1
|
Bhartiya D, Sharma N, Tripathi A, Tripathi A. Do PGCCs in Solid Tumors Appear Due to Treatment-related Stress or Clonal Expansion of CSCs that Survive Oncotherapy? Stem Cell Rev Rep 2025:10.1007/s12015-025-10891-y. [PMID: 40338514 DOI: 10.1007/s12015-025-10891-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
Dedifferentiation of epithelial cells during epithelial-mesenchymal transition (EMT) results in circulating tumor cells (CTCs) that are mobilized singly or in clusters in association with blood cells and results in metastasis. However, lineage tracing studies have failed to delineate any role of EMT during metastasis. Research is also focused on polyploid giant cancer cells (PGCCs) in solid tumors which appear in response to oncotherapy-related stress for their role in metastasis. But how to explain PGCCs role in metastatic tumors in treatment-naïve patients? Studies done using mouse models and clinical samples suggest that cancer initiates due to dysfunctions of tissue-resident, pluripotent very small embryonic-like stem cells (VSELs). VSELs are the most primitive and pluripotent stem cells that exist at top of cellular hierarchy in multiple tissues. They are normally quiescent and undergo asymmetrical cell divisions to give rise to two cells of different sizes and fates including smaller cells to self-renew and bigger tissue-specific progenitors. Progenitors undergo symmetrical cell divisions and clonal expansion (rapid proliferation, endoduplication with incomplete cytokinesis) to form giant cells that further breakdown and differentiate into tissue-specific cell types. Oncotherapy destroys actively dividing cells, but CSCs survive. We hypothesize that excessive self-renewal and clonal expansion of cancer stem cells (CSCs, dysfunctional VSELs) result in multinucleated giant cells (PGCCs) that accumulate as further differentiation into tissue-specific cell types is blocked in cancerous conditions. PGCCS are being reported by multiple groups whereas CSCs remain elusive due to small size and low abundance and actually contribute to both cancer initiation and metastasis.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel, Mumbai, 400013, India.
| | - Nripen Sharma
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel, Mumbai, 400013, India
| | - Anish Tripathi
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel, Mumbai, 400013, India
| | - Ashish Tripathi
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel, Mumbai, 400013, India
- TZAR Labs, 23Ikigai Pte Ltd., 30 Cecil Street, #21-08 Prudential Tower, Singapore, 049712, Singapore
| |
Collapse
|
2
|
Bhartiya D, Dutta S, Tripathi A, Tripathi A. Misconceptions Thrive in the Field of Cancer as Technological Advances Continue to Confuse Stem Cell Biology. Stem Cell Rev Rep 2025:10.1007/s12015-025-10880-1. [PMID: 40238074 DOI: 10.1007/s12015-025-10880-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2025] [Indexed: 04/18/2025]
Abstract
Despite the huge thrust on targeted therapies, cancer survival rates have not improved and both cancer incidence and fatalities continue to rise globally. There is no consensus on how cancer initiates and two contrasting views were published in 2024 regarding cancer initiation. Based on the premise that no stem cells exist in tissues like liver, lungs, and pancreas but they are still affected by cancer; it was suggested that somatic cells dedifferentiate and undergo 'paligenosis' to initiate cancer. The second view discussed that tissue-resident, very small embryonic-like stem cells (VSELs) are vulnerable to extrinsic/intrinsic insults and their dysfunctions initiate cancer. The present article examines the underlying technical reasons that have led to these conflicting views. Scientists have struggled to detect quiescent cancer stem cells (CSCs) that survive chemotherapy, and radiotherapy and escape immunotherapy, cause recurrence and eventually therapeutic resistance leading to death. Lineage tracing studies fail to detect quiescent, acyclic stem cells and instead, the role of actively dividing LGR5+ cells was highlighted for tumor initiation, growth, and metastasis. Similarly, technologies like flow cytometry, and single-cell RNAseq, widely used to comprehend cancer biology, provide insights into cell populations present in abundance. Our article reviews why VSELs/CSCs in the pancreas have remained elusive despite employing advanced technologies, and the critique can be generalized to multiple other organs. This understanding is crucial as it will help to develop better therapeutic strategies for cancer, offer early detection when cancer is a weak disease, and pave the path for prevention over treatment.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel (West), Mumbai, 400013, India.
| | - Shruti Dutta
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel (West), Mumbai, 400013, India
| | - Anish Tripathi
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel (West), Mumbai, 400013, India
| | - Ashish Tripathi
- Epigeneres Biotech Pvt Ltd, Todi Mill Compound, Senapati Bapat Marg, Lower Parel (West), Mumbai, 400013, India
- TZAR Labs, 23Ikigai Pte Ltd., 30 Cecil Street, #21-08 Prudential Tower, Singapore, 049712, Singapore
| |
Collapse
|
3
|
Leck LYW, Abd El-Aziz YS, McKelvey KJ, Park KC, Sahni S, Lane DJR, Skoda J, Jansson PJ. Cancer stem cells: Masters of all traits. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167549. [PMID: 39454969 DOI: 10.1016/j.bbadis.2024.167549] [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: 02/05/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Cancer is a heterogeneous disease, which contributes to its rapid progression and therapeutic failure. Besides interpatient tumor heterogeneity, tumors within a single patient can present with a heterogeneous mix of genetically and phenotypically distinct subclones. These unique subclones can significantly impact the traits of cancer. With the plasticity that intratumoral heterogeneity provides, cancers can easily adapt to changes in their microenvironment and therapeutic exposure. Indeed, tumor cells dynamically shift between a more differentiated, rapidly proliferating state with limited tumorigenic potential and a cancer stem cell (CSC)-like state that resembles undifferentiated cellular precursors and is associated with high tumorigenicity. In this context, CSCs are functionally located at the apex of the tumor hierarchy, contributing to the initiation, maintenance, and progression of tumors, as they also represent the subpopulation of tumor cells most resistant to conventional anti-cancer therapies. Although the CSC model is well established, it is constantly evolving and being reshaped by advancing knowledge on the roles of CSCs in different cancer types. Here, we review the current evidence of how CSCs play a pivotal role in providing the many traits of aggressive tumors while simultaneously evading immunosurveillance and anti-cancer therapy in several cancer types. We discuss the key traits and characteristics of CSCs to provide updated insights into CSC biology and highlight its implications for therapeutic development and improved treatment of aggressive cancers.
Collapse
Affiliation(s)
- Lionel Y W Leck
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Yomna S Abd El-Aziz
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Oral Pathology Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - Kelly J McKelvey
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Kyung Chan Park
- Proteina Co., Ltd./Seoul National University, Seoul, South Korea
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia
| | - Darius J R Lane
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jan Skoda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.
| | - Patric J Jansson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute, Faculty of Medicine and Health, The University of Sydney, St Leonards, NSW, Australia; Cancer Drug Resistance & Stem Cell Program, School of Medical Science, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
4
|
Farmakis D, Stravopodis DJ, Prombona A. TH301 Emerges as a Novel Anti-Oncogenic Agent for Human Pancreatic Cancer Cells: The Dispensable Roles of p53, CRY2 and BMAL1 in TH301-Induced CDKN1A/p21 CIP1/WAF1 Upregulation. Int J Mol Sci 2024; 26:178. [PMID: 39796036 PMCID: PMC11720130 DOI: 10.3390/ijms26010178] [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: 12/02/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Pancreatic Ductal Adeno-Carcinoma (PDAC) is a highly aggressive cancer, with limited treatment options. Disruption of the circadian clock, which regulates key cellular processes, has been implicated in PDAC initiation and progression. Hence, targeting circadian clock components may offer new therapeutic opportunities for the disease. This study investigates the cytopathic effects of TH301, a novel CRY2 stabilizer, on PDAC cells, aiming to evaluate its potential as a novel therapeutic agent. Methods: PDAC cell lines (AsPC-1, BxPC-3 and PANC-1) were treated with TH301, and cell viability, cell cycle progression, apoptosis, autophagy, circadian gene, and protein expression profiles were analyzed, using MTT assay, flow cytometry, Western blotting, and RT-qPCR technologies. Results: TH301 proved to significantly decrease cell viability and to induce cell cycle arrest at the G1-phase across all PDAC cell lines herein examined, especially the AsPC-1 and BxPC-3 ones. It caused dose-dependent apoptosis and autophagy, and it synergized with Chloroquine and Oxaliplatin to enhance anti-oncogenicity. The remarkable induction of p21 by TH301 was shown to follow clock- and p53-independent patterns, thereby indicating the critical engagement of alternative mechanisms. Conclusions: TH301 demonstrates significant anti-cancer activities in PDAC cells, thus serving as a promising new therapeutic agent, which can also synergize with approved treatment schemes by targeting pathways beyond circadian clock regulation. Altogether, TH301 likely opens new therapeutic windows for the successful management of pancreatic cancer in clinical practice.
Collapse
Affiliation(s)
- Danae Farmakis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, Zografou, 157 01 Athens, Greece;
- Laboratory of Chronobiology, Institute of Biosciences and Applications (IBA), National Centre for Scientific Research (NCSR) “Demokritos”, 153 41 Aghia Paraskevi, Greece;
| | - Dimitrios J. Stravopodis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, Zografou, 157 01 Athens, Greece;
| | - Anastasia Prombona
- Laboratory of Chronobiology, Institute of Biosciences and Applications (IBA), National Centre for Scientific Research (NCSR) “Demokritos”, 153 41 Aghia Paraskevi, Greece;
| |
Collapse
|
5
|
Aria H, Azizi M, Nazem S, Mansoori B, Darbeheshti F, Niazmand A, Daraei A, Mansoori Y. Competing endogenous RNAs regulatory crosstalk networks: The messages from the RNA world to signaling pathways directing cancer stem cell development. Heliyon 2024; 10:e35208. [PMID: 39170516 PMCID: PMC11337742 DOI: 10.1016/j.heliyon.2024.e35208] [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: 02/10/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Cancer stem cells (CSCs) are one of the cell types that account for cancer heterogeneity. The cancer cells arrest in G0 and generate non-CSC progeny through self-renewal and pluripotency, resulting in tumor recurrence, metastasis, and resistance to chemotherapy. They can stimulate tumor relapse and re-grow a metastatic tumor. So, CSCs is a promising target for eradicating tumors, and developing an anti-CSCs therapy has been considered. In recent years competing endogenous RNA (ceRNA) has emerged as a significant class of post-transcriptional regulators that affect gene expression via competition for microRNA (miRNA) binding. Furthermore, aberrant ceRNA expression is associated with tumor progression. Recent findings show that ceRNA network can cause tumor progression through the effect on CSCs. To overcome therapeutic resistance due to CSCs, we need to improve our current understanding of the mechanisms by which ceRNAs are implicated in CSC-related relapse. Thus, this review was designed to discuss the role of ceRNAs in CSCs' function. Targeting ceRNAs may open the path for new cancer therapeutic targets and can be used in clinical research.
Collapse
Affiliation(s)
- Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Azizi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Nazem
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behnam Mansoori
- Pediatrics Department, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzaneh Darbeheshti
- Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
6
|
Fatma H, Siddique HR. Cancer cell plasticity, stem cell factors, and therapy resistance: how are they linked? Cancer Metastasis Rev 2024; 43:423-440. [PMID: 37796391 DOI: 10.1007/s10555-023-10144-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Abstract
Cellular plasticity can occur naturally in an organism and is considered an adapting mechanism during the developmental stage. However, abnormal cellular plasticity is observed in different diseased conditions, including cancer. Cancer cell plasticity triggers the stimuli of epithelial-mesenchymal transition (EMT), abnormal epigenetic changes, expression of stem cell factors and implicated signaling pathways, etc., and helps in the maintenance of CSC phenotype. Conversely, CSC maintains the cancer cell plasticity, EMT, and epigenetic plasticity. EMT contributes to increased cell migration and greater diversity within tumors, while epigenetic changes, stem cell factors (OCT4, NANOG, and SOX2), and various signaling pathways allow cancer cells to maintain various phenotypes, giving rise to intra- and inter-tumoral heterogeneity. The intricate relationships between cancer cell plasticity and stem cell factors help the tumor cells adopt drug-tolerant states, evade senescence, and successfully acquire drug resistance with treatment dismissal. Inhibiting molecules/signaling pathways involved in promoting CSCs, cellular plasticity, EMT, and epigenetic plasticity might be helpful for successful cancer therapy management. This review discussed the role of cellular plasticity, EMT, and stem cell factors in tumor initiation, progression, reprogramming, and therapy resistance. Finally, we discussed how the intervention in this axis will help better manage cancers and improve patient survivability.
