1
|
Dey S, Ghosh M, Dev A. Signalling and molecular pathways, overexpressed receptors of colorectal cancer and effective therapeutic targeting using biogenic silver nanoparticles. Gene 2025; 936:149099. [PMID: 39557372 DOI: 10.1016/j.gene.2024.149099] [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: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Increasing morbidity and mortality in CRC is a potential threat to human health. The major challenges for better treatment outcomes are the heterogeneity of CRC cases, complicated molecular pathway cross-talks, the influence of gut dysbiosis in CRC, and the lack of multimodal target-specific drug delivery. The overexpression of many receptors in CRC cells may pave the path for targeting them with multiple ligands. The design of a more target-specific drug-delivery device with multiple ligand-functionalized, green-synthesized silver nanoparticles is highly promising and may also deliver other approved chemotherapeutic agents. This review presents the various aspects of colorectal cancer and over-expressed receptors that can be targeted with appropriate ligands to enhance the specific drug delivery potency of green synthesised silver nanoparticles. This review aims to broaden further research into this multi-ligand functionalised, safer and effective silver nano drug delivery system.
Collapse
Affiliation(s)
- Sandip Dey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India.
| |
Collapse
|
2
|
Saravanan V, Gopalakrishnan V, Mahendran MIMS, Vaithianathan R, Srinivasan S, Boopathy V, Krishnamurthy S. Biofilm mediated integrin activation and directing acceleration of colorectal cancer. APMIS 2024; 132:688-705. [PMID: 39246244 DOI: 10.1111/apm.13466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/15/2024] [Indexed: 09/10/2024]
Abstract
Bacterial biofilm plays a vital role in influencing several diseases, infections, metabolic pathways and communication channels. Biofilm influence over colorectal cancer (CRC) has been a booming area of research interest. The virulence factors of bacterial pathogen have a high tendency to induce metabolic pathway to accelerate CRC. The bacterial species biofilm may induce cancer through regulating the major signalling pathways responsible for cell proliferation, differentiation, survival and growth. Activation of cancer signals may get initiated from the chronic infections through bacterial biofilm species. Integrin mediates in the activation of major pathway promoting cancer. Integrin-mediated signals are expected to be greatly influenced by biofilm. Integrins are identified as an important dimer, whose dysfunction may alter the signalling cascade specially focusing on TGF-β, PI3K/Akt/mToR, MAPK and Wnt pathway. Along with biofilm shield, the tumour gains greater resistance from radiation, chemotherapy and also from other antibiotics. The biofilm barrier is known to cause challenges for CRC patients undergoing treatment.
Collapse
Affiliation(s)
- Vaijayanthi Saravanan
- MGM Advanced Research Institute, Sri Balaji Vidhyapeeth (Deemed to be University), Pondicherry, Tamil Nadu, India
| | - Vinoj Gopalakrishnan
- MGM Advanced Research Institute, Sri Balaji Vidhyapeeth (Deemed to be University), Pondicherry, Tamil Nadu, India
| | | | - Rajan Vaithianathan
- Mahatma Gandhi Medical College and Research Institute, Sri Balaji Vidhyapeeth (Deemed to be University), Pondicherry, Tamil Nadu, India
| | - Sowmya Srinivasan
- Mahatma Gandhi Medical College and Research Institute, Sri Balaji Vidhyapeeth (Deemed to be University), Pondicherry, Tamil Nadu, India
| | | | | |
Collapse
|
3
|
Mei J, Jiang XY, Tian HX, Rong DC, Song JN, Wang L, Chen YS, Wong RCB, Guo CX, Wang LS, Wang LY, Wang PY, Yin JY. Anoikis in cell fate, physiopathology, and therapeutic interventions. MedComm (Beijing) 2024; 5:e718. [PMID: 39286778 PMCID: PMC11401975 DOI: 10.1002/mco2.718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
The extracellular matrix (ECM) governs a wide spectrum of cellular fate processes, with a particular emphasis on anoikis, an integrin-dependent form of cell death. Currently, anoikis is defined as an intrinsic apoptosis. In contrast to traditional apoptosis and necroptosis, integrin correlates ECM signaling with intracellular signaling cascades, describing the full process of anoikis. However, anoikis is frequently overlooked in physiological and pathological processes as well as traditional in vitro research models. In this review, we summarized the role of anoikis in physiological and pathological processes, spanning embryonic development, organ development, tissue repair, inflammatory responses, cardiovascular diseases, tumor metastasis, and so on. Similarly, in the realm of stem cell research focused on the functional evolution of cells, anoikis offers a potential solution to various challenges, including in vitro cell culture models, stem cell therapy, cell transplantation, and engineering applications, which are largely based on the regulation of cell fate by anoikis. More importantly, the regulatory mechanisms of anoikis based on molecular processes and ECM signaling will provide new strategies for therapeutic interventions (drug therapy and cell-based therapy) in disease. In summary, this review provides a systematic elaboration of anoikis, thus shedding light on its future research.
Collapse
Affiliation(s)
- Jie Mei
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Xue-Yao Jiang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Hui-Xiang Tian
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Ding-Chao Rong
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
| | - Jia-Nan Song
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- School of Life Sciences Westlake University Hangzhou Zhejiang China
| | - Luozixian Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Yuan-Shen Chen
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Raymond C B Wong
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Cheng-Xian Guo
- Center of Clinical Pharmacology the Third Xiangya Hospital Central South University Changsha Hunan China
| | - Lian-Sheng Wang
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Lei-Yun Wang
- Department of Pharmacy Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan Hubei Province China
| | - Peng-Yuan Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| |
Collapse
|
4
|
Cloutier G, Beaulieu JF. Reconsideration of the laminin receptor 67LR in colorectal cancer cells. BIOMOLECULES & BIOMEDICINE 2024; 24:1117-1132. [PMID: 38606907 PMCID: PMC11378999 DOI: 10.17305/bb.2024.10323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
The 67 kDa laminin receptor (67LR) was identified as the first laminin receptor shown to be involved in the carcinogenesis of various cancers, including colorectal cancer. While the exact composition of this 67 kDa receptor remains unknown, it has been reported to be formed by the 37 kDa ribosomal protein SA (RPSA) covalently attached to another unidentified protein. The goal of this study was to clarify the molecular structure of 67LR to enhance our understanding of its role in malignancies. Using cell fractionation of colorectal cancer cells, the 67 kDa immunoreactive protein corresponding to 67LR was found in the soluble protein fraction, while some of the 37 kDa RPSA exhibited plasma membrane-like properties. Proteomic analysis of the 67 kDa fraction revealed the absence of RPSA but identified the β-galactosidase-related 67 kDa elastin-binding protein (67EBP), another laminin binding receptor which presents amino acid sequence similarities that can explain the immune cross reactivity with RPSA. The downregulation of β-galactosidase through short hairpin RNA (shRNA) led to a reduction in both 67LR and 67EBP immunoreactive proteins, confirming the misidentification of 67LR and 67EBP in colorectal cancer cells. Based on these findings, we propose to redefine the 67LR as the RPSA-containing laminin receptor (RCLR) to avoid confusion with the 67EBP.
Collapse
Affiliation(s)
- Gabriel Cloutier
- Department of Immunology and Cell Biology, Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Jean-François Beaulieu
- Department of Immunology and Cell Biology, Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
5
|
Wang R, Shu RR, Seldin L. Noncanonical functions of adhesion proteins in inflammation. Am J Physiol Cell Physiol 2024; 327:C505-C515. [PMID: 38981610 PMCID: PMC11427013 DOI: 10.1152/ajpcell.00292.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Cell adhesion proteins localize to epithelial and endothelial cell membranes to form junctional complexes between neighboring cells or between cells and the underlying basement membrane. The structural and functional integrities of these junctions are critical to establish cell polarity and maintain tissue barrier function, while also facilitating leukocyte migration and adhesion to sites of inflammation. In addition to their adhesive properties, however, junctional proteins can also serve important noncanonical functions in inflammatory signaling and transcriptional regulation. Intriguingly, recent work has unveiled novel roles for cell adhesion proteins as both signaling initiators and downstream targets during inflammation. In this review, we discuss both the traditional functions of junction proteins in cell adhesion and tissue barrier function as well as their noncanonical signaling roles that have been implicated in facilitating diverse inflammatory pathologies.
