1
|
Caliendo A, Camorani S, Ibarra LE, Pinto G, Agnello L, Albanese S, Caianiello A, Illiano A, Festa R, Ambrosio V, Scognamiglio G, Cantile M, Amoresano A, Fedele M, Zannetti A, Cerchia L. A novel CD44-targeting aptamer recognizes chemoresistant mesenchymal stem-like TNBC cells and inhibits tumor growth. Bioact Mater 2025; 50:443-460. [PMID: 40342488 PMCID: PMC12059597 DOI: 10.1016/j.bioactmat.2025.04.027] [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: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/20/2025] [Indexed: 05/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) represents a significant therapeutic challenge owing to the scarcity of targeted medicines and elevated recurrence rates. We previously reported the development of the nuclease-resistant RNA sTN58 aptamer, which selectively targets TNBC cells. Here, sTN58 aptamer was employed to capture and purify its binding target from the membrane protein fraction of cisplatin-resistant mesenchymal stem-like TNBC cells. Mass spectrometry in conjunction with aptamer binding assays across various cancer cell lines identified CD44 as the cellular target of sTN58. By binding to CD44, sTN58 inhibits the invasive growth and hyaluronic acid-dependent tube formation in chemoresistant TNBC cells, where CD44 serves as a key driver of tumor cell aggressiveness and stem-like plasticity. Moreover, in vivo studies demonstrated the aptamer's high tumor targeting efficacy and its capacity to significantly inhibit tumor growth and lung metastases following intravenous administration in mice with orthotopic TNBC. Overall, our findings reveal the striking potential of sTN58 as a targeting reagent for the recognition and therapy of cancers overexpressing CD44.
Collapse
Affiliation(s)
- Alessandra Caliendo
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Simona Camorani
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Luis Exequiel Ibarra
- Institute of Environmental Biotechnology and Health (INBIAS), National University of Rio Cuarto (UNRC), National Council for Scientific and Technological Research (CONICET), Río Cuarto, X5800BIA, Argentina
| | - Gabriella Pinto
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Lisa Agnello
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Sandra Albanese
- Institute of Biostructures and Bioimaging, National Research Council, 80145, Naples, Italy
| | - Antonietta Caianiello
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Anna Illiano
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Rosaria Festa
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Vincenzo Ambrosio
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Giosuè Scognamiglio
- Institutional Biobank-Scientific Directorate, National Cancer Institute INT-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Monica Cantile
- Institutional Biobank-Scientific Directorate, National Cancer Institute INT-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Angela Amoresano
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Monica Fedele
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging, National Research Council, 80145, Naples, Italy
| | - Laura Cerchia
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| |
Collapse
|
2
|
Kevat S, Mistry A, Oza N, Majmudar M, Patel N, Shah R, Ramachandran AV, Chauhan R, Haque S, Parashar NC, Tuli HS, Parashar G. Cancer Stem Cell Regulation as a Target of Therapeutic Intervention: Insights into Breast, Cervical and Lung Cancer. Cell Biochem Biophys 2025; 83:1521-1535. [PMID: 39843681 DOI: 10.1007/s12013-025-01666-w] [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] [Accepted: 01/03/2025] [Indexed: 01/24/2025]
Abstract
Cancer Stem Cells (CSCs) play an important role in the development, resistance, and recurrence of many malignancies. These subpopulations of tumor cells have the potential to self-renew, differentiate, and resist conventional therapy, highlighting their importance in cancer etiology. This review explores the regulatory mechanisms of CSCs in breast, cervical, and lung cancers, highlighting their plasticity, self-renewal, and differentiation capabilities. CD44+/CD24- cells are a known marker for breast CSCs. Markers like as CD133 and ALDH have been discovered in cervical cancer CSCs. Similarly, in lung cancer, CSCs identified by CD44, CD133, and ALDH are linked to aggressive tumor behavior and poor therapy results. The commonalities between these tumors highlight the general necessity of targeting CSCs in treatment efforts. However, the intricacies of CSC activity, such as their interaction with the tumor microenvironment and particular signaling pathways differ between cancer types, demanding specialized methods. Wnt/β-catenin, Notch, and Hedgehog pathways are one of the essential signaling pathways, targeting them, may show ameliorative effects on breast, lung and cervical carcinomas and their respective CSCs. Pre-clinical data suggests targeting specific signaling pathways can eliminate CSCs, but ongoing clinical trials are on utilizing signaling pathway inhibitors in patients. In recent studies it has been reported that CAR T based targeting of specific markers may be used as combination therapy. Ongoing research related to nanobiotechnology can also play a significant role in diagnosis and treatment purpose targeting CSCs, as nanomaterials can be used for precise targeting and identification of CSCs. Further research into the targeting of signaling pathways and its precursors could prove to be right step into directing therapies towards CSCs for cancer therapy.
Collapse
Affiliation(s)
- Sakshi Kevat
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Archie Mistry
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Naman Oza
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Mohit Majmudar
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Netra Patel
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Rushabh Shah
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - A V Ramachandran
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- School Of Medicine, Universidad Espiritu Santo, Samborondon, Ecuador
| | | | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar (Deemed to be University), Ambala, Haryana, India
| | - Gaurav Parashar
- Division of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara, Gujarat, India.
| |
Collapse
|
3
|
Xiao M, Chen S, Yang Y, Hu K, Song Y, Hou Z, Sun S, Yang L. Potential of natural polysaccharide for ovarian cancer therapy. Carbohydr Polym 2025; 348:122946. [PMID: 39567158 DOI: 10.1016/j.carbpol.2024.122946] [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: 07/15/2024] [Revised: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024]
Abstract
Ovarian cancer, characterized by high lethality, presents a significant clinical challenge. The standard first-line treatment is surgery and chemotherapy; however, postoperative chemotherapy is often ineffective and associated with severe side effects and the development of drug resistance. Consequently, there is an urgent need for innovative drug delivery strategies to enhance therapeutic efficacy. Natural polysaccharide polymers with high bioactivity have been extensively investigated for use alone or as adjuvants to chemotherapy and radiotherapy, and also for the preparation of efficient delivery systems for ovarian cancer therapy. This paper aims to review recent advances in the application of natural polysaccharides, including hyaluronic acid, chitosan, alginate, and cellulose, in the therapy of ovarian cancer. This paper primarily discusses the anti-tumor properties inherent to these natural polysaccharide polymers and offers a summary of their role in delivery systems used in ovarian cancer therapy.
Collapse
Affiliation(s)
- Miaomiao Xiao
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Siwen Chen
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Center for Molecular Science and Engineering, College of Science, Northeastern University, Shenyang 110819, China
| | - Yaochen Yang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Ke Hu
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yantao Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110002, China
| | - Zhipeng Hou
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| | - Siyu Sun
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Liqun Yang
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| |
Collapse
|
4
|
Kinoshita J, Doden K, Sakimura Y, Hayashi S, Saito H, Tsuji T, Yamamoto D, Moriyama H, Minamoto T, Inaki N. Crosstalk Between Omental Adipose-Derived Stem Cells and Gastric Cancer Cells Regulates Cancer Stemness and Chemotherapy Resistance. Cancers (Basel) 2024; 16:4275. [PMID: 39766174 PMCID: PMC11674675 DOI: 10.3390/cancers16244275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Peritoneal metastasis (PM) remains a major challenge in patients with gastric cancer (GC) and occurs preferentially in adipose-rich organs, such as the omentum. Adipose-derived stem cells (ASCs) may influence cancer behavior. This study aimed to investigate whether ASCs isolated from the omentum can act as progenitors of cancer-associated fibroblasts (CAFs) and analyze their effects on the cancer stem cell (CSC) niche and the treatment resistance of GC cells. Methods: ASCs were isolated from the human omentum and their cellular characteristics were analyzed during co-culturing with GC cells. Results: ASCs express CAF markers and promote desmoplasia in cancer stroma in a mouse xenograft model. When co-cultured with GC cells, ASCs enhanced the sphere-forming efficiency of MKN45 and MKN74 cells. ASCs increased IL-6 secretion and enhanced the expression of Nanog and CD44v6 in GC cells; however, these changes were suppressed by the inhibition of IL-6. Xenograft mouse models co-inoculated with MKN45 cells and ASCs showed enhanced CD44v6 and Nanog expression and markedly reduced apoptosis induced by 5-FU treatment. Conclusion: This study improves our understanding of ASCs' role in PM treatment resistance and has demonstrated the potential for new treatment strategies targeting ASCs.
Collapse
Affiliation(s)
- Jun Kinoshita
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| | - Kenta Doden
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| | - Yusuke Sakimura
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| | - Saki Hayashi
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| | - Hiroto Saito
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| | - Toshikatsu Tsuji
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| | - Daisuke Yamamoto
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| | - Hideki Moriyama
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| | - Toshinari Minamoto
- Japan Community Health Care Organization Kanazawa Hospital, Kanazawa 920-8610, Japan;
- Department of Molecular and Cellular Pathology, Kanazawa University, Kanazawa 920-8640, Japan
| | - Noriyuki Inaki
- Department of Gastrointestinal Surgery, Kanazawa University, Kanazawa 920-8641, Japan; (K.D.); (Y.S.); (S.H.); (H.S.); (T.T.); (D.Y.); (H.M.); (N.I.)
| |
Collapse
|
5
|
Sonnentag SJ, Ibrahim NSM, Orian-Rousseau V. CD44: a stemness driver, regulator, and marker-all in one? Stem Cells 2024; 42:1031-1039. [PMID: 39364735 DOI: 10.1093/stmcls/sxae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/23/2024] [Indexed: 10/05/2024]
Abstract
Although the concept of cancer stem cells is still controversial, previous studies have shown that blood cancers, as well as specific types of solid cancers such as colorectal cancer, rely on stem cells during the onset of tumor growth and further tumor development. Moreover, resistance to therapeutic treatment in leukemias such as acute myeloid leukemia and in colorectal cancer can be attributed to a small population of cells with stemness properties known as minimal residual disease. In this review, we look back on the discovery of cancer stem cells and the contribution of the findings in blood cancer to a parallel discovery in solid cancers. We focus on CD44 as a stem cell marker, both in blood cancers and in several types of solid cancers, particularly of the gastrointestinal tract. This review highlights newly discovered molecular mechanisms of action of CD44 which indicate that CD44 has indeed a function in stemness, stem cell maintenance, and drug resistance. We attempt here to make the link between the functions of CD44 isoforms in stemness and their involvement in specific steps of tumor growth and metastasis.
Collapse
Affiliation(s)
- Steffen J Sonnentag
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems-Functional Molecular Systems, Kaiserstraße 12, 76131 Karlsruhe, Germany
| | - Nagwa S M Ibrahim
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems-Functional Molecular Systems, Kaiserstraße 12, 76131 Karlsruhe, Germany
| | - Veronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems-Functional Molecular Systems, Kaiserstraße 12, 76131 Karlsruhe, Germany
| |
Collapse
|
6
|
Gunjkar S, Gupta U, Nair R, Paul P, Aalhate M, Mahajan S, Maji I, Chourasia MK, Guru SK, Singh PK. The Neoteric Paradigm of Biomolecule-Functionalized Albumin-Based Targeted Cancer Therapeutics. AAPS PharmSciTech 2024; 25:265. [PMID: 39500822 DOI: 10.1208/s12249-024-02977-6] [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: 07/31/2024] [Accepted: 10/17/2024] [Indexed: 12/12/2024] Open
Abstract
Albumin is a nature-derived, versatile protein carrier, that has been explored extensively by researchers for anticancer drug delivery due to its role in enhancing drug stability, solubility, circulation time, targeting capabilities, and overall therapeutic efficacy. Albumin nanoparticles possess inherent biocompatibility, biodegradability, and passive tumor-targeting ability due to the enhanced permeability and retention effect. However, non-specific accumulation of cytotoxic agents in healthy tissues remains a challenge. In this paper, the functionalization of albumin nanoparticles using various biomolecules including antibodies, nucleic acids, proteins and peptides, vitamins, chondroitin sulfate, hyaluronic acid, and lactobionic acid have been discussed which enables specific recognition and binding to cancer cells. Furthermore, we highlight the supremacy of such a targeted approach in tumor-specific drug delivery, minimization of off-target effects, potential improvement in therapeutic efficacy, cellular internalization, reduced side effects, and better clinical outcomes. This review centers on how they have revolutionized the field of biomedical research and tuned into an excellent targeted approach. In conclusion, this review highlights in detail the role of albumin as a nanocarrier for tumor-targeted delivery using biomolecules as ligands.
