1
|
Singh P, Metkari SM, Tripathi A, Bhartiya D. Reversing Uteropathies Including Cancer-Like Changes in Mice by Transplanting Mesenchymal Stromal Cells or XAR Treatment. Stem Cell Rev Rep 2024; 20:258-282. [PMID: 37779174 DOI: 10.1007/s12015-023-10632-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
Pluripotent, very small embryonic-like stem cells (VSELs) and tissue-committed 'progenitors' termed endometrial stem cells (EnSCs) are reported in mouse uterus. They express gonadal and gonadotropin hormone receptors and thus are vulnerable to early-life endocrine insults. Neonatal exposure of mouse pups to endocrine disruption cause stem/progenitor cells to undergo epigenetic changes, excessive self-renewal, and blocked differentiation that results in various uteropathies including non-receptive endometrium, hyperplasia, endometriosis, adenomyosis, and cancer-like changes in adult life. Present study investigated reversal of these uteropathies, by normalizing functions of VSELs and EnSCs. Two strategies were evaluated including (i) transplanting mesenchymal stromal cells (provide paracrine support) on D60 or (ii) oral administration of XAR (epigenetic regulator) daily from days 60-100 and effects were studied later in 100 days old mice. Results show normalization of stem/progenitor cells (Oct-4, Oct-4A, Sox-2, Nanog) and Wnt signalling (Wnt-4, β-catenin, Axin-2) specific transcripts. Flow cytometry results showed reduced numbers of 2-6 µm, LIN-CD45-SCA-1 + VSELs. Hyperplasia (Ki67) of epithelial (Pax-8, Foxa-2) and myometrial (α-Sma, Tgf-β) cells was reduced, adenogenesis (differentiation of glands) was restored, endometrial receptivity and differentiation (LIF, c-KIT, SOX-9, NUMB) and stromal cells niche (CD90, VIMENTIN, Pdgfra, Vimentin) were improved, cancer stem cells markers (OCT-4, CD166) were reduced while tumor suppressor genes (PTEN, P53) and epigenetic regulators (Ezh-2, Sirt-1) were increased. To conclude, normalizing VSELs/EnSCs to manage uteropathies provides a novel basis for initiating clinical studies. The study falls under the umbrella of United Nations Sustainable Development Goal 3 to ensure healthy lives and well-being for all of all ages.
Collapse
Affiliation(s)
- Pushpa Singh
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive & Child Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| | - S M Metkari
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive & Child Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| | - Anish Tripathi
- Epigeneres Biotech Pvt Ltd, Lower Parel, Mumbai, 400 013, India
| | - Deepa Bhartiya
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive & Child Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India.
- Epigeneres Biotech Pvt Ltd, Lower Parel, Mumbai, 400 013, India.
| |
Collapse
|
2
|
Dzul Keflee R, Hoong Leong K, Ogawa S, Bignon J, Chiang Chan M, Weng Kong K. Overview of the multifaceted resistances toward EGFR-TKIs and new chemotherapeutic strategies in non-small cell lung cancer. Biochem Pharmacol 2022; 205:115262. [PMID: 36191627 DOI: 10.1016/j.bcp.2022.115262] [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: 06/17/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/02/2022]
Abstract
The role of epidermal growth factor receptor (EGFR) in non-small cell lung cancer (NSCLC) has been vastly studied over the last decade. This has led to the rapid development of many generations of EGFR tyrosine kinase inhibitors (EGFR-TKIs). However, patients treated with third-generation TKIs (osimertinib, avitinib and rociletinib) targeting the EGFR T790M mutation have shown emerging resistances and relapses. Therefore, further molecular understanding of NSCLC mutations, bypass signalling, tumour microenvironment and the existence of cancer stem cells to overcome such resistances is warranted. This will pave the way for designing novel and effective chemotherapies to improve patients' overall survival. In this review, we provide an overview of the multifaceted mechanism of resistances towards EGFR-TKIs, as well as the challenges and perspectives that should be addressed in strategising chemotherapeutic treatments to overcome the ever evolving and adaptive nature of NSCLC.
Collapse
Affiliation(s)
- Rashidi Dzul Keflee
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kok Hoong Leong
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Satoshi Ogawa
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Jerome Bignon
- Institut de Chimie des Substances Naturelles CNRS UPR 2301, Université Paris Saclay, Gif-sur-Yvette, France
| | - Mun Chiang Chan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kin Weng Kong
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
3
|
Ferragut F, Vachetta VS, Troncoso MF, Rabinovich GA, Elola MT. ALCAM/CD166: A pleiotropic mediator of cell adhesion, stemness and cancer progression. Cytokine Growth Factor Rev 2021; 61:27-37. [PMID: 34272152 DOI: 10.1016/j.cytogfr.2021.07.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022]
Abstract
Activated Leukocyte Cell Adhesion Molecule (ALCAM/CD166) is a glycoprotein involved in homotypic and heterotypic cell adhesion. ALCAM can be proteolytically cleaved at the cell surface by metalloproteases, which generate shedding of its ectodomain. In various tumors, ALCAM is overexpressed and serves as a valuable prognostic marker of disease progression. Moreover, CD166 has been identified as a putative cancer stem cell marker in particular cancers. Herein, we summarize biochemical aspects of ALCAM, including structure, proteolytic shedding, alternative splicing, and specific ligands, and integrate this information with biological functions of this glycoprotein including cell adhesion, migration and invasion. In addition, we discuss different patterns of ALCAM expression in distinct tumor types and its contribution to tumor progression. Finally, we highlight the role of ALCAM as a cancer stem cell marker and introduce current clinical trials associated with this molecule. Future studies are needed to define the value of shed ALCAM in biofluids or ALCAM isoform expression as prognostic biomarkers in tumor progression.
