1
|
Ramilo Amor A, Enlund S, Sinha I, Jiang Q, Hermanson O, Nilsson A, Shirazi Fard S, Holm F. A distinct alternative mRNA splicing profile identifies the oncogenic CD44 transcript variant 3 in KMT2A-rearranged pediatric T-cell acute lymphoblastic leukemia cells. Exp Hematol 2025; 145:104712. [PMID: 39793727 DOI: 10.1016/j.exphem.2025.104712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 01/13/2025]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL), which constitutes of 10-15% of all pediatric acute lymphoblastic leukemia (ALL) cases, is known for its complex pathology due to pervasive genetic and chromosomal abnormalities. Although most children are successfully cured, chromosomal rearrangements involving the KMT2A gene is considered a poor prognostic factor. In a cohort of 171 pediatric T-ALL samples, we have studied differences in gene and splice variant patterns in KMT2A-rearranged (KMT2A-r) T-ALL compared with KMT2A-negative (KMT2A-wt) T-ALL samples. Our results have identified a distinct gene expression and splice variant expression pattern in pediatric KMT2A-r patient samples including significant expression of splicing regulatory markers ESRP1 and MBNL3. Additionally, the prosurvival long transcript variant of BCL2 were upregulated in KMT2A-r compared with KMT2A-wt T-ALL samples. Lastly, increased levels of activating methylation in the promoter region of CD44 were identified followed by an upregulation of the oncogenic transcript variant CD44v3 in KMT2A-r T-ALL. Together, this suggests that CD44v3 could play a potential role as gene expression-based risk stratification of KMT2A-r T-ALL and could possibly serve as a therapeutic target using splicing modulators.
Collapse
Affiliation(s)
- Amanda Ramilo Amor
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Sabina Enlund
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Indranil Sinha
- Department of Molecular Biosciences, Wenner-Gren Institutet, Stockholm University, Stockholm, Sweden
| | - Qingfei Jiang
- Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Ola Hermanson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Nilsson
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Shahrzad Shirazi Fard
- Division of Pediatric Oncology and Surgery, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Frida Holm
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
2
|
Mosoane B, McCabe M, Jackson BS, Dlamini Z. CD44 Variant Expression in Follicular Cell-Derived Thyroid Cancers: Implications for Overcoming Multidrug Resistance. Molecules 2025; 30:1899. [PMID: 40363706 PMCID: PMC12073131 DOI: 10.3390/molecules30091899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Thyroid cancer (TC) is a significant global health issue that exhibits notable heterogeneity in incidence and outcomes. In low-resource settings such as Africa, delayed diagnosis and limited healthcare access exacerbate mortality rates. Among follicular cell-derived thyroid cancers-including papillary (PTC), follicular (FTC), anaplastic (ATC), and poorly differentiated (PDTC) subtypes-the role of CD44 variants has emerged as a critical factor influencing tumor progression and multidrug resistance (MDR). CD44, a transmembrane glycoprotein, and its splice variants (CD44v) mediate cell adhesion, migration, and survival, contributing to cancer stem cell (CSC) maintenance and therapy resistance. Differential expression patterns of CD44 isoforms across TC subtypes have shown diagnostic, prognostic, and therapeutic implications. Specifically, CD44v6 expression in PTC has been correlated with metastasis and aggressive tumor behavior, while in FTC, its expression aids in distinguishing malignant from benign lesions. Furthermore, CD44 contributes to MDR through enhanced drug efflux via ABC transporters, apoptosis evasion, and CSC maintenance via the Wnt/β-catenin and PI3K/Akt pathways. Targeted therapies against CD44 such as monoclonal antibodies, hyaluronic acid-based nanocarriers, and gene-editing technologies hold promise in overcoming MDR. However, despite the mounting evidence supporting CD44-targeted strategies in various cancers, research on this therapeutic potential in TC remains limited. This review synthesizes existing knowledge on CD44 variant expression in follicular cell-derived thyroid cancers and highlights potential therapeutic strategies to mitigate MDR, particularly in high-burden regions, thereby improving patient outcomes and survival.
Collapse
Affiliation(s)
- Benny Mosoane
- Department of Anatomical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Michelle McCabe
- Department of Anatomical Pathology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| | - Brandon S. Jackson
- Breast and Endocrine Unit, Department of General Surgery, University of Pretoria, Kalafong Provincial Tertiary Hospital, Pretoria 0001, South Africa;
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0001, South Africa
| |
Collapse
|
3
|
Sencha LM, Karpova MA, Dobrynina OE, Balalaeva IV. Cell-type dependent effect of 3D collagen matrix on cancer cell resistance to suboptimal conditions: the case of serum deprivation, glucose starvation, and hypoxia. Tissue Cell 2025; 93:102719. [PMID: 39823703 DOI: 10.1016/j.tice.2024.102719] [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: 09/03/2024] [Revised: 12/13/2024] [Accepted: 12/29/2024] [Indexed: 01/20/2025]
Abstract
The extracellular matrix (ECM) and its primary chemical components, including collagen, play a pivotal role in carcinogenesis and tumor progression. The ECM actively regulates cell proliferation, migration, and, importantly, resistance to various adverse factors. It is widely recognized as a key factor in modifying the resistance of tumor cells to various treatment modalities and cytotoxic compounds. However, the role of the ECM in tumor cell adaptation to nutritional deficiencies and hypoxic conditions remains significantly less studied. Since it is generally accepted that tumor cells resistance increases when cultured in a three-dimensional matrix, we sought to experimentally test the universality of this statement. In this work, we analyzed the responses of tumor cells with varying origins and proliferative activities, including human bladder carcinoma, epidermoid carcinoma, and ovarian carcinoma, to deprivation of serum, glucose and oxygen. We compared cell resistance to suboptimal conditions when cultured in a monolayer on tissue culture (TC)-treated polystyrene, on collagen-coated surfaces, or within a three-dimensional hydrogel composed of collagen type I. All three cell lines were stably transfected with fluorescent protein genes. To register the cell growth dynamics, we used a fluorescence-based technique that allows long-term quantitative observations without disrupting the hydrogel. The analyzed cell lines demonstrated different patterns of relative sensitivity to suboptimal conditions. We revealed that the direction and intensity of the collagen matrix effect depend on the cell type. Slowly proliferating ovarian carcinoma cells showed no noticeable changes in their behavior when cultured in a gel compared to a monolayer. In the case of bladder carcinoma, we registered predominantly resistance-stimulating effect of the collagen matrix, but it was significant only under serum deprivation. The most pronounced effect of collagen was registered for epidermoid carcinoma. Importantly, this effect was ambivalent: gel-embedded cells demonstrated significantly enhanced resistance to serum deprivation, but, at the same time, they were more responsive to glucose starvation and hypoxic conditions. We attribute the registered phenomenon to the individual characteristics of tumor cells with different origins and metabolic activities.
Collapse
Affiliation(s)
- Ludmila M Sencha
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Maria A Karpova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga E Dobrynina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Irina V Balalaeva
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.
| |
Collapse
|
4
|
Falougy M, Taubitz C, Ragab M, Patil A, Jensen J, Hoppe S, Kümpers C, Ribbat-Idel J, Rades D, Hakim SG. Prognostic Value of SOX2 and NANOG Expression in Recurrent Oral Squamous Cell Carcinoma. Cancers (Basel) 2025; 17:1181. [PMID: 40227667 PMCID: PMC11987986 DOI: 10.3390/cancers17071181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Recurrent oral squamous cell carcinoma (re-OSCC) poses a serious therapeutic challenge and is linked to poor survival outcomes. SOX2 and NANOG, key transcription factors in cancer stem cell biology, may drive tumor progression and therapy resistance. However, their prognostic value in re-OSCC and their relationship to adjuvant therapy remain unclear. METHODS We retrospectively analyzed a single-center cohort of 94 patients with re-OSCC treated with curative intent via (1) surgery alone, (2) surgery plus adjuvant radiotherapy (RT), or (3) surgery plus adjuvant radiochemotherapy (RCT). Tissue microarrays (TMAs) were constructed from matched primary and recurrent tumors and immunohistochemical (IHC) staining for SOX2, and NANOG was quantified using H-scores. Post-recurrence overall survival (prOS) and post-recurrence disease-free survival (prDFS) were evaluated using Kaplan-Meier analysis and Cox proportional hazards models. RESULTS SOX2 expression and survival: Elevated SOX2 expression (H-score > 14) in re-OSCC was significantly associated with improved prOS (p = 0.013) and prDFS (p = 0.026). Notably, patients who had received adjuvant therapy (particularly RCT) showed higher SOX2 levels in recurrent tumors compared to those treated with surgery alone. NANOG expression and therapy: NANOG expression declined markedly from primary to recurrent tumors (median H-score 42.2 vs. 8.7; p < 0.001). This decline was most pronounced in patients treated with surgery alone. Despite this dynamic change, NANOG expression did not correlate significantly with prOS or prDFS. Other prognostic factors include advanced tumor stage (rT2-rT4) and lymph node involvement (rN+/x)m which remained significant predictors of worse survival in the recurrent setting, regardless of adjuvant therapy. CONCLUSION SOX2 overexpression in re-OSCC correlates with better survival, suggesting a unique prognostic role distinct from primary disease. Adjuvant therapy, especially RCT, appears to maintain or elevate SOX2 levels, potentially contributing to improved treatment response. In contrast, although NANOG expression decreases in recurrence, particularly in patients who undergo surgery alone, it does not significantly affect survival outcomes. These findings underscore the importance of context-specific biomarker assessments and provide a rationale for incorporating SOX2 status into personalized treatment strategies for re-OSCC.
Collapse
Affiliation(s)
- Mohamed Falougy
- Department of Oral and Maxillofacial Surgery, Head and Neck Cancer Center, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany (S.G.H.)
| | - Clara Taubitz
- Department of Oral and Maxillofacial Surgery, Head and Neck Cancer Center, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany (S.G.H.)
| | - Mohab Ragab
- Department of Internal Medicine II, Klinikum Rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, 81675 Munich, Germany
| | - Akshay Patil
- Derby Clinical Trials Support Unit, University Hospitals of Derby and Burton NHS Foundation Trust, Derby DE22 3NE, UK
| | - Justus Jensen
- Department of Oral and Maxillofacial Surgery, Head and Neck Cancer Center, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany (S.G.H.)
| | - Steffen Hoppe
- Department of Oral and Maxillofacial Surgery, Head and Neck Cancer Center, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany (S.G.H.)
| | - Christiane Kümpers
- Institute of Pathology, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany
| | - Julika Ribbat-Idel
- Institute of Pathology, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany
| | - Dirk Rades
- Department of Radiation Oncology, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany
| | - Samer George Hakim
- Department of Oral and Maxillofacial Surgery, Head and Neck Cancer Center, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany (S.G.H.)
- Department of Oral and Maxillofacial Surgery, Helios Medical Center Schwerin, 19055 Schwerin, Germany
| |
Collapse
|
5
|
Le Minh G, Merzy J, Esquea EM, Ahmed NN, Young RG, Sharp RJ, Dhameliya TT, Agana B, Lee MH, Bethard JR, Comte-Walters S, Ball LE, Reginato MJ. GATAD2B O-GlcNAcylation Regulates Breast Cancer Stem-like Potential and Drug Resistance. Cells 2025; 14:398. [PMID: 40136647 PMCID: PMC11941746 DOI: 10.3390/cells14060398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/22/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The growth of breast tumors is driven and controlled by a subpopulation of cancer cells resembling adult stem cells, which are called cancer stem-like cells (CSCs). In breast cancer, the function and maintenance of CSCs are influenced by protein O-GlcNAcylation and the enzyme responsible for this post-translational modification, O-GlcNAc transferase (OGT). However, the mechanism of CSCs regulation by OGT and O-GlcNAc cycling in breast cancer is still unclear. Analysis of the proteome and O-GlcNAcome, revealed GATAD2B, a component of the Nucleosome Remodeling and Deacetylase (NuRD) complex, as a substrate regulated by OGT. Reducing GATAD2B genetically impairs mammosphere formation, decreases expression of self-renewal factors and CSCs population. O-GlcNAcylation of GATAD2B at the C-terminus protects GATAD2B from ubiquitination and proteasomal degradation in breast cancer cells. We identify ITCH as a novel E3 ligase for GATAD2B and show that targeting ITCH genetically increases GATAD2B levels and increases CSCs phenotypes. Lastly, we show that overexpression of wild-type GATAD2B, but not the mutant lacking C-terminal O-GlcNAc sites, promotes mammosphere formation, expression of CSCs factors and drug resistance. Together, we identify a key role of GATAD2B and ITCH in regulating CSCs in breast cancer and GATAD2B O-GlcNAcylation as a mechanism regulating breast cancer stem-like populations and promoting chemoresistance.
Collapse
Affiliation(s)
- Giang Le Minh
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (G.L.M.)
| | - Jessica Merzy
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (G.L.M.)
| | - Emily M. Esquea
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (G.L.M.)
| | - Nusaiba N. Ahmed
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (G.L.M.)
| | - Riley G. Young
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (G.L.M.)
| | - Ryan J. Sharp
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (G.L.M.)
| | - Tejsi T. Dhameliya
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (G.L.M.)
| | - Bernice Agana
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA (L.E.B.)
| | - Mi-Hye Lee
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA (L.E.B.)
| | - Jennifer R. Bethard
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA (L.E.B.)
| | - Susana Comte-Walters
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA (L.E.B.)
| | - Lauren E. Ball
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA (L.E.B.)
| | - Mauricio J. Reginato
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA 19102, USA; (G.L.M.)
- Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
6
|
Yanova M, Stepanova E, Maltseva D, Tonevitsky A. CD44 variant exons induce chemoresistance by modulating cell death pathways. Front Cell Dev Biol 2025; 13:1508577. [PMID: 40114966 PMCID: PMC11924683 DOI: 10.3389/fcell.2025.1508577] [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: 10/10/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025] Open
Abstract
Cancer chemoresistance presents a challenge in oncology, often leading to treatment failure and disease progression. CD44, a multifunctional cell surface glycoprotein, has garnered attention for its involvement in various aspects of cancer biology. Through alternative splicing, CD44 can form isoforms with the inclusion of only standard exons, typical for normal tissue, or with the addition of variant exons, frequently expressed in cancer tissue and associated with chemoresistance. The functions of CD44 involved in regulation of cancer signaling pathways are being actively studied, and the significance of specific variant exons in modulating cell death pathways, central to the response of cancer cells to chemotherapy, begins to become apparent. This review provides a comprehensive analysis of the association of CD44 variant exons/total CD44 with clinical outcomes of patients undergoing chemotherapy. The role of CD44 variant exons v6, v9 and others with a significant effect on patient chemotherapy outcomes by means of key cellular death pathways such as apoptosis, ferroptosis and autophagy modulation is further identified, and their impact on drug resistance is highlighted. An overview of clinical trials aimed at targeting variant exon-containing isoforms is provided, and possible directions for further development of CD44-targeted therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Maria Yanova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Evgeniya Stepanova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Diana Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
7
|
Pashkina E, Bykova M, Berishvili M, Lazarev Y, Kozlov V. Hyaluronic Acid-Based Drug Delivery Systems for Cancer Therapy. Cells 2025; 14:61. [PMID: 39851489 PMCID: PMC11764402 DOI: 10.3390/cells14020061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/26/2025] Open
Abstract
In recent years, hyaluronic acid (HA) has attracted increasing attention as a promising biomaterial for the development of drug delivery systems. Due to its unique properties, such as high biocompatibility, low toxicity, and modifiability, HA is becoming a basis for the creation of targeted drug delivery systems, especially in the field of oncology. Receptors for HA overexpressed in subpopulations of cancer cells, and one of them, CD44, is recognized as a molecular marker for cancer stem cells. This review examines the role of HA and its receptors in health and tumors and analyzes existing HA-based delivery systems and their use in various types of cancer. The development of new HA-based drug delivery systems will bring new opportunities and challenges to anti-cancer therapy.
Collapse
Affiliation(s)
- Ekaterina Pashkina
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
- Department of Clinical Immunology, Novosibirsk State Medical University, 52, Krasny Prospect, 630091 Novosibirsk, Russia
| | - Maria Bykova
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | - Maria Berishvili
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
| | - Yaroslav Lazarev
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 2, Pirogova Street, 630090 Novosibirsk, Russia
| | - Vladimir Kozlov
- Research Institute of Fundamental and Clinical Immunology, 14, Yadrintsevskaya St., 630099 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 2, Pirogova Street, 630090 Novosibirsk, Russia
| |
Collapse
|
8
|
Dakal TC, Bhushan R, Xu C, Gadi BR, Cameotra SS, Yadav V, Maciaczyk J, Schmidt‐Wolf IGH, Kumar A, Sharma A. Intricate relationship between cancer stemness, metastasis, and drug resistance. MedComm (Beijing) 2024; 5:e710. [PMID: 39309691 PMCID: PMC11416093 DOI: 10.1002/mco2.710] [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: 03/13/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/25/2024] Open
Abstract
Cancer stem cells (CSCs) are widely acknowledged as the drivers of tumor initiation, epithelial-mesenchymal transition (EMT) progression, and metastasis. Originating from both hematologic and solid malignancies, CSCs exhibit quiescence, pluripotency, and self-renewal akin to normal stem cells, thus orchestrating tumor heterogeneity and growth. Through a dynamic interplay with the tumor microenvironment (TME) and intricate signaling cascades, CSCs undergo transitions from differentiated cancer cells, culminating in therapy resistance and disease recurrence. This review undertakes an in-depth analysis of the multifaceted mechanisms underlying cancer stemness and CSC-mediated resistance to therapy. Intrinsic factors encompassing the TME, hypoxic conditions, and oxidative stress, alongside extrinsic processes such as drug efflux mechanisms, collectively contribute to therapeutic resistance. An exploration into key signaling pathways, including JAK/STAT, WNT, NOTCH, and HEDGEHOG, sheds light on their pivotal roles in sustaining CSCs phenotypes. Insights gleaned from preclinical and clinical studies hold promise in refining drug discovery efforts and optimizing therapeutic interventions, especially chimeric antigen receptor (CAR)-T cell therapy, cytokine-induced killer (CIK) cell therapy, natural killer (NK) cell-mediated CSC-targeting and others. Ultimately use of cell sorting and single cell sequencing approaches for elucidating the fundamental characteristics and resistance mechanisms inherent in CSCs will enhance our comprehension of CSC and intratumor heterogeneity, which ultimately would inform about tailored and personalized interventions.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology LabDepartment of BiotechnologyMohanlal Sukhadia UniversityUdaipurRajasthanIndia
| | - Ravi Bhushan
- Department of ZoologyM.S. CollegeMotihariBiharIndia
| | - Caiming Xu
- Department of General SurgeryThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research InstituteCity of HopeMonroviaCaliforniaUSA
| | - Bhana Ram Gadi
- Stress Physiology and Molecular Biology LaboratoryDepartment of BotanyJai Narain Vyas UniversityJodhpurRajasthanIndia
| | | | - Vikas Yadav
- School of Life SciencesJawaharlal Nehru UniversityNew DelhiIndia
| | - Jarek Maciaczyk
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
| | - Ingo G. H. Schmidt‐Wolf
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| | - Abhishek Kumar
- Manipal Academy of Higher EducationManipalKarnatakaIndia
- Institute of BioinformaticsInternational Technology ParkBangaloreIndia
| | - Amit Sharma
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
9
|
Acharya SK, Shai S, Choon YF, Gunardi I, Hartanto FK, Kadir K, Roychoudhury A, Amtha R, Vincent-Chong VK. Cancer Stem Cells in Oral Squamous Cell Carcinoma: A Narrative Review on Experimental Characteristics and Methodological Challenges. Biomedicines 2024; 12:2111. [PMID: 39335624 PMCID: PMC11429394 DOI: 10.3390/biomedicines12092111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Cancer stem cells (CSCs) represent a subpopulation of cancer cells that are believed to initiate and drive cancer progression. In animal models, xenotransplanted CSCs have demonstrated the ability to produce tumors. Since their initial isolation in blood cancers, CSCs have been identified in various solid human cancers, including oral squamous cell carcinoma (OSCC). In addition to their tumorigenic properties, dysregulated stem-cell-related signaling pathways-Wnt family member (Wnt), neurogenic locus notch homolog protein (Notch), and hedgehog-have been shown to endow CSCs with characteristics like self-renewal, phenotypic plasticity, and chemoresistance, contributing to recurrence and treatment failure. Consequently, CSCs have become targets for new therapeutic agents, with some currently in different phases of clinical trials. Notably, small molecule inhibitors of the hedgehog signaling pathway, such as vismodegib and glasdegib, have been approved for the treatment of basal cell carcinoma and acute myeloid leukemia, respectively. Other strategies for eradicating CSCs include natural compounds, nano-drug delivery systems, targeting mitochondria and the CSC microenvironment, autophagy, hyperthermia, and immunotherapy. Despite the extensive documentation of CSCs in OSCC since its first demonstration in head and neck (HN) SCC in 2007, none of these novel pharmacological approaches have yet entered clinical trials for OSCC patients. This narrative review summarizes the in vivo and in vitro evidence of CSCs and CSC-related signaling pathways in OSCC, highlighting their role in promoting chemoresistance and immunotherapy resistance. Additionally, it addresses methodological challenges and discusses future research directions to improve experimental systems and advance CSC studies.
Collapse
Affiliation(s)
- Surendra Kumar Acharya
- Department of Oral Medicine, Radiology and Surgery, Faculty of Dentistry, Lincoln University College, Petaling Jaya 47301, Selangor, Malaysia
| | - Saptarsi Shai
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Yee Fan Choon
- Department of Oral and Maxillofacial Surgical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom 42610, Selangor, Malaysia;
| | - Indrayadi Gunardi
- Oral Medicine Department, Faculty of Dentistry, Universitas Trisakti, Jakarta 11440, Indonesia; (I.G.); (F.K.H.)
| | - Firstine Kelsi Hartanto
- Oral Medicine Department, Faculty of Dentistry, Universitas Trisakti, Jakarta 11440, Indonesia; (I.G.); (F.K.H.)
| | - Kathreena Kadir
- Department of Oral and Maxillofacial Clinical Sciences, Faculty of Dentistry, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Ajoy Roychoudhury
- Department of Oral and Maxillofacial Surgery, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Rahmi Amtha
- Oral Medicine Department, Faculty of Dentistry, Universitas Trisakti, Jakarta 11440, Indonesia; (I.G.); (F.K.H.)
| | - Vui King Vincent-Chong
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
10
|
Díaz MA, Fusco M, Benítez CA, Gayet F, García L, Victoria L, Jaramillo S, Bayo J, Zubieta MR, Rizzo MM, Piccioni F, Malvicini M. Targeting hyaluronan metabolism-related molecules associated with resistant tumor-initiating cells potentiates chemotherapy efficacy in lung cancer. Sci Rep 2024; 14:16803. [PMID: 39039104 PMCID: PMC11263553 DOI: 10.1038/s41598-024-66914-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/05/2024] [Indexed: 07/24/2024] Open
Abstract
The success of chemotherapy regimens in patients with non-small cell lung cancer (NSCLC) could be restricted at least in part by cancer stem cells (CSC) niches within the tumor microenvironment (TME). CSC express CD133, CD44, CD47, and SOX2, among other markers and factors. Analysis of public data revealed that high expression of hyaluronan (HA), the main glycosaminoglycan of TME, correlated positively with CSC phenotype and decreased disease-free interval in NSCLC patients. We aimed to cross-validate these findings on human and murine lung cancer cells and observed that CD133 + CSC differentially expressed higher levels of HA, HAS3, ABCC5, SOX2, and CD47 (p < 0.01). We modulated HA expression with 4-methylumbelliferone (4Mu) and detected an increase in sensitivity to paclitaxel (Pa). We evaluated the effect of 4Mu + chemotherapy on survival, HA metabolism, and CSC profile. The combination of 4Mu with Pa reduced the clonogenic and tumor-forming ability of CSC. Pa-induced HAS3, ABCC5, SOX2, and CD47 expression was mitigated by 4Mu. Pa + 4Mu combination significantly reduced in vivo tumor growth, enhancing animal survival and restoring the CSC profile in the TME to basal levels. Our results suggest that HA is involved in lung CSC phenotype and chemosensitivity, and its modulation by 4Mu improves treatment efficacy to inhibit tumor progression.
Collapse
Affiliation(s)
- Marco Aurelio Díaz
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar, Argentina
| | - Mariel Fusco
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar, Argentina
| | - Constanza Arriola Benítez
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar, Argentina
| | - Fernando Gayet
- Servicio de Oncología, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Ludmila García
- Laboratorio Central, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Lucia Victoria
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar, Argentina
| | - Sebastián Jaramillo
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar, Argentina
| | - Juan Bayo
- Programa de Hepatología Experimental y Terapia Génica, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Tecnicas, Pilar, Argentina
| | | | - Manglio M Rizzo
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar, Argentina
- Servicio de Oncología, Hospital Universitario Austral, Buenos Aires, Argentina
| | - Flavia Piccioni
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar, Argentina.
| | - Mariana Malvicini
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional, Universidad Austral-Consejo Nacional de Investigaciones Científicas y Técnicas, Pilar, Argentina.
| |
Collapse
|
11
|
Liang KY, Su NY, Yang HP, Hsieh PL, Fang CY, Tsai LL, Liao YW, Liu CM, Yu CC. Gastric adenocarcinoma predictive long intergenic noncoding RNA (GAPLINC) promotes oral cancer stemness by acting as a molecular sponge of miR331-3p. J Dent Sci 2024; 19:1389-1395. [PMID: 39035323 PMCID: PMC11259681 DOI: 10.1016/j.jds.2024.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/29/2024] [Indexed: 07/23/2024] Open
Abstract
Background/purpose Accumulating evidence has suggested that treatment failure of cancer therapy can be attributed to cancer stem cells (CSCs). Among numerous regulators of cancer stemness, non-coding RNAs (ncRNAs) have gained significant attention recently. In this study, we examined the role of gastric adenocarcinoma predictive long intergenic noncoding RNA (GAPLINC) in oral CSCs (OCSCs). Materials and methods RNA Sequencing and quantitative real-time polymerase chain reaction (qRT-PCR) were used to determine the expression of GAPLINC. Flow cytometry and sphere-forming assay were exploited to isolate OCSCs. Measurement of aldehyde dehydrogenase 1 (ALDH1) activity, CD44 expressing cells, and various phenotypic assays, such as self-renewal, migration, invasion, and colony-forming abilities, were conducted in CSCs of two types of oral cancer cells (SAS and GNM) following the knockdown of GAPLINC. A luciferase reporter was also carried out to validate the direct interaction between GAPLINC and microRNA (miR)-331-3p. Results Our results showed that GAPLINC was overexpressed in OCSCs from patient-derived and oral cancer cell lines. We demonstrated that silencing of GAPLINC in OCSCs downregulated various CSC hallmarks, such as ALDH1 activity, percentage of CD44-expressing cells, self-renewal capacity, and colony-forming ability. Moreover, our results revealed that the effect of GAPLINC on cancer stemness was mediated by direct repression of miR-331-3p. Conclusion These data have potential clinical implications in that we unraveled the aberrant upregulation of GAPLINC and demonstrated that suppression of GAPLINC may reduce cancer stemness via sequestering miR-331-3p.