Collapse
Affiliation(s)
- Swati Gunjkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Ujala Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Rahul Nair
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Priti Paul
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Indrani Maji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India.
| |
Collapse
|
7
|
Mehner LM, Munoz-Sagredo L, Sonnentag SJ, Treffert SM, Orian-Rousseau V. Targeting CD44 and other pleiotropic co-receptors as a means for broad inhibition of tumor growth and metastasis. Clin Exp Metastasis 2024; 41:599-611. [PMID: 38761292 PMCID: PMC11499327 DOI: 10.1007/s10585-024-10292-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/02/2024] [Indexed: 05/20/2024]
Abstract
Although progress has been made in the treatment of cancer, particularly for the four major types of cancers affecting the lungs, colon, breast and prostate, resistance to cancer treatment often emerges upon inhibition of major signaling pathways, which leads to the activation of additional pathways as a last-resort survival mechanism by the cancer cells. This signaling plasticity provides cancer cells with a level of operational freedom, reducing treatment efficacy. Plasticity is a characteristic of cancer cells that are not only able to switch signaling pathways but also from one cellular state (differentiated cells to stem cells or vice versa) to another. It seems implausible that the inhibition of one or a few signaling pathways of heterogeneous and plastic tumors can sustain a durable effect. We propose that inhibiting molecules with pleiotropic functions such as cell surface co-receptors can be a key to preventing therapy escape instead of targeting bona fide receptors. Therefore, we ask the question whether co-receptors often considered as "accessory molecules" are an overlooked key to control cancer cell behavior.
Collapse
Affiliation(s)
- Lisa-Marie Mehner
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Leonel Munoz-Sagredo
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
- School of Medicine, Universidad de Valparaiso, Valparaiso, Chile
| | - Steffen Joachim Sonnentag
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Sven Máté Treffert
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Véronique Orian-Rousseau
- Institute of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, Karlsruhe, Germany.
| |
Collapse
|
8
|
Purić E, Nilsson UJ, Anderluh M. Galectin-8 inhibition and functions in immune response and tumor biology. Med Res Rev 2024; 44:2236-2265. [PMID: 38613488 DOI: 10.1002/med.22041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 03/03/2024] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Galectins are among organisms' most abundantly expressed lectins (carbohydrate-binding proteins) that specifically bind β-galactosides. They act not only outside the cell, where they bind to extracellular matrix glycans, but also inside the cell, where they have a significant impact on signaling pathways. Galectin-8 is a galectin family protein encoded by the LGALS8 gene. Its role is evident in both T- and B-cell immunity and in the innate immune response, where it acts directly on dendritic cells and induces some pro-inflammatory cytokines. Galectin-8 also plays an important role in the defense against bacterial and viral infections. It is known to promote antibacterial autophagy by recognizing and binding glycans present on the vacuolar membrane, thus acting as a danger receptor. The most important role of galectin-8 is the regulation of cancer growth, metastasis, tumor progression, and tumor cell survival. Importantly, the expression of galectins is typically higher in tumor tissues than in noncancerous tissues. In this review article, we focus on galectin-8 and its function in immune response, microbial infections, and cancer. Given all of these functions of galectin-8, we emphasize the importance of developing new and selective galectin-8 inhibitors and report the current status of their development.
Collapse
Affiliation(s)
- Edvin Purić
- Department of Pharmaceutical Chemistry, University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Ulf J Nilsson
- Department of Chemistry, Lund University, Lund, Sweden
| | - Marko Anderluh
- Department of Pharmaceutical Chemistry, University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| |
Collapse
|
9
|
Sanwick AM, Chaple IF. Targeted radionuclide therapy for head and neck squamous cell carcinoma: a review. Front Oncol 2024; 14:1445191. [PMID: 39239273 PMCID: PMC11374632 DOI: 10.3389/fonc.2024.1445191] [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: 06/06/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a type of head and neck cancer that is aggressive, difficult to treat, and often associated with poor prognosis. HNSCC is the sixth most common cancer worldwide, highlighting the need to develop novel treatments for this disease. The current standard of care for HNSCC usually involves a combination of surgical resection, radiation therapy, and chemotherapy. Chemotherapy is notorious for its detrimental side effects including nausea, fatigue, hair loss, and more. Radiation therapy can be a challenge due to the anatomy of the head and neck area and presence of normal tissues. In addition to the drawbacks of chemotherapy and radiation therapy, high morbidity and mortality rates for HNSCC highlight the urgent need for alternative treatment options. Immunotherapy has recently emerged as a possible treatment option for cancers including HNSCC, in which monoclonal antibodies are used to help the immune system fight disease. Combining monoclonal antibodies approved by the US Food and Drug Administration, such as cetuximab and pembrolizumab, with radiotherapy or platinum-based chemotherapy for patients with locally advanced, recurrent, or metastatic HNSCC is an accepted first-line therapy. Targeted radionuclide therapy can potentially be used in conjunction with the first-line therapy, or as an additional treatment option, to improve patient outcomes and quality of life. Epidermal growth factor receptor is a known molecular target for HNSCC; however, other targets such as human epidermal growth factor receptor 2, human epidermal growth factor receptor 3, programmed cell death protein 1, and programmed death-ligand 1 are emerging molecular targets for the diagnosis and treatment of HNSCC. To develop successful radiopharmaceuticals, it is imperative to first understand the molecular biology of the disease of interest. For cancer, this understanding often means detection and characterization of molecular targets, such as cell surface receptors, that can be used as sensitive targeting agents. The goal of this review article is to explore molecular targets for HNSCC and dissect previously conducted research in nuclear medicine and provide a possible path forward for the development of novel radiopharmaceuticals used in targeted radionuclide therapy for HNSCC, which has been underexplored to date.
Collapse
Affiliation(s)
- Alexis M Sanwick
- Department of Nuclear Engineering, University of Tennessee, Knoxville, TN, United States
| | - Ivis F Chaple
- Department of Nuclear Engineering, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
10
|
Vasilevska J, Cheng PF, Lehmann J, Ramelyte E, Gómez JM, Dimitriou F, Sella F, Ferretti D, Salas-Bastos A, Jordaan WS, Levesque MP, Dummer R, Sommer L. Monitoring melanoma patients on treatment reveals a distinct macrophage population driving targeted therapy resistance. Cell Rep Med 2024; 5:101611. [PMID: 38942020 PMCID: PMC11293307 DOI: 10.1016/j.xcrm.2024.101611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/09/2024] [Accepted: 05/20/2024] [Indexed: 06/30/2024]
Abstract
Resistance to targeted therapy remains a major clinical challenge in melanoma. To uncover resistance mechanisms, we perform single-cell RNA sequencing on fine-needle aspirates from resistant and responding tumors of patients undergoing BRAFi/MEKi treatment. Among the genes most prominently expressed in resistant tumors is POSTN, predicted to signal to a macrophage population associated with targeted therapy resistance (TTR). Accordingly, tumors from patients with fast disease progression after therapy exhibit high POSTN expression levels and high numbers of TTR macrophages. POSTN polarizes human macrophages toward a TTR phenotype and promotes resistance to targeted therapy in a melanoma mouse model, which is associated with a phenotype change in intratumoral macrophages. Finally, polarized TTR macrophages directly protect human melanoma cells from MEKi-induced killing via CD44 receptor expression on melanoma cells. Thus, interfering with the protective activity of TTR macrophages may offer a strategy to overcome resistance to targeted therapy in melanoma.
Collapse
Affiliation(s)
- Jelena Vasilevska
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland
| | - Phil Fang Cheng
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Julia Lehmann
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Julia Martínez Gómez
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Florentia Dimitriou
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Federica Sella
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Daria Ferretti
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland
| | | | | | - Mitchell Paul Levesque
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Lukas Sommer
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
11
|
Turkekul K, Erdogan S. Potent Suppression of Prostate Cancer Cell Growth and Eradication of Cancer Stem Cells by CD44-targeted Nanoliposome-quercetin Nanoparticles. J Cancer Prev 2023; 28:160-174. [PMID: 38205358 PMCID: PMC10774486 DOI: 10.15430/jcp.2023.28.4.160] [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: 09/22/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 01/12/2024] Open
Abstract
The bioavailability of quercetin, a natural compound, is hindered by low solubility, limited absorption, and restricted systemic availability. Therefore, encapsulating it in biocompatible nanoparticles presents a promising solution. This study aimed to target prostate cancer stem cells (CSCs) overexpressing CD44+ receptors as well as cancer cells, employing quercetin-loaded hyaluronic acid-modified nanoliposomes (LP-Quer-HA). Synthesized via a green ethanol injection method, these nanoliposomes had an average diameter of 134 nm and an impressive loading efficiency of 96.9%. Human prostate cancer cells were treated with either 10 μM of free quercetin or the same concentration delivered by LP-Quer-HA for 72 hours. Free quercetin reduced androgen-resistant PC3 cell viability by 16%, while LP-Quer-HA significantly increased cell death to 60%. It induced apoptosis, upregulating cytochrome c, Bax, caspases 3 and 8, and downregulating survivin and Bcl-2 expression. Compared to free quercetin, LP-Quer-HA upregulated E-cadherin expression while inhibiting cell migration and reducing the expression of fibronectin, N-cadherin, and MMP9. Treatment of PC3 cell tumor spheroids with LP-Quer-HA decreased the number of CD44 cells and expression of CD44, Oct3/4 and Wnt. Moreover, LP-Quer-HA inhibited p-ERK expression while increasing p38/MAPK and NF-κB protein expression. In androgen-sensitive LNCaP cells, LP-Quer-HA efficacy was notable, reducing cell viability from 10% to 52% compared to free quercetin. Utilizing HA-modified nanoliposomes as a quercetin delivery system enhanced its potency at lower concentrations, reducing the CD44+ cell population and effectively impeding prostate cancer cell proliferation and migration. These findings underscore the potential of quercetin-loaded cationic nanoliposomes as a robust therapeutic approach.
Collapse
Affiliation(s)
- Kader Turkekul
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Turkey
| | - Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Balkan Campus, Edirne, Turkey
| |
Collapse
|
12
|
Mortensen ACL, Berglund H, Segerström L, Walle M, Hofström C, Persson H, Nygren PÅ, Nilvebrant J, Frejd FY, Nestor M. Selection, characterization and in vivo evaluation of novel CD44v6-targeting antibodies for targeted molecular radiotherapy. Sci Rep 2023; 13:20648. [PMID: 38001360 PMCID: PMC10673843 DOI: 10.1038/s41598-023-47891-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/20/2023] [Indexed: 11/26/2023] Open
Abstract
Molecular radiotherapy combines the advantages of systemic administration of highly specific antibodies or peptides and the localized potency of ionizing radiation. A potential target for molecular radiotherapy is the cell surface antigen CD44v6, which is overexpressed in numerous cancers, with limited expression in normal tissues. The aim of the present study was to generate and characterize a panel of human anti-CD44v6 antibodies and identify a suitable candidate for future use in molecular radiotherapy of CD44v6-expressing cancers. Binders were first isolated from large synthetic phage display libraries containing human scFv and Fab antibody fragments. The antibodies were extensively analyzed through in vitro investigations of binding kinetics, affinity, off-target binding, and cell binding. Lead candidates were further subjected to in vivo biodistribution studies in mice bearing anaplastic thyroid cancer xenografts that express high levels of CD44v6. Additionally, antigen-dependent tumor uptake of the lead candidate was verified in additional xenograft models with varying levels of target expression. Interestingly, although only small differences were observed among the top antibody candidates in vitro, significant differences in tumor uptake and retention were uncovered in in vivo experiments. A high-affinity anti-CD44v6 lead drug candidate was identified, mAb UU-40, which exhibited favorable target binding properties and in vivo distribution. In conclusion, a panel of human anti-CD44v6 antibodies was successfully generated and characterized in this study. Through comprehensive evaluation, mAb UU-40 was identified as a promising lead candidate for future molecular radiotherapy of CD44v6-expressing cancers due to its high affinity, excellent target binding properties, and desirable in vivo distribution characteristics.