Collapse
Affiliation(s)
- Ruochong Wang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Raphael R Shu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Lindsey Seldin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| |
Collapse
|
6
|
Oliveira SM, Carvalho PD, Serra-Roma A, Oliveira P, Ribeiro A, Carvalho J, Martins F, Machado AL, Oliveira MJ, Velho S. Fibroblasts Promote Resistance to KRAS Silencing in Colorectal Cancer Cells. Cancers (Basel) 2024; 16:2595. [PMID: 39061234 PMCID: PMC11274566 DOI: 10.3390/cancers16142595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer (CRC) responses to KRAS-targeted inhibition have been limited due to low response rates, the mechanisms of which remain unknown. Herein, we explored the cancer-associated fibroblasts (CAFs) secretome as a mediator of resistance to KRAS silencing. CRC cell lines HCT15, HCT116, and SW480 were cultured either in recommended media or in conditioned media from a normal colon fibroblast cell line (CCD-18Co) activated with rhTGF-β1 to induce a CAF-like phenotype. The expression of membrane stem cell markers was analyzed by flow cytometry. Stem cell potential was evaluated by a sphere formation assay. RNAseq was performed in KRAS-silenced HCT116 colonospheres treated with either control media or conditioned media from CAFs. Our results demonstrated that KRAS-silencing up-regulated CD24 and down-regulated CD49f and CD104 in the three cell lines, leading to a reduction in sphere-forming efficiency. However, CAF-secreted factors restored stem cell marker expression and increased stemness. RNA sequencing showed that CAF-secreted factors up-regulated genes associated with pro-tumorigenic pathways in KRAS-silenced cells, including KRAS, TGFβ, NOTCH, WNT, MYC, cell cycle progression and exit from quiescence, epithelial-mesenchymal transition, and immune regulation. Overall, our results suggest that resistance to KRAS-targeted inhibition might derive not only from cell-intrinsic causes but also from external elements, such as fibroblast-secreted factors.
Collapse
Affiliation(s)
- Susana Mendonça Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- ESS|P.PORTO—Escola Superior de Saúde, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal
| | - Patrícia Dias Carvalho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - André Serra-Roma
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - Patrícia Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - Andreia Ribeiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - Joana Carvalho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| | - Flávia Martins
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ana Luísa Machado
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- FMUP—Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- ESS|P.PORTO—Escola Superior de Saúde, Instituto Politécnico do Porto, Rua Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal
| | - Maria José Oliveira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- FMUP—Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
- INEB—Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-177 Porto, Portugal
| | - Sérgia Velho
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (S.M.O.); (P.O.); (J.C.); (F.M.); (A.L.M.); (M.J.O.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, Universidade do Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
| |
Collapse
|
7
|
Grigoryeva E, Tashireva L, Alifanov V, Savelieva O, Zavyalova M, Menyailo M, Khozyainova A, Denisov EV, Bragina O, Popova N, Cherdyntseva NV, Perelmuter V. Integrin-associated transcriptional characteristics of circulating tumor cells in breast cancer patients. PeerJ 2024; 12:e16678. [PMID: 38250718 PMCID: PMC10800097 DOI: 10.7717/peerj.16678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/23/2023] [Indexed: 01/23/2024] Open
Abstract
Background Integrins enable cell communication with the basal membrane and extracellular matrix, activating signaling pathways and facilitating intracellular changes. Integrins in circulating tumor cells (CTCs) play a significant role in apoptosis evasion and anchor-independent survival. However, the link between CTCs expressing different integrin subunits, their transcriptional profile and, therefore, their functional activity with respect to metastatic potential remains unclear. Methods Single-cell RNA sequencing of CD45-negative cell fraction of breast cancer patients was performed. All CTCs were divided into nine groups according to their integrin profile. Results СTCs without the gene expression of integrins or with the expression of non-complementary α and β subunits that cannot form heterodimers prevailed. Only about 15% of CTCs expressed integrin subunits which can form heterodimers. The transcriptional profile of CTCs appeared to be associated with the spectrum of expressed integrins. The lowest potential activity was observed in CTCs without integrin expression, while the highest frequency of expression of tumor progression-related genes, namely genes of stemness, epithelial-mesenchymal transition (EMT), invasion, proinflammatory chemokines and cytokines as well as laminin subunits, were observed in CTCs co-expressing ITGA6 and ITGB4. Validation on the protein level revealed that the median of integrin β4+ CTCs was higher in patients with more aggressive molecular subtypes as well as in metastatic breast cancer patients. One can expect that CTCs with ITGA6 and ITGB4 expression will have pronounced metastatic potencies manifesting in expression of EMT and stemness-related genes, as well as potential ability to produce chemokine/proinflammatory cytokines and laminins.
Collapse
Affiliation(s)
- Evgeniya Grigoryeva
- The Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- The Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Liubov Tashireva
- The Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Vladimir Alifanov
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Olga Savelieva
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Marina Zavyalova
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Maxim Menyailo
- The Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Anna Khozyainova
- The Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Evgeny V. Denisov
- The Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Olga Bragina
- The Department of Nuclear Therapy and Diagnostics, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Nataliya Popova
- The Department of Chemotherapy, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Nadezhda V. Cherdyntseva
- The Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Vladimir Perelmuter
- The Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
8
|
Jumaniyazova E, Lokhonina A, Dzhalilova D, Kosyreva A, Fatkhudinov T. Role of Microenvironmental Components in Head and Neck Squamous Cell Carcinoma. J Pers Med 2023; 13:1616. [PMID: 38003931 PMCID: PMC10672525 DOI: 10.3390/jpm13111616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/04/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023] Open
Abstract
Head and neck squamous cell cancer (HNSCC) is one of the ten most common malignant neoplasms, characterized by an aggressive course, high recurrence rate, poor response to treatment, and low survival rate. This creates the need for a deeper understanding of the mechanisms of the pathogenesis of this cancer. The tumor microenvironment (TME) of HNSCC consists of stromal and immune cells, blood and lymphatic vessels, and extracellular matrix. It is known that HNSCC is characterized by complex relationships between cancer cells and TME components. TME components and their dynamic interactions with cancer cells enhance tumor adaptation to the environment, which provides the highly aggressive potential of HNSCC and resistance to antitumor therapy. Basic research aimed at studying the role of TME components in HNSCC carcinogenesis may serve as a key to the discovery of both new biomarkers-predictors of prognosis and targets for new antitumor drugs. This review article focuses on the role and interaction with cancer of TME components such as newly formed vessels, cancer-associated fibroblasts, and extracellular matrix.
Collapse
Affiliation(s)
- Enar Jumaniyazova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.L.); (A.K.); (T.F.)
| | - Anastasiya Lokhonina
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.L.); (A.K.); (T.F.)
- Avtsyn Research Institute of Human Morphology of FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997 Moscow, Russia
| | - Dzhuliia Dzhalilova
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.L.); (A.K.); (T.F.)
- Avtsyn Research Institute of Human Morphology of FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Anna Kosyreva
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.L.); (A.K.); (T.F.)
- Avtsyn Research Institute of Human Morphology of FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (A.L.); (A.K.); (T.F.)
- Avtsyn Research Institute of Human Morphology of FSBSI Petrovsky National Research Centre of Surgery, 3 Tsyurupy Street, 117418 Moscow, Russia
| |
Collapse
|
9
|
Li S, Sampson C, Liu C, Piao HL, Liu HX. Integrin signaling in cancer: bidirectional mechanisms and therapeutic opportunities. Cell Commun Signal 2023; 21:266. [PMID: 37770930 PMCID: PMC10537162 DOI: 10.1186/s12964-023-01264-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023] Open
Abstract
Integrins are transmembrane receptors that possess distinct ligand-binding specificities in the extracellular domain and signaling properties in the cytoplasmic domain. While most integrins have a short cytoplasmic tail, integrin β4 has a long cytoplasmic tail that can indirectly interact with the actin cytoskeleton. Additionally, 'inside-out' signals can induce integrins to adopt a high-affinity extended conformation for their appropriate ligands. These properties enable integrins to transmit bidirectional cellular signals, making it a critical regulator of various biological processes.Integrin expression and function are tightly linked to various aspects of tumor progression, including initiation, angiogenesis, cell motility, invasion, and metastasis. Certain integrins have been shown to drive tumorigenesis or amplify oncogenic signals by interacting with corresponding receptors, while others have marginal or even suppressive effects. Additionally, different α/β subtypes of integrins can exhibit opposite effects. Integrin-mediated signaling pathways including Ras- and Rho-GTPase, TGFβ, Hippo, Wnt, Notch, and sonic hedgehog (Shh) are involved in various stages of tumorigenesis. Therefore, understanding the complex regulatory mechanisms and molecular specificities of integrins are crucial to delaying cancer progression and suppressing tumorigenesis. Furthermore, the development of integrin-based therapeutics for cancer are of great importance.This review provides an overview of integrin-dependent bidirectional signaling mechanisms in cancer that can either support or oppose tumorigenesis by interacting with various signaling pathways. Finally, we focus on the future opportunities for emergent therapeutics based on integrin agonists. Video Abstract.