Collapse
Affiliation(s)
- Kuang-Yuan Liang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Kaohsiung Armed Forces General Hospital Gangshan Branch, Kaohsiung, Taiwan
| | - Ni-Yu Su
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsiu-Pin Yang
- Department of Dentistry, Kaohsiung Armed Forces General Hospital Gangshan Branch, Kaohsiung, Taiwan
| | - Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Yuan Fang
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lo-Lin Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Research and Education, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan, Taiwan
| | - Yi-Wen Liao
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Ming Liu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
12
|
Wu S, Tan Y, Li F, Han Y, Zhang S, Lin X. CD44: a cancer stem cell marker and therapeutic target in leukemia treatment. Front Immunol 2024; 15:1354992. [PMID: 38736891 PMCID: PMC11082360 DOI: 10.3389/fimmu.2024.1354992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
CD44 is a ubiquitous leukocyte adhesion molecule involved in cell-cell interaction, cell adhesion, migration, homing and differentiation. CD44 can mediate the interaction between leukemic stem cells and the surrounding extracellular matrix, thereby inducing a cascade of signaling pathways to regulate their various behaviors. In this review, we focus on the impact of CD44s/CD44v as biomarkers in leukemia development and discuss the current research and prospects for CD44-related interventions in clinical application.
Collapse
Affiliation(s)
- Shuang Wu
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yicheng Tan
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
| | - Fanfan Li
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yixiang Han
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shenghui Zhang
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaofei Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
13
|
Ziranu P, Pretta A, Aimola V, Cau F, Mariani S, D’Agata AP, Codipietro C, Rizzo D, Dell’Utri V, Sanna G, Moledda G, Cadoni A, Lai E, Puzzoni M, Pusceddu V, Castagnola M, Scartozzi M, Faa G. CD44: A New Prognostic Marker in Colorectal Cancer? Cancers (Basel) 2024; 16:1569. [PMID: 38672650 PMCID: PMC11048923 DOI: 10.3390/cancers16081569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/19/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Cluster of differentiation 44 (CD44) is a non-kinase cell surface glycoprotein. It is overexpressed in several cell types, including cancer stem cells (CSCs). Cells overexpressing CD44 exhibit several CSC traits, such as self-renewal, epithelial-mesenchymal transition (EMT) capability, and resistance to chemo- and radiotherapy. The role of CD44 in maintaining stemness and the CSC function in tumor progression is accomplished by binding to its main ligand, hyaluronan (HA). The HA-CD44 complex activates several signaling pathways that lead to cell proliferation, adhesion, migration, and invasion. The CD44 gene regularly undergoes alternative splicing, resulting in the standard (CD44s) and variant (CD44v) isoforms. The different functional roles of CD44s and specific CD44v isoforms still need to be fully understood. The clinicopathological impact of CD44 and its isoforms in promoting tumorigenesis suggests that CD44 could be a molecular target for cancer therapy. Furthermore, the recent association observed between CD44 and KRAS-dependent carcinomas and the potential correlations between CD44 and tumor mutational burden (TMB) and microsatellite instability (MSI) open new research scenarios for developing new strategies in cancer treatment. This review summarises current research regarding the different CD44 isoform structures, their roles, and functions in supporting tumorigenesis and discusses its therapeutic implications.
Collapse
Affiliation(s)
- Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Valentina Aimola
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (V.A.); (F.C.)
| | - Flaviana Cau
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (V.A.); (F.C.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Alessandra Pia D’Agata
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Claudia Codipietro
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Daiana Rizzo
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Veronica Dell’Utri
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Giorgia Sanna
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Giusy Moledda
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Andrea Cadoni
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Massimo Castagnola
- Proteomics Laboratory, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, 00013 Rome, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Gavino Faa
- Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy;
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
14
|
Han Y, Ki CS. Effect of Matrix Stiffness and Hepatocyte Growth Factor on Small Cell Lung Cancer Cells in Decellularized Extracellular Matrix-Based Hydrogels. Macromol Biosci 2024; 24:e2300356. [PMID: 37877161 DOI: 10.1002/mabi.202300356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/13/2023] [Indexed: 10/26/2023]
Abstract
Small cell lung cancer (SCLC) is one of lethal cancers resulting in very low 5-year-survival rate. Although its clinical treatment largely relies on chemotherapy, SCLC cell physiology in three-dimenstional (3D) matrix has been less explored. In this work, the tumor microenvironment is reconstructed with decellularized porcine pulmonary extracellular matrix (dECM) with hyaluronic acid. To modulate matrix stiffness, the methacrylate groups are introduced into both dECM and hyaluronic acid, followed by photocrosslinking with photoinitiator. The stiffness of the resulting dECM-based hydrogel covers the stiffness of normal or cancerous tissue with varying dECM content. The proliferation and cancer stem cell marker expression of encapsulated SCLC cells are promoted in a compliant hydrogel matrix, which has a low shear modulus similar to that of the normal tissue. The hepatocyte growth factor (HGF) that induces SCLC cell invasion and chemoresistance markedly increases invasiveness and gene expression levels of CD44 and Sox2 in the hydrogel matrix. In addition, HGF treatment causes higher resistance against anticancer drugs (cisplatin and paclitaxel) in the 3D microenvironment. These findings indicate that malignant SCLC can be recapitulated in a pulmonary dECM-based matrix.
Collapse
Affiliation(s)
- Yoobin Han
- Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul, 08826, Republic of Korea
| | - Chang Seok Ki
- Department of Agriculture, Forestry and Bioresources, Seoul National University, Seoul, 08826, Republic of Korea
- Research Institute of Agriculture and Life Science, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
15
|
Luo X, Wang J, Chen Y, Zhou X, Shao Z, Liu K, Shang Z. Melatonin inhibits the stemness of head and neck squamous cell carcinoma by modulating HA synthesis via the FOSL1/HAS3 axis. J Pineal Res 2024; 76:e12940. [PMID: 38402581 DOI: 10.1111/jpi.12940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/24/2023] [Accepted: 01/04/2024] [Indexed: 02/27/2024]
Abstract
Hyaluronic acid (HA) is a glycosaminoglycan and the main component of the extracellular matrix (ECM), which has been reported to interact with its receptor CD44 to play critical roles in the self-renewal and maintenance of cancer stem cells (CSCs) of multiple malignancies. Melatonin is a neuroendocrine hormone with pleiotropic antitumor properties. However, whether melatonin could regulate HA accumulation in the ECM to modulate the stemness of head and neck squamous cell carcinoma (HNSCC) remains unknown. In this study, we found that melatonin suppressed CSC-related markers, such as CD44, of HNSCC cells and decreased the tumor-initiating frequency of CSCs in vivo. In addition, melatonin modulated HA synthesis of HNSCC cells by downregulating the expression of hyaluronan synthase 3 (HAS3). Further study showed that the Fos-like 1 (FOSL1)/HAS3 axis mediated the inhibitory effects of melatonin on HA accumulation and stemness of HNSCC in a receptor-independent manner. Taken together, melatonin modulated HA synthesis through the FOSL1/HAS3 axis to inhibit the stemness of HNSCC cells, which elucidates the effect of melatonin on the ECM and provides a novel perspective on melatonin in HNSCC treatment.
Collapse
Affiliation(s)
- Xinyue Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jingjing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiaocheng Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhe Shao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ke Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial-Head and Neck oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Todoroki K, Abe Y, Matsuo K, Nomura H, Kawahara A, Nakamura Y, Nakamura M, Seki N, Kusukawa J. Prognostic effect of programmed cell death ligand 1/programmed cell death 1 expression in cancer stem cells of human oral squamous cell carcinoma. Oncol Lett 2024; 27:79. [PMID: 38249811 PMCID: PMC10797318 DOI: 10.3892/ol.2024.14213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
The relationship between cancer stem cells (CSCs) in oral squamous cell carcinoma (OSCC) and programmed cell death ligand 1 (PD-L1)/programmed cell death 1 (PD-1) remains unclear. Therefore, the present study aimed to clarify the association between the CD44v3high/CD24low immunophenotype of CSCs in OSCC and PD-L1/PD-1 co-expression, and to assess the prognostic effect of CSCs in terms of immune checkpoint molecules. Formalin-fixed, paraffin-embedded tissue samples and clinicopathological data from 168 patients with OSCC were retrospectively retrieved. Immunohistochemical staining and reverse transcription quantitative polymerase chain reaction were applied to a tissue microarray of the invasive front of each case. Semi-automated cell counting was used to assess CD44v3, CD24, PD-L1 and PD-1 expression by immunohistochemistry (IHC) using a digital image analysis program. Associations between immunological markers and clinicopathological variables were estimated. Patients with the CSC immunophenotype CD44v3high/CD24low, and patients with a high PD-L1/PD-1-positive cell density in the tumor parenchyma and stroma had significantly lower survival rates. Furthermore, patients with the CSC immunophenotype (CD44v3high/CD24low) and high PD-L1/PD-1 co-expression had even lower survival rates (P<0.01, log-rank test). Notably, there was a positive correlation between CD44v3 and PD-L1 expression (τ=0.1096, P=0.0366, Kendall rank correlation coefficient) and a negative correlation between CD24 and PD-1 expression (τ=-0.1387, P=0.0089, Kendall rank correlation coefficient). Additionally, the high CD44v3 expression group, as determined by IHC, exhibited significantly decreased expression of U2 small nuclear RNA auxiliary factor 1 (U2AF1) at the mRNA level compared with that in the low CD44v3 expression group (P<0.001, Mann-Whitney U test), and U2AF1 and PD-L1 mRNA expression exhibited a significant negative correlation (τ=-0.3948, P<0.001, Kendall rank correlation coefficient). In conclusion, CSCs in OSCC may evade host immune mechanisms and maintain CSC stemness via PD-L1/PD-1 co-expression, resulting in unfavorable clinical outcomes.
Collapse
Affiliation(s)
- Keita Todoroki
- Dental and Oral Medical Center, Kurume University, School of Medicine, Kurume, Fukuoka 830-0011, Japan
- Department of Dental and Oral Surgery, Takagi Hospital, Kouhoukai Medical Corporation, Okawa, Fukuoka 831-0016, Japan
| | - Yushi Abe
- Dental and Oral Medical Center, Kurume University, School of Medicine, Kurume, Fukuoka 830-0011, Japan
- Department of Dental and Oral Surgery, Takagi Hospital, Kouhoukai Medical Corporation, Okawa, Fukuoka 831-0016, Japan
| | - Katsuhisa Matsuo
- Dental and Oral Medical Center, Kurume University, School of Medicine, Kurume, Fukuoka 830-0011, Japan
- Department of Dental and Oral Surgery, Takagi Hospital, Kouhoukai Medical Corporation, Okawa, Fukuoka 831-0016, Japan
| | - Hidetoshi Nomura
- Dental and Oral Medical Center, Kurume University, School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Yoshiaki Nakamura
- Department of Dentistry and Oral Surgery, Oita Saiseikai Hita Hospital, Hita, Oita 877-1292, Japan
| | - Moriyoshi Nakamura
- Dental and Oral Medical Center, Kurume University, School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Naoko Seki
- Dental and Oral Medical Center, Kurume University, School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Jingo Kusukawa
- Dental and Oral Medical Center, Kurume University, School of Medicine, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
17
|
Zhang S, Yang R, Ouyang Y, Shen Y, Hu L, Xu C. Cancer stem cells: a target for overcoming therapeutic resistance and relapse. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0333. [PMID: 38164743 PMCID: PMC10845928 DOI: 10.20892/j.issn.2095-3941.2023.0333] [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: 09/04/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Cancer stem cells (CSCs) are a small subset of cells in cancers that are thought to initiate tumorous transformation and promote metastasis, recurrence, and resistance to treatment. Growing evidence has revealed the existence of CSCs in various types of cancers and suggested that CSCs differentiate into diverse lineage cells that contribute to tumor progression. We may be able to overcome the limitations of cancer treatment with a comprehensive understanding of the biological features and mechanisms underlying therapeutic resistance in CSCs. This review provides an overview of the properties, biomarkers, and mechanisms of resistance shown by CSCs. Recent findings on metabolic features, especially fatty acid metabolism and ferroptosis in CSCs, are highlighted, along with promising targeting strategies. Targeting CSCs is a potential treatment plan to conquer cancer and prevent resistance and relapse in cancer treatment.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610042, China
| | - Rui Yang
- Department of Ultrasound in Medicine, Chengdu Wenjiang District People’s Hospital, Chengdu 611130, China
| | - Yujie Ouyang
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yang Shen
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- School of Pharmacy, Macau University of Science and Technology, Macau SAR 999078, China
| | - Lanlin Hu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Yu-Yue Pathology Scientific Research Center, Chongqing 400039, China
- Jinfeng Laboratory, Chongqing 401329, China
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Yu-Yue Pathology Scientific Research Center, Chongqing 400039, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
18
|
Dorna D, Paluszczak J. Targeting cancer stem cells as a strategy for reducing chemotherapy resistance in head and neck cancers. J Cancer Res Clin Oncol 2023; 149:13417-13435. [PMID: 37453969 PMCID: PMC10587253 DOI: 10.1007/s00432-023-05136-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE Resistance to chemotherapy and radiotherapy is the primary cause of a poor prognosis in oncological patients. Researchers identified many possible mechanisms involved in gaining a therapy-resistant phenotype by cancer cells, including alterations in intracellular drug accumulation, detoxification, and enhanced DNA damage repair. All these features are characteristic of stem cells, making them the major culprit of chemoresistance. This paper reviews the most recent evidence regarding the association between the stemness phenotype and chemoresistance in head and neck cancers. It also investigates the impact of pharmacologically targeting cancer stem cell populations in this subset of malignancies. METHODS This narrative review was prepared based on the search of the PubMed database for relevant papers. RESULTS Head and neck cancer cells belonging to the stem cell population are distinguished by the high expression of certain surface proteins (e.g., CD10, CD44, CD133), pluripotency-related transcription factors (SOX2, OCT4, NANOG), and increased activity of aldehyde dehydrogenase (ALDH). Chemotherapy itself increases the percentage of stem-like cells. Importantly, the intratumor heterogeneity of stem cell subpopulations reflects cell plasticity which has great importance for chemoresistance induction. CONCLUSIONS Evidence points to the advantage of combining classical chemotherapeutics with stemness modulators thanks to the joint targeting of the bulk of proliferating tumor cells and chemoresistant cancer stem cells, which could cause recurrence.