Collapse
Affiliation(s)
- A C L Mortensen
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden.
| | - H Berglund
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| | - L Segerström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| | - M Walle
- Drug Discovery and Development Platform, Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - C Hofström
- Drug Discovery and Development Platform, Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - H Persson
- Drug Discovery and Development Platform, Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - P-Å Nygren
- Drug Discovery and Development Platform, Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - J Nilvebrant
- Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - F Y Frejd
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| | - M Nestor
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| |
Collapse
|
13
|
Agnihotri TG, Salave S, Shinde T, Srikanth I, Gyanani V, Haley JC, Jain A. Understanding the role of endothelial cells in brain tumor formation and metastasis: a proposition to be explored for better therapy. JOURNAL OF THE NATIONAL CANCER CENTER 2023; 3:222-235. [PMID: 39035200 PMCID: PMC11256543 DOI: 10.1016/j.jncc.2023.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 07/23/2024] Open
Abstract
Glioblastoma is one of the most devastating central nervous system disorders. Being a highly vascular brain tumor, it is distinguished by aberrant vessel architecture. This lends credence to the idea that endothelial cells (ECs) linked with glioblastoma vary fundamentally from ECs seen in the healthy human brain. To effectively design an antiangiogenic treatment, it is crucial to identify the functional and phenotypic characteristics of tumor-associated ECs. The ECs associated with glioblastoma are less prone to apoptosis than control cells and are resistant to cytotoxic treatments. Additionally, ECs associated with glioblastoma migrate more quickly than control ECs and naturally produce large amounts of growth factors such as endothelin-1, interleukin-8, and vascular endothelial growth factor (VEGF). For designing innovative antiangiogenic drugs that particularly target tumor-related ECs in gliomas, it is critical to comprehend these distinctive features of ECs associated with gliomas. This review discusses the process of angiogenesis, other factors involved in the genesis of tumors, and the possibility of ECs as a potential target in combating glioblastoma. It also sheds light on the association of tumor microenvironment and ECs with immunotherapy. This review, thus gives us the hope that neuro endothelial targeting with growth factors and angiogenesis regulators combined with gene therapy would open up new doorways and change our traditional perspective of treating cancer.
Collapse
Affiliation(s)
- Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| | - Sagar Salave
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| | - Tanuja Shinde
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| | - Induri Srikanth
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| | - Vijay Gyanani
- Long Acting Drug Delivery, Celanese Corporation, Irving, United States
| | - Jeffrey C. Haley
- Long Acting Drug Delivery, Celanese Corporation, Irving, United States
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, India
| |
Collapse
|
14
|
Bashkeran T, Kamaruddin AH, Ngo TX, Suda K, Umakoshi H, Watanabe N, Nadzir MM. Niosomes in cancer treatment: A focus on curcumin encapsulation. Heliyon 2023; 9:e18710. [PMID: 37593605 PMCID: PMC10428065 DOI: 10.1016/j.heliyon.2023.e18710] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/19/2023] Open
Abstract
Curcumin is widely used as a therapeutic drug for cancer treatment. However, its limited absorption and rapid excretion are the major therapeutic limitations to its clinical use. Using niosomes as a curcumin delivery system is a cheap, easy, and less toxic strategy for enhancing the absorption of curcumin by cells and delaying its excretion. Thus, there is a vital need to explore curcumin niosomes to configure the curcumin to suitably serve and aid current pharmacokinetics in treatments for cancer. To date, no comprehensive review has focused on the cytotoxic effects of curcumin niosomes on malignant cells. Thus, this review provides a critical analysis of the curcumin niosomes in cancer treatment, formulations of curcumin niosomes, characterizations of curcumin niosomes, and factors influencing their performance. The findings from this review article can strongly accelerate the understanding of curcumin niosomes and pave a brighter direction towards advances in the pharmaceutical, biotechnology, and medical industries.
Collapse
Affiliation(s)
- Thaaranni Bashkeran
- School of Chemical Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| | - Azlina Harun Kamaruddin
- School of Chemical Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| | - Trung Xuan Ngo
- Rohto Pharmaceutical Co., Ltd., Basic Research Division, Research Village Kyoto, 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| | - Kazuma Suda
- Rohto Pharmaceutical Co., Ltd., Basic Research Division, Research Village Kyoto, 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| | - Hiroshi Umakoshi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, 560-8531, Japan
| | - Nozomi Watanabe
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, 560-8531, Japan
| | - Masrina Mohd Nadzir
- School of Chemical Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| |
Collapse
|
15
|
Liu B, Song A, Gui P, Wang J, Pan Y, Li C, Li S, Zhang Y, Jiang T, Xu Y, Pei D, Song J. Long noncoding RNA LINC01594 inhibits the CELF6-mediated splicing of oncogenic CD44 variants to promote colorectal cancer metastasis. Cell Death Dis 2023; 14:427. [PMID: 37452042 PMCID: PMC10349055 DOI: 10.1038/s41419-023-05924-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 06/16/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023]
Abstract
Long noncoding RNAs (lncRNAs) play critical roles in tumorigenesis and tumor metastasis. However, the underlying mechanisms of lncRNAs in colorectal cancer (CRC) need further exploration. By using data from The Cancer Genome Atlas (TCGA) and GEO databases, we identified a novel CRC-related lncRNA, LINC01594, that is significantly upregulated in CRC and associated with poor prognosis. In vitro and in vivo, gain- and loss-of-function experiments demonstrated that LINC01594 promotes metastasis in CRC. LINC01594 functions as a DNMT1 scaffold, increasing the level of CELF6 promoter methylation. LINC01594 also competitively binds the transcription factor p53, decreasing CELF6 expression. This inhibited the exon skipping of CD44 V4-V7 induced by CELF6. In summary, this study highlights a novel CRC biomarker and therapeutic target, LINC01594, and the findings suggest that the LINC01594-CELF6-CD44 axis might serve as a biomarker and therapeutic target in CRC.
Collapse
Affiliation(s)
- Bowen Liu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University. No. 99, Huaihai West Road, Quanshan District, Xuzhou, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China
| | - Angxi Song
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China
| | - Pengkun Gui
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China
| | - Jin Wang
- Department of Pathology, Xuzhou Medical University. No. 209, Tongshan Road, Yunlong District, Xuzhou, 221004, China
| | - Yaojie Pan
- Department of Medical Oncology, Zhejiang Provincial People's Hospital. No. 158, Shangtang Road, Xiacheng District, Zhejiang, 310000, China
| | - Chao Li
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University. No. 99, Huaihai West Road, Quanshan District, Xuzhou, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China
| | - Shuai Li
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University. No. 99, Huaihai West Road, Quanshan District, Xuzhou, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China
| | - Yi Zhang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University. No. 99, Huaihai West Road, Quanshan District, Xuzhou, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University. No. 99, Huaihai West Road, Quanshan District, Xuzhou, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University. No. 99, Huaihai West Road, Quanshan District, Xuzhou, 221006, China
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China
| | - Dongsheng Pei
- Department of Pathology, Xuzhou Medical University. No. 209, Tongshan Road, Yunlong District, Xuzhou, 221004, China.
| | - Jun Song
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University. No. 99, Huaihai West Road, Quanshan District, Xuzhou, 221006, China.
- Institute of Digestive Diseases, Xuzhou Medical University. No. 84, Huaihai West Road, Quanshan District, Xuzhou, 221002, China.
| |
Collapse
|
16
|
Kuo YC, Kou HW, Hsu CP, Lo CH, Hwang TL. Identification and Clinical Significance of Pancreatic Cancer Stem Cells and Their Chemotherapeutic Drug Resistance. Int J Mol Sci 2023; 24:ijms24087331. [PMID: 37108495 PMCID: PMC10138402 DOI: 10.3390/ijms24087331] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Pancreatic cancer ranks in the 10th-11th position among cancers affecting men in Taiwan, besides being a rather difficult-to-treat disease. The overall 5-year survival rate of pancreatic cancer is only 5-10%, while that of resectable pancreatic cancer is still approximately 15-20%. Cancer stem cells possess intrinsic detoxifying mechanisms that allow them to survive against conventional therapy by developing multidrug resistance. This study was conducted to investigate how to overcome chemoresistance and its mechanisms in pancreatic cancer stem cells (CSCs) using gemcitabine-resistant pancreatic cancer cell lines. Pancreatic CSCs were identified from human pancreatic cancer lines. To determine whether CSCs possess a chemoresistant phenotype, the sensitivity of unselected tumor cells, sorted CSCs, and tumor spheroid cells to fluorouracil (5-FU), gemcitabine (GEM), and cisplatin was analyzed under stem cell conditions or differentiating conditions. Although the mechanisms underlying multidrug resistance in CSCs are poorly understood, ABC transporters such as ABCG2, ABCB1, and ABCC1 are believed to be responsible. Therefore, we measured the mRNA expression levels of ABCG2, ABCB1, and ABCC1 by real-time RT-PCR. Our results showed that no significant differences were found in the effects of different concentrations of gemcitabine on CSCs CD44+/EpCAM+ of various PDAC cell line cultures (BxPC-3, Capan-1, and PANC-1). There was also no difference between CSCs and non-CSCs. Gemcitabine-resistant cells exhibited distinct morphological changes, including a spindle-shaped morphology, the appearance of pseudopodia, and reduced adhesion characteristics of transformed fibroblasts. These cells were found to be more invasive and migratory, and showed increased vimentin expression and decreased E-cadherin expression. Immunofluorescence and immunoblotting experiments demonstrated increased nuclear localization of total β-catenin. These alterations are hallmarks of epithelial-to-mesenchymal transition (EMT). Resistant cells showed activation of the receptor protein tyrosine kinase c-Met and increased expression of the stem cell marker cluster of differentiation (CD) 24, CD44, and epithelial specific antigen (ESA). We concluded that the expression of the ABCG2 transporter protein was significantly higher in CD44+ and EpCAM+ CSCs of PDAC cell lines. Cancer stem-like cells exhibited chemoresistance. Gemcitabine-resistant pancreatic tumor cells were associated with EMT, a more aggressive and invasive phenotype of numerous solid tumors. Increased phosphorylation of c-Met may also be related to chemoresistance, and EMT and could be used as an attractive adjunctive chemotherapeutic target in pancreatic cancer.
Collapse
Affiliation(s)
- Yu-Chi Kuo
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Hao-Wei Kou
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Chih-Po Hsu
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Chih-Hong Lo
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| | - Tsann-Long Hwang
- Department of Surgery, Chang Gung Memorial Hospital, Chang Gung University, Lin-Kou, Taoyuan 333, Taiwan
| |
Collapse
|
17
|
Gheybi E, Asoodeh A, Amani J. Preparation of chitosan nanoparticle containing recombinant CD44v antigen and evaluation of its immunization capacity against breast cancer in BALB/c mice. BMC Cancer 2023; 23:134. [PMID: 36759786 PMCID: PMC9912563 DOI: 10.1186/s12885-023-10614-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE(S) Breast tumors show heterogeneity containing cancer stem cells as a small subpopulation of a tumor mass. CD44 as a cancer stem cells antigen is abnormally expressed by carcinomas of epithelial origin. Also, overexpression of CD44 variable isoforms (CD44v) is associated with malignancy in breast cancer. In the present research, our objective was to evaluate the immunogenicity of prepared nanoparticles containing a novel recombinant CD44v (rCD44v) protein in the mouse model. MATERIALS AND METHODS CD44 gene was expressed in E. coli BL21 DE3 using the pET28a-CD44 vector. The expressed rCD44v protein was purified, encapsulated into the chitosan nanoparticles, and administered to BALB/c mice. ELISA was used to evaluate the immunoglobulin levels of immunized animals. For challenge experiment, 2 × 106 4T1-CD44 tumor cells were injected subcutaneously in mice, and tumor size, necrosis, and metastases were measured. Finally, cell proliferation assay, cytokines assay, and neutralization assay of the mouse anti-rCD44v on the human breast cancer cell line were examined. RESULTS The measured size of chitosan-rCD44v nanoparticles was 146.5 nm. Recombinant CD44v encapsulated by chitosan nanoparticles increases immunological responses via the adjuvant nature of chitosan nanoparticles. In the immunized mice, IgG and IgA titers were significantly increased. Tumor growth in injection and nano-injection test groups compared with the mice control groups displayed a significant reduction (P < 0.05). A high amount of splenocytes secreting IFNγ and IL-17 was seen in immunized mice with rCD44v (P < 0.05). Furthermore, a smaller size of lung metastases compared to the control mice groups was detected. CONCLUSION The encapsulated rCD44v within the chitosan nanoparticles induced a significant immune response in mice and can establish significant protection against breast cancer. Therefore, it can be considered a vaccine candidate for breast cancer therapeutic modalities.