Collapse
Affiliation(s)
- Homa Fatma
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, UP, 202002, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, UP, 202002, India.
| |
Collapse
|
7
|
Philipp LM, Yesilyurt UU, Surrow A, Künstner A, Mehdorn AS, Hauser C, Gundlach JP, Will O, Hoffmann P, Stahmer L, Franzenburg S, Knaack H, Schumacher U, Busch H, Sebens S. Epithelial and Mesenchymal-like Pancreatic Cancer Cells Exhibit Different Stem Cell Phenotypes Associated with Different Metastatic Propensities. Cancers (Basel) 2024; 16:686. [PMID: 38398077 PMCID: PMC10886860 DOI: 10.3390/cancers16040686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is mostly diagnosed at advanced or even metastasized stages, limiting the prognoses of patients. Metastasis requires high tumor cell plasticity, implying phenotypic switching in response to changing environments. Here, epithelial-mesenchymal transition (EMT), being associated with an increase in cancer stem cell (CSC) properties, and its reversion are important. Since it is poorly understood whether different CSC phenotypes exist along the EMT axis and how these impact malignancy-associated properties, we aimed to characterize CSC populations of epithelial and mesenchymal-like PDAC cells. Single-cell cloning revealed CSC (Holoclone) and non-CSC (Paraclone) clones from the PDAC cell lines Panc1 and Panc89. The Panc1 Holoclone cells showed a mesenchymal-like phenotype, dominated by a high expression of the stemness marker Nestin, while the Panc89 Holoclone cells exhibited a SOX2-dominated epithelial phenotype. The Panc89 Holoclone cells showed enhanced cell growth and a self-renewal capacity but slow cluster-like invasion. Contrarily, the Panc1 Holoclone cells showed slower cell growth and self-renewal ability but were highly invasive. Moreover, cell variants differentially responded to chemotherapy. In vivo, the Panc1 and Panc89 cell variants significantly differed regarding the number and size of metastases, as well as organ manifestation, leading to different survival outcomes. Overall, these data support the existence of different CSC phenotypes along the EMT axis in PDAC, manifesting different metastatic propensities.
Collapse
Affiliation(s)
- Lisa-Marie Philipp
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Umut-Ulas Yesilyurt
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Arne Surrow
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Anne-Sophie Mehdorn
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Charlotte Hauser
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Jan-Paul Gundlach
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, UKSH, Campus Kiel, 24105 Kiel, Germany
| | - Olga Will
- Molecular Imaging North Competence Center, Clinic of Radiology and Neuroradiology, Kiel University, UKSH, Campus Kiel, 24118 Kiel, Germany
| | - Patrick Hoffmann
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Lea Stahmer
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology, Kiel University, 24118 Kiel, Germany
| | - Hendrike Knaack
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
- Academic Affairs Office, Hannover Medical School, 30625 Hannover, Germany
| | - Udo Schumacher
- Department of Anatomy and Experimental Morphology, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, 23538 Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, 23562 Lübeck, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, 23562 Kiel, Germany
| |
Collapse
|
8
|
Huang JZ, Li JD, Chen G, He RQ. Identification of the key genes and mechanisms associated with transcatheter arterial chemoembolisation refractoriness in hepatocellular carcinoma. World J Clin Oncol 2024; 15:62-88. [PMID: 38292662 PMCID: PMC10823944 DOI: 10.5306/wjco.v15.i1.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Transcatheter arterial embolisation (TACE) is the primary treatment for intermediate-stage hepatocellular carcinoma (HCC) patients while some HCC cases have shown resistance to TACE. AIM To investigate the key genes and potential mechanisms correlated with TACE refractoriness in HCC. METHODS The microarray datasets of TACE-treated HCC tissues, HCC and non-HCC tissues were collected by searching multiple public databases. The respective differentially expressed genes (DEGs) were attained via limma R package. Weighted gene co-expression network analysis was employed for identifying the significant modules related to TACE non-response. TACE refractoriness-related genes were obtained by intersecting up-regulated TACE-associated and HCC-associated DEGs together with the genes in significant modules related to TACE non-response. The key genes expression in the above two pairs of samples was compared respectively via Wilcoxon tests and standard mean differences model. The prognostic value of the key genes was evaluated by Kaplan-Meier curve. Multivariate analysis was utilised to investigate the independent prognostic factor in key genes. Single-cell RNA (scRNA) sequencing analysis was conducted to explore the cell types in HCC. TACE refractoriness-related genes activity was calculated via AUCell packages. The CellChat R package was used for the investigation of the cell-cell communication between the identified cell types. RESULTS HCC tissues of TACE non-responders (n = 66) and TACE responders (n = 81), HCC (n = 3941) and non-HCC (n = 3443) tissues were obtained. The five key genes, DLG associated protein 5 (DLGAP5), Kinesin family member 20A (KIF20A), Assembly factor for spindle microtubules (ASPM), Kinesin family member 11 (KIF11) and TPX2 microtubule nucleation factor (TPX2) in TACE refractoriness-related genes, were identified. The five key genes were all up-regulated in the TACE non-responders group and the HCC group. High expression of the five key genes predicted poor prognosis in HCC. Among the key genes, TPX2 was an independent prognostic factor. Four cell types, hepatocytes, embryonic stem cells, T cells and B cells, were identified in the HCC tissues. The TACE refractoriness-related genes expressed primarily in hepatocytes and embryonic stem cells. Hepatocytes, as the providers of ligands, had the strongest interaction with embryonic stem cells that provided receptors. CONCLUSION Five key genes (DLGAP5, KIF20A, ASPM, KIF11 and TPX2) were identified as promoting refractory TACE. Hepatocytes and embryonic stem cells were likely to boost TACE refractoriness.
Collapse
Affiliation(s)
- Jie-Zhuang Huang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Di Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
9
|
Tayanloo-Beik A, Hamidpour SK, Nikkhah A, Arjmand R, Mafi AR, Rezaei-Tavirani M, Larijani B, Gilany K, Arjmand B. DNA Damage Responses, the Trump Card of Stem Cells in the Survival Game. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1470:165-188. [PMID: 37923882 DOI: 10.1007/5584_2023_791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2023]
Abstract
Stem cells, as a group of undifferentiated cells, are enriched with self-renewal and high proliferative capacity, which have attracted the attention of many researchers as a promising approach in the treatment of many diseases over the past years. However, from the cellular and molecular point of view, the DNA repair system is one of the biggest challenges in achieving therapeutic goals through stem cell technology. DNA repair mechanisms are an advantage for stem cells that are constantly multiplying to deal with various types of DNA damage. However, this mechanism can be considered a trump card in the game of cell survival and treatment resistance in cancer stem cells, which can hinder the curability of various types of cancer. Therefore, getting a deep insight into the DNA repair system can bring researchers one step closer to achieving major therapeutic goals. The remarkable thing about the DNA repair system is that this system is not only under the control of genetic factors, but also under the control of epigenetic factors. Therefore, it is necessary to investigate the role of the DNA repair system in maintaining the survival of cancer stem cells from both aspects.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Amirabbas Nikkhah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasta Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Rezazadeh Mafi
- Department of Radiation Oncology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Kambiz Gilany
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Chehelgerdi M, Behdarvand Dehkordi F, Chehelgerdi M, Kabiri H, Salehian-Dehkordi H, Abdolvand M, Salmanizadeh S, Rashidi M, Niazmand A, Ahmadi S, Feizbakhshan S, Kabiri S, Vatandoost N, Ranjbarnejad T. Exploring the promising potential of induced pluripotent stem cells in cancer research and therapy. Mol Cancer 2023; 22:189. [PMID: 38017433 PMCID: PMC10683363 DOI: 10.1186/s12943-023-01873-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 11/30/2023] Open
Abstract
The advent of iPSCs has brought about a significant transformation in stem cell research, opening up promising avenues for advancing cancer treatment. The formation of cancer is a multifaceted process influenced by genetic, epigenetic, and environmental factors. iPSCs offer a distinctive platform for investigating the origin of cancer, paving the way for novel approaches to cancer treatment, drug testing, and tailored medical interventions. This review article will provide an overview of the science behind iPSCs, the current limitations and challenges in iPSC-based cancer therapy, the ethical and social implications, and the comparative analysis with other stem cell types for cancer treatment. The article will also discuss the applications of iPSCs in tumorigenesis, the future of iPSCs in tumorigenesis research, and highlight successful case studies utilizing iPSCs in tumorigenesis research. The conclusion will summarize the advancements made in iPSC-based tumorigenesis research and the importance of continued investment in iPSC research to unlock the full potential of these cells.
Collapse
Affiliation(s)
- Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fereshteh Behdarvand Dehkordi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Hamidreza Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | | - Mohammad Abdolvand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Sharareh Salmanizadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar-Jereeb Street, Isfahan, 81746-73441, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saba Ahmadi
- Department of Molecular and Medical Genetics, Tbilisi State Medical University, Tbilisi, Georgia
| | - Sara Feizbakhshan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saber Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Nasimeh Vatandoost
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tayebeh Ranjbarnejad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
11
|
He XF, Hu X, Wen GJ, Wang Z, Lin WJ. O-GlcNAcylation in cancer development and immunotherapy. Cancer Lett 2023; 566:216258. [PMID: 37279852 DOI: 10.1016/j.canlet.2023.216258] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/30/2023] [Indexed: 06/08/2023]
Abstract
O-linked β-D-N-acetylglucosamine (O-GlcNAc), as a posttranslational modification (PTM), is a reversible reaction that attaches β-N-GlcNAc to Ser/Thr residues on specific proteins by O-GlcNAc transferase (OGT). O-GlcNAcase (OGA) removes the O-GlcNAc from O-GlcNAcylated proteins. O-GlcNAcylation regulates numerous cellular processes, including signal transduction, the cell cycle, metabolism, and energy homeostasis. Dysregulation of O-GlcNAcylation contributes to the development of various diseases, including cancers. Accumulating evidence has revealed that higher expression levels of OGT and hyper-O-GlcNAcylation are detected in many cancer types and governs glucose metabolism, proliferation, metastasis, invasion, angiogenesis, migration and drug resistance. In this review, we describe the biological functions and molecular mechanisms of OGT- or O-GlcNAcylation-mediated tumorigenesis. Moreover, we discuss the potential role of O-GlcNAcylation in tumor immunotherapy. Furthermore, we highlight that compounds can target O-GlcNAcylation by regulating OGT to suppress oncogenesis. Taken together, targeting protein O-GlcNAcylation might be a promising strategy for the treatment of human malignancies.
Collapse
Affiliation(s)
- Xue-Fen He
- Department of Obstetrics and Gynecology, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, 325000, Zhejiang, China
| | - Xiaoli Hu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gao-Jing Wen
- Department of Obstetrics and Gynecology, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, 325000, Zhejiang, China
| | - Zhiwei Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Anhui, China; Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Wen-Jing Lin
- Department of Obstetrics and Gynecology, Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
12
|
Luque-Martínez T. Sensibilidad y elasticidad de las universidades españolas a los indicadores del Ranking de Shanghái (ARWU). REVISTA ESPANOLA DE DOCUMENTACION CIENTIFICA 2023. [DOI: 10.3989/redc.2023.1.1947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
El ranking de Shanghái (ARWU) es uno de los más conocidos e influyentes, buena prueba es su presencia en medios tradicionales y digitales cuando se publica cada año, siendo de los más utilizados y referenciados en la gestión universitaria. Este trabajo analiza la sensibilidad y la elasticidad de los indicadores que lo integran, distinguiendo entre indicadores de naturaleza personal e institucional. El análisis se centra en las universidades españolas dentro del millar recogidas en 2021. El resultado identifica, y cuantifica, las universidades más y menos sensibles y elásticas a los diferentes indicadores, tanto respecto a la posición como a la puntuación. Además se identifican cinco grupos homogéneos de universidades en función de su elasticidad. El estudio prueba la gran heterogeneidad de cada indicador para cada universidad. Esto es útil para realizar benchmarking entre universidades e identificar acciones de mejora para gestionar la presencia en el ranking.
Collapse
|
13
|
Culig L, Chu X, Bohr VA. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res Rev 2022; 78:101636. [PMID: 35490966 PMCID: PMC9168971 DOI: 10.1016/j.arr.2022.101636] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 12/11/2022]
Abstract
Adult neurogenesis, the process by which neurons are generated in certain areas of the adult brain, declines in an age-dependent manner and is one potential target for extending cognitive healthspan. Aging is a major risk factor for neurodegenerative diseases and, as lifespans are increasing, these health challenges are becoming more prevalent. An age-associated loss in neural stem cell number and/or activity could cause this decline in brain function, so interventions that reverse aging in stem cells might increase the human cognitive healthspan. In this review, we describe the involvement of adult neurogenesis in neurodegenerative diseases and address the molecular mechanistic aspects of neurogenesis that involve some of the key aggregation-prone proteins in the brain (i.e., tau, Aβ, α-synuclein, …). We summarize the research pertaining to interventions that increase neurogenesis and regulate known targets in aging research, such as mTOR and sirtuins. Lastly, we share our outlook on restoring the levels of neurogenesis to physiological levels in elderly individuals and those with neurodegeneration. We suggest that modulating neurogenesis represents a potential target for interventions that could help in the fight against neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Luka Culig
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xixia Chu
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
14
|
Yu W, Ma Y, Roy SK, Srivastava R, Shankar S, Srivastava RK. Ethanol exposure of human pancreatic normal ductal epithelial cells induces EMT phenotype and enhances pancreatic cancer development in KC (Pdx1-Cre and LSL-Kras G12D ) mice. J Cell Mol Med 2021; 26:399-409. [PMID: 34859959 PMCID: PMC8743655 DOI: 10.1111/jcmm.17092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/17/2021] [Accepted: 11/19/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol is a risk factor for pancreatic cancer. However, the molecular mechanism by which chronic alcohol consumption influences pancreatic cancer development is not well understood. We have recently demonstrated that chronic ethanol exposure of pancreatic normal ductal epithelial cells (HPNE) induces cellular transformation by generating cancer stem cells (CSCs). Here, we examined whether chronic ethanol treatment induces epithelial–mesenchymal transition in HPNE cells and promotes pancreatic cancer development in KC (Pdx1‐Cre, and LSL‐KrasG12D) mice. Our data demonstrate that chronic ethanol exposure of HPNE cells induces SATB2 gene and those cells became highly motile. Ethanol treatment of HPNE cells results in downregulation of E‐Cadherin and upregulation of N‐Cadherin, Snail, Slug, Zeb1, Nanog and BMI‐1. Suppression of SATB2 expression in ethanol‐transformed HPNE cells inhibits EMT phenotypes. KC mice fed with an ethanol‐containing diet show enhanced pancreatic cancer growth and development than those fed with a control diet. Pancreas isolated from KC mice fed with an ethanol‐containing diet show higher expression of stem cell markers (CD133, CD44, CD24), pluripotency‐maintaining factors (cMyc, KLF4, SOX‐2, and Oct‐4), N‐Cadherin, EMT‐transcription factors (Snail, Slug, and Zeb1), and lower expression of E‐cadherin than those isolated from mice fed with a control diet. Furthermore, pancreas isolated from KC mice fed with an ethanol‐containing diet show higher expression of inflammatory cytokines (TNF‐α, IL‐6, and IL‐8) and PTGS‐2 (COX‐2) gene than those isolated from mice fed with a control diet. These data suggest that chronic alcohol consumption may contribute to pancreatic cancer development by generating inflammatory signals and CSCs.