Collapse
Affiliation(s)
- Dawid Dorna
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Ul. Święcickiego 4, 60-781 Poznan, Poland
| | - Jarosław Paluszczak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Ul. Święcickiego 4, 60-781 Poznan, Poland
| |
Collapse
|
19
|
Kalli M, Poskus MD, Stylianopoulos T, Zervantonakis IK. Beyond matrix stiffness: targeting force-induced cancer drug resistance. Trends Cancer 2023; 9:937-954. [PMID: 37558577 PMCID: PMC10592424 DOI: 10.1016/j.trecan.2023.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/27/2023] [Accepted: 07/13/2023] [Indexed: 08/11/2023]
Abstract
During tumor progression, mechanical abnormalities in the tumor microenvironment (TME) trigger signaling pathways in cells that activate cellular programs, resulting in tumor growth and drug resistance. In this review, we describe mechanisms of action for anti-cancer therapies and mechanotransduction programs that regulate cellular processes, including cell proliferation, apoptosis, survival and phenotype switching. We discuss how the therapeutic response is impacted by the three main mechanical TME abnormalities: high extracellular matrix (ECM) composition and stiffness; interstitial fluid pressure (IFP); and elevated mechanical forces. We also review drugs that normalize these abnormalities or block mechanosensors and mechanotransduction pathways. Finally, we discuss current challenges and perspectives for the development of new strategies targeting mechanically induced drug resistance in the clinic.
Collapse
Affiliation(s)
- Maria Kalli
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Matthew D Poskus
- Department of Bioengineering and Hillman Cancer Center, University of Pittsburgh, PA, USA
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | | |
Collapse
|
20
|
Kummer S, Klang A, Strohmayer C, Walter I, Jindra C, Kneissl S, Brandt S. Feline SCCs of the Head and Neck Display Partial Epithelial-Mesenchymal Transition and Harbor Stem Cell-like Cancer Cells. Pathogens 2023; 12:1288. [PMID: 38003753 PMCID: PMC10674711 DOI: 10.3390/pathogens12111288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Squamous cell carcinoma of the head and neck (HNSCC) is a malignant cancer disease in humans and animals. There is ample evidence that the high plasticity of cancer cells, i.e., their ability to switch from an epithelial to a mesenchymal, endothelial, and stem cell-like phenotype, chiefly contributes to progression, metastasis, and multidrug resistance of human HNSCCs. In feline HNSCC, the field of cancer cell plasticity is still unexplored. In this study, fourteen feline HNSCCs with a known feline papillomavirus (FPV) infection status were subjected to histopathological grading and subsequent screening for expression of epithelial, mesenchymal, and stem cell markers by immunohistochemistry (IHC) and immunofluorescence staining (IF). Irrespective of the FPV infection status, all tumors except one corresponded to high-grade, invasive lesions and concurrently expressed epithelial (keratins, E-cadherin, β-catenin) and mesenchymal (vimentin, N-cadherin, CD146) proteins. This finding is indicative for partial epithelial-mesenchymal transition (pEMT) events in the lesions, as similarly described for human HNSCCs. IF double staining revealed the presence of CD44/CD271 double-positive cells notably within the tumors' invasive fronts that likely correspond to cancer stem cells. Taken together, the obtained findings suggest that feline HNSCCs closely resemble their human counterparts with respect to tumor cell plasticity.
Collapse
Affiliation(s)
- Stefan Kummer
- VetCore Facility for Research, University of Veterinary Medicine, 1210 Vienna, Austria; (S.K.); (I.W.)
| | - Andrea Klang
- Institute of Pathology, Department of Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Carina Strohmayer
- Clinical Unit of Diagnostic Imaging, Department for Companion Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria; (C.S.); (S.K.)
| | - Ingrid Walter
- VetCore Facility for Research, University of Veterinary Medicine, 1210 Vienna, Austria; (S.K.); (I.W.)
- Institute of Morphology, Department of Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Christoph Jindra
- Research Group Oncology (RGO), Clinical Unit of Equine Surgery, Department for Companion Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria;
- Division of Molecular Oncology and Hematology, Karl Landsteiner University of Health Sciences, 3500 Krems an der Donau, Austria
| | - Sibylle Kneissl
- Clinical Unit of Diagnostic Imaging, Department for Companion Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria; (C.S.); (S.K.)
| | - Sabine Brandt
- Research Group Oncology (RGO), Clinical Unit of Equine Surgery, Department for Companion Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria;
| |
Collapse
|
21
|
Skandalis SS. CD44 Intracellular Domain: A Long Tale of a Short Tail. Cancers (Basel) 2023; 15:5041. [PMID: 37894408 PMCID: PMC10605500 DOI: 10.3390/cancers15205041] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
CD44 is a single-chain transmembrane receptor that exists in multiple forms due to alternative mRNA splicing and post-translational modifications. CD44 is the main cell surface receptor of hyaluronan as well as other extracellular matrix molecules, cytokines, and growth factors that play important roles in physiological processes (such as hematopoiesis and lymphocyte homing) and the progression of various diseases, the predominant one being cancer. Currently, CD44 is an established cancer stem cell marker in several tumors, implying a central functional role in tumor biology. The present review aims to highlight the contribution of the CD44 short cytoplasmic tail, which is devoid of any enzymatic activity, in the extraordinary functional diversity of the receptor. The interactions of CD44 with cytoskeletal proteins through specific structural motifs within its intracellular domain drives cytoskeleton rearrangements and affects the distribution of organelles and transport of molecules. Moreover, the CD44 intracellular domain specifically interacts with various cytoplasmic effectors regulating cell-trafficking machinery, signal transduction pathways, the transcriptome, and vital cell metabolic pathways. Understanding the cell type- and context-specificity of these interactions may unravel the high complexity of CD44 functions and lead to novel improved therapeutic interventions.
Collapse
Affiliation(s)
- Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
22
|
Inoue A, Ohnishi T, Nishikawa M, Ohtsuka Y, Kusakabe K, Yano H, Tanaka J, Kunieda T. A Narrative Review on CD44's Role in Glioblastoma Invasion, Proliferation, and Tumor Recurrence. Cancers (Basel) 2023; 15:4898. [PMID: 37835592 PMCID: PMC10572085 DOI: 10.3390/cancers15194898] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
High invasiveness is a characteristic of glioblastoma (GBM), making radical resection almost impossible, and thus, resulting in a tumor with inevitable recurrence. GBM recurrence may be caused by glioma stem-like cells (GSCs) that survive many kinds of therapy. GSCs with high expression levels of CD44 are highly invasive and resistant to radio-chemotherapy. CD44 is a multifunctional molecule that promotes the invasion and proliferation of tumor cells via various signaling pathways. Among these, paired pathways reciprocally activate invasion and proliferation under different hypoxic conditions. Severe hypoxia (0.5-2.5% O2) upregulates hypoxia-inducible factor (HIF)-1α, which then activates target genes, including CD44, TGF-β, and cMET, all of which are related to tumor migration and invasion. In contrast, moderate hypoxia (2.5-5% O2) upregulates HIF-2α, which activates target genes, such as vascular endothelial growth factor (VEGF)/VEGFR2, cMYC, and cyclin D1. All these genes are related to tumor proliferation. Oxygen environments around GBM can change before and after tumor resection. Before resection, the oxygen concentration at the tumor periphery is severely hypoxic. In the reparative stage after resection, the resection cavity shows moderate hypoxia. These observations suggest that upregulated CD44 under severe hypoxia may promote the migration and invasion of tumor cells. Conversely, when tumor resection leads to moderate hypoxia, upregulated HIF-2α activates HIF-2α target genes. The phenotypic transition regulated by CD44, leading to a dichotomy between invasion and proliferation according to hypoxic conditions, may play a crucial role in GBM recurrence.
Collapse
Affiliation(s)
- Akihiro Inoue
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| | - Takanori Ohnishi
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
- Department of Neurosurgery, Advanced Brain Disease Center, Washoukai Sadamoto Hospital, 1-6-1 Takehara, Matsuyama 790-0052, Ehime, Japan
| | - Masahiro Nishikawa
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| | - Yoshihiro Ohtsuka
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| | - Kosuke Kusakabe
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicene, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (H.Y.); (J.T.)
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicene, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (H.Y.); (J.T.)
| | - Takeharu Kunieda
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| |
Collapse
|
23
|
Bhattacharyya M, Jariyal H, Srivastava A. Hyaluronic acid: More than a carrier, having an overpowering extracellular and intracellular impact on cancer. Carbohydr Polym 2023; 317:121081. [PMID: 37364954 DOI: 10.1016/j.carbpol.2023.121081] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/28/2023]
Abstract
Hyaluronic acid (HA), also named hyaluronan, is an omnipresent component of the tissue microenvironment. It is extensively used to formulate targeted drug delivery systems for cancer. Although HA itself has pivotal influences in various cancers, its calibers are somewhat neglected when using it as delivering platform to treat cancer. In the last decade, multiple studies revealed roles of HA in cancer cell proliferation, invasion, apoptosis, and dormancy through pathways like mitogen-activated protein kinase-extracellular signal-regulated kinase (MAPK/ERK), P38, and nuclear factor kappa-light chain-enhancer of activated B cells (NFκB). A more fascinating fact is that the distinct molecular weight (MW) of HA exerts disparate effects on the same type of cancer. Its overwhelming use in cancer therapy and other therapeutic products make collective research on the sundry impact of it on various types of cancer, an essential aspect to be considered in all of these domains. Even the development of new therapies against cancer needed meticulous studies on HA because of its divergence of activity based on MW. This review will provide painstaking insight into the extracellular and intracellular bioactivity of HA, its modified forms, and its MW in cancers, which may improve the management of cancer.
Collapse
Affiliation(s)
- Medha Bhattacharyya
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat, India
| | - Heena Jariyal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat, India
| | - Akshay Srivastava
- Department of Medical Device, National Institute of Pharmaceutical Education and Research-Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
24
|
Dai Y, Wu Z, Chen Y, Ye X, Wang C, Zhu H. OCT4's role and mechanism underlying oral squamous cell carcinoma. J Zhejiang Univ Sci B 2023; 24:796-806. [PMID: 37701956 PMCID: PMC10500100 DOI: 10.1631/jzus.b2200602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/06/2023] [Indexed: 09/14/2023]
Abstract
Oral squamous cell carcinoma (OSCC), a common malignancy of the head and neck, ranks sixth worldwide in terms of cancers with the most negative impact, owing to tumor relapse rates, cervical lymphnode metastasis, and the lack of an efficacious systemic therapy. Its prognosis is poor, and its mortality rate is high. Octamer-binding transcription factor 4 (OCT4) is a member of the Pit-Oct-Unc (POU) family and is a key reprogramming factor that produces a marked effect in preserving the pluripotency and self-renewal state of embryonic stem cells (ESCs). According to recent studies, OCT4 participates in retaining the survival of OSCC cancer stem cells (CSCs), which has far-reaching implications for the occurrence, recurrence, metastasis, and prognosis of oral carcinogenesis. Therefore, we summarize the structure, subtypes, and function of OCT4 as well as its role in the occurrence, progression, and prognosis of OSCC.