Collapse
Affiliation(s)
- Elaheh Gheybi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Cellular and Molecular Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
- Cellular and Molecular Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Zhang H, Cao H, Luo H, Zhang N, Wang Z, Dai Z, Wu W, Liu G, Xie Z, Cheng Q, Cheng Y. RUNX1/CD44 axis regulates the proliferation, migration, and immunotherapy of gliomas: A single-cell sequencing analysis. Front Immunol 2023; 14:1086280. [PMID: 36776876 PMCID: PMC9909339 DOI: 10.3389/fimmu.2023.1086280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Glioma is one of the most common, primary, and lethal adult brain tumors because of its extreme aggressiveness and poor prognosis. Several recent studies relevant to the immune function of CD44, a transmembrane glycoprotein as a significant hyaluronic acid receptor, have achieved great success, revealing the critical role of CD44 in immune infiltration in gliomas. The overexpression of CD44 has been verified to correlate with cancer aggressiveness and migration, while the clinical and immune features of CD44 expression have not yet been thoroughly characterized in gliomas. METHODS Molecular and clinical data of glioma collected from publicly available genomic databases were analyzed. RESULTS CD44 was up-expressed in malignant gliomas, notably in the 1p/19q non-codeletion cases, isocitrate dehydrogenase (IDH) wild-type, and mesenchymal subtypes in GBM samples. CD44 expression level strongly correlates with stromal and immune cells, mainly infiltrating the glioma microenvironment by single-cell sequencing analysis. Meanwhile, CD44 can be a promising biomarker in predicting immunotherapy responses and mediating the expression of PD-L1. Finally, RUNX1/CD44 axis could promote the proliferation and migration of gliomas. CONCLUSIONS Therefore, CD44 was responsible for glioma growth and progression. It could potentially lead to a novel target for glioma immunotherapy or a prognostic biomarker.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Psychiatry, Brain Hospital of Hunan Province, The Second People’s Hospital of Hunan Province, Changsha, China
- The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Hong Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wantao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zongyi Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Caracciolo D, Mancuso A, Polerà N, Froio C, D'Aquino G, Riillo C, Tagliaferri P, Tassone P. The emerging scenario of immunotherapy for T-cell Acute Lymphoblastic Leukemia: advances, challenges and future perspectives. Exp Hematol Oncol 2023; 12:5. [PMID: 36624522 PMCID: PMC9828428 DOI: 10.1186/s40164-022-00368-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a challenging pediatric and adult haematologic disease still associated with an unsatisfactory cure rate. Unlike B-ALL, the availability of novel therapeutic options to definitively improve the life expectancy for relapsed/resistant patients is poor. Indeed, the shared expression of surface targets among normal and neoplastic T-cells still limits the efficacy and may induce fratricide effects, hampering the use of innovative immunotherapeutic strategies. However, novel monoclonal antibodies, bispecific T-cell engagers (BTCEs), and chimeric antigen receptors (CAR) T-cells recently showed encouraging results and some of them are in an advanced stage of pre-clinical development or are currently under investigation in clinical trials. Here, we review this exciting scenario focusing on most relevant advances, challenges, and perspectives of the emerging landscape of immunotherapy of T-cell malignancies.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Caterina Froio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Giuseppe D'Aquino
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | | | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Epithelial-Mesenchymal Transition Induced in Cancer Cells by Adhesion to Type I Collagen. Int J Mol Sci 2022; 24:ijms24010198. [PMID: 36613638 PMCID: PMC9820580 DOI: 10.3390/ijms24010198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is an important biological process that is physiologically observed during development, wound healing, and cancer invasion. During EMT induction, cancer cells lose their epithelial properties owing to various tumor microenvironmental factors and begin to exhibit mesenchymal properties, such as loss of apical-basal polarity, weakened intercellular adhesion, and promotion of single cell migration. Several factors, including growth factor stimulation and adhesion to type I collagen (Col-I), induce EMT in cancer cells. Cells adhere to Col-I via specific receptors and induce EMT by activating outside-in signals. In vivo, Col-I molecules often form fibrils, which then assemble into supramolecular structures (gel form). Col-I also self-assembles in vitro under physiological conditions. Notably, Col-I can be used as a culture substrate in both gel and non-gel forms, and the gel formation state of Col-I affects cell fate. Although EMT can be induced in both forms of Col-I, the effects of gel formation on EMT induction remain unclear and somewhat inconsistent. Therefore, this study reviews the relationship between Col-I gel-forming states and EMT induction in cancer cells.
Collapse
|
21
|
Hamdy NM, Eskander G, Basalious EB. Insights on the Dynamic Innovative Tumor Targeted-Nanoparticles-Based Drug Delivery Systems Activation Techniques. Int J Nanomedicine 2022; 17:6131-6155. [PMID: 36514378 PMCID: PMC9741821 DOI: 10.2147/ijn.s386037] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer conventional chemotherapeutic drugs novel formula progress, nowadays, uses nano technology for targeted drug delivery, specifically tailored to overcome therapeutic agents' delivery challenges. Polymer drug delivery systems (DDS) play a crucial role in minimizing off-target side effects arising when using standard cytotoxic drugs. Using nano-formula for targeted localized action, permits using larger effective cytotoxic doses on a single special spot, that can seriously cause harm if it was administered systemically. Therefore, various nanoparticles (NPs) specifically have attached groups for targeting capabilities, not seen in bulk materials, which then need activation. In this review, we will present a simple innovative, illustrative, in a cartoon-way, enumeration of NP anti-cancer drug targeting delivery system activation-types. Area(s) covered in this review are the mechanisms of various NP activation techniques.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Georgette Eskander
- Faculty of Pharmacy, Ain Shams University, Postgraduate Student, Cairo, Egypt
| | - Emad B Basalious
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
22
|
Xie X, Li Y, Lian S, Lu Y, Jia L. Cancer metastasis chemoprevention prevents circulating tumour cells from germination. Signal Transduct Target Ther 2022; 7:341. [PMID: 36184654 PMCID: PMC9526788 DOI: 10.1038/s41392-022-01174-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/19/2022] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
The war against cancer traces back to the signature event half-a-century ago when the US National Cancer Act was signed into law. The cancer crusade costs trillions with disappointing returns, teasing the possibility of a new breakthrough. Cure for cancer post-metastases still seems tantalisingly out of reach. Once metastasized, cancer-related death is extremely difficult, if not impossible, to be reversed. Here we present cancer pre-metastasis chemoprevention strategy that can prevent circulating tumour cells (CTCs) from initiating metastases safely and effectively, and is disparate from the traditional cancer chemotherapy and cancer chemoprevention. Deep learning of the biology of CTCs and their disseminating organotropism, complexity of their adhesion to endothelial niche reveals that if the adhesion of CTCs to their metastasis niche (the first and the most important part in cancer metastatic cascade) can be pharmaceutically interrupted, the lethal metastatic cascade could be prevented from getting initiated. We analyse the key inflammatory and adhesive factors contributing to CTC adhesion/germination, provide pharmacological fundamentals for abortifacients to intervene CTC adhesion to the distant metastasis sites. The adhesion/inhibition ratio (AIR) is defined for selecting the best cancer metastasis chemopreventive candidates. The successful development of such new therapeutic modalities for cancer metastasis chemoprevention has great potential to revolutionise the current ineffective post-metastasis treatments.
Collapse
Affiliation(s)
- Xiaodong Xie
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Yumei Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Shu Lian
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Yusheng Lu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Lee Jia
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China. .,Cancer Metastasis Alert and Prevention Center, College of Chemistry, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University, Fuzhou, Fujian, 350116, China.
| |
Collapse
|
23
|
Li D, Li X, Zhao J, Tan F. Advances in nuclear medicine-based molecular imaging in head and neck squamous cell carcinoma. J Transl Med 2022; 20:358. [PMID: 35962347 PMCID: PMC9373390 DOI: 10.1186/s12967-022-03559-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/25/2022] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are often aggressive, making advanced disease very difficult to treat using contemporary modalities, such as surgery, radiation therapy, and chemotherapy. However, targeted therapy, e.g., cetuximab, an epidermal growth factor receptor inhibitor, has demonstrated survival benefit in HNSCC patients with locoregional failure or distant metastasis. Molecular imaging aims at various biomarkers used in targeted therapy, and nuclear medicine-based molecular imaging is a real-time and non-invasive modality with the potential to identify tumor in an earlier and more treatable stage, before anatomic-based imaging reveals diseases. The objective of this comprehensive review is to summarize recent advances in nuclear medicine-based molecular imaging for HNSCC focusing on several commonly radiolabeled biomarkers. The preclinical and clinical applications of these candidate imaging strategies are divided into three categories: those targeting tumor cells, tumor microenvironment, and tumor angiogenesis. This review endeavors to expand the knowledge of molecular biology of HNSCC and help realizing diagnostic potential of molecular imaging in clinical nuclear medicine.
Collapse
Affiliation(s)
- Danni Li
- Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
| | - Xuran Li
- Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fei Tan
- Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China. .,The Royal College of Surgeons in Ireland, Dublin, Ireland. .,The Royal College of Surgeons of England, London, UK.
| |
Collapse
|
24
|
CD44 Contributes to the Regulation of MDR1 Protein and Doxorubicin Chemoresistance in Osteosarcoma. Int J Mol Sci 2022; 23:ijms23158616. [PMID: 35955749 PMCID: PMC9368984 DOI: 10.3390/ijms23158616] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma is the most common type of pediatric bone tumor. Despite great advances in chemotherapy during the past decades, the survival rates of osteosarcoma patients remain unsatisfactory. Drug resistance is one of the main reasons, leading to treatment failure and poor prognosis. Previous reports correlated expression of cluster of differentiation 44 (CD44) with drug resistance and poor survival of osteosarcoma patients, however the underlying mechanisms are poorly defined. Here, we investigated the role of CD44 in the regulation of drug chemoresistance, using osteosarcoma cells isolated from mice carrying a mutation of the tumor suppressor neurofibromatosis type 2 (Nf2) gene. CD44 expression was knocked-down in the cells using CRISPR/Cas9 approach. Subsequently, CD44 isoforms and mutants were re-introduced to investigate CD44-dependent processes. Sensitivity to doxorubicin was analyzed in the osteosarcoma cells with modified CD44 expression by immunoblot, colony formation- and WST-1 assay. To dissect the molecular alterations induced by deletion of Cd44, RNA sequencing was performed on Cd44-positive and Cd44-negative primary osteosarcoma tissues isolated from Nf2-mutant mice. Subsequently, expression of candidate genes was evaluated by quantitative reverse transcription PCR (qRT-PCR). Our results indicate that CD44 increases the resistance of osteosarcoma cells to doxorubicin by up-regulating the levels of multidrug resistance (MDR) 1 protein expression, and suggest the role of proteolytically released CD44 intracellular domain, and hyaluronan interactions in this process. Moreover, high throughput sequencing analysis identified differential regulation of several apoptosis-related genes in Cd44-positive and -negative primary osteosarcomas, including p53 apoptosis effector related to PMP-22 (Perp). Deletion of Cd44 in osteosarcoma cells led to doxorubicin-dependent p53 activation and a profound increase in Perp mRNA expression. Overall, our results suggest that CD44 might be an important regulator of drug resistance and suggest that targeting CD44 can sensitize osteosarcoma to standard chemotherapy.
Collapse
|
25
|
Ghatak S, Hascall VC, Karamanos N, Markwald RR, Misra S. Interplay Between Chemotherapy-Activated Cancer Associated Fibroblasts and Cancer Initiating Cells Expressing CD44v6 Promotes Colon Cancer Resistance. Front Oncol 2022; 12:906415. [PMID: 35982950 PMCID: PMC9380598 DOI: 10.3389/fonc.2022.906415] [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: 03/28/2022] [Accepted: 06/22/2022] [Indexed: 11/14/2022] Open
Abstract
Cancer-initiating cells (CICs) drive colorectal tumor growth by their supportive niches where CICs interact with multiple cell types within the microenvironment, including cancer-associated fibroblasts (CAFs). We investigated the interplay between the CICs and the clinically relevant chemotherapeutic FOLFOX that creates the persistent tumorigenic properties of colorectal CICs, and stimulates the microenvironmental factors derived from the CAFs. We found that the CICs expressing an immunophenotype (CD44v6[+]) promote FOLFOX-resistance and that the CIC-immunophenotype was enhanced by factors secreted by CAFs after FOLFOX treatment These secreted factors included periostin, IL17A and WNT3A, which induced CD44v6 expression by activating WNT3A/β-catenin signaling. Blocking the interaction between CICs with any of these CAF-derived factors through tissue-specific conditional silencing of CD44v6 significantly reduced colorectal tumorigenic potential. To achieve this, we generated two unique vectors (floxed-pSico-CD44v6 shRNA plus Fabpl-Cre) that were encapsulated into transferrin coated PEG-PEI/(nanoparticles), which when introduced in vivo reduced tumor growth more effectively than using CD44v6-blocking antibodies. Notably, this tissue-specific conditional silencing of CD44v6 resulted in long lasting effects on self-renewal and tumor growth associated with a positive feedback loop linking WNT3A signaling and alternative-splicing of CD44. These findings have crucial clinical implications suggesting that therapeutic approaches for modulating tumor growth that currently focus on cell-autonomous mechanisms may be too limited and need to be broadened to include mechanisms that recognize the interplay between the stromal factors and the subsequent CIC-immunophenotype enrichment. Thus, more specific therapeutic approaches may be required to block a chemotherapy induced remodeling of a microenvironment that acts as a paracrine regulator to enrich CD44v6 (+) in colorectal CICs.