Collapse
Affiliation(s)
- Siyi Li
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Chibuzo Sampson
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Changhao Liu
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Hai-Long Piao
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, Shenyang, 110122, China.
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Cancer Research Institute, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
10
|
Khademi R, Malekzadeh H, Bahrami S, Saki N, Khademi R, Villa-Diaz LG. Regulation and Functions of α6-Integrin (CD49f) in Cancer Biology. Cancers (Basel) 2023; 15:3466. [PMID: 37444576 DOI: 10.3390/cancers15133466] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Over the past decades, our knowledge of integrins has evolved from being understood as simple cell surface adhesion molecules to receptors that have a complex range of intracellular and extracellular functions, such as delivering chemical and mechanical signals to cells. Consequently, they actively control cellular proliferation, differentiation, and apoptosis. Dysregulation of integrin signaling is a major factor in the development and progression of many tumors. Many reviews have covered the broader integrin family in molecular and cellular studies and its roles in diseases. Nevertheless, further understanding of the mechanisms specific to an individual subunit of different heterodimers is more useful. Thus, we describe the current understanding of and exploratory investigations on the α6-integrin subunit (CD49f, VLA6; encoded by the gene itga6) in normal and cancer cells. The roles of ITGA6 in cell adhesion, stemness, metastasis, angiogenesis, and drug resistance, and as a diagnosis biomarker, are discussed. The role of ITGA6 differs based on several features, such as cell background, cancer type, and post-transcriptional alterations. In addition, exosomal ITGA6 also implies metastatic organotropism. The importance of ITGA6 in the progression of a number of cancers, including hematological malignancies, suggests its potential usage as a novel prognostic or diagnostic marker and useful therapeutic target for better clinical outcomes.
Collapse
Affiliation(s)
- Rahele Khademi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
| | - Hossein Malekzadeh
- Department of Oral Medicine, Faculty of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Sara Bahrami
- Resident of Restorative Dentistry, Qazvin University of Medical Sciences, Qazvin 3419759811, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Reyhane Khademi
- Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno_TACT), Universal Scientific Education and Research Network (USERN), Tehran 1419733151, Iran
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 6135715794, Iran
- Department of Medical Laboratory Sciences, School of Para-Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz 6135715794, Iran
| | - Luis G Villa-Diaz
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, USA
- Department of Bioengineering, Oakland University, Rochester, MI 48309, USA
| |
Collapse
|
11
|
Antona A, Leo G, Favero F, Varalda M, Venetucci J, Faletti S, Todaro M, Mazzucco E, Soligo E, Saglietti C, Stassi G, Manfredi M, Pelicci G, Corà D, Valente G, Capello D. Targeting lysine-specific demethylase 1 (KDM1A/LSD1) impairs colorectal cancer tumorigenesis by affecting cancer cells stemness, motility, and differentiation. Cell Death Discov 2023; 9:201. [PMID: 37385999 DOI: 10.1038/s41420-023-01502-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/12/2023] [Accepted: 06/19/2023] [Indexed: 07/01/2023] Open
Abstract
Among all cancers, colorectal cancer (CRC) is the 3rd most common and the 2nd leading cause of death worldwide. New therapeutic strategies are required to target cancer stem cells (CSCs), a subset of tumor cells highly resistant to present-day therapy and responsible for tumor relapse. CSCs display dynamic genetic and epigenetic alterations that allow quick adaptations to perturbations. Lysine-specific histone demethylase 1A (KDM1A also known as LSD1), a FAD-dependent H3K4me1/2 and H3K9me1/2 demethylase, was found to be upregulated in several tumors and associated with a poor prognosis due to its ability to maintain CSCs staminal features. Here, we explored the potential role of KDM1A targeting in CRC by characterizing the effect of KDM1A silencing in differentiated and CRC stem cells (CRC-SCs). In CRC samples, KDM1A overexpression was associated with a worse prognosis, confirming its role as an independent negative prognostic factor of CRC. Consistently, biological assays such as methylcellulose colony formation, invasion, and migration assays demonstrated a significantly decreased self-renewal potential, as well as migration and invasion potential upon KDM1A silencing. Our untargeted multi-omics approach (transcriptomic and proteomic) revealed the association of KDM1A silencing with CRC-SCs cytoskeletal and metabolism remodeling towards a differentiated phenotype, supporting the role of KDM1A in CRC cells stemness maintenance. Also, KDM1A silencing resulted in up-regulation of miR-506-3p, previously reported to play a tumor-suppressive role in CRC. Lastly, loss of KDM1A markedly reduced 53BP1 DNA repair foci, implying the involvement of KDM1A in the DNA damage response. Overall, our results indicate that KDM1A impacts CRC progression in several non-overlapping ways, and therefore it represents a promising epigenetic target to prevent tumor relapse.
Collapse
Affiliation(s)
- Annamaria Antona
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy.
| | - Giovanni Leo
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Francesco Favero
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, Department of Translational Medicine, Università del Piemonte Orientale, Corso Trieste 15/A, 28100, Novara, Italy
| | - Marco Varalda
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Jacopo Venetucci
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Stefania Faletti
- Department of Experimental Oncology, IRCCS, European Institute of Oncology, Via Adamello 16, 20139, Milano, Italy
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche 2, 90127, Palermo, Italy
| | - Eleonora Mazzucco
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Enrica Soligo
- Pathology Unit, Ospedale Sant'Andrea, Corso Mario Abbiate 21, 13100, Vercelli, Italy
| | - Chiara Saglietti
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences, Università di Palermo, Via del Vespro 131, 90127, Palermo, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, Department of Translational Medicine, Università del Piemonte Orientale, Corso Trieste 15/A, 28100, Novara, Italy
| | - Giuliana Pelicci
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
- Department of Experimental Oncology, IRCCS, European Institute of Oncology, Via Adamello 16, 20139, Milano, Italy
| | - Davide Corà
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases, Department of Translational Medicine, Università del Piemonte Orientale, Corso Trieste 15/A, 28100, Novara, Italy
| | - Guido Valente
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
- Pathology Unit, Ospedale Sant'Andrea, Corso Mario Abbiate 21, 13100, Vercelli, Italy
| | - Daniela Capello
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100, Novara, Italy
| |
Collapse
|
12
|
Liu Y, Ge X, Li C, Xue T. Derivation and characterization of new cell line from intestine of turbot (Scophthalmus maximus). In Vitro Cell Dev Biol Anim 2023; 59:153-162. [PMID: 36809593 PMCID: PMC10073165 DOI: 10.1007/s11626-022-00746-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/09/2022] [Indexed: 02/23/2023]
Abstract
A continuous intestine cell line from turbot (Scophthalmus maximus) designated as SMI was established utilizing the tissue explant technique. Primary SMI cell was cultured at 24 °C in a medium with 20% fetal bovine serum (FBS), then subcultured in 10% FBS after 10 passages. Impacts of medium or temperature on the growth of SMI were examined and the results indicated it grew well in DMEM supplemented with 10% FBS at 24 °C. The SMI cell line was subcultured more than 60 times. Karyotyping, chromosome number, and ribosomal RNA genotyping analysis revealed that SMI had a modal diploid chromosome number of 44 and originated from turbot. After being transfected with pEGFP-N1 and FAM-siRNA, a large number of green fluorescence signals were observed in SMI, indicating that SMI could be used as an ideal platform to explore gene function in vitro. In addition, the expression of epithelium-associated genes such as itga6, itgb4, gja1, claudin1, zo-1, and E-cadherin in SMI suggested the SMI had some characteristics of epidermal cells. The upregulation of immune-associated genes such as TNF-β, NF-κB, and IL-1β in SMI after stimulation with pathogen-associated molecular patterns suggested the SMI might exhibit immune functions similar to the intestinal epithelium in vivo.