Collapse
Affiliation(s)
- Yuwei Dai
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ziqiong Wu
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yitong Chen
- School of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xinjian Ye
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Disease of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Chaowei Wang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huiyong Zhu
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
25
|
Tolue Ghasaban F, Maharati A, Zangouei AS, Zangooie A, Moghbeli M. MicroRNAs as the pivotal regulators of cisplatin resistance in head and neck cancers. Cancer Cell Int 2023; 23:170. [PMID: 37587481 PMCID: PMC10428558 DOI: 10.1186/s12935-023-03010-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/28/2023] [Indexed: 08/18/2023] Open
Abstract
Although, there is a high rate of good prognosis in early stage head and neck tumors, about half of these tumors are detected in advanced stages with poor prognosis. A combination of chemotherapy, radiotherapy, and surgery is the treatment option in head and neck cancer (HNC) patients. Although, cisplatin (CDDP) as the first-line drug has a significant role in the treatment of HNC patients, CDDP resistance can be observed in a large number of these patients. Therefore, identification of the molecular mechanisms involved in CDDP resistance can help to reduce the side effects and also provides a better therapeutic management. MicroRNAs (miRNAs) as the post-transcriptional regulators play an important role in drug resistance. Therefore, in the present review we investigated the role of miRNAs in CDDP response of head and neck tumors. It has been reported that the miRNAs exerted their roles in CDDP response by regulation of signaling pathways such as WNT, NOTCH, PI3K/AKT, TGF-β, and NF-kB as well as apoptosis, autophagy, and EMT process. The present review paves the way to suggest a non-invasive miRNA based panel marker for the prediction of CDDP response among HNC patients. Therefore, such diagnostic miRNA based panel marker reduces the CDDP side effects and improves the clinical outcomes of these patients following an efficient therapeutic management.
Collapse
Affiliation(s)
- Faezeh Tolue Ghasaban
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Zangooie
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Student research committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
26
|
Fu Y, Zha J, Wu Q, Tang Y, Wang W, Zhou Q, Jiang L. Stromal micropapillary pattern and CD44s expression predict worse outcome in lung adenocarcinomas with micropapillary pattern. Pathol Res Pract 2023; 248:154595. [PMID: 37343380 DOI: 10.1016/j.prp.2023.154595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/09/2023] [Accepted: 06/05/2023] [Indexed: 06/23/2023]
Abstract
OBJECTIVES This study aims to investigate the clinicopathologic characteristics of lung adenocarcinoma with micropapillary pattern (MPP) and the expression of CD44s and CD44v6 in MPP. METHODS A total of 202 patients diagnosed with primary lung adenocarcinoma with MPP were included. We estimated the proportion of MPP in each tumor tissue and divided MPP into aerogenous micropapillary pattern (AMP) and stromal micropapillary pattern (SMP). The expression of CD44s and CD44v6 was estimated by immunohistochemical staining. Clinicopathologic data were collected from the patients' medical records. We also collected patients' follow-up data and used PFS (progression-free survival) as a survival indicator. RESULTS Lung adenocarcinoma with MPP had a high risk of pleural invasion, lymph node metastasis, in advanced TNM stage, and a high rate of EGFR mutation. The presence of SMP indicated a higher rate of pleural invasion, lymphovascular invasion, lymph node metastasis, and a worse PFS compared with pure AMP. We found high expression of CD44s in micropapillary, especially in AMP, while the absence of CD44s expression indicated shorter survival, which was an independent unfavorable factor for PFS. CONCLUSIONS Lung adenocarcinoma with micropapillary pattern indicated an unfavorable prognosis, which had two different pattens, AMP and SMP. SMP indicated a worse survival than AMP, and was an independent unfavorable factor for PFS. So, AMP/SMP subclassification is necessary to evaluate patient's prognosis. Furthermore, the absent expression of CD44s in micropapillary indicated shorter survival, especially in patients with EGFR mutation. Herein, CD44s may be a biological marker for micropapillary lung adenocarcinoma.
Collapse
Affiliation(s)
- Yiyun Fu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Junmei Zha
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Wu
- Department of Pathology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuan Tang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Weiya Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Zhou
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
27
|
Bei Y, He J, Dong X, Wang Y, Wang S, Guo W, Cai C, Xu Z, Wei J, Liu B, Zhang N, Shen P. Targeting CD44 Variant 5 with an Antibody-Drug Conjugate Is an Effective Therapeutic Strategy for Intrahepatic Cholangiocarcinoma. Cancer Res 2023; 83:2405-2420. [PMID: 37205633 PMCID: PMC10345965 DOI: 10.1158/0008-5472.can-23-0510] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/06/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023]
Abstract
UNLABELLED Intrahepatic cholangiocarcinoma (ICC) is the second most frequent type of primary liver cancer. ICC is among the deadliest malignancies, highlighting that novel treatments are urgently needed. Studies have shown that CD44 variant isoforms, rather than the CD44 standard isoform, are selectively expressed in ICC cells, providing an opportunity for the development of an antibody-drug conjugate (ADC)-based targeted therapeutic strategy. In this study, we observed the specific expression of CD44 variant 5 (CD44v5) in ICC tumors. CD44v5 protein was expressed on the surface of most ICC tumors (103 of 155). A CD44v5-targeted ADC, H1D8-DC (H1D8-drug conjugate), was developed that comprises a humanized anti-CD44v5 mAb conjugated to the microtubule inhibitor monomethyl auristatin E (MMAE) via a cleavable valine-citrulline-based linker. H1D8-DC exhibited efficient antigen binding and internalization in cells expressing CD44v5 on the cell surface. Because of the high expression of cathepsin B in ICC cells, the drug was preferentially released in cancer cells but not in normal cells, thus inducing potent cytotoxicity at picomolar concentrations. In vivo studies showed that H1D8-DC was effective against CD44v5-positive ICC cells and induced tumor regression in patient-derived xenograft models, whereas no significant adverse toxicities were observed. These data demonstrate that CD44v5 is a bona fide target in ICC and provide a rationale for the clinical investigation of a CD44v5-targeted ADC-based approach. SIGNIFICANCE Elevated expression of CD44 variant 5 in intrahepatic cholangiocarcinoma confers a targetable vulnerability using the newly developed antibody-drug conjugate H1D8-DC, which induces potent growth suppressive effects without significant toxicity.
Collapse
Affiliation(s)
- Yuncheng Bei
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, PR China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, PR China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xuhui Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, PR China
| | - Yuxin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, PR China
| | - Sijie Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, PR China
| | - Wan Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, PR China
| | - Chengjie Cai
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, PR China
| | - Zhiye Xu
- Department of Clinical Laboratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jia Wei
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, PR China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and Clinical Cancer Institute of Nanjing University, Nanjing, Jiangsu, PR China
| | - Nan Zhang
- Centre of Micro/Nano Manufacturing Technology (MNMT-Dublin), School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland
| | - Pingping Shen
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, PR China
- Shenzhen Research Institute of Nanjing University, Shenzhen, PR China
| |
Collapse
|
28
|
Suzuki H, Kitamura K, Goto N, Ishikawa K, Ouchida T, Tanaka T, Kaneko MK, Kato Y. A Novel Anti-CD44 Variant 3 Monoclonal Antibody C 44Mab-6 Was Established for Multiple Applications. Int J Mol Sci 2023; 24:8411. [PMID: 37176118 PMCID: PMC10179237 DOI: 10.3390/ijms24098411] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Cluster of differentiation 44 (CD44) promotes tumor progression through the recruitment of growth factors and the acquisition of stemness, invasiveness, and drug resistance. CD44 has multiple isoforms including CD44 standard (CD44s) and CD44 variants (CD44v), which have common and unique functions in tumor development. Therefore, elucidating the function of each CD44 isoform in a tumor is essential for the establishment of CD44-targeting tumor therapy. We have established various anti-CD44s and anti-CD44v monoclonal antibodies (mAbs) through the immunization of CD44v3-10-overexpressed cells. In this study, we established C44Mab-6 (IgG1, kappa), which recognized the CD44 variant 3-encoded region (CD44v3), as determined via an enzyme-linked immunosorbent assay. C44Mab-6 reacted with CD44v3-10-overexpressed Chinese hamster ovary (CHO)-K1 cells (CHO/CD44v3-10) or some cancer cell lines (COLO205 and HSC-3) via flow cytometry. The apparent KD of C44Mab-6 for CHO/CD44v3-10, COLO205, and HSC-3 was 1.5 × 10-9 M, 6.3 × 10-9 M, and 1.9 × 10-9 M, respectively. C44Mab-6 could detect the CD44v3-10 in Western blotting and stained the formalin-fixed paraffin-embedded tumor sections in immunohistochemistry. These results indicate that C44Mab-6 is useful for detecting CD44v3 in various experiments and is expected for the application of tumor diagnosis and therapy.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (K.K.); (N.G.); (K.I.); (T.O.); (T.T.); (M.K.K.)
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Kaishi Kitamura
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (K.K.); (N.G.); (K.I.); (T.O.); (T.T.); (M.K.K.)
| | - Nohara Goto
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (K.K.); (N.G.); (K.I.); (T.O.); (T.T.); (M.K.K.)
| | - Kenichiro Ishikawa
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (K.K.); (N.G.); (K.I.); (T.O.); (T.T.); (M.K.K.)
| | - Tsunenori Ouchida
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (K.K.); (N.G.); (K.I.); (T.O.); (T.T.); (M.K.K.)
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Tomohiro Tanaka
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (K.K.); (N.G.); (K.I.); (T.O.); (T.T.); (M.K.K.)
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Mika K. Kaneko
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (K.K.); (N.G.); (K.I.); (T.O.); (T.T.); (M.K.K.)
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Yukinari Kato
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan; (K.K.); (N.G.); (K.I.); (T.O.); (T.T.); (M.K.K.)
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
29
|
Betulinic Acid Inhibits the Stemness of Gastric Cancer Cells by Regulating the GRP78-TGF-β1 Signaling Pathway and Macrophage Polarization. Molecules 2023; 28:molecules28041725. [PMID: 36838713 PMCID: PMC9964887 DOI: 10.3390/molecules28041725] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Cancer stemness is the process by which cancer cells acquire chemoresistance and self-renewal in the tumor microenvironment. Glucose-regulated protein 78 (GRP78) is a biomarker for gastric cancer and is involved in cancer stemness. By inducing cancer stemness in various types of cancer, the polarization of macrophages into tumor-associated macrophages (TAMs) controls tumor progression. Betulinic acid (BA) is a bioactive natural compound with anticancer properties. However, whether GRP78 regulates TAM-mediated cancer stemness in the tumor microenvironment and whether BA inhibits GRP78-mediated cancer stemness in gastric cancer remain unknown. In this study, we investigated the role of GRP78 in gastric cancer stemness in a tumor microenvironment regulated by BA. The results indicated that BA inhibited not only GRP78-mediated stemness-related protein expression and GRP78-TGF-β-mediated macrophage polarization into TAMs, but also TAM-mediated cancer stemness. Therefore, BA is a promising candidate for clinical application in combination-chemotherapy targeting cancer stemness.
Collapse
|
30
|
Giraud J, Seeneevassen L, Rousseau B, Bouriez D, Sifré E, Giese A, Nguyen TL, Tiffon C, Lippi Y, Azzi-Martin L, Pannequin J, Ménard A, Bessède E, Staedel C, Mégraud F, Belleannée G, Lehours P, Gronnier C, Dubus P, Varon C. CD44v3 is a marker of invasive cancer stem cells driving metastasis in gastric carcinoma. Gastric Cancer 2023; 26:234-249. [PMID: 36528833 PMCID: PMC9950191 DOI: 10.1007/s10120-022-01357-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/27/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cancer stem cells (CSCs) are at the origin of tumour initiation and progression in gastric adenocarcinoma (GC). However, markers of metastasis-initiating cells remain unidentified in GC. In this study, we characterized CD44 variants expressed in GC and evaluated the tumorigenic and metastatic properties of CD44v3+ cells and their clinical significance in GC patients. METHODS Using GC cell lines and patient-derived xenografts, we evaluated CD44+ and CD44v3+ GC cells molecular signature and their tumorigenic, chemoresistance, invasive and metastatic properties, and expression in patients-derived tissues. RESULTS CD44v3+ cells, which represented a subpopulation of CD44+ cells, were detected in advanced preneoplastic lesions and presented CSCs chemoresistance and tumorigenic properties in vitro and in vivo. Molecular and functional analyses revealed two subpopulations of gastric CSCs: CD44v3+ CSCs with an epithelial-mesenchymal transition (EMT)-like signature, and CD44+/v3- CSCs with an epithelial-like signature; both were tumorigenic but CD44v3+ cells showed higher invasive and metastatic properties in vivo. CD44v3+ cells detected in the primary tumours of GC patients were associated with a worse prognosis. CONCLUSION CD44v3 is a marker of a subpopulation of CSCs with metastatic properties in GC. The identification of metastasis-initiating cells in GC represents a major advance for further development of anti-metastatic therapeutic strategies.