Collapse
Affiliation(s)
- Shibnath Ghatak
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department Natural Sciences, Trident Technical College, North Charleston, SC, United States
| | - Vincent C. Hascall
- Department of Biomedical Engineering/ND20, Cleveland Clinic, Cleveland, OH, United States
| | - Nikos Karamanos
- Department of Chemistry, University of Patras, Matrix Pathobiology Research Group, Patras, Greece
| | - Roger R. Markwald
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
| | - Suniti Misra
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Department Natural Sciences, Trident Technical College, North Charleston, SC, United States
| |
Collapse
|
26
|
Responses to the Tepotinib in Gastric Cancers with MET Amplification or MET Exon 14 Skipping Mutations and High Expression of Both PD-L1 and CD44. Cancers (Basel) 2022; 14:cancers14143444. [PMID: 35884507 PMCID: PMC9318186 DOI: 10.3390/cancers14143444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/30/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
Both MET exon 14 skipping mutation (METex14SM) and high copy-number variation (CNV) lead to enhanced carcinogenesis; additionally, programmed-death ligand 1 (PD-L1) is often upregulated in cancers. In this study, we characterized the expression of MET (including METex14SM), PD-L1, and CD44 in human gastric cancer (GC) cells as well as the differential susceptibility of these cells to tepotinib. Tepotinib treatments inhibited the growth of five GC cells in a dose-dependent manner with a concomitant induction of cell death. Tepotinib treatments also significantly reduced the expression of phospho-MET, total MET, c-Myc, VEGFR2, and Snail protein in SNU620, MKN45, and Hs746T cells. Notably, tepotinib significantly reduced the expression of CD44 and PD-L1 in METex14SM Hs746T cells. By contrast, tepotinib was only slightly active against SNU638 and KATO III cells. Migration was reduced to a greater extent in the tepotinib-treated group than in the control group. Tepotinib may have therapeutic effects on c-MET-amplified GC, a high expression of both PD-L1 and CD44, and METex14SM. Clinical studies are needed to confirm these therapeutic effects.
Collapse
|
27
|
Wang J, Xie D, Wu H, Li Y, Wan C. Ferroptosis-related local immune cytolytic activity in tumor microenvironment of basal cell and squamous cell carcinoma. Aging (Albany NY) 2022; 14:3956-3972. [PMID: 35501667 PMCID: PMC9134950 DOI: 10.18632/aging.204057] [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: 11/01/2021] [Accepted: 04/11/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Ferroptosis, a recently discovered form of cell death, whose role in basal cell carcinoma (BCC) and squamous cell carcinoma (SCC) has not been well disclosed. To improve our understanding of the differences in tumor progression and therapeutic effects between BCC and SCC, and to find potential therapeutic targets, this study systematically analyzed ferroptosis-related genes (FRGs) and their associated local immune cytolytic activity (LICA) and tumor microenvironment (TME) metabolic function differences. METHODS Two bulk RNA-seq datasets, GSE7553 and GSE125285, from the Gene Expression Omnibus database were compared within and between groups to screen for common differentially expressed genes (DEGs) for enrichment analysis. The currently recognized FRGs in DEGs gene set were selected as the targets to analyze their correlation and difference in LICA and TME metabolic functions. And validated using immune cell populations from another single-cell RNA-seq (scRNA-seq) dataset (GSE123813) to accurately understand the difference in LICA. All of the gene sets for functional enrichment analysis comes from published results and MSigDB database. RESULTS Ten FRGs were used to further analyze the differences in LICA and TME metabolic functions between BCC and SCC. In the SCC samples, LICA (e.g. Treg, CCR, Cytolytic activity, etc.) and TME metabolic functions (e.g. lipid and energy, etc.) were significantly related to ferroptosis genes (e.g. SLC1A5, CD44, NQO1, HMOX1 and STEAP3), and the ferroptosis potential index were also significantly higher than that in the BCC samples. Finally, based on these ten FRGs and related enrichment results, we postulated a model of NQO1 homeostasis regulated by FRGs during induction of ferroptosis in SCC. CONCLUSIONS The results showed that three FRGs, SLC1A5, CD44 and NQO1, have significant potential in targeted therapies for SCC chemotherapy resistance. And two FRGs, STEAP3 and HMOX1, formed a synergistic effect on the occurrence of ferroptosis in tumor cells. Our findings can be used as the main research materials for metastasis and chemotherapy resistance in SCC patients.
Collapse
Affiliation(s)
- Jianqiao Wang
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Dong Xie
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Hongxuan Wu
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yuchen Li
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Chuan Wan
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
28
|
Qiu S, Iimori M, Edahiro K, Fujimoto Y, Matsuoka K, Oki E, Maehara Y, Mori M, Kitao H. A CD44 variant is essential for Slug-dependent vimentin gene expression to acquire TGF-β1-induced tumor cell motility. Cancer Sci 2022; 113:2654-2667. [PMID: 35363934 PMCID: PMC9357650 DOI: 10.1111/cas.15353] [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: 11/14/2021] [Revised: 03/02/2022] [Accepted: 03/07/2022] [Indexed: 11/28/2022] Open
Abstract
CD44 is a widely expressed polymorphic adhesion molecule that has pleiotropic functions in development and tumor progression. Its mRNA undergoes alternative splicing to generate multiple variant (CD44v) isoforms, although the function of each CD44v isoform is not fully elucidated. Here, we show that CD44v plays an important role in the induction of vimentin expression upon transforming growth factor‐β1 (TGF‐β1)‐induced epithelial–mesenchymal transition (EMT). Among multiple CD44v isoforms expressed in NUGC3 gastric cancer cells, CD44v8‐10 and CD44v3,8‐10 are involved in the acquisition of migratory and invasive properties associated with TGF‐β1‐induced EMT, and only CD44v3,8‐10 induces the transcription of vimentin mediated by the EMT transcription factor Slug. In primary tumor specimens obtained from patients with gastric cancer, CD44‐containing variant exon 9 (CD44v9) expression and EMT features [E‐cadherin(−)vimentin(+)] were significantly correlated, and EMT features in the cells expressing CD44v9 were associated with tumor invasion depth, lymph node metastasis, and pStage, which indicate invasive and metastatic properties, and poor prognosis. These results indicate that certain CD44v isoforms promote tumor cell motility and metastasis in gastric cancer in association with EMT features, and CD44v3,8‐10 may contribute to these clinical characteristics.
Collapse
Affiliation(s)
- Shichao Qiu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Iimori
- Department of Molecular Cancer Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Keitaro Edahiro
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshiaki Fujimoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiko Maehara
- Kyushu Central Hospital of the Mutual Aid Association of Public School Teachers, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Graduate School of Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Hiroyuki Kitao
- Department of Molecular Cancer Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
29
|
Zou W, Zhang Y, Bai G, Zhuang J, Wei L, Wang Z, Sun M, Wang J. siRNA-induced CD44 knockdown suppresses the proliferation and invasion of colorectal cancer stem cells through inhibiting epithelial-mesenchymal transition. J Cell Mol Med 2022; 26:1969-1978. [PMID: 35229451 PMCID: PMC8980945 DOI: 10.1111/jcmm.17221] [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: 07/27/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 12/04/2022] Open
Abstract
CD44 has shown prognostic values and promising therapeutic potential in multiple human cancers; however, the effects of CD44 silencing on biological behaviors of cancer stem cells (CSCs) have not been fully understood in colorectal cancer. To examine the contribution of siRNA‐induced knockdown of CD44 to the biological features of colorectal CSCs, colorectal CSCs HCT116‐CSCs were generated, and CD44 was knocked down in HCT116‐CSCs using siRNA. The proliferation, migration and invasion of HCT116‐CSCs were measured, and apoptosis and cell‐cycle analyses were performed. The sensitivity of HCT116‐CSCs to oxaliplatin was tested, and xenograft tumor growth assay was performed to examine the role of CD44 in HCT116‐CSCs tumorigenesis in vivo. In addition, the expression of epithelial–mesenchymal transition (EMT) markers E‐cadherin, N‐cadherin and vimentin was quantified. siRNA‐induced knockdown of CD44 was found to inhibit the proliferation, migration and invasion, induce apoptosis, promote cell‐cycle arrest at the G1/G0 phase and increase the sensitivity of HCT116‐CSCs to oxaliplatin in HCT116‐CSCs, and knockdown of CD44 suppressed in vivo tumorigenesis and intrapulmonary metastasis of HCT116‐CSCs. Moreover, silencing CD44 resulted in EMT inhibition. Our findings demonstrate that siRNA‐induced CD44 knockdown suppresses the proliferation, invasion and in vivo tumorigenesis and metastasis of colorectal CSCs by inhibiting EMT.
Collapse
Affiliation(s)
- Weiyan Zou
- Department of Histology and Embryology, Bengbu Medical College, Bengbu City, China
| | - Yi Zhang
- The Second Department of Surgery, Xiamen Hospital Affiliated to Beijing University of Chinese Medicine, Xiamen City, China
| | - Guangfu Bai
- Department of Emergency, Wuxi Huishan District People's Hospital, Wuxi City, China
| | - Jialu Zhuang
- The Second School of Clinical Medicine, Bengbu Medical College, Bengbu City, China
| | - Lin Wei
- The Second School of Clinical Medicine, Bengbu Medical College, Bengbu City, China
| | - Zishu Wang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Meiqun Sun
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| | - Junbin Wang
- Department of Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, China
| |
Collapse
|
30
|
Xie Z, Gao Y, Ho C, Li L, Jin C, Wang X, Zou C, Mao Y, Wang X, Li Q, Fu D, Zhang YF. Exosome-delivered CD44v6/C1QBP complex drives pancreatic cancer liver metastasis by promoting fibrotic liver microenvironment. Gut 2022; 71:568-579. [PMID: 33827783 DOI: 10.1136/gutjnl-2020-323014] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) shows a remarkable predilection for liver metastasis. Pro-oncogenic secretome delivery and trafficking via exosomes are crucial for pre-metastatic microenvironment formation and metastasis. This study aimed to explore the underlying mechanisms of how PDAC-derived exosomes (Pex) modulate the liver microenvironment and promote metastasis. DESIGN C57BL/6 mice were 'educated' by tail vein Pex injection. The intrasplenic injection liver metastasis and PDAC orthotopic transplantation models were used to evaluate liver metastasis. Stable cell lines CD44v6 (CD44 variant isoform 6) or C1QBP (complement C1q binding protein) knockdown or overexpression was established using lentivirus transfection or gateway systems. A total of 142 patients with PDAC in Huashan Hospital were retrospectively enrolled. Prognosis and liver metastasis were predicted using Kaplan-Meier survival curves and logistic regression models. RESULTS Pex tail vein injection induced the deposition of liver fibrotic extracellular matrix, which promoted PDAC liver metastasis. Specifically, the exosomal CD44v6/C1QBP complex was delivered to the plasma membrane of hepatic satellite cells (HSCs), leading to phosphorylation of insulin-like growth factor 1 signalling molecules, which resulted in HSC activation and liver fibrosis. Expression of Pex CD44v6 and C1QBP in PDAC patients with liver metastasis was significantly higher than in PDAC patients without liver metastasis, and simultaneous high expression of exosomal CD44v6 and C1QBP correlated with a worse prognosis and a higher risk of postoperative PDAC liver metastasis. CONCLUSION The Pex-derived CD44v6/C1QBP complex is essential for the formation of a fibrotic liver microenvironment and PDAC liver metastasis. Highly expressed exosomal CD44v6 and C1QBP are promising biomarkers for predicting prognosis and liver metastasis in patients with PDAC.