Collapse
Affiliation(s)
- Wei Yu
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Yuming Ma
- Kansas City VA Medical Center, Kansas City, Missouri, USA
| | - Sanjit K Roy
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA
| | - Rashmi Srivastava
- Department of Pharmacology, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA
| | - Sharmila Shankar
- Kansas City VA Medical Center, Kansas City, Missouri, USA.,Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA.,Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA.,John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisina, USA.,Southeast Louisiana Veterans Health Care System, New Orleans, Louisina, USA
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, Kansas City, Missouri, USA.,Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA.,Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, Louisina, USA.,Southeast Louisiana Veterans Health Care System, New Orleans, Louisina, USA
| |
Collapse
|
15
|
Patil K, Khan FB, Akhtar S, Ahmad A, Uddin S. The plasticity of pancreatic cancer stem cells: implications in therapeutic resistance. Cancer Metastasis Rev 2021; 40:691-720. [PMID: 34453639 PMCID: PMC8556195 DOI: 10.1007/s10555-021-09979-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
The ever-growing perception of cancer stem cells (CSCs) as a plastic state rather than a hardwired defined entity has evolved our understanding of the functional and biological plasticity of these elusive components in malignancies. Pancreatic cancer (PC), based on its biological features and clinical evolution, is a prototypical example of a CSC-driven disease. Since the discovery of pancreatic CSCs (PCSCs) in 2007, evidence has unraveled their control over many facets of the natural history of PC, including primary tumor growth, metastatic progression, disease recurrence, and acquired drug resistance. Consequently, the current near-ubiquitous treatment regimens for PC using aggressive cytotoxic agents, aimed at ''tumor debulking'' rather than eradication of CSCs, have proven ineffective in providing clinically convincing improvements in patients with this dreadful disease. Herein, we review the key hallmarks as well as the intrinsic and extrinsic resistance mechanisms of CSCs that mediate treatment failure in PC and enlist the potential CSC-targeting 'natural agents' that are gaining popularity in recent years. A better understanding of the molecular and functional landscape of PCSC-intrinsic evasion of chemotherapeutic drugs offers a facile opportunity for treating PC, an intractable cancer with a grim prognosis and in dire need of effective therapeutic advances.
Collapse
Affiliation(s)
- Kalyani Patil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Farheen B Khan
- Department of Biology, College of Science, The United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | - Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar.
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
16
|
Integrated lipidomics and proteomics reveal cardiolipin alterations, upregulation of HADHA and long chain fatty acids in pancreatic cancer stem cells. Sci Rep 2021; 11:13297. [PMID: 34168259 PMCID: PMC8225828 DOI: 10.1038/s41598-021-92752-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer stem cells (PCSCs) play a key role in the aggressiveness of pancreatic ductal adenocarcinomas (PDAC); however, little is known about their signaling and metabolic pathways. Here we show that PCSCs have specific and common proteome and lipidome modulations. PCSCs displayed downregulation of lactate dehydrogenase A chain, and upregulation of trifunctional enzyme subunit alpha. The upregulated proteins of PCSCs are mainly involved in fatty acid (FA) elongation and biosynthesis of unsaturated FAs. Accordingly, lipidomics reveals an increase in long and very long-chain unsaturated FAs, which are products of fatty acid elongase-5 predicted as a key gene. Moreover, lipidomics showed the induction in PCSCs of molecular species of cardiolipin with mixed incorporation of 16:0, 18:1, and 18:2 acyl chains. Our data indicate a crucial role of FA elongation and alteration in cardiolipin acyl chain composition in PCSCs, representing attractive therapeutic targets in PDAC.
Collapse
|
17
|
Mayer P, Kraft A, Witzel HR, Marnet N, Hörner N, Roth W, Heinrich S, Hackert T, Bergmann F, Kauczor HU, Klauss M, Gaida MM. Restricted Water Diffusion in Diffusion-Weighted Magnetic Resonance Imaging in Pancreatic Cancer is Associated with Tumor Hypoxia. Cancers (Basel) 2020; 13:cancers13010089. [PMID: 33396818 PMCID: PMC7801953 DOI: 10.3390/cancers13010089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/21/2020] [Accepted: 12/26/2020] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Pancreatic cancer is characterized by a dense network of connective tissue surrounding clusters of cancer cells, the so-called stroma. This ubiquitous connective tissue impairs the delivery of oxygen to cancer cells. This results in hypoxia, which renders the cancer more aggressive and more resistant to treatment. In the present study, we investigated whether the extent of hypoxia in pancreatic cancer can be predicted by magnetic resonance imaging (MRI), a widely used medical imaging technique. More specifically, we used an MRI sequence which can quantitate the random motion (i.e., diffusion) of water molecules within the cancer tissue, namely diffusion-weighted (DW) MRI. We found that the random motion of water molecules is lower in cancer lesions with high hypoxia compared to those with low hypoxia. The findings from our study imply that DW-MRI can be used to identify pancreatic cancer lesions with high hypoxia which are at high risk for treatment failure. Abstract Hypoxia is a hallmark of pancreatic cancer (PDAC) due to its compact and extensive fibrotic tumor stroma. Hypoxia contributes to high lethality of this disease, by inducing a more malignant phenotype and resistance to radiation and chemotherapy. Thus, non-invasive methods to quantify hypoxia could be helpful for treatment decisions, for monitoring, especially in non-resectable tumors, or to optimize personalized therapy. In the present study, we investigated whether tumor hypoxia in PDAC is reflected by diffusion-weighted magnetic resonance imaging (DW-MRI), a functional imaging technique, frequently used in clinical practice for identification and characterization of pancreatic lesions. DW-MRI assesses the tissue microarchitecture by measuring the diffusion of water molecules, which is more restricted in highly compact tissues. As reliable surrogate markers for hypoxia, we determined Blimp-1 (B-lymphocyte induced maturation protein), a transcription factor, as well as vascular endothelial growth factor (VEGF), which are up-regulated in response to hypoxia. In 42 PDAC patients, we observed a close association between restricted water diffusion in DW-MRI and tumor hypoxia in matched samples, as expressed by high levels of Blimp-1 and VEGF in tissue samples of the respective patients. In summary, our data show that DW-MRI is well suited for the evaluation of tumor hypoxia in PDAC and could potentially be used for the identification of lesions with a high hypoxic fraction, which are at high risk for failure of radiochemotherapy.
Collapse
Affiliation(s)
- Philipp Mayer
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
- Correspondence: ; Tel.: +49-6221-5637-345
| | - Anne Kraft
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Hagen R. Witzel
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Nicole Marnet
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Nina Hörner
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Stefan Heinrich
- Department of Surgery, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany;
| | - Thilo Hackert
- Department of General, Visceral, and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Frank Bergmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
- Clinical Pathology, Klinikum Darmstadt GmbH, 64283 Darmstadt, Germany
| | - Hans-Ulrich Kauczor
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
| | - Miriam Klauss
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
| | - Matthias M. Gaida
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
- Research Center for Immunotherapy, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany
- Joint Unit Immunopathology, Institute of Pathology, University Medical Center, JGU-Mainz and TRON, Translational Oncology at the University Medical Center, JGU-Mainz, 55131 Mainz, Germany
| |
Collapse
|
18
|
Zhang Q, Han Z, Zhu Y, Chen J, Li W. The Role and Specific Mechanism of OCT4 in Cancer Stem Cells: A Review. Int J Stem Cells 2020; 13:312-325. [PMID: 32840233 PMCID: PMC7691851 DOI: 10.15283/ijsc20097] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Recently, evidences show that cancer stem cells (CSCs) are a type of cancer cell group with self-renewal and play a huge role in tumor recurrence, metastasis, and drug resistance. Finding new treatment directions and targets for cancer prognosis and reducing mortality has become a top priority. OCT4, as a transcription factor, participates in maintaining the stem characteristics of CSCs, but the mechanism of OCT4 is often overlooked. In this review, we try to illustrate the mechanism by which OCT4 plays a role in CSCs from the perspective of genetic modification of OCT4, non-coding RNA, complexes and signaling pathways associated with OCT4. Our ultimate goal is to provide new targets for cancer treatment to prolong the survival of cancer patients.
Collapse
Affiliation(s)
- Qi Zhang
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Zhenzhen Han
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yanbo Zhu
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jingcheng Chen
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Wei Li
- Stem Cell and Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Huang T, Song X, Xu D, Tiek D, Goenka A, Wu B, Sastry N, Hu B, Cheng SY. Stem cell programs in cancer initiation, progression, and therapy resistance. Am J Cancer Res 2020; 10:8721-8743. [PMID: 32754274 PMCID: PMC7392012 DOI: 10.7150/thno.41648] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
Over the past few decades, substantial evidence has convincingly revealed the existence of cancer stem cells (CSCs) as a minor subpopulation in cancers, contributing to an aberrantly high degree of cellular heterogeneity within the tumor. CSCs are functionally defined by their abilities of self-renewal and differentiation, often in response to cues from their microenvironment. Biological phenotypes of CSCs are regulated by the integrated transcriptional, post-transcriptional, metabolic, and epigenetic regulatory networks. CSCs contribute to tumor progression, therapeutic resistance, and disease recurrence through their sustained proliferation, invasion into normal tissue, promotion of angiogenesis, evasion of the immune system, and resistance to conventional anticancer therapies. Therefore, elucidation of the molecular mechanisms that drive cancer stem cell maintenance, plasticity, and therapeutic resistance will enhance our ability to improve the effectiveness of targeted therapies for CSCs. In this review, we highlight the key features and mechanisms that regulate CSC function in tumor initiation, progression, and therapy resistance. We discuss factors for CSC therapeutic resistance, such as quiescence, induction of epithelial-to-mesenchymal transition (EMT), and resistance to DNA damage-induced cell death. We evaluate therapeutic approaches for eliminating therapy-resistant CSC subpopulations, including anticancer drugs that target key CSC signaling pathways and cell surface markers, viral therapies, the awakening of quiescent CSCs, and immunotherapy. We also assess the impact of new technologies, such as single-cell sequencing and CRISPR-Cas9 screening, on the investigation of the biological properties of CSCs. Moreover, challenges remain to be addressed in the coming years, including experimental approaches for investigating CSCs and obstacles in therapeutic targeting of CSCs.
Collapse
|
20
|
Ebisu Y, Ishida M, Mizokami T, Kita M, Okada H, Tsuta K. Immunohistochemical analysis of SOX2 expression in small-cell neuroendocrine carcinoma of the endometrium. Mol Clin Oncol 2020; 13:115-118. [PMID: 32714533 PMCID: PMC7366239 DOI: 10.3892/mco.2020.2051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 04/10/2020] [Indexed: 12/15/2022] Open
Abstract
Small-cell neuroendocrine carcinoma (NEC) of the endometrium is an extremely rare and highly aggressive carcinoma. Sex-determining region Y-box 2 (SOX2) is a master transcription factor regulating the self-renewal, maintenance of stem cell properties and pluripotency of embryonic stem cells, and recent studies revealed that SOX2 plays important roles in cancer growth and progression in several types of carcinomas, including small-cell neuroendocrine carcinoma (NEC) of the lung and oesophagus. Few studies to date have analysed the association between SOX2 and endometrioid carcinoma, whereas the expression of SOX2 in small-cell NEC of the endometrium has not been investigated. The aim of the present study was to analyse the expression status of SOX2, p16 and paired-box gene (PAX) 8, a useful Müllerian marker, in endometrial small-cell NEC. A total of 4 patients with small-cell NEC of the endometrium were enrolled (median age, 70 years). Immunohistochemical studies revealed SOX2 expression in 3 patients and p16 expression in all patients. No patients exhibited positive immunoreactivity for PAX8. SOX2 expression has been reported to be associated with the pathogenesis of small-cell NEC of the oesophagus. Therefore, the results of the present study indicated that SOX2 expression plays an important role in the development of small-cell NEC of the endometrium and the oesophagus. Moreover, expression of p16 and loss of PAX8 do not indicate the origin of small-cell NEC of the endometrium.