Collapse
Affiliation(s)
- Julie Giraud
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Lornella Seeneevassen
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Benoit Rousseau
- Animal Facility, University of Bordeaux, 33076 Bordeaux, France
| | - Damien Bouriez
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,Department of Digestive Surgery, Haut-Lévêque Hospital, 33000 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France
| | - Elodie Sifré
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Alban Giese
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Tra Ly Nguyen
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Camille Tiffon
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Yannick Lippi
- Toxalim Research Centre in Food Toxicology, Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Lamia Azzi-Martin
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Julie Pannequin
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Armelle Ménard
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Emilie Bessède
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France
| | - Cathy Staedel
- INSERM U1212, ARNA, University of Bordeaux, 33076 Bordeaux, France
| | - Francis Mégraud
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France ,Centre National de Référence des Campylobacters et Helicobacters, Pellegrin Hospital, 33076 Bordeaux, France
| | - Geneviève Belleannée
- CHU Bordeaux, 33076 Bordeaux, France ,Department of Histology and Pathology, Haut-Lévêque Hospital, 33000 Bordeaux, France
| | - Philippe Lehours
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France ,Centre National de Référence des Campylobacters et Helicobacters, Pellegrin Hospital, 33076 Bordeaux, France
| | - Caroline Gronnier
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,Department of Digestive Surgery, Haut-Lévêque Hospital, 33000 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France
| | - Pierre Dubus
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076 Bordeaux, France ,CHU Bordeaux, 33076 Bordeaux, France ,Department of Histology and Pathology, Haut-Lévêque Hospital, 33000 Bordeaux, France
| | - Christine Varon
- INSERM U1312, Bordeaux Institute of Oncology, University of Bordeaux, 146 rue Leo Saignat, 33076, Bordeaux, France.
| |
Collapse
|
31
|
Verma AH, Haldavnekar R, Venkatakrishnan K, Tan B. Dual-Purpose 3D-Silica Nanostructure Matrix for Rapid Epigenetic Reprogramming of Tumor Cell to Cancer Stem Cell Spheroid. SMALL METHODS 2023; 7:e2200798. [PMID: 36424183 DOI: 10.1002/smtd.202200798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/14/2022] [Indexed: 06/16/2023]
Abstract
Cancer stem cells (CSCs), a rare subpopulation responsible for tumorigenesis and therapeutic resistance, are difficult to characterize and isolate. Conventional method of growing CSCs takes up to 2-8 weeks inhibiting the rate of research. Therefore, rapid reprogramming (RR) of tumor cells into CSCs is crucial to accelerate the stem cell oncology research. The current RR techniques cannot be utilized for CSC RR due to many limitations posed due to isolation requirements resulting in loss of vital data. Hence, a technique that can induce CSC RR without the need for isolation procedures is needed. Here, fabrication of a 3D-silica nanostructured extracellular matrix for RR and in situ monitoring is reported. The RR is tested using three preclinical cancer models. The 3D matrix and a zeta potential study confirm an intense material-cellular interaction resulting in the enhanced expressions of surface and epigenetic biomarkers. Cancer cells require only 3-day period to form CSC spheroids with 3D-silica extracellular matrix. Real-time single-cell monitoring of the methylene blue-induced photodynamic demonstrates the dual functionality. To the authors' knowledge, this is the first study to demonstrate a CSC epigenetic reprogramming using nanostructures. These findings may pave the path for accelerating the stem cell research in oncology.
Collapse
Affiliation(s)
- Anish Hiresha Verma
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, ON, M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University and St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
| | - Rupa Haldavnekar
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, ON, M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University and St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
| | - Krishnan Venkatakrishnan
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, ON, M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University and St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Ultrashort Laser Nanomanufacturing Research Facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
| | - Bo Tan
- Keenan Research Center for Biomedical Science, Unity Health Toronto, Toronto, ON, M5B 1W8, Canada
- Institute for Biomedical Engineering, Science and Technology (I BEST), Partnership between Toronto Metropolitan University and St. Michael's Hospital, Toronto, ON, M5B 1W8, Canada
- Nano-Bio Interface facility, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
- Nano-characterization Laboratory, Faculty of Engineering and Architectural Sciences, Toronto Metropolitan University, Toronto, ON, M5B 2K3, Canada
| |
Collapse
|
32
|
Kim EY, Lee SU, Kim YH. 1,2,3,4,6-Penta- O-galloyl-β-D-glucose Inhibits CD44v3, a cancer stem cell marker, by regulating its transcription factor, in human pancreatic cancer cell line. Anim Cells Syst (Seoul) 2022; 26:328-337. [PMID: 36605595 PMCID: PMC9809349 DOI: 10.1080/19768354.2022.2152864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Inhibition of cluster of differentiation 44 (CD44), a pancreatic cancer stem cell (CSC) marker, is a potential treatment for pancreatic ductal adenocarcinoma (PDAC). In this study, we evaluated the effect of 1,2,3,4,6-penta-O-galloyl-β-D-glucose (PGG), a gallotannin contained in various medicinal plants, on CD44 standard (CD44s) and CD44 variant 3 (CD44v3) in Mia-PaCa-2, human pancreatic cancer cells and explored the underlying mechanisms. PGG showed cytotoxic effects and inhibited the proliferation of Mia-PaCa-2 cells. It also inhibited clonogenic activity, adhesion to fibronectin, and cell migration, which are characteristics of CSCs. PGG inhibited the expression of CD44s and CD44v3 by inducing the phosphorylation of p53 and suppressing NF-κB and Foxo3. Inhibition of Foxo3 induces CD44v3 ubiquitination. Indeed, PGG increased proteasome activity and promoted CD44v3 ubiquitination. PGG downregulated the CSC regulatory factors Nanog, Oct-4, and Sox-2, which act downstream of CD44v3 signaling. These data indicate that PGG may have therapeutic effects in pancreatic cancer mediated by inhibition of CSC markers.
Collapse
Affiliation(s)
- Eun-Young Kim
- Department of Food and Nutrition, Daegu University, Gyeongsan-si, Republic of Korea
| | - Seong-Uk Lee
- Department of Food and Nutrition, Daegu University, Gyeongsan-si, Republic of Korea
| | - Yoon Hee Kim
- Department of Food and Nutrition, Daegu University, Gyeongsan-si, Republic of Korea, Yoon Hee Kim Department of Food and Nutrition, Daegu University, 201, Daegudae-ro, Gyeongsan-si, Gyeongsangbuk-do38453, Republic of Korea
| |
Collapse
|
33
|
Guo Q, Yang C, Gao F. The state of CD44 activation in cancer progression and therapeutic targeting. FEBS J 2022; 289:7970-7986. [PMID: 34478583 DOI: 10.1111/febs.16179] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/20/2021] [Accepted: 09/02/2021] [Indexed: 01/14/2023]
Abstract
CD44, a non-kinase transmembrane glycoprotein, is ubiquitously expressed on various types of cells, especially cancer stem cells (CSCs), and has been implicated in cancer onset and aggressiveness. The major ligand for the CD44, hyaluronan (HA), binds to and interacts with CD44, which in turn triggers downstream signaling cascades, thereby promoting cellular behaviors such as proliferation, motility, invasiveness and chemoresistance. The CD44-HA interaction is cell-specific and strongly affected by the state of CD44 activation. Therefore, the binding of HA to CD44 is essential for the activation of CD44 during which the detailed regulatory mechanism needs to be clarified. Different CD44 activation states distribute in human carcinoma and normal tissue; however, whether CD44 activation is a critical requirement for tumor initiation, progression and notorious CSC properties remains to be clarified. A deeper understanding of the regulation of CD44 activation may facilitate the development of novel targeted drugs in the future. Here, we review the current findings concerning the states of CD44 activation on the cell surface, the underlying regulatory mechanisms of CD44 activation, the known role for CD44 activation in tumor progression and CSC hallmarks, as well as the potential of HA-coated nanoparticle for targeting activated CD44 for cancer therapy.
Collapse
Affiliation(s)
- Qian Guo
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cuixia Yang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Feng Gao
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
34
|
Vishnubalaji R, Shaath H, Al-Alwan M, Abdelalim EM, Alajez NM. Reciprocal interplays between MicroRNAs and pluripotency transcription factors in dictating stemness features in human cancers. Semin Cancer Biol 2022; 87:1-16. [PMID: 36354097 DOI: 10.1016/j.semcancer.2022.10.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
The interplay between microRNAs (miRNAs) and pluripotency transcription factors (TFs) orchestrates the acquisition of cancer stem cell (CSC) features during the course of malignant transformation, rendering them essential cancer cell dependencies and therapeutic vulnerabilities. In this review, we discuss emerging themes in tumor heterogeneity, including the clonal evolution and the CSC models and their implications in resistance to cancer therapies, and then provide thorough coverage on the roles played by key TFs in maintaining normal and malignant stem cell pluripotency and plasticity. In addition, we discuss the reciprocal interactions between miRNAs and MYC, OCT4, NANOG, SOX2, and KLF4 pluripotency TFs and their contributions to tumorigenesis. We provide our view on the potential to interfere with key miRNA-TF networks through the use of RNA-based therapeutics as single agents or in combination with other therapeutic strategies, to abrogate the CSC state and render tumor cells more responsive to standard and targeted therapies.
Collapse
Affiliation(s)
- Radhakrishnan Vishnubalaji
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Hibah Shaath
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Monther Al-Alwan
- Stem Cell and Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; College of Medicine, Al-Faisal University, Riyadh 11533, Saudi Arabia
| | - Essam M Abdelalim
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Nehad M Alajez
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar; College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
| |
Collapse
|
35
|
Mirzaei S, Paskeh MDA, Entezari M, Mirmazloomi SR, Hassanpoor A, Aboutalebi M, Rezaei S, Hejazi ES, Kakavand A, Heidari H, Salimimoghadam S, Taheriazam A, Hashemi M, Samarghandian S. SOX2 function in cancers: Association with growth, invasion, stemness and therapy response. Biomed Pharmacother 2022; 156:113860. [DOI: 10.1016/j.biopha.2022.113860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/29/2022] Open
|
36
|
Cell adhesion molecule CD44v10 promotes stem-like properties in triple-negative breast cancer cells via glucose transporter GLUT1-mediated glycolysis. J Biol Chem 2022; 298:102588. [PMID: 36243113 PMCID: PMC9647553 DOI: 10.1016/j.jbc.2022.102588] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/02/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
Cell adhesion molecule CD44v8-10 is associated with tumor ste0mness and malignancy; however, whether CD44v10 alone confers these properties is unknown. Here, we demonstrated that CD44v10 promotes stemness and chemoresistance of triple-negative breast cancers (TNBCs) individually. Next, we identified that genes differentially expressed in response to ectopic expression of CD44v10 are mostly related to glycolysis. Further, we showed that CD44v10 upregulates glucose transporter 1 to facilitate glycolysis by activating the MAPK/ERK and PI3K/AKT signaling pathways. This glycolytic reprogramming induced by CD44v10 contributes to the stem-like properties of TNBC cells and confers resistance to paclitaxel treatment. Notably, we determined that the knockdown of glucose transporter 1 could attenuate the enhanced effects of CD44v10 on glycolysis, stemness, and paclitaxel resistance. Collectively, our findings provide novel insights into the function of CD44v10 in TNBCs and suggest that targeting CD44v10 may contribute to future clinical therapy.
Collapse
|
37
|
Thankappan P, Augustine PI, Shaga IB, Nirmal RM, Joseph TI, Girish KL. Immunohistochemical expression of putative cancer stem cell markers aldehyde dehydrogenase 1, SOX2, CD44 and OCT4 in different grades of oral epithelial dysplasia. J Oral Maxillofac Pathol 2022; 26:440-446. [PMID: 37082056 PMCID: PMC10112113 DOI: 10.4103/jomfp.jomfp_166_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/18/2021] [Indexed: 04/22/2023] Open
Abstract
Objectives The hypothesized existence of cancer stem cells (CSC) and its markers aldehyde dehydrogenase 1 (ALDH1), CD44, SOX2 and OCT4 in oral dysplastic tissues provides the potential for a more reliable assessment of malignant transformation of oral epithelial dysplasia (OED). Thus, the present study is intended to evaluate the immunohistochemical expression of four different CSC markers ALDH1, CD44, SOX2 and OCT4 in different grades of OED and to investigate the co-expression of these putative stem cell markers in OED. Subjects and Methods A total of 35 samples of varying grades of OED which included 7 mild, 11 moderate and 17 severe dysplasia samples and 10 samples of normal oral mucosa without dysplasia were used. Four sections each from all 45 samples were stained with ALDH1, CD44, SOX2 and OCT4, respectively, by immunohistochemistry. The acquired data were analyzed and evaluated using the Chi-square test and unpaired t-test and the P < 0.05 was taken significant. Results ALDH1 and SOX2 expression percentages showed statistically significant differences among study groups (P < 0.05). Statistical comparison of percentage expression of CD44 and OCT4 between OED and normal was nonsignificant (P > 0.05). Co-expression of all four markers was found in 15 cases of OED with none of the normal cases showing co-expression. Conclusion The expression of CSC markers in OED and normal mucosa differs significantly with co-expression of all four markers located only in dysplastic tissues. Until now, no single protein marker has been able to unequivocally identify the CSCs. Thus, a panel of putative CSC markers will help in identifying the patients with high risk for malignant transformation in OED.