Collapse
Affiliation(s)
- Zhibo Xie
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Children's Hospital, Shanghai, China
- Department of Pancreatic Surgery, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Ya Gao
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Chiakang Ho
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Lequn Li
- Department of Hepatobiliary and Pancreas Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi, China
| | - Chen Jin
- Department of Pancreatic Surgery, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Xiaoyi Wang
- Department of Pancreatic Surgery, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Caifeng Zou
- Department of Pancreatic Surgery, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Yishen Mao
- Department of Pancreatic Surgery, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Xiaobo Wang
- Department of Hepatobiliary and Pancreas Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi, China
| | - Qingfeng Li
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Deliang Fu
- Department of Pancreatic Surgery, Huashan Hospital Fudan University, Shanghai, Shanghai, China
| | - Yi-Fan Zhang
- Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| |
Collapse
|
31
|
Jia Y, Wei Z, Zhang S, Yang B, Li Y. Instructive Hydrogels for Primary Tumor Cell Culture: Current Status and Outlook. Adv Healthc Mater 2022; 11:e2102479. [PMID: 35182456 DOI: 10.1002/adhm.202102479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/07/2022] [Indexed: 02/06/2023]
Abstract
Primary tumor organoids (PTOs) growth in hydrogels have emerged as an important in vitro model that recapitulates many characteristics of the native tumor tissue, and have important applications in fundamental cancer research and for the development of useful therapeutic treatment. This paper begins with reviewing the methods of isolation of primary tumor cells. Then, recent advances on the instructive hydrogels as biomimetic extracellular matrix for primary tumor cell culture and construction of PTO models are summarized. Emerging microtechnology for growth of PTOs in microscale hydrogels and the applications of PTOs are highlighted. This paper concludes with an outlook on the future directions in the investigation of instructive hydrogels for PTO growth.
Collapse
Affiliation(s)
- Yiyang Jia
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University 2699 Qianjin Street Changchun 130012 China
| | - Zhentong Wei
- Department of Oncologic Gynecology The First Hospital of Jilin University Changchun 130021 China
| | - Songling Zhang
- Department of Oncologic Gynecology The First Hospital of Jilin University Changchun 130021 China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University 2699 Qianjin Street Changchun 130012 China
- Joint Laboratory of Opto‐Functional Theranostics in Medicine and Chemistry The First Hospital of Jilin University Changchun 130021 China
| | - Yunfeng Li
- State Key Laboratory of Supramolecular Structure and Materials College of Chemistry Jilin University 2699 Qianjin Street Changchun 130012 China
- Joint Laboratory of Opto‐Functional Theranostics in Medicine and Chemistry The First Hospital of Jilin University Changchun 130021 China
| |
Collapse
|
32
|
Multifunctional nanoparticles recruiting hyaluronic acid ligand and polyplexes containing low molecular weight protamine and ATP-Sensitive DNA motif for doxorubicin delivery. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
33
|
Icardi A, Lompardia SL, Papademetrio DL, Rosales P, Díaz M, Pibuel MA, Alaniz L, Alvarez E. Hyaluronan in the Extracellular Matrix of Hematological and Solid Tumors. Its Biological Effects. BIOLOGY OF EXTRACELLULAR MATRIX 2022:161-196. [DOI: 10.1007/978-3-030-99708-3_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
34
|
Hassn Mesrati M, Syafruddin SE, Mohtar MA, Syahir A. CD44: A Multifunctional Mediator of Cancer Progression. Biomolecules 2021; 11:1850. [PMID: 34944493 PMCID: PMC8699317 DOI: 10.3390/biom11121850] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/23/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022] Open
Abstract
CD44, a non-kinase cell surface transmembrane glycoprotein, has been widely implicated as a cancer stem cell (CSC) marker in several cancers. Cells overexpressing CD44 possess several CSC traits, such as self-renewal and epithelial-mesenchymal transition (EMT) capability, as well as a resistance to chemo- and radiotherapy. The CD44 gene regularly undergoes alternative splicing, resulting in the standard (CD44s) and variant (CD44v) isoforms. The interaction of such isoforms with ligands, particularly hyaluronic acid (HA), osteopontin (OPN) and matrix metalloproteinases (MMPs), drive numerous cancer-associated signalling. However, there are contradictory results regarding whether high or low CD44 expression is associated with worsening clinicopathological features, such as a higher tumour histological grade, advanced tumour stage and poorer survival rates. Nonetheless, high CD44 expression significantly contributes to enhanced tumourigenic mechanisms, such as cell proliferation, metastasis, invasion, migration and stemness; hence, CD44 is an important clinical target. This review summarises current research regarding the different CD44 isoform structures and their roles and functions in supporting tumourigenesis and discusses CD44 expression regulation, CD44-signalling pathways and interactions involved in cancer development. The clinical significance and prognostic value of CD44 and the potential of CD44 as a therapeutic target in cancer are also addressed.
Collapse
Affiliation(s)
- Malak Hassn Mesrati
- Nanobiotechnology Research Group, Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400 UPM, Selangor, Malaysia;
| | - Saiful Effendi Syafruddin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (S.E.S.); (M.A.M.)
| | - M. Aiman Mohtar
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (S.E.S.); (M.A.M.)
| | - Amir Syahir
- Nanobiotechnology Research Group, Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400 UPM, Selangor, Malaysia;
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400 UPM, Selangor, Malaysia
| |
Collapse
|
35
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 505] [Impact Index Per Article: 126.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany
- Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
36
|
Lee JW, Lee HY. Targeting Cancer Stem Cell Markers or Pathways: A Potential Therapeutic Strategy for Oral Cancer Treatment. Int J Stem Cells 2021; 14:386-399. [PMID: 34711702 PMCID: PMC8611309 DOI: 10.15283/ijsc21084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/14/2021] [Accepted: 06/05/2021] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) are a small subset of cancer cells with stem cell-like properties, self-renewal potential, and differentiation capacity into multiple cell types. Critical genetic alterations or aberrantly activated signaling pathways associated with drug resistance and recurrence have been observed in multiple types of CSCs. In this context, CSCs are considered to be responsible for tumor initiation, growth, progression, therapeutic resistance, and metastasis. Therefore, to effectively eradicate CSCs, tremendous efforts have been devoted to identify specific target molecules that play a critical role in regulating their distinct functions and to develop novel therapeutics, such as proteins, monoclonal antibodies, selective small molecule inhibitors, and small antisense RNA (asRNA) drugs. Similar to other CSC types, oral CSCs can be characterized by certain pluripotency-associated markers, and oral CSCs can also survive and form 3D tumor spheres in suspension culture conditions. These oral CSC-targeting therapeutics selectively suppress specific surface markers or key signaling components and subsequently inhibit the stem-like properties of oral CSCs. A large number of new therapeutic candidates have been tested, and some products are currently in the pre-clinical or clinical development phase. In the present study, we review new oral CSC-targeted therapeutic strategies and discuss the various specific CSC surface markers and key signaling components involved in the stem-like properties, growth, drug resistance, and tumorigenicity of oral CSCs.
Collapse
Affiliation(s)
- Jin Woo Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, Korea
| | - Hwa-Yong Lee
- Department of Biomedical Science, Jungwon University, Goesan, Korea.,Division of Science Education, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
37
|
An intelligent cell-selective polymersome-DM1 nanotoxin toward triple negative breast cancer. J Control Release 2021; 340:331-341. [PMID: 34774889 DOI: 10.1016/j.jconrel.2021.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/02/2021] [Accepted: 11/09/2021] [Indexed: 12/22/2022]
Abstract
Antibody-drug conjugates (ADCs) are among the most significant advances in clinical cancer treatments, however, they are haunted with fundamental issues like low drug/antibody ratio (DAR), need of large amount of antibody, and complex chemistry. Targeted nanomedicines while offering a promising alternative to ADCs are afflicted with drug leakage and inferior cancer-specificity. Herein, we developed an intelligent cell-selective nanotoxin based on anti-CD44 antibody-polymersome-DM1 conjugates (aCD44-AP-DM1) for potent treatment of solid tumors. DM1 was simultaneously coupled to vesicular membrane via disulfide bonds during self-assembly and anti-CD44 antibody was facilely clicked onto polymersome surface, tailor-making an optimal aCD44-AP-DM1 with a controlled antibody density of 5.0, extraordinary DAR of 275, zero drug leakage and rapid reduction-responsive DM1 release. aCD44-AP-DM1 displayed a high specificity and exceptional cytotoxicity toward MDA-MB-231 triple negative breast cancer, SMMC-7721 hepatocellular carcinoma and A549 non-small cell lung cancer cells with half-maximal inhibitory concentrations (IC50) of 21.4, 3.7 and 64.6 ng/mL, respectively, 3.6-47.2-fold exceeding non-targeted P-DM1. Intriguingly, the systemic administration of aCD44-AP-DM1 significantly suppressed subcutaneous MDA-MB-231 tumor xenografts in nude mice while intratumoral injection achieved complete tumor eradication in four out of five mice, without causing toxicity. This intelligent cell-selective nanotoxin has emerged as a better platform over ADCs for targeted cancer therapy.
Collapse
|
38
|
Functionalized niosomes as a smart delivery device in cancer and fungal infection. Eur J Pharm Sci 2021; 168:106052. [PMID: 34740786 DOI: 10.1016/j.ejps.2021.106052] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/13/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022]
Abstract
Various diseases remain untreated due to lack of suitable therapeutic moiety or a suitable drug delivery device, especially where toxicities and side effects are the primary reason for concern. Cancer and fungal infections are diseases where treatment schedules are not completed due to severe side effects or lengthy treatment protocols. Advanced treatment approaches such as active targeting and inhibition of angiogenesis may be preferred method for the treatment for malignancy over the conventional method. Niosomes may be a better alternative drug delivery carrier for various therapeutic moieties (either hydrophilic or hydrophobic) and also due to ease of surface modification, non-immunogenicity and economical. Active targeting approach may be done by targeting the receptors through coupling of suitable ligand on niosomal surface. Moreover, various receptors (CD44, folate, epidermal growth factor receptor (EGFR) & Vascular growth factor receptor (VGFR)) expressed by malignant cells have also been reviewed. The preparation of suitable niosomal formulation also requires considerable attention, and its formulation depends upon various factors such as selection of non-ionic surfactant, method of fabrication, and fabrication parameters. A combination therapy (dual drug and immunotherapy) has been proposed for the treatment of fungal infection with special consideration for surface modification with suitable ligand on niosomal surface to sensitize the receptors (C-type lectin receptors, Toll-like receptors & Nucleotide-binding oligomerization domain-like receptors) present on immune cells involved in fungal immunity. Certain gene silencing concept has also been discussed as an advanced alternative treatment for cancer by silencing the mRNA at molecular level using short interfering RNA (si-RNA).
Collapse
|
39
|
Gorczyca G, Wartalski K, Wiater J, Samiec M, Tabarowski Z, Duda M. Anabolic Steroids-Driven Regulation of Porcine Ovarian Putative Stem Cells Favors the Onset of Their Neoplastic Transformation. Int J Mol Sci 2021; 22:ijms222111800. [PMID: 34769230 PMCID: PMC8583785 DOI: 10.3390/ijms222111800] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Nandrolone (Ndn) and boldenone (Bdn), the synthetic testosterone analogues with strong anabolic effects, despite being recognized as potentially carcinogenic compounds, are commonly abused by athletes and bodybuilders, which includes women, worldwide. This study tested the hypothesis that different doses of Ndn and Bdn can initiate neoplastic transformation of porcine ovarian putative stem cells (poPSCs). Immunomagnetically isolated poPSCs were expanded ex vivo in the presence of Ndn or Bdn, for 7 and 14 days. Results show that pharmacological doses of both Ndn and Bdn, already after 7 days of poPSCs culture, caused a significant increase of selected, stemness-related markers of cancer cells: CD44 and CD133. Notably, Ndn also negatively affected poPSCs growth not only by suppressing their proliferation and mitochondrial respiration but also by inducing apoptosis. This observation shows, for the first time, that chronic exposure to Ndn or Bdn represents a precondition that might enhance risk of poPSCs neoplastic transformation. These studies carried out to accomplish detailed molecular characterization of the ex vivo expanded poPSCs and their potentially cancerous derivatives (PCDs) might be helpful to determine their suitability as nuclear donor cells (NDCs) for further investigations focused on cloning by somatic cell nuclear transfer (SCNT). Such investigations might also be indispensable to estimate the capabilities of nuclear genomes inherited from poPSCs and their PCDs to be epigenetically reprogrammed (dedifferentiated) in cloned pig embryos generated by SCNT. This might open up new possibilities for biomedical research aimed at more comprehensively recognizing genetic and epigenetic mechanisms underlying not only tumorigenesis but also reversal/retardation of pro-tumorigenic intracellular events.