Collapse
Affiliation(s)
- Yiping Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xuefeng Ge
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Ting Xue
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
13
|
Single-Cell and Transcriptome-Based Immune Cell-Related Prognostic Model in Clear Cell Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2023; 2023:5355269. [PMID: 36925653 PMCID: PMC10014191 DOI: 10.1155/2023/5355269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/26/2022] [Accepted: 10/11/2022] [Indexed: 03/09/2023]
Abstract
Traditional studies mostly focus on the role of single gene in regulating clear cell renal cell carcinoma (ccRCC), while it ignores the impact of tumour heterogeneity on disease progression. The purpose of this study is to construct a prognostic risk model for ccRCC by analysing the differential marker genes related to immune cells in the single-cell database to provide help in clinical diagnosis and targeted therapy. Single-cell data and ligand-receptor relationship pair data were downloaded from related publications, and ccRCC phenotype and expression profile data were downloaded from TCGA and CPTAC. Based on the DEGs of each cluster acquired from single-cell data, immune cell marker genes, and ligand-receptor gene data, we constructed a multilayer network. Then, the genes in the network and the genes in TCGA were used to construct the WGCNA network, which screened out prognosis-associated genes for subsequent analysis. Finally, a prognostic risk scoring model was obtained, and CPTAC data showed that the effectiveness of this model was good. A nomogram based on the predictive model for predicting the overall survival was established, and internal validation was performed well. Our findings suggest that the predictive model built and based on the immune cell scRNA-seq will enable us to judge the prognosis of patients with ccRCC and provide more accurate directions for basic relevant research and clinical practice.
Collapse
|
14
|
Martel R, Shen ML, DeCorwin-Martin P, de Araujo LO, Juncker D. Extracellular Vesicle Antibody Microarray for Multiplexed Inner and Outer Protein Analysis. ACS Sens 2022; 7:3817-3828. [PMID: 36515500 PMCID: PMC9791990 DOI: 10.1021/acssensors.2c01750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteins are found both outside and inside of extracellular vesicles (EVs) and govern the properties and functions of EVs, while also constituting a signature of the cell of origin and of biological function and disease. Outer proteins on EVs can be directly bound by antibodies to either enrich EVs, or probe the expression of a protein on EVs, including in a combinatorial manner. However, co-profiling of inner proteins remains challenging. Here, we present the high-throughput, multiplexed analysis of EV inner and outer proteins (EVPio). We describe the optimization of fixation and heat-induced protein epitope retrieval for EVs, along with oligo-barcoded antibodies and branched DNA signal amplification for sensitive, multiplexed, and high-throughput assays. We captured four subpopulations of EVs from colorectal cancer (CRC) cell lines HT29 and SW403 based on EpCAM, CD9, CD63, and CD81 expression, and quantified the co-expression of eight outer [integrins (ITGs) and tetraspanins] and four inner (heat shock, endosomal, and inner leaflet) proteins. The differences in co-expression patterns were consistent with the literature and known biological function. In conclusion, EVPio analysis can simultaneously detect multiple inner and outer proteins in EVs immobilized on a surface, opening the way to extensive combinatorial protein profiles for both discovery and clinical translation.
Collapse
Affiliation(s)
- Rosalie Martel
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Molly L. Shen
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Philippe DeCorwin-Martin
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - Lorenna Oliveira
Fernandes de Araujo
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada
| | - David Juncker
- Biomedical
Engineering Department, McGill University, Montreal, Quebec H3A 2B4, Canada,McGill
Genome Centre, McGill University, Montreal, Quebec H3A 0G1, Canada,
| |
Collapse
|
15
|
Combining single-cell tracking and omics improves blood stem cell fate regulator identification. Blood 2022; 140:1482-1495. [PMID: 35820055 PMCID: PMC9523371 DOI: 10.1182/blood.2022016880] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022] Open
Abstract
Molecular programs initiating cell fate divergence (CFD) are difficult to identify. Current approaches usually compare cells long after CFD initiation, therefore missing molecular changes at its start. Ideally, single cells that differ in their CFD molecular program but are otherwise identical are compared early in CFD. This is possible in diverging sister cells, which were identical until their mother's division and thus differ mainly in CFD properties. In asymmetrically dividing cells, divergent daughter fates are prospectively committed during division, and diverging sisters can thus be identified at the start of CFD. Using asymmetrically dividing blood stem cells, we developed a pipeline (ie, trackSeq) for imaging, tracking, isolating, and transcriptome sequencing of single cells. Their identities, kinship, and histories are maintained throughout, massively improving molecular noise filtering and candidate identification. In addition to many identified blood stem CFD regulators, we offer here this pipeline for use in CFDs other than asymmetric division.
Collapse
|
16
|
CircRNA-Associated ceRNA Network Reveals Focal Adhesion and Metabolism Pathways in Neuropathic Pain. JOURNAL OF ONCOLOGY 2022; 2022:7246904. [PMID: 36065302 PMCID: PMC9440820 DOI: 10.1155/2022/7246904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/20/2022] [Accepted: 07/05/2022] [Indexed: 11/17/2022]
Abstract
Background. Increasing evidence has shown that noncoding RNAs perform a remarkable function in neuropathic pain (NP); nonetheless, the mechanisms underlying the modulation of competitive endogenous RNA in NP remain uncertain. The goal of this research was to investigate the molecular processes underlying NP. Methods. We utilized the Gene Expression Omnibus (GEO) to obtain NP-related microarray datasets that included the expression patterns of circular RNAs (circRNAs) and messenger RNAs (mRNAs). Following that, bioinformatics analyses and a molecular biology experiment were carried out. Results. According to the findings, carrying out enrichment studies of the targeted genes had an impact on a variety of NP-related pathways. Notably, we isolated a ceRNA subnetwork incorporating two upregulated circRNAs (Esrrg and Map3k3) which primarily participate in the focal adhesion pathway by regulating Integrin Subunit Beta 4 (ITGB4) and two downregulated circRNAs (Dgkb and Atp2a2), which potentially regulate metabolism-related molecule Lipase A (LIPA). Conclusions. According to our findings, the focal adhesion and metabolic signaling pathways could be critical in the advancement of NP, and some circRNA might regulate this biological process through the ceRNA network, which might offer pertinent insights into the underlying mechanisms.
Collapse
|
17
|
Mattila TT, Patankar M, Väyrynen JP, Klintrup K, Mäkelä J, Tuomisto A, Nieminen P, Mäkinen MJ, Karttunen TJ. Putative anoikis resistant subpopulations are enriched in lymph node metastases and indicate adverse prognosis in colorectal carcinoma. Clin Exp Metastasis 2022; 39:883-898. [PMID: 36018456 PMCID: PMC9637608 DOI: 10.1007/s10585-022-10184-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022]
Abstract
Anoikis refers to apoptosis induced by the loss of contact with the extracellular matrix. Anoikis resistance is essential for metastasis. We have recently shown that it is possible to quantitatively evaluate putative anoikis resistant (AR) subpopulations in colorectal carcinoma (CRC). Abundance of these multi-cell structures is an independent marker of adverse prognosis. Here, we have quantified putative AR subpopulations in lymph node (LN) metastases of CRC and evaluated their prognostic value and relationship with the characteristics of primary tumors. A case series included 137 unselected CRC patients, 54 with LN metastases. Areal densities (structures/mm2) of putative AR structures in primary tumors had been analyzed previously and now were determined from all nodal metastases (n = 183). Areal density of putative AR structures was higher in LN metastases than in primary tumors. Variation of the areal density within different LN metastases of a single patient was lower than between metastases of different patients. Abundance of putative AR structures in LN metastases was associated with shorter cancer specific survival (p = 0.013), and this association was independent of T and N stages. Abundance of putative AR structures in primary tumors and LN metastases had a cumulative adverse effect on prognosis. Enrichment of putative AR subpopulations in LN metastases suggest that in metastasis formation, there is a selection favoring cells capable of forming these structures. Higher intra-case constancy relative to inter-case variation suggests that such selection is stable in metastasis development. Our findings indirectly support the biological validity of our concept of putative AR structures.
