 vs. 76.79%, P < 0.001) patients before match. And this difference persisted after match. CONCLUSIONS Resectable adenosquamous carcinoma patients had higher pleural invasion incidence, poorer differentiation and worse survival compared with squamous cell carcinoma and adenocarcinoma patients. Visceral pleural invasion status and differentiation grade were vital prognostic factors of adenosquamous carcinoma patients on the basis of stage.
Collapse
Affiliation(s)
- Tengyong Wang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| | - Jian Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| | - Yaxin Wang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Quan Zheng
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| | - Zhangyu Lin
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Guangchen Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiandong Mei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Koppenhafer SL, Goss KL, Voigt E, Croushore E, Terry WW, Ostergaard J, Gordon PM, Gordon DJ. Inhibitor of DNA binding 2 (ID2) regulates the expression of developmental genes and tumorigenesis in ewing sarcoma. Oncogene 2022; 41:2873-2884. [PMID: 35422476 PMCID: PMC9107507 DOI: 10.1038/s41388-022-02310-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 11/24/2022]
Abstract
Sarcomas are difficult to treat and the therapy, even when effective, is associated with long-term and life-threatening side effects. In addition, the treatment regimens for many sarcomas, including Ewing sarcoma, rhabdomyosarcoma, and osteosarcoma, are relatively unchanged over the past two decades, indicating a critical lack of progress. Although differentiation-based therapies are used for the treatment of some cancers, the application of this approach to sarcomas has proven challenging. Here, using a CRISPR-mediated gene knockout approach, we show that Inhibitor of DNA Binding 2 (ID2) is a critical regulator of developmental-related genes and tumor growth in vitro and in vivo in Ewing sarcoma tumors. We also identified that homoharringtonine, which is an inhibitor of protein translation and FDA-approved for the treatment of leukemia, decreases the level of the ID2 protein and significantly reduces tumor growth and prolongs mouse survival in an Ewing sarcoma xenograft model. Furthermore, in addition to targeting ID2, homoharringtonine also reduces the protein levels of ID1 and ID3, which are additional members of the ID family of proteins with well-described roles in tumorigenesis, in multiple types of cancer. Overall, these results provide insight into developmental regulation in Ewing sarcoma tumors and identify a novel, therapeutic approach to target the ID family of proteins using an FDA-approved drug.
Collapse
Affiliation(s)
- Stacia L Koppenhafer
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kelli L Goss
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Ellen Voigt
- Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Emma Croushore
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - William W Terry
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA
| | - Jason Ostergaard
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Peter M Gordon
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - David J Gordon
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
46
|
Lee SB, Garofano L, Ko A, D'Angelo F, Frangaj B, Sommer D, Gan Q, Kim K, Cardozo T, Iavarone A, Lasorella A. Regulated interaction of ID2 with the anaphase-promoting complex links progression through mitosis with reactivation of cell-type-specific transcription. Nat Commun 2022; 13:2089. [PMID: 35440621 PMCID: PMC9018835 DOI: 10.1038/s41467-022-29502-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 03/13/2022] [Indexed: 12/05/2022] Open
Abstract
Tissue-specific transcriptional activity is silenced in mitotic cells but it remains unclear whether the mitotic regulatory machinery interacts with tissue-specific transcriptional programs. We show that such cross-talk involves the controlled interaction between core subunits of the anaphase-promoting complex (APC) and the ID2 substrate. The N-terminus of ID2 is independently and structurally compatible with a pocket composed of core APC/C subunits that may optimally orient ID2 onto the APCCDH1 complex. Phosphorylation of serine-5 by CDK1 prevented the association of ID2 with core APC, impaired ubiquitylation and stabilized ID2 protein at the mitosis-G1 transition leading to inhibition of basic Helix-Loop-Helix (bHLH)-mediated transcription. The serine-5 phospho-mimetic mutant of ID2 that inefficiently bound core APC remained stable during mitosis, delayed exit from mitosis and reloading of bHLH transcription factors on chromatin. It also locked cells into a "mitotic stem cell" transcriptional state resembling the pluripotent program of embryonic stem cells. The substrates of APCCDH1 SKP2 and Cyclin B1 share with ID2 the phosphorylation-dependent, D-box-independent interaction with core APC. These results reveal a new layer of control of the mechanism by which substrates are recognized by APC.
Collapse
Affiliation(s)
- Sang Bae Lee
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA.