Collapse
Affiliation(s)
- Gabriela Gorczyca
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
| | - Kamil Wartalski
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7 Street, 31-034 Krakow, Poland; (K.W.); (J.W.)
| | - Jerzy Wiater
- Department of Histology, Faculty of Medicine, Jagiellonian University Medical College, Kopernika 7 Street, 31-034 Krakow, Poland; (K.W.); (J.W.)
| | - Marcin Samiec
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice near Kraków, Poland
- Correspondence: (M.S.); (M.D.)
| | - Zbigniew Tabarowski
- Department of Experimental Hematology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
| | - Małgorzata Duda
- Department of Endocrinology, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland;
- Correspondence: (M.S.); (M.D.)
| |
Collapse
|
40
|
Tan T, Yang Q, Chen D, Zhao J, Xiang L, Feng J, Song X, Fu Y, Gong T. Chondroitin sulfate-mediated albumin corona nanoparticles for the treatment of breast cancer. Asian J Pharm Sci 2021; 16:508-518. [PMID: 34703499 PMCID: PMC8520051 DOI: 10.1016/j.ajps.2021.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/08/2021] [Accepted: 03/10/2021] [Indexed: 12/20/2022] Open
Abstract
Chondroitin sulfate-mediated albumin corona nanoparticles were readily prepared without any chemical reaction, and their active tumor targeting and therapeutic effects were examined. Negatively charged chondroitin sulfate (CS) and positively charged doxorubicin (DOX) self-assembled into nanoparticles (CS-DOX-NPs) via electrostatic interactions. Bovine serum albumin (BSA) was then adsorbed on the surface of CS-DOX-NPs to form albumin corona nanoparticles (BC-DOX-NPs) protected from endogenous proteins. Due to the dual effect of BSA and CS, BC-DOX-NPs interacted with the gp60, SPARC and CD44 receptors on tumor cells, facilitating their rapid and efficient transcytosis and improving their accumulation and uptake within tumor tissues. The simultaneous presence of BSA and CS also allowed BC-DOX-NPs to target CD44 efficiently, leading to greater cellular uptake and cytotoxicity against 4T1 cells than CS-DOX-NPs or free DOX. Intravenous injection of BC-DOX-NPs into orthotopic 4T1 tumor-bearing mice led to greater drug accumulation at the tumor site than with CS-DOX-NPs or free DOX, resulting in significant inhibition of tumor growth and lower exposure of major organs to the drug.
Collapse
Affiliation(s)
- Tiantian Tan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Qin Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.,School of Pharmacy, North Sichuan Medical College, Nanchong 637100, China
| | - Dan Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Juan Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.,Departments of Pharmacy, Mianyang Hospital of Traditional Chinese Medicine, Mianyang 621000, China
| | - Ling Xiang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Jiaxing Feng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Xu Song
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology,West China School of Pharmacy, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
41
|
Takei J, Asano T, Suzuki H, Kaneko MK, Kato Y. Epitope Mapping of the Anti-CD44 Monoclonal Antibody (C 44Mab-46) Using Alanine-Scanning Mutagenesis and Surface Plasmon Resonance. Monoclon Antib Immunodiagn Immunother 2021; 40:219-226. [PMID: 34678095 DOI: 10.1089/mab.2021.0028] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
CD44 is a type I transmembrane protein expressed in various kinds of normal cancer cells, including pancreatic, breast, and oral cancers. CD44 is associated with cancer progression, metastases, and treatment resistance. CD44 consists of 20 exons, and various isoforms exist due to alternative splicing of the central 10 exons. Some splicing variants show cancer-specific expression patterns and are related to prognosis of patients with cancer. Therefore, CD44 targeting therapy has been attracting attention. In a previous study, we established an anti-CD44 monoclonal antibody, C44Mab-46 (IgG1, kappa), useful for flow cytometry, Western blotting, and immunohistochemistry by immunizing mice with CD44v3-10 ectodomain. This study investigated the binding epitope of C44Mab-46 using enzyme-linked immunosorbent assay (ELISA) and the surface plasmon resonance (SPR) with the synthesized peptide. ELISA results using deletion mutants showed that C44Mab-46 reacted with the amino acids (aa) of 161-180 aa of CD44. Further examination of the C44Mab-46 epitope using ELISA with point mutants in 161-180 aa of CD44 demonstrates that the C44Mab-46 epitope comprised Thr174, Asp177, and Val178. The SPR with point mutants in 161-180 aa of CD44 demonstrated that the C44Mab-46 epitope comprises Thr174, Asp175, Asp176, Asp177, and Val178. Together, the C44Mab-46 epitope was determined to be located in exon 5 of CD44.
Collapse
Affiliation(s)
- Junko Takei
- Department of Antibody Drug Development, and Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, and Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, and Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, and Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
42
|
de Carvalho PA, Bonatelli M, Cordeiro MD, Coelho RF, Reis S, Srougi M, Nahas WC, Pinheiro C, Leite KRM. MCT1 expression is independently related to shorter cancer-specific survival in clear cell renal cell carcinoma. Carcinogenesis 2021; 42:1420-1427. [PMID: 34668521 DOI: 10.1093/carcin/bgab100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/05/2021] [Accepted: 10/18/2021] [Indexed: 11/15/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) has been considered a metabolic disease, with loss of von Hippel-Lindau (VHL) gene and consequent overexpression of hypoxia-inducible factor 1 alpha (HIF-1α), which is central for tumor development and progression. Among other effects, HIF-1α is involved in the metabolic reprogramming of cancer cells towards the Warburg effect involved in tumor cell proliferation, migration and survival. In this context, several proteins are expressed by cancer cells, including glucose and lactate transporters as well as different pH regulators. Among them, monocarboxylate transporters (MCTs) can be highlighted. Our aim is to comprehensively analyze the immunoexpression of MCT1, MCT2, MCT4, CD147, CD44, HIF-1α, GLUT1 and CAIX in ccRCC surgical specimens correlating with classical prognostic factors and survival of patients with long follow up. Surgical specimens from 207 patients with ccRCC who underwent radical or partial nephrectomy were used to build a tissue microarray. Immunostaining was categorized into absent/weak or moderate/strong and related to all classic ccRCC prognostic parameters. Kaplan-Meier curves were generated to assess overall and cancer-specific survival, and multivariate analysis was performed to identify independent prognostic factors of survival. Multivariate analysis showed that MCT1 together with tumor size and TNM staging, were independently related to cancer-specific survival. MCT1, CD147, CD44 and GLUT1 expression were significantly associated with poor prognostic factors. We show that MCT1 is an independent prognostic factor for cancer-specific survival in ccRCC justifying the use of new target therapies already being tested in clinical trials.
Collapse
Affiliation(s)
- Paulo Afonso de Carvalho
- Faculdade de Medicina da Universidade de Sao Paulo, Laboratory of Medical Investigation (LIM55)-Urology Department, Sao Paulo, Brazil
- Instituto do Câncer do Estado de Sao Paulo (ICESP), Sao Paulo, Brazil
| | - Murilo Bonatelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Sao Paulo, Brazil
| | | | | | - Sabrina Reis
- Faculdade de Medicina da Universidade de Sao Paulo, Laboratory of Medical Investigation (LIM55)-Urology Department, Sao Paulo, Brazil
| | - Miguel Srougi
- Faculdade de Medicina da Universidade de Sao Paulo, Laboratory of Medical Investigation (LIM55)-Urology Department, Sao Paulo, Brazil
- Instituto do Câncer do Estado de Sao Paulo (ICESP), Sao Paulo, Brazil
| | - Willian Carlos Nahas
- Faculdade de Medicina da Universidade de Sao Paulo, Laboratory of Medical Investigation (LIM55)-Urology Department, Sao Paulo, Brazil
- Instituto do Câncer do Estado de Sao Paulo (ICESP), Sao Paulo, Brazil
| | - Celine Pinheiro
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Sao Paulo, Brazil
- Barretos School of Health Sciences Dr. Paulo Prata-FACISB, Barretos, Sao Paulo, Brazil
| | - Katia Ramos Moreira Leite
- Faculdade de Medicina da Universidade de Sao Paulo, Laboratory of Medical Investigation (LIM55)-Urology Department, Sao Paulo, Brazil
| |
Collapse
|
43
|
NK Cell-Mediated Eradication of Ovarian Cancer Cells with a Novel Chimeric Antigen Receptor Directed against CD44. Biomedicines 2021; 9:biomedicines9101339. [PMID: 34680456 PMCID: PMC8533227 DOI: 10.3390/biomedicines9101339] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/09/2022] Open
Abstract
Ovarian cancer is the most common cause of gynecological cancer-related death in the developed world. Disease recurrence and chemoresistance are major causes of poor survival rates in ovarian cancer patients. Ovarian cancer stem cells (CSCs) were shown to represent a source of tumor recurrence owing to the high resistance to chemotherapy and enhanced tumorigenicity. Chimeric antigen receptor (CAR)-based adoptive immunotherapy represents a promising strategy to reduce the risk for recurrent disease. In this study, we developed a codon-optimized third-generation CAR to specifically target CD44, a marker widely expressed on ovarian cancer cells and associated with CSC-like properties and intraperitoneal tumor spread. We equipped NK-92 cells with the anti-CD44 CAR (CD44NK) and an anti-CD19 control CAR (CD19NK) using lentiviral SIN vectors. Compared to CD19NK and untransduced NK-92 cells, CD44NK showed potent and specific cytotoxic activity against CD44-positive ovarian cancer cell lines (SKOV3 and OVCAR3) and primary ovarian cancer cells harvested from ascites. In contrast, CD44NK had less cytotoxic activity against CD44-negative A2780 cells. Specific activation of engineered NK cells was also demonstrated by interferon-γ (IFNγ) secretion assays. Furthermore, CD44NK cells still demonstrated cytotoxic activity under cisplatin treatment. Most importantly, the simultaneous treatment with CD44NK and cisplatin showed higher anti-tumor activity than sequential treatment.
Collapse
|
44
|
Simultaneous targeting of CD44 and MMP9 catalytic and hemopexin domains as a therapeutic strategy. Biochem J 2021; 478:1139-1157. [PMID: 33600567 PMCID: PMC7959692 DOI: 10.1042/bcj20200628] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/09/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022]
Abstract
Crosstalk of the oncogenic matrix metalloproteinase-9 (MMP9) and one of its ligands, CD44, involves cleavage of CD44 by the MMP9 catalytic domain, with the CD44–MMP9 interaction on the cell surface taking place through the MMP9 hemopexin domain (PEX). This interaction promotes cancer cell migration and invasiveness. In concert, MMP9-processed CD44 induces the expression of MMP9, which degrades ECM components and facilitates growth factor release and activation, cancer cell invasiveness, and metastasis. Since both MMP9 and CD44 contribute to cancer progression, we have developed a new strategy to fully block this neoplastic process by engineering a multi-specific inhibitor that simultaneously targets CD44 and both the catalytic and PEX domains of MMP9. Using a yeast surface display technology, we first obtained a high-affinity inhibitor for the MMP9 catalytic domain, which we termed C9, by modifying a natural non-specific MMP inhibitor, N-TIMP2. We then conjugated C9 via a flexible linker to PEX, thereby creating a multi-specific inhibitor (C9-PEX) that simultaneously targets the MMP9 catalytic and PEX domains and CD44. It is likely that, via its co-localization with CD44, C9-PEX may compete with MMP9 localization on the cell surface, thereby inhibiting MMP9 catalytic activity, reducing MMP9 cellular levels, interfering with MMP9 homodimerization, and reducing the activation of downstream MAPK/ERK pathway signaling. The developed platform could be extended to other oncogenic MMPs as well as to other important target proteins, thereby offering great promise for creating novel multi-specific therapeutics for cancer and other diseases.
Collapse
|
45
|
Bahmani A, Shokri E, Hosseini M, Hosseinkhani S. A fluorescent aptasensor based on copper nanoclusters for optical detection of CD44 exon v10, an important isoform in metastatic breast cancer. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2021; 13:3837-3844. [PMID: 34378562 DOI: 10.1039/d1ay01087e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Recent studies suggest that breast cancer cells express various CD44 isoforms. CD44 is an integral transmembrane protein encoded by a single 20-exon gene. Exon v10 of CD44 plays a critical role in promoting cancer metastasis, so sensitive detection of this isoform helps in early diagnosis of metastatic breast cancer and facilitates the treatment process. This study aimed to use v10-specific aptamers to set up an optical aptasensor based on fluorescent metal nanoclusters. For this purpose, nanoclusters of silver, gold, and copper were prepared by different CD44 v10 DNA aptamers as molecular templates. UV-vis, TEM, and fluorescence spectrometer results confirmed the accuracy and quality of the synthesized aptamer-templated nanoclusters (Apt-NCs). Finally, we compared the performance of the as-prepared Apt-NCs in response to different cultured cell lines. According to the results, the optical response of M-Apt4-CuNCs was more efficient and correlated well with the concentrations of CD44 v10-enriched cells. The detection limit of the aptasensor was 40 ± 5 cells per mL.