Collapse
Affiliation(s)
- Fátima Ferragut
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro C. Paladini, Buenos Aires, Argentina
| | - Vanina S Vachetta
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro C. Paladini, Buenos Aires, Argentina
| | - María F Troncoso
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro C. Paladini, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María T Elola
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Buenos Aires, Argentina; CONICET-Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof. Alejandro C. Paladini, Buenos Aires, Argentina.
| |
Collapse
|
4
|
Lin Z, Wu Z, Luo W. Chimeric Antigen Receptor T-Cell Therapy: The Light of Day for Osteosarcoma. Cancers (Basel) 2021; 13:cancers13174469. [PMID: 34503279 PMCID: PMC8431424 DOI: 10.3390/cancers13174469] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/24/2021] [Accepted: 08/28/2021] [Indexed: 01/14/2023] Open
Abstract
Simple Summary As a novel immunotherapy, chimeric antigen receptor (CAR) T-cell therapy has achieved encouraging results in leukemia and lymphoma. Furthermore, CAR-T cells have been explored in the treatment of osteosarcoma (OS). However, there is no strong comprehensive evidence to support their efficacy. Therefore, we reviewed the current evidence on CAR-T cells for OS to demonstrate their feasibility and provide new options for the treatment of OS. Abstract Osteosarcoma (OS) is the most common malignant bone tumor, arising mainly in children and adolescents. With the introduction of multiagent chemotherapy, the treatments of OS have remarkably improved, but the prognosis for patients with metastases is still poor, with a five-year survival rate of 20%. In addition, adverse effects brought by traditional treatments, including radical surgery and systemic chemotherapy, may seriously affect the survival quality of patients. Therefore, new treatments for OS await exploitation. As a novel immunotherapy, chimeric antigen receptor (CAR) T-cell therapy has achieved encouraging results in treating cancer in recent years, especially in leukemia and lymphoma. Furthermore, researchers have recently focused on CAR-T therapy in solid tumors, including OS. In this review, we summarize the safety, specificity, and clinical transformation of the targets in treating OS and point out the direction for further research.
Collapse
|
5
|
Kim DK, Ham MH, Lee SY, Shin MJ, Kim YE, Song P, Suh DS, Kim JH. CD166 promotes the cancer stem-like properties of primary epithelial ovarian cancer cells. BMB Rep 2021. [PMID: 32843129 PMCID: PMC7781915 DOI: 10.5483/bmbrep.2020.53.12.102] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer stem cells (CSCs) or tumor-initiating cells are thought to play critical roles in tumorigenesis, metastasis, drug resistance, and tumor recurrence. For the diagnosis and targeted therapy of CSCs, the molecular identity of biomarkers or therapeutic targets for CSCs needs to be clarified. In this study, we identified CD166 as a novel marker expressed in the sphere-forming CSC population of A2780 epithelial ovarian cancer cells and primary ovarian cancer cells. The CD166+ cells isolated from A2780 cells and primary ovarian cancer cells highly expressed CSC markers, including ALDH1a1, OCT4, and SOX2, and ABC transporters, which are implicated in the drug resistance of CSCs. The CD166+ cells exhibited enhanced CSC-like properties, such as increased sphere-forming ability, cell migration and adhesion abilities, resistance to conventional anti-cancer drugs, and high tumorigenic potential in a xenograft mouse model. Knockdown of CD166 expression in the sphere-forming ovarian CSCs abrogated their CSC-like properties. Moreover, silencing of CD166 expression in the sphere-forming CSCs suppressed the phosphorylation of focal adhesion kinase, paxillin, and SRC. These results suggest that CD166 plays a key role in the regulation of CSC-like properties and focal adhesion kinase signaling in ovarian cancer.