- Division of Life Sciences, Jeonbuk National University, Jeonju, 54896, Republic of Korea.
| | - Luciano Garofano
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA
| | - Aram Ko
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA
| | - Fulvio D'Angelo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA
| | - Brulinda Frangaj
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA
| | - Danika Sommer
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA
| | - Qiwen Gan
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA
| | - KyeongJin Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Timothy Cardozo
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, 10016, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, 10032, USA.
- Department of Neurology, Columbia University Medical Center, New York, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, 10032, USA.
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York, 10032, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, 10032, USA.
- Department of Pediatrics, Columbia University Medical Center, New York, 10032, USA.
| |
Collapse
|
47
|
Kozlov AP. Mammalian tumor-like organs. 2. Mammalian adipose has many tumor features and obesity is a tumor-like process. Infect Agent Cancer 2022; 17:15. [PMID: 35395810 PMCID: PMC8994355 DOI: 10.1186/s13027-022-00423-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND In previous publications, the author developed the theory of carcino-evo-devo, which predicts that evolutionarily novel organs should recapitulate some features of tumors in their development. MAIN TEXT Mammalian adipose is currently recognized as a multi-depot metabolic and endocrine organ consisting of several adipose tissues. Although lipid-storing cells and proteins are ancient, the adipose organ as a whole is evolutionarily novel to mammals. The adipose expansion has remarkable similarities with the growth of solid tumors. These similarities are the following: (1) The capability to unlimited expansion; (2) Reversible plasticity; (3) Induction of angiogenesis; (4) Chronic inflammation; (5) Remodeling and disfunction; (6) Systemic influence on the organism; (7) Hormone production; (8) Production of miRNAs that influence other tissues; (9) Immunosuppression; (10) DNA damage and resistance to apoptosis; (11) Destructive infiltration in other organs and tissues. These similarities include the majority of "hallmarks of cancer". In addition, lipomas are the most frequent soft tissue tumors, and similar drugs may be used for the treatment of obesity and cancer by preventing infiltration. This raises the possibility that obesity, at least in part, may represent an oncological problem. The existing similarities between adipose and tumors suggest the possible evolutionary origin of mammalian adipose from some ancestral benign mesenchymal hereditary tumors. Indeed, using a transgenic inducible zebrafish tumor model, we described many genes, which originated in fish and were expressed in fish tumors. Their human orthologs LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA acquired functions connected with the adipose organ. They are also involved in tumor development in humans. CONCLUSION If the hypothesis of the evolutionary origin of the adipose organ from the ancestral hereditary tumor is correct, it may open new opportunities to resolve the oncological problem and the problem of the obesity epidemic. New interventions targeting LEP, NOTCH1, SPRY1, PPARG, ID2, and CIDEA gene network, in addition to what already is going on, can be designed for treatment and prevention of both obesity and tumors.
Collapse
Affiliation(s)
- A P Kozlov
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 3, Gubkina Street, Moscow, Russia, 117971.
- Peter the Great St. Petersburg Polytechnic University, 29, Polytekhnicheskaya Street, St. Petersburg, Russia, 195251.
- The Biomedical Center, 8, Viborgskaya Street, St. Petersburg, Russia, 194044.
| |
Collapse
|
48
|
Hongu T, Pein M, Insua-Rodríguez J, Gutjahr E, Mattavelli G, Meier J, Decker K, Descot A, Bozza M, Harbottle R, Trumpp A, Sinn HP, Riedel A, Oskarsson T. Perivascular tenascin C triggers sequential activation of macrophages and endothelial cells to generate a pro-metastatic vascular niche in the lungs. NATURE CANCER 2022; 3:486-504. [PMID: 35469015 PMCID: PMC9046090 DOI: 10.1038/s43018-022-00353-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/24/2022] [Indexed: 02/07/2023]
Abstract
Disseminated cancer cells frequently lodge near vasculature in secondary organs. However, our understanding of the cellular crosstalk invoked at perivascular sites is still rudimentary. Here, we identify intercellular machinery governing formation of a pro-metastatic vascular niche during breast cancer colonization in the lung. We show that specific secreted factors, induced in metastasis-associated endothelial cells (ECs), promote metastasis in mice by enhancing stem cell properties and the viability of cancer cells. Perivascular macrophages, activated via tenascin C (TNC) stimulation of Toll-like receptor 4 (TLR4), were shown to be crucial in niche activation by secreting nitric oxide (NO) and tumor necrosis factor (TNF) to induce EC-mediated production of niche components. Notably, this mechanism was independent of vascular endothelial growth factor (VEGF), a key regulator of EC behavior and angiogenesis. However, targeting both macrophage-mediated vascular niche activation and VEGF-regulated angiogenesis resulted in added potency to curb lung metastasis in mice. Together, our findings provide mechanistic insights into the formation of vascular niches in metastasis.