Collapse
Affiliation(s)
- Taneli T Mattila
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Madhura Patankar
- Division of Gastroenterology and Hepatology, Department of Medicine, Keck School of Medicine of USC, Los Angeles, CA, 90089-0110, USA
| | - Juha P Väyrynen
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Kai Klintrup
- Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland.,Department of Surgery, Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, POB 5000, 90014, Oulu, Finland
| | - Jyrki Mäkelä
- Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland.,Department of Surgery, Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, POB 5000, 90014, Oulu, Finland
| | - Anne Tuomisto
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Pentti Nieminen
- Medical Informatics and Data Analysis Research Group, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - Markus J Mäkinen
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Tuomo J Karttunen
- Department of Pathology, Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland. .,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland.
| |
Collapse
|
18
|
Okamoto T, Natsume Y, Doi M, Nosato H, Iwaki T, Yamanaka H, Yamamoto M, Kawachi H, Noda T, Nagayama S, Sakanashi H, Yao R. Integration of human inspection and AI-based morphological typing of PDOs reveals inter-patient heterogeneity of colorectal cancer. Cancer Sci 2022; 113:2693-2703. [PMID: 35585758 PMCID: PMC9357621 DOI: 10.1111/cas.15396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022] Open
Abstract
Colorectal cancer (CRC) is a heterogenous disease, and patients have differences in therapeutic response. However, the mechanisms underlying inter-patient heterogeneity in the response to chemotherapeutic agents remain to be elucidated, and molecular tumor characteristics are required to select patients for specific therapies. Patient-derived organoids (PDOs) established from CRCs recapitulate various biological characteristics of tumor tissues, including cellular heterogeneity and the response to chemotherapy. PDOs established from CRCs exhibit various morphologies, but there are no criteria for defining these morphologies, which hampers the analysis of their biological significance. Here, we developed an artificial intelligence (AI)-based classifier to categorize PDOs based on microscopic images according to their similarity in appearance and classified tubular adenocarcinoma-derived PDOs into six types. Transcriptome analysis identified differential expression of genes related to cell adhesion in some of the morphological types. Genes involved in ribosome biogenesis were also differentially expressed and were most highly expressed in morphological types exhibiting CRC stem cell properties. We identified an RNA polymerase I inhibitor, CX-5641, to be an upstream regulator of these type-specific gene sets. Notably, PDO types with increased expression of genes involved in ribosome biogenesis were resistant to CX-5461 treatment. Taken together, these results uncover the biological significance of the morphology of PDOs and provide novel indicators by which to categorize CRCs. Therefore, the AI-based classifier is a useful tool to support PDO-based cancer research.
Collapse
Affiliation(s)
- Takuya Okamoto
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan.,Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuko Natsume
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Motomichi Doi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Hirokazu Nosato
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Toshiyuki Iwaki
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Hitomi Yamanaka
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Mayuko Yamamoto
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| | - Hiroshi Kawachi
- Division of Pathology, Cancer Institute; Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tetsuo Noda
- Director's office, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Nagayama
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.,Department of Surgery, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Hidenori Sakanashi
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Ryoji Yao
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research (JFCR), Tokyo, Japan
| |
Collapse
|
19
|
Sharma C, Hemler ME. Antioxidant and Anticancer Functions of Protein Acyltransferase DHHC3. Antioxidants (Basel) 2022; 11:antiox11050960. [PMID: 35624824 PMCID: PMC9137668 DOI: 10.3390/antiox11050960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/29/2022] [Accepted: 05/06/2022] [Indexed: 11/16/2022] Open
Abstract
Silencing of DHHC3, an acyltransferase enzyme in the DHHC family, extensively upregulates oxidative stress (OS). Substrates for DHHC3-mediated palmitoylation include several antioxidant proteins and many other redox regulatory proteins. This helps to explain why DHHC3 ablation upregulates OS. DHHC3 also plays a key role in cancer. DHHC3 ablation leads to diminished xenograft growth of multiple cancer cell types, along with diminished metastasis. Furthermore, DHHC3 protein is upregulated on malignant/metastatic cancer samples, and upregulated gene expression correlates with diminished patient survival in several human cancers. Decreased primary tumor growth due to DHHC3 ablation may be partly explained by an elevated OS → senescence → innate immune cell recruitment mechanism. Elevated OS due to DHHC3 ablation may also contribute to adaptive anticancer immunity and impair tumor metastasis. In addition, DHHC3 ablation disrupts antioxidant protection mechanisms, thus enhancing the efficacy of OS-inducing anticancer drugs. A major focus has thus far been on OS regulation by DHHC3. However, remaining to be studied are multiple DHHC3 substrates that may affect tumor behavior independent of OS. Nonetheless, the currently established properties of DHHC3 make it an attractive candidate for therapeutic targeting in situations in which antioxidant protections need to be downmodulated, and also in cancer.
Collapse
|
20
|
Harryman WL, Marr KD, Nagle RB, Cress AE. Integrins and Epithelial-Mesenchymal Cooperation in the Tumor Microenvironment of Muscle-Invasive Lethal Cancers. Front Cell Dev Biol 2022; 10:837585. [PMID: 35300411 PMCID: PMC8921537 DOI: 10.3389/fcell.2022.837585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/04/2022] [Indexed: 11/18/2022] Open
Abstract
Muscle-invasive lethal carcinomas traverse into and through this specialized biophysical and growth factor enriched microenvironment. We will highlight cancers that originate in organs surrounded by smooth muscle, which presents a barrier to dissemination, including prostate, bladder, esophageal, gastric, and colorectal cancers. We propose that the heterogeneity of cell-cell and cell-ECM adhesion receptors is an important driver of aggressive tumor networks with functional consequences for progression. Phenotype heterogeneity of the tumor provides a biophysical advantage for tumor network invasion through the tensile muscle and survival of the tumor network. We hypothesize that a functional epithelial-mesenchymal cooperation (EMC)exists within the tumor invasive network to facilitate tumor escape from the primary organ, invasion and traversing of muscle, and navigation to metastatic sites. Cooperation between specific epithelial cells within the tumor and stromal (mesenchymal) cells interacting with the tumor is illustrated using the examples of laminin-binding adhesion molecules—especially integrins—and their response to growth and inflammatory factors in the tumor microenvironment. The cooperation between cell-cell (E-cadherin, CDH1) and cell-ECM (α6 integrin, CD49f) expression and growth factor receptors is highlighted within poorly differentiated human tumors associated with aggressive disease. Cancer-associated fibroblasts are examined for their role in the tumor microenvironment in generating and organizing various growth factors. Cellular structural proteins are potential utility markers for future spatial profiling studies. We also examine the special characteristics of the smooth muscle microenvironment and how invasion by a primary tumor can alter this environment and contribute to tumor escape via cooperation between epithelial and stromal cells. This cooperative state allows the heterogenous tumor clusters to be shaped by various growth factors, co-opt or evade immune system response, adapt from hypoxic to normoxic conditions, adjust to varying energy sources, and survive radiation and chemotherapeutic interventions. Understanding the epithelial-mesenchymal cooperation in early tumor invasive networks holds potential for both identifying early biomarkers of the aggressive transition and identification of novel agents to prevent the epithelial-mesenchymal cooperation phenotype. Epithelial-mesenchymal cooperation is likely to unveil new tumor subtypes to aid in selection of appropriate therapeutic strategies.
Collapse
Affiliation(s)
- William L Harryman
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ, United States
| | - Kendra D Marr
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ, United States.,Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ, United States.,Medical Scientist Training Program, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Ray B Nagle
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ, United States.,Department of Pathology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Anne E Cress
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona Cancer Center, Tucson, AZ, United States.,Department of Cellular and Molecular Medicine and Department of Radiation Oncology, College of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
21
|
Ying L, Liang C, Zhang Y, Wang J, Wang C, Xia K, Shi K, Yu C, Yang B, Xu H, Zhang Y, Shu J, Huang X, Xing H, Li F, Zhou X, Chen Q. Enhancement of nucleus pulposus repair by glycoengineered adipose-derived mesenchymal cells. Biomaterials 2022; 283:121463. [DOI: 10.1016/j.biomaterials.2022.121463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 03/01/2022] [Accepted: 03/05/2022] [Indexed: 12/28/2022]
|
22
|
Zhang W, Li Y, Chen G, Yang X, Hu J, Zhang X, Feng G, Wang H. Integrin α6-Targeted Molecular Imaging of Central Nervous System Leukemia in Mice. Front Bioeng Biotechnol 2022; 10:812277. [PMID: 35284414 PMCID: PMC8905628 DOI: 10.3389/fbioe.2022.812277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/17/2022] [Indexed: 11/14/2022] Open
Abstract
Central nervous system leukemia (CNS-L) is caused by leukemic cells infiltrating into the meninges or brain parenchyma and remains the main reason for disease relapse. Currently, it is hard to detect CNS-L accurately by clinically available imaging models due to the relatively low amount of tumor cells, confined blood supply, and the inferior glucose metabolism intensity. Recently, integrin α6-laminin interactions have been identified to mediate CNS-L, which suggests that integrin α6 may be a promising molecular imaging target for the detection of CNS-L. The acute lymphoblastic leukemia (ALL) cell line NALM6 stabled and transfected with luciferase was used to establish the CNS-L mouse model. CNS-L-bearing mice were monitored and confirmed by bioluminescence imaging. Three of our previously developed integrin α6-targeted peptide-based molecular imaging agents, Cy5-S5 for near-infrared fluorescence (NIRF), Gd-S5 for magnetic resonance (MR), and 18F-S5 for positron emission tomography (PET) imaging, were employed for the molecular imaging of these CNS-L-bearing mice. Bioluminescence imaging showed a local intensive signal in the heads among CNS-L-bearing mice; meanwhile, Cy5-S5/NIRF imaging produced intensive fluorescence intensity in the same head regions. Moreover, Gd-S5/MR imaging generated superior MR signal enhancement at the site of meninges, which were located between the skull bone and brain parenchyma. Comparatively, MR imaging with the clinically available MR enhancer Gd-DTPA did not produce the distinguishable MR signal in the same head regions. Additionally, 18F-S5/PET imaging also generated focal radio-concentration at the same head regions, which generated nearly 5-times tumor-to-background ratio compared to the clinically available PET radiotracer 18F-FDG. Finally, pathological examination identified layer-displayed leukemic cells in the superficial part of the brain parenchyma tissue, and immunohistochemical staining confirmed the overexpression of the integrin α6 within the lesion. These findings suggest the potential application of these integrin α6-targeted molecular imaging agents for the accurate detection of CNS-L.