Collapse
Affiliation(s)
- Yusuke Ebisu
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Mitsuaki Ishida
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Tomomi Mizokami
- Department of Obstetrics and Gynaecology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Masato Kita
- Department of Obstetrics and Gynaecology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Hidetaka Okada
- Department of Obstetrics and Gynaecology, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| | - Koji Tsuta
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
21
|
Quan MY, Guo Q, Liu J, Yang R, Bai J, Wang W, Cai Y, Han R, Lv YQ, Ding L, Billadeau DD, Lou Z, Bellusci S, Li X, Zhang JS. An FGFR/AKT/SOX2 Signaling Axis Controls Pancreatic Cancer Stemness. Front Cell Dev Biol 2020; 8:287. [PMID: 32457900 PMCID: PMC7221133 DOI: 10.3389/fcell.2020.00287] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/02/2020] [Indexed: 12/20/2022] Open
Abstract
Cancer stemness is associated with high malignancy and low differentiation, as well as therapeutic resistance of tumors including pancreatic ductal adenocarcinoma (PDAC). Fibroblast growth factors (FGFs) exert pleiotropic effects on a variety of cellular processes and functions including embryonic stem cell pluripotency and cancer cell stemness via the activation of four tyrosine kinase FGF receptors (FGFRs). FGF ligands have been a major component of the cocktail of growth factors contained in the cancer stemness-inducing (CSI) and organoid culture medium. Although FGF/FGFR signaling has been hypothesized to maintain cancer stemness, its function in this process is still unclear. We report that inhibition of FGF/FGFR signaling impairs sphere-forming ability of PDAC in vitro, and knocking down FGFR1 and FGFR2 decreased their tumorigenesis abilities in vivo. Mechanistically, we demonstrated that SOX2 is down-regulated upon loss of FGFR signaling. The overexpression of SOX2 in SOX2-negative cells, which normally do not display stemness capabilities, is sufficient to induce spheroid formation. Additionally, we found that AKT phosphorylation was reduced upon FGFR signaling inhibition. The inhibition of AKT using specific pharmacological inhibitors in the context of CSI medium leads to the loss of spheroid formation associated with loss of SOX2 nuclear expression and increased degradation. We demonstrate that an FGFR/AKT/SOX2 axis controls cancer stemness in PDAC and therefore may represent an important therapeutic target in the fight against this very aggressive form of cancer.
Collapse
Affiliation(s)
- Mei-Yu Quan
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Qiang Guo
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Jiayu Liu
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Ruo Yang
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Jing Bai
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Wei Wang
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Yaxin Cai
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Rui Han
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Yu-Qing Lv
- Center for Precision Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Ding
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Daniel D Billadeau
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Zhenkun Lou
- Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Saverio Bellusci
- Institute of Life Sciences, Wenzhou University, Wenzhou, China.,Cardio-Pulmonary Institute, Member of the German Lung Center, Justus Liebig University Giessen, Giessen, Germany
| | - Xiaokun Li
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China
| | - Jin-San Zhang
- School of Pharmaceutical Sciences and International Collaborative Center on Growth Factor Research, Wenzhou Medical University, Wenzhou, China.,Division of Oncology Research and Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
22
|
Gupta R, Leon F, Thompson CM, Nimmakayala R, Karmakar S, Nallasamy P, Chugh S, Prajapati DR, Rachagani S, Kumar S, Ponnusamy MP. Global analysis of human glycosyltransferases reveals novel targets for pancreatic cancer pathogenesis. Br J Cancer 2020; 122:1661-1672. [PMID: 32203219 PMCID: PMC7251111 DOI: 10.1038/s41416-020-0772-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Several reports have shown the role of glycosylation in pancreatic cancer (PC), but a global systematic screening of specific glycosyltransferases (glycoTs) in its progression remains unknown. METHODS We demonstrate a rigorous top-down approach using TCGA-based RNA-Seq analysis, multi-step validation using RT-qPCR, immunoblots and immunohistochemistry. We identified six unique glycoTs (B3GNT3, B4GALNT3, FUT3, FUT6, GCNT3 and MGAT3) in PC pathogenesis and studied their function using CRISPR/Cas9-based KD systems. RESULTS Serial metastatic in vitro models using T3M4 and HPAF/CD18, generated in house, exhibited decreases in B3GNT3, FUT3 and GCNT3 expression on increasing metastatic potential. Immunohistochemistry identified clinical significance for GCNT3, B4GALNT3 and MGAT3 in PC. Furthermore, the effects of B3GNT3, FUT3, GCNT3 and MGAT3 were shown on proliferation, migration, EMT and stem cell markers in CD18 cell line. Talniflumate, GCNT3 inhibitor, reduced colony formation and migration in T3M4 and CD18 cells. Moreover, we found that loss of GCNT3 suppresses PC progression and metastasis by downregulating cell cycle genes and β-catenin/MUC4 axis. For GCNT3, proteomics revealed downregulation of MUC5AC, MUC1, MUC5B including many other proteins. CONCLUSIONS Collectively, we demonstrate a critical role of O- and N-linked glycoTs in PC progression and delineate the mechanism encompassing the role of GCNT3 in PC.
Collapse
Affiliation(s)
- Rohitesh Gupta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Frank Leon
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Christopher M Thompson
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ramakrishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saswati Karmakar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Palanisamy Nallasamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Seema Chugh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Dipakkumar R Prajapati
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
23
|
Khan MA, Srivastava SK, Zubair H, Patel GK, Arora S, Khushman M, Carter JE, Gorman GS, Singh S, Singh AP. Co-targeting of CXCR4 and hedgehog pathways disrupts tumor-stromal crosstalk and improves chemotherapeutic efficacy in pancreatic cancer. J Biol Chem 2020; 295:8413-8424. [PMID: 32358063 DOI: 10.1074/jbc.ra119.011748] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 04/17/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) remains a therapeutic challenge because of its intrinsic and extrinsic chemoresistance mechanisms. Here, we report that C-X-C motif chemokine receptor 4 (CXCR4) and hedgehog pathways cooperate in PC chemoresistance via bidirectional tumor-stromal crosstalk. We show that when PC cells are co-cultured with pancreatic stellate cells (PSCs) they are significantly more resistant to gemcitabine toxicity than those grown in monoculture. We also demonstrate that this co-culture-induced chemoresistance is abrogated by inhibition of the CXCR4 and hedgehog pathways. Similarly, the co-culture-induced altered expression of genes in PC cells associated with gemcitabine metabolism, antioxidant defense, and cancer stemness is also reversed upon CXCR4 and hedgehog inhibition. We have confirmed the functional impact of these genetic alterations by measuring gemcitabine metabolites, reactive oxygen species production, and sphere formation in vehicle- or gemcitabine-treated monocultures and co-cultured PC cells. Treatment of orthotopic pancreatic tumor-bearing mice with gemcitabine alone or in combination with a CXCR4 antagonist (AMD3100) or hedgehog inhibitor (GDC-0449) displays reduced tumor growth. Notably, we show that the triple combination treatment is the most effective, resulting in nearly complete suppression of tumor growth. Immunohistochemical analysis of Ki67 and cleaved caspase-3 confirm these findings from in vivo imaging and tumor measurements. Our findings provide preclinical and mechanistic evidence that a combination of gemcitabine treatment with targeted inhibition of both the CXCR4 and hedgehog pathways improves outcomes in a PC mouse model.
Collapse
Affiliation(s)
- Mohammad Aslam Khan
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama.,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Sanjeev Kumar Srivastava
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama.,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Haseeb Zubair
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama.,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Girijesh Kumar Patel
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Sumit Arora
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - Moh'd Khushman
- Department of Medical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama
| | - James Elliot Carter
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama
| | | | - Seema Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama.,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Ajay Pratap Singh
- Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama .,Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| |
Collapse
|
24
|
Zhou J, Wang H, Che J, Xu L, Yang W, Li Y, Zhou W. Silencing of microRNA-135b inhibits invasion, migration, and stemness of CD24 +CD44 + pancreatic cancer stem cells through JADE-1-dependent AKT/mTOR pathway. Cancer Cell Int 2020; 20:134. [PMID: 32351328 PMCID: PMC7183669 DOI: 10.1186/s12935-020-01210-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/10/2020] [Indexed: 12/24/2022] Open
Abstract
Background Recent studies have emphasized determining the ability of microRNAs (miRNAs) as crucial regulators in the occurrence and development of pancreatic cancer (PC), which continues to be one of the deadliest malignancies with few effective therapies. The study aimed to investigate the functional role of miR-135b and its associated mechanism to unravel the biological characteristics of tumor growth in pancreatic cancer stem cells (PCSCs). Methods Microarray analyses were initially performed to identify the PC-related miRNAs and genes. The expression of miR-135b and PCSC markers in PC tissues and cells was determined by RT-qPCR and western blot analysis, respectively. The potential gene (JADE-1) that could bind to miR-135b was confirmed by the dual-luciferase reporter assay. To investigate the tumorigenicity, migration, invasion, and stemness of PC cells, several gain-of-function and loss-of-function genetic experiments were conducted. Finally, tumor formation in nude mice was conducted to confirm the results in vivo. Results miR-135b was highly-expressed in PC tissues and PCSCs, which was identified to specifically target JADE-1. The overexpression of miR-135b promoted proliferation, migration, and invasion of PCSC, inhibited cell apoptosis and increased the expression of stemness-related factors (Sox-2, Oct-4, Nanog, Aldh1, and Slug). Moreover, miR-135b could promote the expression of phosphorylated AKT and phosphorylated mTOR in the AKT/mTOR pathway. Additionally, miR-135b overexpression accelerated tumor growth in nude mice. Conclusions Taken together, the silencing of miR-135b promotes the JADE-1 expression, which inactivates the AKT/mTOR pathway and ultimately results in inhibition of self-renewal and tumor growth of PCSCs. Hence, this study contributes to understanding the role of miR-135 in PCSCs and its underlying molecular mechanisms to aid in the development of effective PC therapeutics.
Collapse
Affiliation(s)
- Jingyang Zhou
- 1Class 182, Queen Mary School, Medical Department, Nanchang University, Nanchang, 330031 People's Republic of China
| | - Haihong Wang
- Department of Hepatopancreatobillary Surgery, Xuzhou City Cancer Hospital, No. 131 Huancheng Rd., Gulou District, Xuzhou, 221000 Jiangsu People's Republic of China
| | - Jinhui Che
- Department of Hepatopancreatobillary Surgery, Xuzhou City Cancer Hospital, No. 131 Huancheng Rd., Gulou District, Xuzhou, 221000 Jiangsu People's Republic of China
| | - Lu Xu
- Department of Hepatopancreatobillary Surgery, Xuzhou City Cancer Hospital, No. 131 Huancheng Rd., Gulou District, Xuzhou, 221000 Jiangsu People's Republic of China
| | - Weizhong Yang
- Department of Hepatopancreatobillary Surgery, Xuzhou City Cancer Hospital, No. 131 Huancheng Rd., Gulou District, Xuzhou, 221000 Jiangsu People's Republic of China
| | - Yunjiu Li
- Department of Hepatopancreatobillary Surgery, Xuzhou City Cancer Hospital, No. 131 Huancheng Rd., Gulou District, Xuzhou, 221000 Jiangsu People's Republic of China
| | - Wuyuan Zhou
- Department of Hepatopancreatobillary Surgery, Xuzhou City Cancer Hospital, No. 131 Huancheng Rd., Gulou District, Xuzhou, 221000 Jiangsu People's Republic of China
| |
Collapse
|
25
|
Chong Y, Thakur N, Paik KY, Lee EJ, Kang CS. Prognostic significance of stem cell/ epithelial-mesenchymal transition markers in periampullary/pancreatic cancers: FGFR1 is a promising prognostic marker. BMC Cancer 2020; 20:216. [PMID: 32171280 PMCID: PMC7071628 DOI: 10.1186/s12885-020-6673-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 02/24/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Periampullary cancers (PAC) including pancreatic, ampulla of Vater (AOV), and common bile duct (CBD) cancers are highly aggressive with a lack of useful prognostic markers beyond T stage. However, T staging can be biased due to the anatomic complexity of this region. Recently, several markers related to cancer stem cells and epithelial-mesenchymal transition (EMT) such as octamer transcription factor-4 (Oct4) and fibroblast growth factor receptor 1 (FGFR1) respectively, have been proposed as new promising markers in other solid cancers. The aim of this study was to assess the expression and prognostic significance of stem cell/EMT markers in PACs. METHODS Formalin-fixed, paraffin-embedded tissues of surgically excised PACs from the laboratory archives from 1998 to 2014 were evaluated by immunohistochemical staining for stem cell/EMT markers using tissue microarray. The clinicopathologic parameters were documented and statistically analyzed with the immunohistochemical findings. Survival and recurrence data were collected and analyzed. RESULTS A total of 126 PAC cases were evaluated. The average age was 63 years, with 76 male and 50 female patient samples. Age less than 74 years, AOV cancers, lower T & N stage, lower tumor size, no lymphatic, vascular, perineural invasion and histologic well differentiation, intestinal type, no fibrosis, severe inflammation were significantly associated with the better overall survival High expression levels of FGFR1 as well as CK20, CDX2, and VEGF were significantly related to better overall survival, while other stem cell markers were not related. Similar findings were observed for tumor recurrence using disease-free survival. CONCLUSIONS In addition to other clinicopathologic parameters, severe fibrosis was related to frequent tumor recurrence, and high FGFR1 expression was associated with better overall survival. Histologic changes such as extensive fibrosis need to be investigated further in relation to EMT of PACs.