Collapse
Affiliation(s)
- Tsunaki Hongu
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Maren Pein
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Jacob Insua-Rodríguez
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Ewgenija Gutjahr
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Greta Mattavelli
- Mildred Scheel Early Career Center, University Hospital of Würzburg, Würzburg, Germany
| | - Jasmin Meier
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Kristin Decker
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Arnaud Descot
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Matthias Bozza
- DNA Vector Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - Richard Harbottle
- DNA Vector Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
- DKFZ-ZMBH Alliance, Heidelberg, Germany
- German Cancer Consortium, Heidelberg, Germany
| | - Hans-Peter Sinn
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Angela Riedel
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
- Mildred Scheel Early Career Center, University Hospital of Würzburg, Würzburg, Germany
| | - Thordur Oskarsson
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.
- Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany.
- German Cancer Consortium, Heidelberg, Germany.
- Department of Molecular Oncology and Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
49
|
Togni C, Rom E, Burghardt I, Roth P, Rushing EJ, Weller M, Gramatzki D. Prognostic Relevance of Transforming Growth Factor-β Receptor Expression and Signaling in Glioblastoma, Isocitrate Dehydrogenase-Wildtype. J Neuropathol Exp Neurol 2022; 81:225-235. [PMID: 35190826 DOI: 10.1093/jnen/nlac007] [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: 11/14/2022] Open
Abstract
The transforming growth factor (TGF)-β signaling pathway has been recognized as a major factor in promoting the aggressive behavior of glioblastoma, isocitrate dehydrogenase-wildtype. However, there is little knowledge about the expression of TGF-β receptors in glioblastoma. Here, we studied the expression patterns of TGF-β receptor II (TGFβRII), type I receptors activin receptor-like kinase (ALK)-5, and ALK-1, as well as of the transcriptional regulators inhibitor of differentiation (Id) 2, Id3, and Id4 in human glioblastoma. The expression of TGFβRII, ALK-5, and ALK-1 varied greatly, with TGFβRII and ALK-5 being the most abundant and ALK-1 being the least expressed receptor. None of the 3 receptors was preferentially expressed by tumor vasculature as opposed to the tumor bulk, indicating tumor bulk-governed mechanisms of TGF-β signaling with regard to glioblastoma-associated angiogenesis. A positive correlation was found between ALK-1 and Id2, suggesting that Id2, broadly expressed in the tumor cells, is a downstream target of this receptor-dependent pathway. Furthermore, there was a trend for high expression of ALK-5 or Id2 to be associated with inferior overall survival. Hence, we propose that ALK-5 may be used for patient stratification in future anti-TGF-β treatment trials and that Id2 might be a potential target for anti-TGF-β interventions.
Collapse
Affiliation(s)
- Claudio Togni
- From the Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Emanuel Rom
- From the Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Isabel Burghardt
- From the Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Patrick Roth
- From the Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Elisabeth J Rushing
- Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Michael Weller
- From the Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Dorothee Gramatzki
- From the Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
50
|
Yoshida Y, Yuki K, Dan S, Yamazaki K, Noda M. Suppression of tumor metastasis by a RECK-activating small molecule. Sci Rep 2022; 12:2319. [PMID: 35149728 PMCID: PMC8837781 DOI: 10.1038/s41598-022-06288-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
RECK encodes a membrane-anchored protease-regulator which is often downregulated in a wide variety of cancers, and reduced RECK expression often correlates with poorer prognoses. In mouse models, forced expression of RECK in tumor xenografts results in suppression of tumor angiogenesis, invasion, and metastasis. RECK mutations, however, are rare in cancer genomes, suggesting that agents that re-activate dormant RECK may be of clinical value. We found a potent RECK-inducer, DSK638, that inhibits spontaneous lung metastasis in our mouse xenograft model. Induction of RECK expression involves SP1 sites in its promoter and may be mediated by KLF2. DSK638 also upregulates MXI1, an endogenous MYC-antagonist, and inhibition of metastasis by DSK638 is dependent on both RECK and MXI1. This study demonstrates the utility of our approach (using a simple reporter assay followed by multiple phenotypic assays) and DSK638 itself (as a reference compound) in finding potential metastasis-suppressing drugs.
Collapse
Affiliation(s)
- Yoko Yoshida
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, 606-8501, Japan. .,Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, 135-8550, Japan.
| | - Kanako Yuki
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, 135-8550, Japan
| | - Kanami Yamazaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo, 135-8550, Japan
| | - Makoto Noda
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|