Collapse
Affiliation(s)
- Dae Kyoung Kim
- Departments of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Min Hee Ham
- Departments of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Seo Yul Lee
- Departments of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Min Joo Shin
- Departments of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Ye Eun Kim
- Departments of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Parkyong Song
- Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Dong-Soo Suh
- Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan 50612, Korea
| | - Jae Ho Kim
- Departments of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Korea; Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Korea
| |
Collapse
|
6
|
Collagen type I promotes osteogenic differentiation of amniotic membrane-derived mesenchymal stromal cells in basal and induction media. Biosci Rep 2021; 40:227060. [PMID: 33245097 PMCID: PMC7736623 DOI: 10.1042/bsr20201325] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
Collagen has been widely shown to promote osteogenesis of bone marrow mesenchymal stromal cells (BM-MSCs). Due to the invasive procedure of obtaining BM-MSCs, MSCs from other tissues have emerged as a promising alternative for regenerative therapy. MSCs originated from different sources, exhibiting different differentiation potentials. Therefore, the applicability of collagen type I (COL), combining with amniotic membrane (AM)-MSCs was examined through proliferation and differentiation assays together with the expression of surface markers and genes associated with stemness and differentiation under basal or induction conditions. No increase in cell growth was observed because AM-MSCs might be directed toward spontaneous osteogenesis. This was evidenced by the calcium deposition and elevated expression of osteogenic genes when AM-MSCs were cultured in collagen plate with basal media. Under the osteogenic condition, reciprocal expression of OCN and CEBPA suggested a shift toward adipogenesis. Surprisingly, adipogenic genes were not elevated upon adipogenic induction, although oil droplets deposition was observed. In conclusion, our findings demonstrated that collagen causes spontaneous osteogenesis in AM-MSCs. However, the presence of exogenous inductors could shift the direction of adipo-osteogenic gene regulatory network modulated by collagen.
Collapse
|
7
|
Simões IT, Aranda F, Casadó-Llombart S, Velasco-de Andrés M, Català C, Álvarez P, Consuegra-Fernández M, Orta-Mascaró M, Merino R, Merino J, Alberola-Ila J, González-Aseguinolaza G, Carreras E, Martínez V, Lozano F. Multifaceted effects of soluble human CD6 in experimental cancer models. J Immunother Cancer 2020; 8:jitc-2019-000172. [PMID: 32217757 PMCID: PMC7174071 DOI: 10.1136/jitc-2019-000172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2020] [Indexed: 12/11/2022] Open
Abstract
Background CD6 is a lymphocyte surface co-receptor physically associated with the T-cell receptor (TCR)/CD3 complex at the center of the immunological synapse. There, CD6 assists in cell-to-cell contact stabilization and modulation of activation/differentiation events through interaction with CD166/ALCAM (activated leukocyte cell adhesion molecule), its main reported ligand. While accumulating evidence is attracting new interest on targeting CD6 for therapeutic purposes in autoimmune disorders, little is known on its potential in cancer. In an attempt to elucidate the in vivo relevance of blocking CD6-mediated interactions in health and disease, we explored the consequences of expressing high circulating levels of a soluble form CD6 (sCD6) as a decoy receptor. Methods High sCD6 serum levels were achieved by using transgenic C57BL/6 mice expressing human sCD6 under the control of lymphoid-specific transcriptional elements (shCD6LckEμTg) or wild type either transduced with hepatotropic adeno-associated virus coding for mouse sCD6 or undergoing repeated infusions of recombinant human sCD6 protein. Characterization of sCD6-induced changes was performed by ex vivo flow cytometry and functional analyses of mouse lymphoid organ cells. The in vivo relevance of those changes was explored by challenging mice with subcutaneous or metastatic tumors induced by syngeneic cancer cells of different lineage origins. Results Through a combination of in vitro and in vivo studies, we show that circulating sCD6 expression induces defective regulatory T cell (Treg) generation and function, decreased CD166/ALCAM-mediated tumor cell proliferation/migration and impaired galectin-induced T-cell apoptosis, supporting the fact that sCD6 modulates antitumor lymphocyte effector function and tumorigenesis. Accordingly, sCD6 expression in vivo resulted in delayed subcutaneous tumor growth and/or reduced metastasis on challenge of mice with syngeneic cancer cells. Conclusions Evidence is provided for the disruption of CD6 receptor–ligand interactions as a feasible immunomodulatory approach in cancer.
Collapse
Affiliation(s)
- Inês T Simões
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Fernando Aranda
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Sergi Casadó-Llombart
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - María Velasco-de Andrés
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Cristina Català
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Pilar Álvarez
- Departamento de Biología Molecular, Universidad de Cantabria-IDIVAL, Santander, Cantabria, Spain
| | - Marta Consuegra-Fernández
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Marc Orta-Mascaró
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Ramón Merino
- Instituto de Biomedicina y Biotecnología de Cantabria, CSIC-UC, Santander, Cantabria, Spain
| | - Jesús Merino
- Departamento de Biología Molecular, Universidad de Cantabria-IDIVAL, Santander, Cantabria, Spain
| | - José Alberola-Ila
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | | | - Esther Carreras
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Vanesa Martínez
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Francisco Lozano
- Immunoreceptors del Sistema Innat i Adaptatiu, Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain .,Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Barcelona, Spain.,Servei d'Immunologia, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|