Collapse
Affiliation(s)
- Yosep Chong
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul, 07345 Republic of Korea
| | - Nishant Thakur
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul, 07345 Republic of Korea
| | - Kwang Yeol Paik
- Department of Surgery, College of Medicine, The Catholic University of Korea, Seoul, 07345 Republic of Korea
| | - Eun Jung Lee
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul, 07345 Republic of Korea
- Department of Pathology, Shinwon Medical Foundation, Soha-ro 109 beon-gil, Gwanmyeong-si, 14316 Gyeonggi-do Republic of Korea
| | - Chang Suk Kang
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul, 07345 Republic of Korea
- Department of Pathology, Samkwang Medical Laboratories, 57, Baumoe-ro 41-gil, Seocho-gu, Seoul, 06742 Republic of Korea
| |
Collapse
|
26
|
Cui F, Hao ZX, Li J, Zhang YL, Li XK, He JX. SOX2 mediates cisplatin resistance in small-cell lung cancer with downregulated expression of hsa-miR-340-5p. Mol Genet Genomic Med 2020; 8:e1195. [PMID: 32130794 PMCID: PMC7216814 DOI: 10.1002/mgg3.1195] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/19/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Background This study is aimed to unravel the genetic factors associated with microRNA (miRNA) expression in regulating sex‐determining region Y‐box 2 (SOX2)‐mediated cisplatin resistance in small‐cell lung cancer (SCLC). Methods The relevance of SOX2 expression in SCLC was analyzed in a panel of SCLC cells by quantitative real‐time PCR (qPCR) and western blot (WB). We selected DMS114 cell line, in which SOX2 was amplified via lentiviral vector‐mediated transfection of the SOX2 genes and tested for the half‐maximal inhibitory concentration (IC50) by MTS assay. High‐throughput sequencing and screening of differentially expressed miRNAs between SOX2‐overexpressing and normal control cells were performed. Finally, miRanda software was used to verify the miRNAs bound with SOX2 and qPCR was used to identify the expression of miRNAs which were binding with SOX2. Results Cisplatin‐resistant SOX2‐overexpressing DMS114 cell lines were successfully developed, showing a statistically significant increase in SOX2 expression by qPCR and WB. Our results showed a typically higher IC50 value in SOX2‐overexpressing cells compared with the negative controls. The high‐throughput sequencing analysis revealed that 68 miRNAs were upregulated and 24 miRNAs were downregulated in the SOX2‐overexpressing cells. The 24 downregulated miRNAs were further verified. Of them, a cancer‐related miRNA, hsa‐miR‐340‐5p, showed a higher binding affinity with SOX2 in network regulation mapping, which was also found to be markedly downregulated under qPCR analysis. Conclusion We demonstrated that downregulated expression of hsa‐miR‐340‐5p may affect cisplatin resistance by mediating SOX2 expression in SCLC cells, which may provide a potential target for the therapy of chemoresistant SCLCs.
Collapse
Affiliation(s)
- Fei Cui
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhe-Xue Hao
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jin Li
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ya-Lei Zhang
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xu-Kai Li
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian-Xing He
- Department of Cardiothoracic Surgery, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
27
|
Zhao H, Wu S, Li H, Duan Q, Zhang Z, Shen Q, Wang C, Yin T. ROS/KRAS/AMPK Signaling Contributes to Gemcitabine-Induced Stem-like Cell Properties in Pancreatic Cancer. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:299-312. [PMID: 31508487 PMCID: PMC6726755 DOI: 10.1016/j.omto.2019.07.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
Poor prognosis in pancreatic cancer (PanCa) is partially due to chemoresistance to gemcitabine (GEM). Glucose metabolism has been revealed to contribute to the therapeutic resistance and pluripotent state of PanCa cells. However, few studies have focused on the effects of GEM on cancer cell metabolism, stemness of tumor cells, and molecular mechanisms that critically influence PanCa treatment. We demonstrate that GEM treatment induces metabolic reprogramming, reducing mitochondrial oxidation and upregulating aerobic glycolysis, and promotes stem-like behaviors in cancer cells. Inhibiting aerobic glycolysis suppresses cancer cell stemness and strengthens GEM's cytotoxicity. GEM-induced metabolic reprogramming is KRAS dependent, as knockdown of KRAS reverses the metabolic shift. GEM-induced metabolic reprogramming also activates AMP-activated protein kinase (AMPK), which promotes glycolytic flux and cancer stemness. In addition, GEM-induced reactive oxygen species (ROS) activate the KRAS/AMPK pathway. This effect was validated by introducing exogenous hydrogen peroxide (H2O2). Taken together, these findings reveal a counterproductive GEM effect during PanCa treatment. Regulating cellular redox, targeting KRAS/AMPK signaling, or reversing metabolic reprogramming might be effective approaches to eliminate cancer stem cells (CSCs) and enhance chemosensitivity to GEM to improve the prognosis of PanCa patients.
Collapse
Affiliation(s)
- Hengqiang Zhao
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shihong Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Hehe Li
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Qingke Duan
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Zhengle Zhang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Qiang Shen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunyou Wang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Tao Yin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.,Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| |
Collapse
|
28
|
Hernández-Camarero P, López-Ruiz E, Griñán-Lisón C, García MÁ, Chocarro-Wrona C, Marchal JA, Kenyon J, Perán M. Pancreatic (pro)enzymes treatment suppresses BXPC-3 pancreatic Cancer Stem Cell subpopulation and impairs tumour engrafting. Sci Rep 2019; 9:11359. [PMID: 31388092 PMCID: PMC6684636 DOI: 10.1038/s41598-019-47837-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/23/2019] [Indexed: 01/01/2023] Open
Abstract
Cancer stem cells (CSCs) subpopulation within the tumour is responsible for metastasis and cancer relapse. Here we investigate in vitro and in vivo the effects of a pancreatic (pro)enzyme mixture composed of Chymotrypsinogen and Trypsinogen (PRP) on CSCs derived from a human pancreatic cell line, BxPC3. Exposure of pancreatic CSCs spheres to PRP resulted in a significant decrease of ALDEFLUOR and specific pancreatic CSC markers (CD 326, CD 44 and CxCR4) signal tested by flow cytometry, further CSCs markers expression was also analyzed by western and immunofluorescence assays. PRP also inhibits primary and secondary sphere formation. Three RT2 Profiler PCR Arrays were used to study gene expression regulation after PRP treatment and resulted in, (i) epithelial-mesenchymal transition (EMT) inhibition; (ii) CSCs related genes suppression; (iii) enhanced expression of tumour suppressor genes; (iv) downregulation of migration and metastasis genes and (v) regulation of MAP Kinase Signalling Pathway. Finally, in vivo anti-tumor xenograft studies demonstrated high anti-tumour efficacy of PRP against tumours induced by BxPC3 human pancreatic CSCs. PRP impaired engrafting of pancreatic CSC’s tumours in nude mice and displayed an antigrowth effect toward initiated xenografts. We concluded that (pro)enzymes treatment is a valuable strategy to suppress the CSC population in solid pancreatic tumours.
Collapse
Affiliation(s)
- Pablo Hernández-Camarero
- Department of Health Sciences, University of Jaén, Jaén, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Elena López-Ruiz
- Department of Health Sciences, University of Jaén, Jaén, Spain.,Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - María Ángel García
- Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Department of Biochemistry and Molecular Biology 3 and Immunology, University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Carlos Chocarro-Wrona
- Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine, Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.,Biosanitary Research Institute of Granada (ibs. GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain.,Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain
| | - Julian Kenyon
- The Dove Clinic for Integrated Medicine, Twyford, SO21 1RG, UK.
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, Spain. .,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E-18016, Spain.
| |
Collapse
|
29
|
Yang F, Cui P, Lu Y, Zhang X. Requirement of the transcription factor YB-1 for maintaining the stemness of cancer stem cells and reverting differentiated cancer cells into cancer stem cells. Stem Cell Res Ther 2019; 10:233. [PMID: 31375149 PMCID: PMC6679460 DOI: 10.1186/s13287-019-1360-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/12/2019] [Accepted: 07/24/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Cancer stem cells always express high levels of stemness-associated transcription factors to maintain their features. However, the regulatory mechanism of the stemness of cancer stem cells mediated by transcription factors has not been extensively explored. METHODS The YB-1 gene in cancer stem cells was knocked out by the CRISPR/Cas9 system. The YB-1 knockout cancer stem cells were transfected with a vector expressing YB-1 to rescue YB-1, and then the cell proliferation, cell cycle, apoptosis, and stemness, as well as tumorigenesis in nude mice, were assessed to examine the effect of YB-1 in cancer stem cells. The target genes of YB-1 were confirmed by CHIP-seq. The totipotency or pluripotency of differentiated cancer stem cells were detected by tumorsphere formation assay and quantitative real-time PCR. RESULTS The deletion of YB-1 gene inhibited the proliferation of breast cancer stem cells and melanoma stem cells, leading to cell cycle arrest and apoptosis, and induced irreversible differentiation of cancer stem cells. The tumorigenicity ability of YB-1-deleted cancer stem cells was significantly reduced in vitro and in vivo. The results of ChIP-seq showed that YB-1 maintained the stemness of cancer stem cells by promoting the expressions of stemness-associated genes (FZD-1, p21, GLP-1, GINS1, and Notch2). Furthermore, simultaneous expressions of YB-1 and the other four (SOX2, POU3F2, OCT-4, and OLIG1) or five (SOX2, SALL2, OCT-4, POU3F2, and Bmi-1) transcription factors in YB-1 knockout cancer stem cells restored the stemness of YB-1 knockout cancer stem cells. CONCLUSIONS Our study indicated that YB-1 was required for maintaining the stemness of cancer stem cells and reverting the differentiated tumor cells into cancer stem cells.
Collapse
Affiliation(s)
- Fan Yang
- College of Life Sciences and Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Pei Cui
- College of Life Sciences and Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Yu Lu
- College of Life Sciences and Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Xiaobo Zhang
- College of Life Sciences and Laboratory for Marine Biology and Biotechnology of Qingdao National Laboratory for Marine Science and Technology, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
30
|
Abstract
Cancer-initiating cells (CIC) are the driving force in tumor progression. There is strong evidence that CIC fulfill this task via exosomes (TEX), which modulate and reprogram stroma, nontransformed cells, and non-CIC. Characterization of CIC, besides others, builds on expression of CIC markers, many of which are known as metastasis-associated molecules. We here discuss that the linkage between CIC/CIC-TEX and metastasis-associated molecules is not fortuitously, but relies on the contribution of these markers to TEX biogenesis including loading and TEX target interactions. In addition, CIC markers contribute to TEX binding- and uptake-promoted activation of signaling cascades, transcription initiation, and translational control. Our point of view will be outlined for pancreas and colon CIC highly expressing CD44v6, Tspan8, EPCAM, claudin7, and LGR5, which distinctly but coordinately contribute to tumor progression. Despite overwhelming progress in unraveling the metastatic cascade and the multiple tasks taken over by CIC-TEX, there remains a considerable gap in linking CIC biomarkers, TEX, and TEX-initiated target modulation with metastasis. We will try to outline possible bridges, which could allow depicting pathways for new and expectedly powerful therapeutic interference with tumor progression.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
| | - Margot Zöller
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China.