Collapse
Affiliation(s)
- Amin Bahmani
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
| | - Ehsan Shokri
- Department of Nanotechnology, Agricultural Biotechnology Research Institute of Iran (ABRII), Agricultural Research, Education, and Extension Organization (AREEO), Karaj, Iran
| | - Morteza Hosseini
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
- Department of Pharmaceutical Biomaterials, Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
46
|
Tao J, Feng S, Liu B, Pan J, Li C, Zheng Y. Hyaluronic acid conjugated nitrogen-doped graphene quantum dots for identification of human breast cancer cells. Biomed Mater 2021; 16. [PMID: 34157704 DOI: 10.1088/1748-605x/ac0d93] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/22/2021] [Indexed: 11/12/2022]
Abstract
Accurate distinguish of cancer cells through fluorescence plays an important role in cancer diagnosis. Here we synthesized a blue fluorescent nitrogen-doped graphene quantum dots (N-GQDs) from citric acid and diethylamine via one-step hydrothermal synthesis method which was simple and quick to avoid by-products, and highlighted the binding sites to achieve precise combination. Due to the nitrogen element doping, amide II bond was amply obtained and abundant binding sites were provided for hyaluronic acid (HA) conjugation. N-GQDs solution with different pH value was then conjugated to HA via an amide bond for the recognition of human breast cancer cells (MCF-7 cells), and the formation of amide bond was more favorable under alkaline conditions. HA conjugated N-GQDs (HA-N-GQDs) were combined with CD44 which was over expressed on the surface of MCF-7 cells, resulting in MCF-7 cells performing stronger fluorescence. HA-N-GQDs showed high fluorescence, low toxicity, and good cytocompatibility, which held it play a role in fluorescence imaging for accurate identification of cancer cells.
Collapse
Affiliation(s)
- Junting Tao
- Department of Physics, and Key Laboratory of ATMMT Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Shixuan Feng
- Department of Physics, and Key Laboratory of ATMMT Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Bing Liu
- Department of Physics, and Key Laboratory of ATMMT Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Jiaqi Pan
- Department of Physics, and Key Laboratory of ATMMT Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Chaorong Li
- Department of Physics, and Key Laboratory of ATMMT Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Yingying Zheng
- Department of Physics, and Key Laboratory of ATMMT Ministry of Education, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| |
Collapse
|
47
|
Cavaliere M, Bisogno A, Scarpa A, D'Urso A, Marra P, Colacurcio V, De Luca P, Ralli M, Cassandro E, Cassandro C. Biomarkers of laryngeal squamous cell carcinoma: a review. Ann Diagn Pathol 2021; 54:151787. [PMID: 34242969 DOI: 10.1016/j.anndiagpath.2021.151787] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/30/2021] [Indexed: 10/21/2022]
Abstract
Laryngeal carcinoma is the second common malignancy of the upper aerodigestive tract after lung cancer; in most cases is a squamous cell carcinoma, whose risk factors include tobacco smoking and alcohol consumption. Despite therapeutic progress, the five-year overall survival rate for this malignancy has remained nearly 50% and many patients already present metastasis at the time of diagnosis. To date, there are no tools that predict the evolution of laryngeal carcinoma: in this light, during the last years, many studies were planned with the aim to investigate the role played by different biomarkers expressed by larynx cancer, which can help make an early diagnosis, predict disease evolution and direct therapeutic choice. This review aims to summarize these markers and correlating them with disease evolution.
Collapse
Affiliation(s)
- Matteo Cavaliere
- Department of Medicine and Surgery, University of Salerno, Via Salvador Allende 43, 84081 Baronissi, Salerno, Italy
| | - Antonella Bisogno
- Department of Medicine and Surgery, University of Salerno, Via Salvador Allende 43, 84081 Baronissi, Salerno, Italy.
| | - Alfonso Scarpa
- Department of Medicine and Surgery, University of Salerno, Via Salvador Allende 43, 84081 Baronissi, Salerno, Italy
| | - Alessia D'Urso
- Department of Medicine and Surgery, University of Salerno, Via Salvador Allende 43, 84081 Baronissi, Salerno, Italy
| | - Pasquale Marra
- Department of Medicine and Surgery, University of Salerno, Via Salvador Allende 43, 84081 Baronissi, Salerno, Italy
| | - Vito Colacurcio
- Department of Medicine and Surgery, University of Salerno, Via Salvador Allende 43, 84081 Baronissi, Salerno, Italy
| | - Pietro De Luca
- Department of Medicine and Surgery, University of Salerno, Via Salvador Allende 43, 84081 Baronissi, Salerno, Italy
| | - Massimo Ralli
- Department of Sense Organs, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy.
| | - Ettore Cassandro
- Department of Medicine and Surgery, University of Salerno, Via Salvador Allende 43, 84081 Baronissi, Salerno, Italy.
| | - Claudia Cassandro
- Surgical Sciences Department, University of Turin, Corso Dogliotti 14, 10124 Turin, Italy
| |
Collapse
|
48
|
Leyva-González CA, Salas-Treviño D, Contreras-Torres FF, Loera-Arias MDJ, Gómez-Tristán CA, Piña-Mendoza EI, García-Rivas GDJ, Guillén-Meléndez GA, Montes-de-Oca-Luna R, Saucedo-Cárdenas O, Soto-Domínguez A. Hyaluronate Functionalized Multi-Wall Carbon Nanotubes Loaded with Carboplatin Enhance Cytotoxicity on Human Cancer Cell Lines. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3622. [PMID: 34209588 PMCID: PMC8269704 DOI: 10.3390/ma14133622] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/18/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023]
Abstract
Cancer is a major global public health problem and conventional chemotherapy has several adverse effects and deficiencies. As a valuable option for chemotherapy, nanomedicine requires novel agents to increase the effects of antineoplastic drugs in multiple cancer models. Since its discovery, carbon nanotubes (CNTs) are intensively investigated for their use as carriers in drug delivery applications. This study shows the development of a nanovector generated with commercial carbon nanotubes (cCNTs) that were oxidized (oxCNTs) and chemically functionalized with hyaluronic acid (HA) and loaded with carboplatin (CPT). The nanovector, oxCNTs-HA-CPT, was used as a treatment against HeLa and MDA-MB-231 human tumor cell lines. The potential antineoplastic impact of the fabricated nanovector was evaluated in human cervical adenocarcinoma (HeLa) and mammary adenocarcinoma (MDA-MB-231). The oxCNTs-HA-CPT nanovector demonstrate to have a specific antitumor effect in vitro. The functionalization with HA allows that nanovector bio-directed towards tumor cells, while the toxicity effect is attributed mainly to CPT in a dose-dependent manner.
Collapse
Affiliation(s)
- César Adrián Leyva-González
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
| | - Daniel Salas-Treviño
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
| | | | - María de Jesús Loera-Arias
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
| | - Christian Alexis Gómez-Tristán
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
| | - Edgar Iván Piña-Mendoza
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
| | | | - Gloria Arely Guillén-Meléndez
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
| | - Roberto Montes-de-Oca-Luna
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
| | - Odila Saucedo-Cárdenas
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
- Departamento de Genética Molecular, Centro de Investigación Biomédica del Noreste (CIBIN) del IMSS, Monterrey C.P. 64720, Mexico
| | - Adolfo Soto-Domínguez
- Departamento de Histología, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey C.P. 64460, Mexico; (C.A.L.-G.); (D.S.-T.); (M.d.J.L.-A.); (C.A.G.-T.); (E.I.P.-M.); (G.A.G.-M.); (R.M.-d.-O.-L.)
| |
Collapse
|
49
|
Fontanella RA, Sideri S, Di Stefano C, Catizone A, Di Agostino S, Angelini DF, Guerrera G, Battistini L, Battafarano G, Del Fattore A, Campese AF, Padula F, De Cesaris P, Filippini A, Riccioli A. CD44v8-10 is a marker for malignant traits and a potential driver of bone metastasis in a subpopulation of prostate cancer cells. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0495. [PMID: 34018387 PMCID: PMC8330537 DOI: 10.20892/j.issn.2095-3941.2020.0495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Bone metastasis is a clinically important outcome of prostate carcinoma (PC). We focused on the phenotypic and functional characterization of a particularly aggressive phenotype within the androgen-independent bone metastasis-derived PC3 cell line. These cells, originated from the spontaneous conversion of a CD44-negative subpopulation, stably express the CD44v8-10 isoform (CD44v8-10pos) and display stem cell-like features and a marked invasive phenotype in vitro that is lost upon CD44v8-10 silencing. METHODS Flow cytometry, enzyme-linked immunoassay, immunofluorescence, and Western blot were used for phenotypic and immunologic characterization. Real-time quantitative polymerase chain reaction and functional assays were used to assess osteomimicry. RESULTS Analysis of epithelial-mesenchymal transition markers showed that CD44v8-10pos PC3 cells surprisingly display epithelial phenotype and can undergo osteomimicry, acquiring bone cell phenotypic and behavioral traits. Use of specific siRNA evidenced the ability of CD44v8-10 variant to confer osteomimetic features, hence the potential to form bone-specific metastasis. Moreover, the ability of tumors to activate immunosuppressive mechanisms which counteract effective immune responses is a sign of the aggressiveness of a tumor. Here we report that CD44v8-10pos cells express programmed death ligand 1, a negative regulator of anticancer immunity, and secrete exceptionally high amounts of interleukin-6, favoring osteoclastogenesis and immunosuppression in bone microenvironment. Notably, we identified a novel pathway activated by CD44v8-10, involving tafazzin (TAZ) and likely the Wnt/TAZ axis, known to play a role in upregulating osteomimetic genes. CONCLUSIONS CD44v8-10 could represent a marker of a more aggressive bone metastatic PC population exerting a driver role in osteomimicry in bone. A novel link between TAZ and CD44v8-10 is also shown.
Collapse
Affiliation(s)
- Rosaria A. Fontanella
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University, Rome 00161, Italy
| | - Silvia Sideri
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University, Rome 00161, Italy
| | - Chiara Di Stefano
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University, Rome 00161, Italy
| | - Angiolina Catizone
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University, Rome 00161, Italy
| | - Silvia Di Agostino
- Department of Health Sciences School of Medicine – “Magna Graecia” University of Catanzaro, Catanzaro 88100, Italy
| | | | | | | | - Giulia Battafarano
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Rome 00146, Italy
| | - Andrea Del Fattore
- Bone Physiopathology Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Rome 00146, Italy
| | | | - Fabrizio Padula
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University, Rome 00161, Italy
| | - Paola De Cesaris
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila 67100, Italy
| | - Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University, Rome 00161, Italy
| | - Anna Riccioli
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Unit of Histology and Medical Embryology, Sapienza University, Rome 00161, Italy
| |
Collapse
|
50
|
Gao N, Fan B, Li L, Sun X, Wang X, Ma H, Wei Q, Ju H. Label-Free Antifouling Photoelectrochemical Sensing Strategy for Detecting Breast Tumor Cells Based on Ligand-Receptor Interactions. ACS APPLIED BIO MATERIALS 2021; 4:4479-4485. [PMID: 35006860 DOI: 10.1021/acsabm.1c00215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biomarker expression both on the cell surface and in serum is directly related to the pathological process of tumor. Based on the interaction between the ligand and the protein receptor, a label-free photoelectrochemical (PEC) biosensing interface with good antifouling ability was proposed for tumor cell detection. TiO2 nanotube (NT) arrays were used as the substrate to enhance the ability of the biosensor to capture the target. Mercapto-terminated 8-arm poly(ethylene glycol) was introduced onto the electrode surface by the deposition of Au nanoparticles on TiO2 NTs, creating an antifouling molecular layer. The recognition ligand hyaluronic acid (HA) was functionalized by dopamine and introduced onto the sensing surface based on the unique chelating interaction between the catechol group and the titanium atom. Benefitting from the specific recognition of HA with CD44 and the 3D porous structures of NTs, the constructed PEC biosensor showed excellent abilities toward the detection of MDA-MB-231 breast tumor cells and the soluble form of CD44. The ligand-receptor PEC sensing strategy has promising potential for the detection of tumor cells and protein biomarkers.
Collapse
Affiliation(s)
- Ning Gao
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Bobo Fan
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Li Li
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Xiaojun Sun
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Xueying Wang
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Hongmin Ma
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Qin Wei
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| | - Huangxian Ju
- Key Laboratory of Interfacial Reaction & Sensing Analysis in Universities of Shandong, School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, P. R. China
| |
Collapse
|