Collapse
Affiliation(s)
- Wenbiao Zhang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yongjiang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guanjun Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaochun Yang
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Junfeng Hu
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaofei Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- *Correspondence: Xiaofei Zhang, ; Guokai Feng, ; Hua Wang,
| | - Guokai Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- *Correspondence: Xiaofei Zhang, ; Guokai Feng, ; Hua Wang,
| | - Hua Wang
- Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
- *Correspondence: Xiaofei Zhang, ; Guokai Feng, ; Hua Wang,
| |
Collapse
|
23
|
Mesenchymal stem cell (MSC)-derived exosomes as novel vehicles for delivery of miRNAs in cancer therapy. Cancer Gene Ther 2022; 29:1105-1116. [PMID: 35082400 DOI: 10.1038/s41417-022-00427-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/11/2021] [Accepted: 01/14/2022] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are known as promising sources for cancer therapy and can be utilized as vehicles in cancer gene therapy. MSC-derived exosomes are central mediators in the therapeutic functions of MSCs, known as the novel cell-free alternatives to MSC-based cell therapy. MSC-derived exosomes show advantages including higher safety as well as more stability and convenience for storage, transport and administration compared to MSCs transplant therapy. Unmodified MSC-derived exosomes can promote or inhibit tumors while modified MSC-derived exosomes are involved in the suppression of cancer development and progression via the delivery of several therapeutics molecules including chemotherapeutic drugs, miRNAs, anti-miRNAs, specific siRNAs, and suicide gene mRNAs. In most malignancies, dysregulation of miRNAs not only occurs as a consequence of cancer progression but also is directly involved during tumor initiation and development due to their roles as oncogenes (oncomiRs) or tumor suppressors (TS-miRNAs). MiRNA restoration is usually achieved by overexpression of TS-miRNAs using synthetic miRNA mimics and viral vectors or even downregulation of oncomiRs using anti-miRNAs. Similar to other therapeutic molecules, the efficacy of miRNAs restoration in cancer therapy depends on the effectiveness of the delivery system. In the present review, we first provided an overview of the properties and potentials of MSCs in cancer therapy as well as the application of MSC-derived exosomes in cancer therapy. Finally, we specifically focused on harnessing the MSC-derived exosomes for the aim of miRNA delivery in cancer therapy.
Collapse
|
24
|
Yu J, Yang K, Zheng J, Sun X, Zhao W. Establishment of a novel prognostic signature based on an identified expression profile of integrin superfamily to predict overall survival of patients with colorectal adenocarcinoma. Gene 2022; 808:145990. [PMID: 34624456 DOI: 10.1016/j.gene.2021.145990] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/14/2021] [Accepted: 10/01/2021] [Indexed: 12/02/2022]
Abstract
The abnormal expression of integrin superfamily members commonly related to kinds of malignancies. However, the role of integrins in predicting the prognosis of cancers is still little known, especially for colorectal cancer that is one of the leading causes of cancer-related death. RNA-seq data and clinical features of colorectal adenocarcinoma (COAD) patients were derived from The Cancer Genome Atlas (TCGA), used to analyze the expression pattern and genomic alterations of integrin genes in the COAD cohort. Unsupervised hierarchical clustering divided COAD patients into two clusters (clusters 1 & 2), and we observed that patients in cluster 2 with high expressions of most integrin genes had worse clinical features and shorter overall survival (a median OS: 67.25 months vs 99.93 months, p = 0.012), compared to those in cluster 1. Combined with univariate Cox regression analysis, Pearson Correlation Coefficients (PCC), and Principal Component Analysis (PCA), an integrin-related signature was established, including ITGA1, ITGA5, ITGA7, ITGA11, ITGAX, ITGAM, ITGB1, and ITGB5. And the AUC values for OS at 1, 3, and 5 years was 0.61, 0.59, and 0.56, further demonstrating the predicting capacity of our signature. Furthermore, overexpression of which also significantly correlated with poorer prognosis of colon cancer patients in a separate validation cohort, GSE17536 (p < 0.05). Meanwhile, the AUC values for OS in the validation cohort at 1, 3, and 5 years was 0.62, 0.59, and 0.59. Additionally, enrichment analysis indicated significant differences between cluster 1 and cluster 2 in the biological processes of cell adhesion, signal transduction, extracellular matrix, immune system, and in tumor microenvironment (TME), which were crucial to the progression of tumor. The findings supplied compelling evidence that our signature could be a novel prognostic biomarker for COAD patients, and these genes had the potential to be therapeutic targets.
Collapse
Affiliation(s)
- Junhui Yu
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| | - Kui Yang
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| | - Jianbao Zheng
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| | - Wei Zhao
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, People's Republic of China.
| |
Collapse
|
25
|
Hou S, Wang J, Li W, Hao X, Hang Q. Roles of Integrins in Gastrointestinal Cancer Metastasis. Front Mol Biosci 2021; 8:708779. [PMID: 34869579 PMCID: PMC8634653 DOI: 10.3389/fmolb.2021.708779] [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: 05/12/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Integrins are a large family of heterodimeric transmembrane receptors which mediate cell adhesion and transmit signals to the cell interior. The mechanistic roles of integrins have long been an enigma in cancer, given its complexity in regulating different cellular behaviors. Recently, however, increasing research is providing new insights into its function and the underlying mechanisms, which collectively include the influences of altered integrin expression on the aberrant signaling pathways and cancer progression. Many studies have also demonstrated the potentiality of integrins as therapeutic targets in cancer treatment. In this review, we have summarized these recent reports and put a particular emphasis on the dysregulated expression of integrins and how they regulate related signaling pathways to facilitate the metastatic progression of gastrointestinal cancer, including gastric cancer (GC) and colorectal cancer (CRC), which will address the crucial roles of integrins in gastrointestinal cancer.
Collapse
Affiliation(s)
- Sicong Hou
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jiaxin Wang
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Wenqian Li
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Xin Hao
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Qinglei Hang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
26
|
Hu LF, Lan HR, Huang D, Li XM, Jin KT. Personalized Immunotherapy in Colorectal Cancers: Where Do We Stand? Front Oncol 2021; 11:769305. [PMID: 34888246 PMCID: PMC8649954 DOI: 10.3389/fonc.2021.769305] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer death in the world. Immunotherapy using monoclonal antibodies, immune-checkpoint inhibitors, adoptive cell therapy, and cancer vaccines has raised great hopes for treating poor prognosis metastatic CRCs that are resistant to the conventional therapies. However, high inter-tumor and intra-tumor heterogeneity hinder the success of immunotherapy in CRC. Patients with a similar tumor phenotype respond differently to the same immunotherapy regimen. Mutation-based classification, molecular subtyping, and immunoscoring of CRCs facilitated the multi-aspect grouping of CRC patients and improved immunotherapy. Personalized immunotherapy using tumor-specific neoantigens provides the opportunity to consider each patient as an independent group deserving of individualized immunotherapy. In the recent decade, the development of sequencing and multi-omics techniques has helped us classify patients more precisely. The expansion of such advanced techniques along with the neoantigen-based immunotherapy could herald a new era in treating heterogeneous tumors such as CRC. In this review article, we provided the latest findings in immunotherapy of CRC. We elaborated on the heterogeneity of CRC patients as a bottleneck of CRC immunotherapy and reviewed the latest advances in personalized immunotherapy to overcome CRC heterogeneity.