- Pancreas Section, University Hospital of Surgery, Heidelberg, Germany.
| |
Collapse
|
31
|
Ma Y, Yu W, Shrivastava A, Srivastava RK, Shankar S. Inhibition of pancreatic cancer stem cell characteristics by α-Mangostin: Molecular mechanisms involving Sonic hedgehog and Nanog. J Cell Mol Med 2019; 23:2719-2730. [PMID: 30712329 PMCID: PMC6433724 DOI: 10.1111/jcmm.14178] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 12/31/2018] [Accepted: 01/02/2019] [Indexed: 12/21/2022] Open
Abstract
The current investigation was intended to elucidate the molecular mechanism of α‐Mangostin in the regulation of pancreatic cancer stem cell (CSC) characteristics. Here, we demonstrate that α‐Mangostin inhibited cell proliferation in pancreatic CSCs and cancer cell lines while it showed no effect on human pancreatic normal ductal epithelial cells. Also, α‐Mangostin inhibited colony formation and induced apoptosis in these cells. Further, α‐Mangostin inhibited the self‐renewal capacity of CSCs isolated from human primary tumours and KrasG12D mice. Furthermore, α‐Mangostin inhibited the invasive and metastatic ability of pancreatic CSCs by suppressing the epithelial‐to‐mesenchymal transition (EMT) via up‐regulation of E‐cadherin and down‐regulation of mesenchymal phenotype by inhibiting N‐cadherin, Snail and Slug expression. Interestingly, the pluripotency maintaining factors and CSC markers were inhibited by α‐Mangostin thus suggesting that α‐Mangostin can target CSCs to inhibit pancreatic cancer effectively. Gli signalling plays a crucial role in the self‐renewal and pluripotency of CSCs. α‐Mangostin inhibited the Gli transcription and the expression of Gli target genes (Nanog, Oct4, c‐Myc, Sox‐2 and KLF4) in CSCs. Using ChIP assay, we demonstrated that Nanog could directly bind to promoters of Cdk2, Cdk6, FGF4, c‐Myc and α‐Mangostin inhibited Nanog binding to these promoters. Conversely, the inhibitory effects of the α‐Mangostin on CSC proliferation and Gli or Nanog transcription and their targets were abrogated by either enforced activation of sonic hedgehog (Shh) or by the overexpression of Nanog. Taken together, our studies suggest that α‐Mangostin may act as Gli inhibitor and establishes the pre‐clinical significance of α‐Mangostin for the prevention and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yiming Ma
- Kansas City VA Medical Center, Kansas City, Missouri
| | - Wei Yu
- Kansas City VA Medical Center, Kansas City, Missouri
| | - Anju Shrivastava
- Department of Oncology, St. Joseph's Hospital and Medical Center, Phoenix, Arizona
| | - Rakesh K Srivastava
- Kansas City VA Medical Center, Kansas City, Missouri.,Department of Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, Missouri
| | - Sharmila Shankar
- Kansas City VA Medical Center, Kansas City, Missouri.,Department of Pathology, School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri
| |
Collapse
|
32
|
Zhang Z, Han H, Rong Y, Zhu K, Zhu Z, Tang Z, Xiong C, Tao J. Hypoxia potentiates gemcitabine-induced stemness in pancreatic cancer cells through AKT/Notch1 signaling. J Exp Clin Cancer Res 2018; 37:291. [PMID: 30486896 PMCID: PMC6263055 DOI: 10.1186/s13046-018-0972-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/19/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Profound chemoresistance remains an intractable obstacle in pancreatic cancer treatment. Pancreatic cancer stem cells (CSCs) and the ubiquitous hypoxic niche have been proposed to account for drug resistance. However, the mechanism involved requires further exploration. This study investigated whether the hypoxic niche enhances gemcitabine-induced stemness and acquired resistance in pancreatic cancer cells by activating the AKT/Notch1 signaling cascade. The therapeutic effects of blockading this signaling cascade on gemcitabine-enriched CSCs were also investigated. METHODS The expression levels of CSC-associated markers Bmi1 and Sox2 as well as those of proteins involved in AKT/Notch1 signaling were measured by Western blot analysis. The expression level of the pancreatic CSC marker CD24 was measured by flow cytometry. Change in gemcitabine sensitivity was evaluated by the MTT assay. The ability of sphere formation was tested by the sphere-forming assay in stem cell medium. The ability of migration and invasion was detected by the transwell migration/invasion assay. A mouse xenograft model of pancreatic cancer was established to determine the effect of Notch1 inhibition on the killing effect of gemcitabine in vivo. The ability of metastasis was investigated by an in vivo lung metastasis assay. RESULTS Gemcitabine promoted pancreatic cancer cell stemness and associated malignant phenotypes such as enhanced migration, invasion, metastasis, and chemoresistance. The AKT/Notch1 signaling cascade was activated after gemcitabine treatment and mediated this process. Blockading this pathway enhanced the killing effect of gemcitabine in vivo. However, supplementation with hypoxia treatment synergistically enhanced the AKT/Notch1 signaling pathway and collaboratively promoted gemcitabine-induced stemness. CONCLUSIONS These findings demonstrate a novel mechanism of acquired gemcitabine resistance in pancreatic cancer cells through induction of stemness, which was mediated by the activation of AKT/Notch1 signaling and synergistically aggravated by the ubiquitous hypoxic niche. Our results might provide new insights for identifying potential targets for reversing chemoresistance in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Zhengle Zhang
- Department of Pancreatic Surgery, Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060 Hubei Province China
| | - Han Han
- Department of Dermatology, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014 Hubei Province China
| | - Yuping Rong
- Department of Pancreatic Surgery, Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060 Hubei Province China
| | - Kongfan Zhu
- Department of Pancreatic Surgery, Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060 Hubei Province China
| | - Zhongchao Zhu
- Department of Pancreatic Surgery, Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060 Hubei Province China
| | - Zhigang Tang
- Department of Pancreatic Surgery, Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060 Hubei Province China
| | - Chenglong Xiong
- Department of Pancreatic Surgery, Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060 Hubei Province China
| | - Jing Tao
- Department of Pancreatic Surgery, Renmin Hospital, Wuhan University, 238 Jiefang Road, Wuhan, 430060 Hubei Province China
| |
Collapse
|
33
|
Dhar D, Deep G, Kumar S, Wempe MF, Raina K, Agarwal C, Agarwal R. Bitter melon juice exerts its efficacy against pancreatic cancer via targeting both bulk and cancer stem cells. Mol Carcinog 2018; 57:1166-1180. [PMID: 29727019 PMCID: PMC6118209 DOI: 10.1002/mc.22833] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer (PanC) is one of the deadliest malignancies worldwide and frontline treatment with gemcitabine becomes eventually ineffective due to increasing PanC resistance, suggesting additional approaches are needed to manage PanC. Recently, we have shown the efficacy of bitter melon juice (BMJ) against PanC cells, including those resistant to gemcitabine. As cancer stem cells (CSCs) are actively involved in PanC initiation, progression, relapse and drug-resistance, here we assessed BMJ ability in targeting pancreatic cancer-associated cancer stem cells (PanC-CSCs). We found BMJ efficacy against CD44+ /CD24+ /EpCAMhigh enriched PanC-CSCs in spheroid assays; BMJ also increased the sensitivity of gemcitabine-resistant PanC-CSCs. Exogenous addition of BMJ to PanC-CSC generated spheroids (not pre-exposed to BMJ) also significantly reduced spheroid number and size. Mechanistically, BMJ effects were associated with a decrease in the expression of genes and proteins involved in PanC-CSC renewal and proliferation. Specifically, immunofluorescence staining showed that BMJ decreases protein expression/nuclear localization of CSC-associated transcription factors SOX2, OCT4 and NANOG, and CSC marker CD44. Immunohistochemical analysis of MiaPaCa2 xenografts from BMJ treated animals also showed a significant decrease in the levels of CSC-associated transcription factors. Together, these results show BMJ potential in targeting PanC-CSC pool and associated regulatory pathways, suggesting the need for further investigation of its efficacy against PanC growth and progression including gemcitabine-resistant PanC.
Collapse
Affiliation(s)
- Deepanshi Dhar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO
| | - Gagan Deep
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO
| | - Sushil Kumar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO
| | - Michael F. Wempe
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO
- University of Colorado Cancer Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Komal Raina
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO
- University of Colorado Cancer Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO
- University of Colorado Cancer Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, Aurora, CO
- University of Colorado Cancer Center, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
34
|
Harbuzariu A, Oprea-Ilies GM, Gonzalez-Perez RR. The Role of Notch Signaling and Leptin-Notch Crosstalk in Pancreatic Cancer. MEDICINES (BASEL, SWITZERLAND) 2018; 5:medicines5030068. [PMID: 30004402 PMCID: PMC6164868 DOI: 10.3390/medicines5030068] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/27/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023]
Abstract
There is accumulating evidence that deregulated Notch signaling affects cancer development, and specifically pancreatic cancer (PC) progression. Notch canonical and non-canonical signaling has diverse impact on PC. Moreover, the actions of RBP-Jk (nuclear partner of activated Notch) independent of Notch signaling pathway seem to affect differently cancer progression. Recent data show that in PC and other cancer types the adipokine leptin can modulate Notch/RBP-Jk signaling, thereby, linking the pandemic obesity with cancer and chemoresistance. The potential pivotal role of leptin on PC, and its connection with Notch signaling and chemoresistance are still not completely understood. In this review, we will describe the most important aspects of Notch-RBP-Jk signaling in PC. Further, we will discuss on studies related to RBP-Jk-independent Notch and Notch-independent RPB-Jk signaling. We will also discuss on the novel crosstalk between leptin and Notch in PC and its implications in chemoresistance. The effects of leptin-Notch/RBP-Jk signaling on cancer cell proliferation, apoptosis, and drug resistance require more investigation. Data from these investigations could help to open unexplored ways to improve PC treatment success that has shown little progress for many years.
Collapse
Affiliation(s)
- Adriana Harbuzariu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA.
| | | | - Ruben R Gonzalez-Perez
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA.
| |
Collapse
|
35
|
Pan P, Chen T, Zhang Y, Qi Z, Qin J, Cui G, Guo X. LIN28A inhibits lysosome‑associated membrane glycoprotein 1 protein expression in embryonic stem and bladder cancer cells. Mol Med Rep 2018; 18:399-406. [PMID: 29749495 DOI: 10.3892/mmr.2018.8965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 03/14/2018] [Indexed: 11/06/2022] Open
Abstract
Tumor cells and embryonic stem cells (ESCs) have similar transcription mechanisms. LIN28A is an important factor in tumor cells and ESCs, it is an inhibitor of intracellular endoplasmic reticulum (ER)‑related protein translation in ESCs. The present study aimed to examine the effects of LIN28A on an ER‑related protein, lysosome‑associated membrane glycoprotein 1 (LAMP1), in human bladder cancer cells and mouse (m)ESCs, using reverse transcription‑quantitative polymerase chain reaction and western blotting to detect the expression of LAMP1 mRNA and protein, respectively, following LIN28A knockdown. LIN28A was revealed to promote the proliferation, migration and invasion in human bladder cancer cells. These data suggested similarities between ESC cells and cancer cells and may provide novel ideas for the use of induced embryonic stem cell differentiation to treat tumors.
Collapse
Affiliation(s)
- Peng Pan
- Department of Pathology, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| | - Ting Chen
- Department of Pathology, Guangzhou Medical University, Guangzhou, Guangdong 510182, P.R. China
| | - Yanmin Zhang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Shenzhen, Guangdong 518036, P.R. China
| | - Zhengyu Qi
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Shenzhen, Guangdong 518036, P.R. China
| | - Jie Qin
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Shenzhen, Guangdong 518036, P.R. China
| | - Guanghui Cui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Shenzhen, Guangdong 518036, P.R. China
| | - Xin Guo
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Shenzhen, Guangdong 518036, P.R. China
| |
Collapse
|
36
|
Emerging functional markers for cancer stem cell-based therapies: Understanding signaling networks for targeting metastasis. Semin Cancer Biol 2018; 53:90-109. [PMID: 29966677 DOI: 10.1016/j.semcancer.2018.06.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
Abstract
Metastasis is one of the most challenging issues in cancer patient management, and effective therapies to specifically target disease progression are missing, emphasizing the urgent need for developing novel anti-metastatic therapeutics. Cancer stem cells (CSCs) gained fast attention as a minor population of highly malignant cells within liquid and solid tumors that are responsible for tumor onset, self-renewal, resistance to radio- and chemotherapies, and evasion of immune surveillance accelerating recurrence and metastasis. Recent progress in the identification of their phenotypic and molecular characteristics and interactions with the tumor microenvironment provides great potential for the development of CSC-based targeted therapies and radical improvement in metastasis prevention and cancer patient prognosis. Here, we report on newly uncovered signaling mechanisms controlling CSC's aggressiveness and treatment resistance, and CSC-specific agents and molecular therapeutics, some of which are currently under investigation in clinical trials, gearing towards decisive functional CSC intrinsic or surface markers. One special research focus rests upon subverted regulatory pathways such as insulin-like growth factor 1 receptor signaling and its interactors in metastasis-initiating cell populations directly related to the gain of stem cell- and EMT-associated properties, as well as key components of the E2F transcription factor network regulating metastatic progression, microenvironmental changes, and chemoresistance. In addition, the study provides insight into systems biology tools to establish complex molecular relationships behind the emergence of aggressive phenotypes from high-throughput data that rely on network-based analysis and their use to investigate immune escape mechanisms or predict clinical outcome-relevant CSC receptor signaling signatures. We further propose that customized vector technologies could drastically enhance systemic drug delivery to target sites, and summarize recent progress and remaining challenges. This review integrates available knowledge on CSC biology, computational modeling approaches, molecular targeting strategies, and delivery techniques to envision future clinical therapies designed to conquer metastasis-initiating cells.