Collapse
Affiliation(s)
- Prasanth Thankappan
- Department of Oral and Maxillofacial Pathology, Sree Mookambika Institute of Dental Sciences, Kanyakumari, Tamil Nadu, India
| | - Percy Ida Augustine
- Department of Oral and Maxillofacial Pathology, Sree Mookambika Institute of Dental Sciences, Kanyakumari, Tamil Nadu, India
| | - I Bevin Shaga
- Department of Orthodontics and Dentofacial Orthopedics, Rajas Dental College, Tirunelveli, Tamil Nadu, India
| | - R Madhavan Nirmal
- Department of Oral and Maxillofacial Pathology, Rajah Muthiah Dental College and Hospital, Annamalai University, Chidambaram, Tamil Nadu, India
| | - T Isaac Joseph
- Department of Oral and Maxillofacial Pathology, Sree Mookambika Institute of Dental Sciences, Kanyakumari, Tamil Nadu, India
| | - KL Girish
- Department of Oral and Maxillofacial Pathology, Sree Mookambika Institute of Dental Sciences, Kanyakumari, Tamil Nadu, India
| |
Collapse
|
38
|
Nandi SK, Pradhan A, Das B, Das B, Basu S, Mallick B, Dutta A, Sarkar DK, Mukhopadhyay A, Mukhopadhyay S, Bhattacharya R. Kaempferol attenuates viability of ex-vivo cultured post-NACT breast tumor explants through downregulation of p53 induced stemness, inflammation and apoptosis evasion pathways. Pathol Res Pract 2022; 237:154029. [PMID: 35961057 DOI: 10.1016/j.prp.2022.154029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 01/15/2023]
Abstract
Early onset of chemotherapy evasion is a therapeutic challenge. Chemotherapy-induced upregulation of stem cell markers imparts invasiveness and metastatic property to the resident tumor. The efficacy of Kaempferol in attenuating epithelial to mesenchymal transition has earlier been established in the breast cancer cell. In our study population, progression-free survival was observed to be statistically more significant in post-NACT low-grade tumors than the high-grade tumors. Further, in post-NACT TNBCs, high-grade tumors showed a preponderance of strong nuclear p53 expression and very low expression of Caspase 3, indicating that, altered p53 expression predisposes these tumors to apoptosis escape and up-regulation of stemness markers. Herein, we report the robust efficacy of Kaempferol on ex-vivo grown breast tumors, derived from post-NACT TNBC patients, through downregulation of nuclear p53, CD44, ALDH1, NANOG, MDR1, Ki67, BCL2 and upregulation of Caspase 3. Such tumors also showed concurrent deregulated RNA and protein expression of CD44, NANOG, ALDH1 and MDR1 with upregulation of Caspase 3 and cleaved Caspase 3, upon Kaempferol treatment. Validation of efficacy of the treatment dosage of Kaempferol through immunophenotyping on MDA-MB-231, suggested that Kaempferol at its IC-50 dosage was effective against CD44 and CD326 positive breast cancer through deregulating their expression. Protein-protein interaction network through STRING pathway analysis and co-expression study of candidate proteins showed the highest degree of co-expression of p53 and KI-67, CD44, NF- kappaB, ALDH1, NANOG, MDR1, and BCL2. Thus, potentially targetable oncogenic protein markers, that are susceptible to downregulation by Kaempferol, provides insight into biomarker-driven therapeutic approaches with it.
Collapse
Affiliation(s)
- Sourav Kumar Nandi
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata 700094, India
| | - Ayan Pradhan
- Department of General Surgery, Institute of Post graduate Medical Education & Research and SSKM Hospital, 244B AJC Bose Road, Kolkata 700020, India
| | - Basudeb Das
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Biswajit Das
- Department of Pathology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata 700094, India
| | - Sudarshana Basu
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata 700094, India
| | - Bibekanand Mallick
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Amitava Dutta
- Department of Pathology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata 700094, India
| | - Diptendra Kumar Sarkar
- Department of General Surgery, Institute of Post graduate Medical Education & Research and SSKM Hospital, 244B AJC Bose Road, Kolkata 700020, India
| | - Ashis Mukhopadhyay
- Department of Haematooncology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata 700094, India
| | - Soma Mukhopadhyay
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata 700094, India.
| | - Rittwika Bhattacharya
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata 700094, India.
| |
Collapse
|
39
|
Wang Z, Wang J, Zhao H, Zhao T, Chen Y, Jiang M, Zhang S, Wei Y, Zhang J, Zhou Y, Shi S, Fu Z, Yang Y, Zhang Y, Yang L, Que J, Liu K. Targeting the SOX2/PARP1 complex to intervene in the growth of esophageal squamous cell carcinoma. Biomed Pharmacother 2022; 153:113309. [PMID: 35738180 DOI: 10.1016/j.biopha.2022.113309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
Elevated SOX2 protein levels are closely correlated with the increased incidence of esophageal squamous cell carcinoma (ESCC). However, establishing effective target measures for ESCC treatments continue to be researched. It has been previously proposed that SOX2 represents a potential therapeutic target for ESCC. Here, we found that the enzyme Poly(ADP-Ribose) polymerase 1 (PARP1) enriched in ESCCs interact with SOX2. Inhibition of PARP1 with 3-aminobenzamide (3-ABA) or shRNA knockdown reduced the proliferation of ESCCs, accompanied by decreased protein levels of SOX2. RNA sequencing demonstrated that PARP1 inhibition affected multiple signaling pathways involved in cancer cell proliferation. Additionally, 3-ABA synergistically suppressed the growth of ESCC cells when combined with cisplatin, and metformin potentiated the suppressive effect of 3-ABA on ESCC cell growth. Together these findings suggest that targeting SOX2 binding partner PARP1 provides a possible avenue to treat patients with high levels of SOX2.
Collapse
Affiliation(s)
- Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Junkai Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yunyun Chen
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Ming Jiang
- Department of Gastroenterology of The Children's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Shihui Zhang
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Yuxuan Wei
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiaying Zhang
- School of Life Science, Xiamen University, Xiamen, Fujian 361102, China
| | - Yijian Zhou
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhichao Fu
- Department of radiotherapy, 900 Hospital of the Joint Logistics Team (Dongfang Hospital, Xiamen University), Fuzhou, Fujian 350025, China
| | - Yaxin Yang
- Department of Biology, University of Rochester, NY 14627, USA
| | - Yujun Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Ling Yang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
40
|
Vasefifar P, Motafakkerazad R, Maleki LA, Najafi S, Ghrobaninezhad F, Najafzadeh B, Alemohammad H, Amini M, Baghbanzadeh A, Baradaran B. Nanog, as a key cancer stem cell marker in tumor progression. Gene X 2022; 827:146448. [PMID: 35337852 DOI: 10.1016/j.gene.2022.146448] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/16/2022] [Accepted: 03/18/2022] [Indexed: 12/20/2022] Open
Abstract
Cancer stem cells (CSCs) are a small population of malignant cells that induce tumor onset and development. CSCs share similar features with normal stem cells in the case of self-renewal and differentiation. They also contribute to chemoresistance and metastasis of cancer cells, leading to therapeutic failure. To identify CSCs, multiple cell surface markers have been characterized, including Nanog, which is found at high levels in different cancers. Recent studies have revealed that Nanog upregulation has a substantial association with the advanced stages and poor prognosis of malignancies, playing a pivotal role through tumorigenesis of multiple human cancers, including leukemia, liver, colorectal, prostate, ovarian, lung, head and neck, brain, pancreatic, gastric and breast cancers. Nanog through different signaling pathways, like JAK/STAT and Wnt/β-catenin pathways, induces stemness, self-renewal, metastasis, invasiveness, and chemoresistance of cancer cells. Some of these signaling pathways are common in various types of cancers, but some have been found in one or two cancers. Therefore, this review aimed to focus on the function of Nanog in multiple cancers based on recent studies surveying the suitable approaches to target Nanog and inhibit CSCs residing in tumors to gain favorable results from cancer treatments.
Collapse
Affiliation(s)
- Parisa Vasefifar
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Basira Najafzadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Hajar Alemohammad
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
41
|
Luo Y, Liang F, Wan X, Liu S, Fu L, Mo J, Meng X, Mo Z. Hyaluronic Acid Facilitates Angiogenesis of Endothelial Colony Forming Cell Combining With Mesenchymal Stem Cell via CD44/ MicroRNA-139-5p Pathway. Front Bioeng Biotechnol 2022; 10:794037. [PMID: 35350177 PMCID: PMC8957954 DOI: 10.3389/fbioe.2022.794037] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/19/2022] [Indexed: 12/13/2022] Open
Abstract
Stem cells and progenitor cells have been identified as potential new therapeutic options for severe limb ischemia to induce angiogenesis, and hyaluronic acid (HA) is commonly applied as a biomaterial in tissue engineering. However, the efficiency of HA combined with human umbilical cord blood-derived endothelial colony forming cells (ECFCs) and human umbilical-derived mesenchymal stem cells (MSCs) on angiogenesis is unclear. In the present study, we showed that HA promoted angiogenesis induced by MSCs-ECFCs in Matrigel plugs and promoted blood perfusion of murine ischemic muscles. Laser confocal microscopy revealed that human-derived cells grew into the host vasculature and formed connections, as shown by mouse-specific CD31+/human-specific CD31+ double staining. In vitro assays revealed that HA supported cell proliferation and migration, enhanced CD44 expression and reduced microRNA (miR)-139-5p expression. Further analysis revealed that miR-139-5p expression was negatively regulated by CD44 in ECFCs. Flow cytometry assays showed that HA increased CD31 positive cells proportion in MSC-ECFC and could be reversed by miR-139-5p mimics transfection. Moreover, the improvement of MSC-ECFC proliferation and migration induced by HA could be blocked by upregulation of miR-139-5p expression. In conclusion, HA facilitates angiogenesis of MSCs-ECFCs, and this positive effect be associated with activation of the CD44/miR-139-5p pathway, providing a promising strategy for improving severe limb ischemia.
Collapse
Affiliation(s)
- Yufang Luo
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
| | - Fang Liang
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
| | - Shengping Liu
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
| | - Lanfang Fu
- Department of Endocrinology, Haikou People’s Hospital and Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Jiake Mo
- School of Medicine, Hunan Normal University, Changsha, China
| | - Xubiao Meng
- Department of Endocrinology, Haikou People’s Hospital and Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
- *Correspondence: Xubiao Meng, ; Zhaohui Mo,
| | - Zhaohui Mo
- Department of Endocrinology, Third Xiangya Hospital of Central South University and Diabetic Foot Research Center of Central South University, Changsha, China
- *Correspondence: Xubiao Meng, ; Zhaohui Mo,
| |
Collapse
|
42
|
Li X, Li K, Li M, Lin X, Mei Y, Huang X, Yang H. Chemoresistance Transmission via Exosome-Transferred MMP14 in Pancreatic Cancer. Front Oncol 2022; 12:844648. [PMID: 35223528 PMCID: PMC8865617 DOI: 10.3389/fonc.2022.844648] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies. Gemcitabine is the most commonly used chemotherapy for the treatment of PDAC, but the development of drug resistance still remains challenging. Recently, exosomes have emerged as important mediators for intercellular communication. Exosomes affect recipient cells’ behavior through the engulfed cargos, however the specific cargos responsible for gemcitabine resistance in PDAC are poorly understood. Here, we reported that exosomes could transfer gemcitabine resistance via a metalloproteinase 14 (MMP14)-dependent mechanism. MMP14 was identified as a major differentially secreted protein from the gemcitabine-resistant PDAC cells by comparative secretome. It was packaged into the exosomes and transmitted from the chemoresistant cells to the sensitive ones. The exosome-transferred MMP14 could enhance drug resistance and promotes the sphere-formation and migration abilities of the recipient sensitive PDAC cells. Mechanically, exosome-transferred MMP14 promotes the stability of CD44, the cancer stem cell marker in the recipient cells. Our results indicate that MMP14 is a key player for exosome-mediated transfer of gemcitabine resistance, thus targeting MMP14 in exosomes may represent a novel strategy to limit gemcitabine resistance in PDAC.