Collapse
Affiliation(s)
- Li-Feng Hu
- Department of Colorectal Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Dong Huang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xue-Min Li
- Department of Hepatobiliary Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
27
|
Proteomic characterization of GSK3β knockout shows altered cell adhesion and metabolic pathway utilisation in colorectal cancer cells. PLoS One 2021; 16:e0246707. [PMID: 34739494 PMCID: PMC8570494 DOI: 10.1371/journal.pone.0246707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 10/01/2021] [Indexed: 11/22/2022] Open
Abstract
Glycogen-specific kinase (GSK3β) is an integral regulator of the Wnt signalling pathway as well as many other diverse signalling pathways and processes. Dys-regulation of GSK3β is implicated in many different pathologies, including neurodegenerative disorders as well as many different tumour types. In the context of tumour development, GSK3β has been shown to play both oncogenic and tumour suppressor roles, depending upon tissue, signalling environment or disease progression. Although multiple substrates of the GSK3β kinase have been identified, the wider protein networks within which GSK3β participates are not well known, and the consequences of these interactions not well understood. In this study, LC-MS/MS expression analysis was performed using knockout GSK3β colorectal cancer cells and isogenic controls in colorectal cancer cell lines carrying dominant stabilizing mutations of β-catenin. Consistent with the role of GSK3β, we found that β-catenin levels and canonical Wnt activity are unaffected by knockout of GSK3β and therefore used this knockout cell model to identify other processes in which GSK3β is implicated. Quantitative proteomic analysis revealed perturbation of proteins involved in cell-cell adhesion, and we characterized the phenotype and altered proteomic profiles associated with this. We also characterized the perturbation of metabolic pathways resulting from GSK3β knockout and identified defects in glycogen metabolism. In summary, using a precision colorectal cancer cell-line knockout model with constitutively activated β-catenin we identified several of the diverse pathways and processes associated with GSK3β function.
Collapse
|
28
|
Przygodzka P, Sochacka E, Soboska K, Pacholczyk M, Papiewska-Pająk I, Przygodzki T, Płociński P, Ballet S, De Prins A, Boncela J. Neuromedin U induces an invasive phenotype in CRC cells expressing the NMUR2 receptor. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:283. [PMID: 34493299 PMCID: PMC8422652 DOI: 10.1186/s13046-021-02073-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022]
Abstract
Background Successful colorectal cancer (CRC) therapy often depends on the accurate identification of primary tumours with invasive potential. There is still a lack of identified pathological factors associated with disease recurrence that could help in making treatment decisions. Neuromedin U (NMU) is a secretory neuropeptide that was first isolated from the porcine spinal cord, and it has emerged as a novel factor involved in the tumorigenesis and/or metastasis of many types of cancers. Previously associated with processes leading to CRC cell invasiveness, NMU has the potential to be a marker of poor outcome, but it has not been extensively studied in CRC. Methods Data from The Cancer Genome Atlas (TCGA) were used to analyse NMU and NMU receptor (NMUR1 and NMUR2) expression in CRC tissues vs. normal tissues, and real-time PCR was used for NMU and NMU receptor expression analysis. NMU protein detection was performed by immunoblotting. Secreted NMU was immunoprecipitated from cell culture-conditioned media and analysed by immunoblotting and protein sequencing. DNA demethylation by 5-aza-CdR was used to analyse the regulation of NMUR1 and NMUR2 expression. NMU receptor activity was monitored by detecting calcium mobilisation in cells loaded with fluo-4, and ERK1/2 kinase activation was detected after treatment with NMU or receptor agonist. Cell migration and invasion were investigated using membrane filters. Integrin expression was evaluated by flow cytometry. Results The obtained data revealed elevated expression of NMU and NMUR2 in CRC tissue samples and variable expression in the analysed CRC cell lines. We have shown, for the first time, that NMUR2 activation induces signalling in CRC cells and that NMU increases the motility and invasiveness of NMUR2-positive CRC cells and increases prometastatic integrin receptor subunit expression. Conclusions Our results show the ability of CRC cells to respond to NMU via activation of the NMUR2 receptor, which ultimately leads to a shift in the CRC phenotype towards a more invasive phenotype. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02073-8.
Collapse
Affiliation(s)
- Patrycja Przygodzka
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland.
| | - Ewelina Sochacka
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland.,Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Kamila Soboska
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland.,Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Marcin Pacholczyk
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 16, 44-100, Gliwice, Poland
| | | | - Tomasz Przygodzki
- Department of Haemostasis and Haemostatic Disorders, Chair of Biomedical Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-235, Lodz, Poland
| | - Przemysław Płociński
- Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237, Lodz, Poland
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - An De Prins
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Joanna Boncela
- Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232, Lodz, Poland
| |
Collapse
|
29
|
Kamalkazemi E, Abedi-Gaballu F, Mohammad Hosseini TF, Mohammadi A, Mansoori B, Dehghan G, Baradaran B, Sheibani N. Glimpse into Cellular Internalization and Intracellular Trafficking of Lipid-Based Nanoparticles in Cancer Cells. Anticancer Agents Med Chem 2021; 22:1897-1912. [PMID: 34488605 DOI: 10.2174/1871520621666210906101421] [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: 12/18/2020] [Revised: 06/14/2021] [Accepted: 06/27/2021] [Indexed: 11/22/2022]
Abstract
Lipid-based nanoparticles as drug delivery carriers have been mainly used for delivery of anti-cancer therapeutic agents. Lipid-based nanoparticles, due to their smaller particle size and similarity to cell membranes, are readily internalized into cancer cells. Interestingly, cancer cells also overexpress receptors for specific ligands including folic acid, hyaluronic acid, and transferrin on their surface. This allows the use of these ligands for surface modification of the lipid-based nanoparticle. These modifications then allow the specific recognition of these ligand-coated nanoparticles by their receptors on cancer cells allowing the targeted gradual intracellular accumulation of the functionalized nanoplatforms. These interactions could eventually enhance the internalization of desired drugs via increasing ligand-receptor mediated cellular uptake of the nanoplatforms. The cellular internalization of the nanoplatforms also varies and depends on their physicochemical properties including particle size, zeta potential, and shape. The cellular uptake is also influenced by the types of ligand internalization pathway utilized by cells such as phagocytosis, macropinocytosis, and multiple endocytosis pathways. In this review, we will classify and discuss lipid based nanoparticles engineered to express specific ligands, and are recognized by their receptors on cancer cell, and their cellular internalization pathways. Moreover, the intracellular fate of nanoparticles decorated with specific ligands and the best internalization pathways (caveolae mediated endocytosis) for safe cargo delivery will be discussed.
Collapse
Affiliation(s)
- Elham Kamalkazemi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | | | | | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Nader Sheibani
- Departments of Ophthalmology and Visual Sciences, Biomedical Engineering, and Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI . United States
| |
Collapse
|
30
|
Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, Qiao Y. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther 2021; 6:153. [PMID: 33888679 PMCID: PMC8062524 DOI: 10.1038/s41392-021-00544-0] [Citation(s) in RCA: 340] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/17/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is one of the major components of tumors that plays multiple crucial roles, including mechanical support, modulation of the microenvironment, and a source of signaling molecules. The quantity and cross-linking status of ECM components are major factors determining tissue stiffness. During tumorigenesis, the interplay between cancer cells and the tumor microenvironment (TME) often results in the stiffness of the ECM, leading to aberrant mechanotransduction and further malignant transformation. Therefore, a comprehensive understanding of ECM dysregulation in the TME would contribute to the discovery of promising therapeutic targets for cancer treatment. Herein, we summarized the knowledge concerning the following: (1) major ECM constituents and their functions in both normal and malignant conditions; (2) the interplay between cancer cells and the ECM in the TME; (3) key receptors for mechanotransduction and their alteration during carcinogenesis; and (4) the current therapeutic strategies targeting aberrant ECM for cancer treatment.
Collapse
Affiliation(s)
- Jiacheng Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Lele Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Dalong Wan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shengzhang Lin
- School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310000, China.
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China.