Collapse
|
37
|
Harbuzariu A, Rampoldi A, Daley-Brown DS, Candelaria P, Harmon TL, Lipsey CC, Beech DJ, Quarshie A, Ilies GO, Gonzalez-Perez RR. Leptin-Notch signaling axis is involved in pancreatic cancer progression. Oncotarget 2018; 8:7740-7752. [PMID: 27999190 PMCID: PMC5352357 DOI: 10.18632/oncotarget.13946] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 12/07/2016] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer (PC) shows a high death rate. PC incidence and prognosis are affected by obesity, a pandemic characterized by high levels of leptin. Notch is upregulated by leptin in breast cancer. Thus, leptin and Notch crosstalk could influence PC progression. Here we investigated in PC cell lines (BxPC-3, MiaPaCa-2, Panc-1, AsPC-1), derived tumorspheres and xenografts whether a functional leptin-Notch axis affects PC progression and expansion of pancreatic cancer stem cells (PCSC). PC cells and tumorspheres were treated with leptin and inhibitors of Notch (gamma-secretase inhibitor, DAPT) and leptin (iron oxide nanoparticle-leptin peptide receptor antagonist 2, IONP-LPrA2). Leptin treatment increased cell cycle progression and proliferation, and the expression of Notch receptors, ligands and targeted molecules (Notch1-4, DLL4, JAG1, Survivin and Hey2), PCSC markers (CD24/CD44/ESA, ALDH, CD133, Oct-4), ABCB1 protein, as well as tumorsphere formation. Leptin-induced effects on PC and tumorspheres were decreased by IONP-LPrA2 and DAPT. PC cells secreted leptin and expressed the leptin receptor, OB-R, which indicates a leptin autocrine/paracrine signaling loop could also affect tumor progression. IONP-LPrA2 treatment delayed the onset of MiaPaCa-2 xenografts, and decreased tumor growth and the expression of proliferation and PCSC markers. Present data suggest that leptin-Notch axis is involved in PC. PC has no targeted therapy and is mainly treated with chemotherapy, whose efficiency could be decreased by leptin and Notch activities. Thus, the leptin-Notch axis could be a novel therapeutic target, particularly for obese PC patients.
Collapse
Affiliation(s)
- Adriana Harbuzariu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310 USA
| | - Antonio Rampoldi
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310 USA
| | - Danielle S Daley-Brown
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310 USA
| | - Pierre Candelaria
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310 USA
| | - Tia L Harmon
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310 USA
| | - Crystal C Lipsey
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310 USA
| | - Derrick J Beech
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, 30310 USA
| | - Alexander Quarshie
- Biomedical Informatics Program and Master of Science in Clinical Research Program, Clinical Research Center, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Gabriela Oprea Ilies
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Grady Memorial Hospital, Atlanta, GA, 30303 USA
| | - Ruben R Gonzalez-Perez
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310 USA
| |
Collapse
|
38
|
Abstract
Pancreatic cancer is an aggressive malignancy with poor survival and high mortality rate with 250 000 deaths per year worldwide. The unique pancreatic cancer microenvironment serves as a major obstacle in the effective treatment of this malignancy. The microenvironment consists not only of pancreatic ductal adenocarcinoma cells but also comprises cells of pancreatic cancer stellate, vascular, and immune origin combined with a dense extracellular matrix containing collagen. The aforementioned pathology leads to an increased intratumor pressure combined with an erratic vascular proliferation within the tumor causing hypoxia and decreased drug delivery. This has led both scientists and clinicians to develop and study drugs with unique mechanisms of action to target the pancreatic cancer microenvironment. Herein, we discuss the pancreatic cancer hypoxic microenvironment, development of hypoxia-activated prodrugs, and results of trials utilizing those drugs to target pancreatic cancer.
Collapse
|
39
|
Pützer BM, Solanki M, Herchenröder O. Advances in cancer stem cell targeting: How to strike the evil at its root. Adv Drug Deliv Rev 2017; 120:89-107. [PMID: 28736304 DOI: 10.1016/j.addr.2017.07.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/10/2017] [Accepted: 07/16/2017] [Indexed: 12/18/2022]
Abstract
Cancer progression to metastatic stages is still unmanageable and the promise of effective anti-metastatic therapy remains largely unmet, emphasizing the need to develop novel therapeutics. The special focus here is on cancer stem cells (CSC) as the seed of tumor initiation, epithelial-mesenchymal transition, chemoresistance and, as a consequence, drivers of metastatic dissemination. We report on targeted therapies gearing towards the CSC's internal and membrane-anchored markers using agents such as antibody derivatives, nucleic therapeutics, small molecules and genetic payloads. Another emphasis lies on novel proceedings envisaged to deliver current and prospective therapies to the target sites using newest viral and non-viral vector technologies. In this review, we summarize recent progress and remaining challenges in therapeutic strategies to combat CSC.
Collapse
Affiliation(s)
- Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany.
| | - Manish Solanki
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| | - Ottmar Herchenröder
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| |
Collapse
|
40
|
Li J, Sun P, Yue Z, Zhang D, You K, Wang J. miR-144-3p Induces Cell Cycle Arrest and Apoptosis in Pancreatic Cancer Cells by Targeting Proline-Rich Protein 11 Expression via the Mitogen-Activated Protein Kinase Signaling Pathway. DNA Cell Biol 2017; 36:619-626. [PMID: 28574724 DOI: 10.1089/dna.2017.3656] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Jian Li
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Peisheng Sun
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhongyi Yue
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Dezhong Zhang
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Kun You
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jianguo Wang
- Department of General Surgery, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
41
|
Mohanta S, Siddappa G, Valiyaveedan SG, Dodda Thimmasandra Ramanjanappa R, Das D, Pandian R, Khora SS, Kuriakose MA, Suresh A. Cancer stem cell markers in patterning differentiation and in prognosis of oral squamous cell carcinoma. Tumour Biol 2017. [PMID: 28631562 DOI: 10.1177/1010428317703656] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Differentiation is a major histological parameter determining tumor aggressiveness and prognosis of the patient; cancer stem cells with their slow dividing and undifferentiated nature might be one of the factors determining the same. This study aims to correlate cancer stem cell markers (CD44 and CD147) with tumor differentiation and evaluate their subsequent effect on prognosis. Immunohistochemical analysis in treatment naïve oral cancer patients (n = 53) indicated that the expression of CD147 was associated with poorly differentiated squamous cell carcinoma and moderately differentiated squamous cell carcinoma (p < 0.01). Furthermore, co-expression analysis showed that 45% each of moderately differentiated squamous cell carcinoma and poorly differentiated squamous cell carcinoma patients were CD44high/CD147high as compared to only 10% of patients with well-differentiated squamous cell carcinoma. A three-way analysis indicated that differentiation correlated with recurrence and survival (p < 0.05) in only the patients with CD44high/CD147high cohort. Subsequently, relevance of these cancer stem cell markers in patterning the differentiation characteristics was evaluated in oral squamous cell carcinoma cell lines originating from different grades of oral cancer. Flowcytometry-based analysis indicated an increase in CD44+/CD147+ cells in cell lines of poorly differentiated squamous cell carcinoma (94.35 ± 1.14%, p < 0.001) and moderately differentiated squamous cell carcinoma origin (93.49 ± 0.47%, p < 0.001) as compared to cell line of well-differentiated squamous cell carcinoma origin (23.12% ± 0.49%). Expression profiling indicated higher expression of cancer stem cell and epithelial-mesenchymal transition markers in SCC029B (poorly differentiated squamous cell carcinoma originated; p ≤ 0.001), which was further translated into increased spheroid formation, migration, and invasion (p < 0.001) as compared to cell line of well-differentiated squamous cell carcinoma origin. This study suggests that CD44 and CD147 together improve the prognostic efficacy of tumor differentiation; in vitro results further point out that these markers might be determinant of differentiation characteristics, imparting properties of increased self-renewal, migration, and invasion.
Collapse
Affiliation(s)
- Simple Mohanta
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India.,2 School of Bio Sciences & Technology, Vellore Institute of Technology (VIT) University, Vellore, India.,3 Department of Head and Neck Oncology, Mazumdar Shaw Medical Center, Narayana Hrudayalaya, Bangalore, India
| | - Gangotri Siddappa
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India
| | - Sindhu Govindan Valiyaveedan
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India
| | - Ravindra Dodda Thimmasandra Ramanjanappa
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India.,3 Department of Head and Neck Oncology, Mazumdar Shaw Medical Center, Narayana Hrudayalaya, Bangalore, India
| | - Debashish Das
- 4 Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Bangalore, India
| | - Ramanan Pandian
- 5 GROW Lab, Narayana Nethralaya Foundation, Bangalore, India
| | - Samanta Sekhar Khora
- 2 School of Bio Sciences & Technology, Vellore Institute of Technology (VIT) University, Vellore, India
| | - Moni Abraham Kuriakose
- 3 Department of Head and Neck Oncology, Mazumdar Shaw Medical Center, Narayana Hrudayalaya, Bangalore, India.,6 Mazumdar Shaw Medical Centre-Roswell Park Collaboration Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Amritha Suresh
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India.,6 Mazumdar Shaw Medical Centre-Roswell Park Collaboration Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
42
|
Mooney B, Abdul-Raof N, Tian YI, Xie Y. Restriction of Cancer Metastatic Potential Using Embryonic Stem Cells Encapsulated in Alginate Hydrogel Microstrands. ACS Biomater Sci Eng 2017; 3:1769-1779. [DOI: 10.1021/acsbiomaterials.7b00237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Bridget Mooney
- Nanobioscience, Colleges
of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, New York 12203, United States
| | - Nurazhani Abdul-Raof
- Nanobioscience, Colleges
of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, New York 12203, United States
| | - Yangzi Isabel Tian
- Nanobioscience, Colleges
of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, New York 12203, United States
| | - Yubing Xie
- Nanobioscience, Colleges
of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, New York 12203, United States
| |
Collapse
|
43
|
Fu Z, Li G, Li Z, Wang Y, Zhao Y, Zheng S, Ye H, Luo Y, Zhao X, Wei L, Liu Y, Lin Q, Zhou Q, Chen R. Endogenous miRNA Sponge LincRNA-ROR promotes proliferation, invasion and stem cell-like phenotype of pancreatic cancer cells. Cell Death Discov 2017; 3:17004. [PMID: 28580169 PMCID: PMC5447127 DOI: 10.1038/cddiscovery.2017.4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/24/2016] [Accepted: 12/29/2016] [Indexed: 01/02/2023] Open
Abstract
The long intergenic non-coding RNA, regulator of reprogramming (linc-ROR) is an oncogene and plays a key role in the embryonic stem cell maintenance and is involved in cancer progression. The objective of this study was to analyze linc-ROR expression in pancreatic ductal adenocarcinoma (PDAC) and determine the regulation effects of linc-ROR on proliferation and invasion of cancer cells, as well as properties of cancer stem-like cells (CSLCs). In this study, we found that linc-ROR was up-regulated in PDAC tissues and related to poor prognosis. Linc-ROR knockdown in pancreatic cancer cells inhibited cell growth and arrested in G1 phrase. Suppressed linc-ROR expression also attenuated cancer cell migration, invasion, and epithelial-mesenchymal transition. We observed that linc-ROR expression was increased in CSLCs. Importantly, linc-ROR knockdown impaired the properties and tumorigenesis of pancreatic CSLCs in vivo. Mechanistically, we found that linc-ROR functioned as a competing endogenous RNA (ceRNA) to several tumor suppressor microRNAs, particularly some members of let-7 family. We conclude that, as a crucial oncogene, linc-ROR promotes cell proliferation, invasiveness and contributes to stem cell properties of CSLCs in PDAC via acting as a ceRNA to regulate function of microRNAs. The linc-ROR is a potential therapeutic target for PDAC.
Collapse
Affiliation(s)
- Zhiqiang Fu
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guolin Li
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihua Li
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingxue Wang
- Department of Endocrinology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Yue Zhao
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shangyou Zheng
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huilin Ye
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuming Luo
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaohui Zhao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lusheng Wei
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yimin Liu
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing Lin
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quanbo Zhou
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rufu Chen
- Department of Hepato-Pancreato-Billiary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
44
|
Bhartiya D. Stem cells to replace or regenerate the diabetic pancreas: Huge potential & existing hurdles. Indian J Med Res 2017; 143:267-74. [PMID: 27241638 PMCID: PMC4892071 DOI: 10.4103/0971-5916.182615] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Various stem cell sources are being explored to treat diabetes since the proof-of-concept for cell therapy was laid down by transplanting cadaveric islets as a part of Edmonton protocol in 2000. Human embryonic stem (hES) cells derived pancreatic progenitors have got US-FDA approval to be used in clinical trials to treat type 1 diabetes mellitus (T1DM). However, these progenitors more closely resemble their foetal counterparts and thus whether they will provide long-term regeneration of adult human pancreas remains to be demonstrated. In addition to lifestyle changes and administration of insulin sensitizers, regeneration of islets from endogenous pancreatic stem cells may benefit T2DM patients. The true identity of pancreatic stem cells, whether these exist or not, whether regeneration involves reduplication of existing islets or ductal epithelial cells transdifferentiate, remains a highly controversial area. We have recently demonstrated that a novel population of very small embryonic-like stem cells (VSELs) is involved during regeneration of adult mouse pancreas after partial-pancreatectomy. VSELs (pluripotent stem cells in adult organs) should be appreciated as an alternative for regenerative medicine as these are autologous (thus immune rejection issues do not exist) with no associated risk of teratoma formation. T2DM is a result of VSELs dysfunction with age and uncontrolled proliferation of VSELs possibly results in pancreatic cancer. Extensive brainstorming and financial support are required to exploit the potential of endogenous VSELs to regenerate the pancreas in a patient with diabetes.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health (ICMR), Mumbai, India
| |
Collapse
|
45
|
Freitag D, McLean AL, Simon M, Koch A, Grube S, Walter J, Kalff R, Ewald C. NANOG overexpression and its correlation with stem cell and differentiation markers in meningiomas of different WHO grades. Mol Carcinog 2017; 56:1953-1964. [PMID: 28345785 DOI: 10.1002/mc.22653] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 03/10/2017] [Accepted: 03/23/2017] [Indexed: 01/15/2023]
Abstract
NANOG, as a key regulator of pluripotency and acting synergistically with other factors, has been described as a crucial transcription factor in various types of cancer. In meningiomas the expression of this marker has not yet been described. With our study, we aimed to identify and localize NANOG and other possible markers of pluripotency, stem cell properties and differentiation in meningioma tissue, to elucidate a possible effect on tumorigenesis. The gene expression levels of NANOG (NANOG1 and NANOGP8), SOX2, OCT4, KLF4, ABCG2, CMYC, MSI1, CD44, NOTCH1, NES, SALL4B, TP53, and EPAS1 were quantitatively examined using RT-qPCR in 33 surgical specimens of low- (WHO grade I) as well as in high-grade (WHO grade II/III) meningiomas with dural tissue as reference. Immunofluorescence co-localization analysis following confocal fluorescence microscopy for NANOG, OCT4, SOX2, Nestin, KI-67, and CD44 was also performed. There was a significant overexpression of NANOG, MSI1, and EPAS1 and a downregulation of NES in all examined tumors. Subgroup analysis (WHO grade I versus grade II/III) revealed differences in the expression of NANOG, CD44, and MSI1. We found 1% NANOG-positive (NANOG+) cells in low-grade and 2% in grade II/III meningiomas co-expressing the other mentioned markers in various compositions. In particular, NANOG+ cells expressing SOX2 and OCT4 were successfully identified (26% low-grade versus 20% high-grade). Our data reveal an overexpression of NANOG and other markers of pluripotency and stemness in meningiomas. Such potentially pluripotent "stem cell-like" cells may have an impact on tumorigenesis and progression in human meningiomas.