Collapse
Affiliation(s)
- Xinyuan Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kai Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Mengmeng Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiaoyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yu Mei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xuemei Huang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Huanjie Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
43
|
Pouremamali F, Vahedian V, Hassani N, Mirzaei S, Pouremamali A, Kazemzadeh H, Faridvand Y, Jafari-gharabaghlou D, Nouri M, Maroufi NF. The role of SOX family in cancer stem cell maintenance: With a focus on SOX2. Pathol Res Pract 2022; 231:153783. [DOI: 10.1016/j.prp.2022.153783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/17/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023]
|
44
|
Strohmayer C, Klang A, Kummer S, Walter I, Jindra C, Weissenbacher-Lang C, Redmer T, Kneissl S, Brandt S. Tumor Cell Plasticity in Equine Papillomavirus-Positive Versus-Negative Squamous Cell Carcinoma of the Head and Neck. Pathogens 2022; 11:pathogens11020266. [PMID: 35215208 PMCID: PMC8875230 DOI: 10.3390/pathogens11020266] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 11/29/2022] Open
Abstract
Squamous cell carcinoma of the head and neck (HNSCC) is a common malignant tumor in humans and animals. In humans, papillomavirus (PV)-induced HNSCCs have a better prognosis than papillomavirus-unrelated HNSCCs. The ability of tumor cells to switch from epithelial to mesenchymal, endothelial, or therapy-resistant stem-cell-like phenotypes promotes disease progression and metastasis. In equine HNSCC, PV-association and tumor cell phenotype switching are poorly understood. We screened 49 equine HNSCCs for equine PV (EcPV) type 2, 3 and 5 infection. Subsequently, PV-positive versus -negative lesions were analyzed for expression of selected epithelial (keratins, β-catenin), mesenchymal (vimentin), endothelial (COX-2), and stem-cell markers (CD271, CD44) by immunohistochemistry (IHC) and immunofluorescence (IF; keratins/vimentin, CD44/CD271 double-staining) to address tumor cell plasticity in relation to PV infection. Only EcPV2 PCR scored positive for 11/49 equine HNSCCs. IHC and IF from 11 EcPV2-positive and 11 EcPV2-negative tumors revealed epithelial-to-mesenchymal transition events, with vimentin-positive cells ranging between <10 and >50%. CD44- and CD271-staining disclosed the intralesional presence of infiltrative tumor cell fronts and double-positive tumor cell subsets independently of the PV infection status. Our findings are indicative of (partial) epithelial–mesenchymal transition events giving rise to hybrid epithelial/mesenchymal and stem-cell-like tumor cell phenotypes in equine HNSCCs and suggest CD44 and CD271 as potential malignancy markers that merit to be further explored in the horse.
Collapse
Affiliation(s)
- Carina Strohmayer
- Clinical Unit of Diagnostic Imaging, Department for Companion Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria; (C.S.); (S.K.)
| | - Andrea Klang
- Institute of Pathology, Department of Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria; (A.K.); (C.W.-L.)
| | - Stefan Kummer
- VetCore Facility for Research, University of Veterinary Medicine, 1210 Vienna, Austria; (S.K.); (I.W.)
| | - Ingrid Walter
- VetCore Facility for Research, University of Veterinary Medicine, 1210 Vienna, Austria; (S.K.); (I.W.)
- Institute of Morphology, Department of Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Christoph Jindra
- Research Group Oncology (RGO), Clinical Unit of Equine Surgery, Department for Companion Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Christiane Weissenbacher-Lang
- Institute of Pathology, Department of Pathobiology, University of Veterinary Medicine, 1210 Vienna, Austria; (A.K.); (C.W.-L.)
| | - Torben Redmer
- Institute of Medical Biochemistry, Department of Biomedical Sciences, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Sibylle Kneissl
- Clinical Unit of Diagnostic Imaging, Department for Companion Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria; (C.S.); (S.K.)
| | - Sabine Brandt
- Research Group Oncology (RGO), Clinical Unit of Equine Surgery, Department for Companion Animals and Horses, University of Veterinary Medicine, 1210 Vienna, Austria;
- Correspondence: ; Tel.: +43-12-5077-5308
| |
Collapse
|
45
|
Kołodziej-Rzepa M, Biesaga B, Janecka-Widła A, Mucha-Małecka A. Lack of CD44 and Sox-2 Overexpression as Two Independent Favourable Prognostic Factors in HPV Positive Patients with Oropharyngeal Cancers. Pathobiology 2022; 89:205-213. [PMID: 35078199 DOI: 10.1159/000521292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/01/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION In our earlier publications, in the group of 63 patients with oropharyngeal cancer, we have found HPV16 infection (assessed by qPCR) in 25 tumours (39.7%), immunohistochemical overexpression of CD44, CD98, ALDH1/2 and Nanog in, respectively: 43 (68.2%), 30 (47.6%), 33 (52.4%), and 53 (84.1%) cancers. Analysing CD44, CD98, ALDH1/2, we have also shown that lack of CD44 overexpression indicates excellent prognosis in patients with HPV16 positivity. The aim of the present study was to compare prognostic potential of Nanog, Oct3/4, Sox-2 expression in relation to CD44, CD98, ALDH1/2 immunoreactivity (assessed by us earlier) and clinicopathological features in the subgroups of patients: with HPV16 positivity and HPV16 negativity. METHODS Status of Oct3/4 and Sox-2 expression was assessed for 63 patients with oropharyngeal cancers based on immunohistochemistry. In survival analysis, two endpoints were applied: overall survival (OS) and disease-free survival (DFS). RESULTS Overexpression of Oct3/4 and Sox-2 was found in 0 (0.0%) and 27 (42.9%) of patients. In the subgroup with HPV16 positivity, the DFS for patients with lack of Sox-2 overexpression was significantly (p = 0.003) higher than for patients with Sox-2 overexpression. In the subgroup with HPV16 negativity, Nanog and Sox-2 immunoexpression did not significantly influence OS and DFS. In multivariate analysis performed for the subgroup with HPV16 positivity, lack of CD44 overexpression (p = 0.012) and lack of Sox-2 overexpression (p = 0.027) were positive independent prognostic factors. CONCLUSION Based on CD44 and Sox-2 immunoreactivity, it is possible to differentiate the prognosis of HPV16-positive patients with oropharyngeal cancers.
Collapse
Affiliation(s)
- Marta Kołodziej-Rzepa
- Department of General, Oncological and Vascular Surgery, 5th Military Clinical Hospital in Cracow, Cracow, Poland
| | - Beata Biesaga
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland.,Department of Tumour Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, Cracow, Poland
| | - Anna Janecka-Widła
- Department of Tumour Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, Cracow, Poland
| | - Anna Mucha-Małecka
- Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Cracow Branch, Cracow, Poland
| |
Collapse
|
46
|
microRNA-21 Regulates Stemness in Pancreatic Ductal Adenocarcinoma Cells. Int J Mol Sci 2022; 23:ijms23031275. [PMID: 35163198 PMCID: PMC8835847 DOI: 10.3390/ijms23031275] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common and aggressive type of pancreatic cancer (PCa) with a low survival rate. microRNAs (miRs) are endogenous, non-coding RNAs that moderate numerous biological processes. miRs have been associated with the chemoresistance and metastasis of PDAC and the presence of a subpopulation of highly plastic "stem"-like cells within the tumor, known as cancer stem cells (CSCs). In this study, we investigated the role of miR-21, which is highly expressed in Panc-1 and MiaPaCa-2 PDAC cells in association with CSCs. Following miR-21 knockouts (KO) from both MiaPaCa-2 and Panc-1 cell lines, reversed expressions of epithelial-mesenchymal transition (EMT) and CSCs markers were observed. The expression patterns of key CSC markers, including CD44, CD133, CX-C chemokine receptor type 4 (CXCR4), and aldehyde dehydrogenase-1 (ALDH1), were changed depending on miR-21 status. miR-21 (KO) suppressed cellular invasion of Panc-1 and MiaPaCa-2 cells, as well as the cellular proliferation of MiaPaCa-2 cells. Our data suggest that miR-21 is involved in the stemness of PDAC cells, may play roles in mesenchymal transition, and that miR-21 poses as a novel, functional biomarker for PDAC aggressiveness.
Collapse
|
47
|
Epi-miRNAs: Regulators of the Histone Modification Machinery in Human Cancer. JOURNAL OF ONCOLOGY 2022; 2022:4889807. [PMID: 35087589 PMCID: PMC8789461 DOI: 10.1155/2022/4889807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022]
Abstract
Cancer is a leading cause of death and disability worldwide. Epigenetic deregulation is one of the most critical mechanisms in carcinogenesis and can be classified into effects on DNA methylation and histone modification. MicroRNAs are small noncoding RNAs involved in fine-tuning their target genes after transcription. Various microRNAs control the expression of histone modifiers and are involved in a variety of cancers. Therefore, overexpression or downregulation of microRNAs can alter cell fate and cause malignancies. In this review, we discuss the role of microRNAs in regulating the histone modification machinery in various cancers, with a focus on the histone-modifying enzymes such as acetylases, deacetylases, methyltransferases, demethylases, kinases, phosphatases, desumoylases, ubiquitinases, and deubiquitinases. Understanding of microRNA-related aberrations underlying histone modifiers in pathogenesis of different cancers can help identify novel therapeutic targets or early detection approaches that allow better management of patients or monitoring of treatment response.
Collapse
|
48
|
Ketkar M, Dutt S. Epigenetic Regulation Towards Acquired Drug Resistance in Cancer. Subcell Biochem 2022; 100:473-502. [PMID: 36301503 DOI: 10.1007/978-3-031-07634-3_14] [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] [Indexed: 06/16/2023]
Abstract
Therapy resistance remains the most challenging obstacle in cancer treatment. Substantial efforts and evidences have accumulated over decades suggesting not only genetic but non-genomic mechanisms underlying this adaptation of tumor cells. Alterations in epigenome can have a fundamental effect on cellular functions and response to stresses like anticancer therapy. This chapter discusses the principal mechanisms by which epigenetic modifications in the genome and transcriptome aid tumor cells toward acquisition of resistance to chemotherapy.
Collapse
Affiliation(s)
- Madhura Ketkar
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India.
- Homi Bhabha National Institute, Mumbai, India.
- ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra, India.
| |
Collapse
|
49
|
Joshi J, Patel H, Bhavnagari H, Tarapara B, Pandit A, Shah F. Eliminating Cancer Stem-Like Cells in Oral Cancer by Targeting Elementary Signaling Pathways. Crit Rev Oncog 2022; 27:65-82. [PMID: 37199303 DOI: 10.1615/critrevoncog.2022047207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Oral cancer is a heterogeneous, aggressive, and complex entity. Current major treatment options for the disease are surgery, chemo, and/or radiotherapy either alone or in combination with each other. Each treatment method has its own limitations such as a significant journey with deformities and a protracted rehabilitation process leading to loss of self-esteem, loss of tolerance, and therapeutic side effects. Conventional therapies are frequently experienced with regimen resistance and recurrence attributed to the cancer stem cells (CSCs). Given that CSCs exert their tumorigenesis by affecting several cellular and molecular targets and pathways an improved understanding of CSCs' actions is required. Hence, more research is recommended to fully understand the fundamental mechanisms driving CSC-mediated treatment resistance. Despite the difficulties and disagreements surrounding the removal of CSCs from solid tumors, a great amount of knowledge has been derived from the characterization of CSCs. Various efforts have been made to identify the CSCs using several cell surface markers. In the current review, we will discuss numerous cell surface markers such as CD44, ALDH1, EPCAM, CD24, CD133, CD271, CD90, and Cripto-1 for identifying and isolating CSCs from primary oral squamous cell carcinoma (OSCC). Further, a spectrum of embryonic signaling pathways has been thought to be the main culprit of CSCs' active state in cancers, resulting in conventional therapeutic resistance. Hence, we discuss the functional and molecular bases of several signaling pathways such as the Wnt/beta;-catenin, Notch, Hedgehog, and Hippo pathways and their associations with disease aggressiveness. Moreover, numerous inhibitors targeting the above mentioned signaling pathways have already been identified and some of them are already undergoing clinical trials. Hence, the present review encapsulates the characterization and effectiveness of the prospective potential targeted therapies for eradicating CSCs in oral cancers.
Collapse
Affiliation(s)
- Jigna Joshi
- Molecular Diagnostic and Research Lab-III, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Hitarth Patel
- Molecular Diagnostic and Research Lab-III, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Hunayna Bhavnagari
- Molecular Diagnostic and Research Lab-III, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Bhoomi Tarapara
- Molecular Diagnostic and Research Lab-III, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Apexa Pandit
- Molecular Diagnostic and Research Lab-III, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Franky Shah
- Molecular Diagnostic and Research Lab-III, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| |
Collapse
|
50
|
Ebrahimi N, Rezanejad H, Asadi MH, Vallian S. LncRNA LOC100507144 acts as a novel regulator of CD44/Nanog/Sox2/miR-302/miR-21 axis in colorectal cancer. Biofactors 2022; 48:164-180. [PMID: 34882869 DOI: 10.1002/biof.1813] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022]
Abstract
Long noncoding RNAs (lncRNAs) appear as vital regulators and biomarkers in many human cancers. LOC100507144 is a validated lncRNA located in the neighborhood of CD44 in a head-to-head configuration, and its expression and function in cancer cells are still unknown. This research aimed to find out more about the expression and function of this lncRNA in colorectal cancer (CRC). Our expression data represented that the expression of LOC100507144 transcript was substantially higher in tumors with advanced stages, lymph node metastasis, and vascular invasion. Loss-of-function examinations demonstrated that LOC100507144 contributed to CRC cell proliferation by restricting apoptosis, cellular senescence, and promoting cell cycle. Gain-of-function experiments also confirmed these results. Our data illustrated that LOC100507144 enhanced the migration and the epithelial to mesenchymal transition (EMT) of CRC cells, accompanied by the generation of cells with stemness characteristics. Our findings revealed that the knocking-down of LOC100507144 inhibited the expression of crucial stemness factors, including CD44, Nanog, and Sox2, and accordingly resulted in suppressing their targets, miR-302 and miR-21. Overall, the current study's findings for the first time reveal that LOC100507144 could enhance CRC progression and metastasis through regulation of the CD44/Nanog/Sox2/miR-302/miR-21 axis.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Hajar Rezanejad
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Malek Hossein Asadi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Sadeq Vallian
- Division of Genetics, Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|