Collapse
Affiliation(s)
- Diana Freitag
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Aaron Lawson McLean
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Michèle Simon
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.,Department of Neurosurgery, Vivantes Klinikum im Friedrichshain, Berlin, Germany
| | - Arend Koch
- Department of Neuropathology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne Grube
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Jan Walter
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Rolf Kalff
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Christian Ewald
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.,Department of Neurosurgery, Städtisches Klinikum Brandenburg, Brandenburg an der Havel, Germany
| |
Collapse
|
46
|
Strnadel J, Choi S, Fujimura K, Wang H, Zhang W, Wyse M, Wright T, Gross E, Peinado C, Park HW, Bui J, Kelber J, Bouvet M, Guan KL, Klemke RL. eIF5A-PEAK1 Signaling Regulates YAP1/TAZ Protein Expression and Pancreatic Cancer Cell Growth. Cancer Res 2017; 77:1997-2007. [PMID: 28381547 PMCID: PMC5392372 DOI: 10.1158/0008-5472.can-16-2594] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/05/2016] [Accepted: 12/30/2016] [Indexed: 01/16/2023]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), mutant KRAS stimulates the translation initiation factor eIF5A and upregulates the focal adhesion kinase PEAK1, which transmits integrin and growth factor signals mediated by the tumor microenvironment. Although eIF5A-PEAK1 signaling contributes to multiple aggressive cancer cell phenotypes, the downstream signaling processes that mediate these responses are uncharacterized. Through proteomics and informatic analyses of PEAK1-depleted PDAC cells, we defined protein translation, cytoskeleton organization, and cell-cycle regulatory pathways as major pathways controlled by PEAK1. Biochemical and functional studies revealed that the transcription factors YAP1 and TAZ are key targets of eIF5A-PEAK1 signaling. YAP1/TAZ coimmunoprecipitated with PEAK1. Interfering with eIF5A-PEAK1 signaling in PDAC cells inhibited YAP/TAZ protein expression, decreasing expression of stem cell-associated transcription factors (STF) including Oct4, Nanog, c-Myc, and TEAD, thereby decreasing three-dimensional (3D) tumor sphere growth. Conversely, amplified eIF5A-PEAK1 signaling increased YAP1/TAZ expression, increasing expression of STF and enhancing 3D tumor sphere growth. Informatic interrogation of mRNA sequence databases revealed upregulation of the eIF5A-PEAK1-YAP1-TEAD signaling module in PDAC patients. Taken together, our findings indicate that eIF5A-PEAK1-YAP signaling contributes to PDAC development by regulating an STF program associated with increased tumorigenicity. Cancer Res; 77(8); 1997-2007. ©2017 AACR.
Collapse
Affiliation(s)
- Jan Strnadel
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Sunkyu Choi
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Ken Fujimura
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Huawei Wang
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Wei Zhang
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Meghan Wyse
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Tracy Wright
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Emilie Gross
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Carlos Peinado
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Hyun Woo Park
- Moores Cancer Center, University of California, San Diego, La Jolla, California
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Jack Bui
- Department of Pathology, University of California, San Diego, La Jolla, California
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Jonathan Kelber
- Department of Biology, California State University Northridge, Northridge, California
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, La Jolla, California
| | - Kun-Liang Guan
- Moores Cancer Center, University of California, San Diego, La Jolla, California
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| | - Richard L Klemke
- Department of Pathology, University of California, San Diego, La Jolla, California.
- Moores Cancer Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
47
|
Candelaria PV, Rampoldi A, Harbuzariu A, Gonzalez-Perez RR. Leptin signaling and cancer chemoresistance: Perspectives. World J Clin Oncol 2017; 8:106-119. [PMID: 28439492 PMCID: PMC5385432 DOI: 10.5306/wjco.v8.i2.106] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/20/2016] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity is a major health problem and currently is endemic around the world. Obesity is a risk factor for several different types of cancer, significantly promoting cancer incidence, progression, poor prognosis and resistance to anti-cancer therapies. The study of this resistance is critical as development of chemoresistance is a serious drawback for the successful and effective drug-based treatments of cancer. There is increasing evidence that augmented adiposity can impact on chemotherapeutic treatment of cancer and the development of resistance to these treatments, particularly through one of its signature mediators, the adipokine leptin. Leptin is a pro-inflammatory, pro-angiogenic and pro-tumorigenic adipokine that has been implicated in many cancers promoting processes such as angiogenesis, metastasis, tumorigenesis and survival/resistance to apoptosis. Several possible mechanisms that could potentially be developed by cancer cells to elicit drug resistance have been suggested in the literature. Here, we summarize and discuss the current state of the literature on the role of obesity and leptin on chemoresistance, particularly as it relates to breast and pancreatic cancers. We focus on the role of leptin and its significance in possibly driving these proposed chemoresistance mechanisms, and examine its effects on cancer cell survival signals and expansion of the cancer stem cell sub-populations.
Collapse
|
48
|
Wang P, Wan WW, Xiong SL, Feng H, Wu N. Cancer stem-like cells can be induced through dedifferentiation under hypoxic conditions in glioma, hepatoma and lung cancer. Cell Death Discov 2017; 3:16105. [PMID: 28179999 PMCID: PMC5253691 DOI: 10.1038/cddiscovery.2016.105] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/15/2016] [Accepted: 12/17/2016] [Indexed: 12/11/2022] Open
Abstract
Traditional studies have shown that transcription factors, including SOX-2, OCT-4, KLF-4, Nanog and Lin-28A, contribute to the dedifferentiation and reprogramming process in normal tissues. Hypoxia is a physiological phenomenon that exists in tumors and promotes the expression of SOX-2, OCT-4, KLF-4, Nanog and Lin-28A. Therefore, an interesting question is whether hypoxia as a stimulating factor promotes the process of dedifferentiation and induces the formation of cancer stem-like cells. Studies have shown that OCT-4 and Nanog overexpression induced the formation of cancer stem cell-like cells through dedifferentiation and enhanced malignancy in lung adenocarcinoma, and reprogramming SOX-2 in pancreatic cancer cells also promoted the dedifferentiation process. Therefore, we investigated this phenomenon in glioma, lung cancer and hepatoma cells and found that the transcription factors mentioned above were highly expressed under hypoxic conditions and induced the formation of spheres, which exhibited asymmetric division and cell cycle arrest. The dedifferentiation process induced by hypoxia highlights a new pattern of cancer development and recurrence, demonstrating that all kinds of cancer cells and the hypoxic microenvironment should be taken into consideration when developing tumor therapies.
Collapse
Affiliation(s)
- Pan Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University , Chongqing 400038, China
| | - Wen-Wu Wan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University , Chongqing 400038, China
| | | | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University , Chongqing 400038, China
| | - Nan Wu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University , Chongqing 400038, China
| |
Collapse
|
49
|
Zhou S, Abdouh M, Arena V, Arena M, Arena GO. Reprogramming Malignant Cancer Cells toward a Benign Phenotype following Exposure to Human Embryonic Stem Cell Microenvironment. PLoS One 2017; 12:e0169899. [PMID: 28068409 PMCID: PMC5222525 DOI: 10.1371/journal.pone.0169899] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 12/23/2016] [Indexed: 12/21/2022] Open
Abstract
The embryonic microenvironment is well known to be non-permissive for tumor development because early developmental signals naturally suppress the expression of proto-oncogenes. In an analogous manner, mimicking an early embryonic environment during embryonic stem cell culture has been shown to suppress oncogenic phenotypes of cancer cells. Exosomes derived from human embryonic stem cells harbor substances that mirror the content of the cells of origin and have been reported to reprogram hematopoietic stem/progenitor cells via horizontal transfer of mRNA and proteins. However, the possibility that these embryonic stem cells-derived exosomes might be the main effectors of the anti-tumor effect mediated by the embryonic stem cells has not been explored yet. The present study aims to investigate whether exosomes derived from human embryonic stem cells can reprogram malignant cancer cells to a benign stage and reduce their tumorigenicity. We show that the embryonic stem cell-conditioned medium contains factors that inhibit cancer cell growth and tumorigenicity in vitro and in vivo. Moreover, we demonstrate that exosomes derived from human embryonic stem cells display anti-proliferation and pro-apoptotic effects, and decrease tumor size in a xenograft model. These exosomes are also able to transfer their cargo into target cancer cells, inducing a dose-dependent increase in SOX2, OCT4 and Nanog proteins, leading to a dose-dependent decrease of cancer cell growth and tumorigenicity. This study shows for the first time that human embryonic stem cell-derived exosomes play an important role in the tumor suppressive activity displayed by human embryonic stem cells.
Collapse
Affiliation(s)
- Shufeng Zhou
- Cancer Research Program, McGill University Health Centre-Research Institute, Montreal, Canada
- Department of Experimental Surgery, Montreal General Hospital, McGill University, Montreal, Canada
| | - Mohamed Abdouh
- Cancer Research Program, McGill University Health Centre-Research Institute, Montreal, Canada
| | - Vincenzo Arena
- Deparment of Obstetrics and Gynecology, Santo Bambino Hospital, Catania, Italy
| | - Manuel Arena
- Department of Surgical Sciences, Organ Transplantation and Advances Technologies, University of Catania, Catania, Italy
| | - Goffredo Orazio Arena
- Cancer Research Program, McGill University Health Centre-Research Institute, Montreal, Canada
- Department of Surgery, St. Mary Hospital, McGill University, Montreal, Canada
- * E-mail:
| |
Collapse
|
50
|
Jiang Z, Liu Y, Wang C. Oncogenic NanogP8 expression regulates cell proliferation and migration through the Akt/mTOR signaling pathway in human gastric cancer - SGC-7901cell line. Onco Targets Ther 2016; 9:4859-66. [PMID: 27563247 PMCID: PMC4984828 DOI: 10.2147/ott.s97861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Although elevated expression of NanogP8 has been detected in many human tumor tissues, its role in gastric tumorigenesis remains unclear. Therefore, this study aimed to investigate the function and regulatory mechanism of NanogP8 in gastric cancer. METHODS In this study, NanogP8 cDNA was amplified by real time polymerase chain reaction from the human gastric cancer cell line SGC-7901. The shRNA for RNA interference was established. The NanogP8, pAkt, Akt, pERK, ERK, p-mTOR, and mTOR proteins were detected by using the Western blot assay. Cell viability was evaluated by using the CCK-8 assay. Cell migration and invasion were also examined by using the transwell assay. RESULTS The results indicated that the NanogP8 overexpression promoted proliferation and migration of SGC-7901 cell line, whereas its ablation exerted opposite effects. Interestingly, NanogP8 activated Akt, a key mediator of survival signals, and without affecting total Akt protein level. The NanogP8-increased gastric cell proliferation was downregulated by Akt inhibition. Our results further showed that increasing NanogP8 expression in human gastric cancer cells promoted cell proliferation by activating the AKT/mTOR pathway and further maintained gastric cell survival. CONCLUSION Our findings extend the knowledge regarding the oncogenic functions and proved that the NanogP8 regulates cell proliferation and migration by Akt/mTOR signaling pathway in human gastric cancer SGC-7901cell line.
Collapse
Affiliation(s)
- Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People's Republic of China
| | - Yao Liu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People's Republic of China
| | - Chuan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, People's Republic of China
| |
Collapse
|