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China.
| |
Collapse
|
31
|
Bu T, Wang L, Wu X, Li L, Mao B, Wong CKC, Perrotta A, Silvestrini B, Sun F, Cheng CY. A laminin-based local regulatory network in the testis that supports spermatogenesis. Semin Cell Dev Biol 2021; 121:40-52. [PMID: 33879391 DOI: 10.1016/j.semcdb.2021.03.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
In adult rat testes, the basement membrane is structurally constituted by laminin and collagen chains that lay adjacent to the blood-testis barrier (BTB). It plays a crucial scaffolding role to support spermatogenesis. On the other hand, laminin-333 comprised of laminin-α3/ß3/γ3 at the apical ES (ectoplasmic specialization, a testis-specific cell-cell adherens junction at the Sertoli cell-step 8-19 spermatid interface) expressed by spermatids serves as a unique cell adhesion protein that forms an adhesion complex with α6ß1-integrin expressed by Sertoli cells to support spermiogenesis. Emerging evidence has shown that biologically active fragments are derived from basement membrane and apical ES laminin chains through proteolytic cleavage mediated by matrix metalloproteinase 9 (MMP9) and MMP2, respectively. Two of these laminin bioactive fragments: one from the basement membrane laminin-α2 chain called LG3/4/5-peptide, and one from the apical ES laminin-γ3 chain known as F5-peptide, are potent regulators that modify cell adhesion function at the Sertoli-spermatid interface (i.e., apical ES) but also at the Sertoli cell-cell interface designated basal ES at the blood-testis barrier (BTB) with contrasting effects. These findings not only highlight the physiological significance of these bioactive peptides that create a local regulatory network to support spermatogenesis, they also open a unique area of research. For instance, it is likely that several other bioactive peptides remain to be identified. These bioactive peptides including their downstream signaling proteins and cascades should be studied collectively in future investigations to elucidate the underlying mechanism(s) by which they coordinate with each other to maintain spermatogenesis. This is the goal of this review.
Collapse
Affiliation(s)
- Tiao Bu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Xiaolong Wu
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Baiping Mao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Chris K C Wong
- Department of Biology, Croucher Institute for Environmental Sciences, Hong Kong Baptist University, Kowloon, Hong Kong, China
| | - Adolfo Perrotta
- Department of Translational & Precision Medicine, La Sapienza University of Rome, 00185 Rome, Italy
| | | | - Fei Sun
- Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Institute of Reproductive Medicine, Nantong University School of Medicine, Nantong, Jiangsu 226001, China.
| |
Collapse
|
32
|
An autophagy-related prognostic signature associated with immune microenvironment features of uveal melanoma. Biosci Rep 2021; 41:228037. [PMID: 33682883 PMCID: PMC7982771 DOI: 10.1042/bsr20203812] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/02/2021] [Accepted: 03/04/2021] [Indexed: 12/22/2022] Open
Abstract
Autophagy is involved in cancer initiation and progression but its role in uveal melanoma (UM) was rarely investigated. Herein, we built an autophagy-related gene (ARG) risk model of UM patients by univariate Cox regression and least absolute shrinkage and selection operator (Lasso) regression model and filtrated out nine prognostic ARGs in The Cancer Genome Atlas (TCGA) cohort. Survival and Receiver Operating Characteristic (ROC) Curve analysis in the TCGA and other four independent UM cohorts (GSE22138, GSE27831, GSE44295 and GSE84976) proved that the ARG-signature possessed robust and steady prognosis predictive ability. We calculated risk scores for patients included in our study and patients with higher risk scores showed worse clinical outcomes. We found the expressions of the nine ARGs were significantly associated with clinical and molecular features (including risk score) and overall survival (OS) of UM patients. Furthermore, we utilized univariate and multivariate Cox regression analyses to determine the independent prognostic ability of the ARG-signature. Functional enrichment analysis showed the ARG-signature was correlated with several immune-related processes and pathways like T-cell activation and T-cell receptor signaling pathway. Gene set enrichment analysis (GSEA) found tumor hallmarks including angiogenesis, IL6-JAK-STAT3-signaling, reactive oxygen species pathway and oxidative phosphorylation were enriched in high-risk UM patients. Finally, infiltrations of several immune cells and immune-related scores were found significantly associated with the ARG-signature. In conclusion, the ARG-signature might be a strong predictor for evaluating the prognosis and immune infiltration of UM patients.
Collapse
|
33
|
Shahraki N, Mehrabian A, Amiri-Darban S, Moosavian SA, Jaafari MR. Preparation and characterization of PEGylated liposomal Doxorubicin targeted with leptin-derived peptide and evaluation of their anti-tumor effects, in vitro and in vivo in mice bearing C26 colon carcinoma. Colloids Surf B Biointerfaces 2021; 200:111589. [PMID: 33545570 DOI: 10.1016/j.colsurfb.2021.111589] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/18/2021] [Accepted: 01/20/2021] [Indexed: 12/16/2022]
Abstract
Employing targeting ligands on the surface of liposomes has the great potential to improve therapeutic efficacy and decreases off-target effects of liposomal formulations. In the present study, a leptin-derived peptide (Lp31) was evaluated to optimize the therapeutic efficacy of PEGylated liposomal Doxorubicin (PLD, Caelyx®). Leptin is an appetite regulatory hormone that is secreted into the blood circulation by the adipose tissue and it functions via its over expressed receptors (Ob-R) in a wide variety of cancers. Lp31, as targeting ligand, was conjugated to Maleimide-PEG2000-DSPE and then post-inserted into Caelyx. The anti-tumor activity and therapeutic efficacy of leptin modified Caelyx were evaluated and compared with Caelyx. The in vitro experiments demonstrated enhanced cytotoxicity and cellular uptake of Lp31-targeted Caelyx in C26 cell line compared to Caelyx. In BALB/c mice bearing C-26 murine carcinoma, Lp31 modified Caelyx groups exhibited significantly higher doxorubicin concentration at tumor tissue. Furthermore, Lp31 modified Caelyx at the dose of 10 mg/kg resulted in significant tumor growth inhibition and enhanced survival time compared to Caelyx. According to these results, the novel Lp31-liposomal doxorubicin offers great promise for the treatment of colon cancer and merits further investigation.
Collapse
Affiliation(s)
- Naghmeh Shahraki
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Mehrabian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shahrazad Amiri-Darban
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Alia Moosavian
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
34
|
Ghaffari S, Hanson C, Schmidt RE, Bouchonville KJ, Offer SM, Sinha S. An integrated multi-omics approach to identify regulatory mechanisms in cancer metastatic processes. Genome Biol 2021; 22:19. [PMID: 33413550 PMCID: PMC7789593 DOI: 10.1186/s13059-020-02213-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Metastatic progress is the primary cause of death in most cancers, yet the regulatory dynamics driving the cellular changes necessary for metastasis remain poorly understood. Multi-omics approaches hold great promise for addressing this challenge; however, current analysis tools have limited capabilities to systematically integrate transcriptomic, epigenomic, and cistromic information to accurately define the regulatory networks critical for metastasis. RESULTS To address this limitation, we use a purposefully generated cellular model of colon cancer invasiveness to generate multi-omics data, including expression, accessibility, and selected histone modification profiles, for increasing levels of invasiveness. We then adopt a rigorous probabilistic framework for joint inference from the resulting heterogeneous data, along with transcription factor binding profiles. Our approach uses probabilistic graphical models to leverage the functional information provided by specific epigenomic changes, models the influence of multiple transcription factors simultaneously, and automatically learns the activating or repressive roles of cis-regulatory events. Global analysis of these relationships reveals key transcription factors driving invasiveness, as well as their likely target genes. Disrupting the expression of one of the highly ranked transcription factors JunD, an AP-1 complex protein, confirms functional relevance to colon cancer cell migration and invasion. Transcriptomic profiling confirms key regulatory targets of JunD, and a gene signature derived from the model demonstrates strong prognostic potential in TCGA colorectal cancer data. CONCLUSIONS Our work sheds new light into the complex molecular processes driving colon cancer metastasis and presents a statistically sound integrative approach to analyze multi-omics profiles of a dynamic biological process.
Collapse
Affiliation(s)
- Saba Ghaffari
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, USA
| | - Casey Hanson
- Department of Genetics, Stanford University, Stanford, USA
| | - Remington E Schmidt
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Gonda 19-476, 200 First St SW, Rochester, MN, 55905, USA
| | - Kelly J Bouchonville
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Gonda 19-476, 200 First St SW, Rochester, MN, 55905, USA
| | - Steven M Offer
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Gonda 19-476, 200 First St SW, Rochester, MN, 55905, USA.
| | - Saurabh Sinha
- Department of Computer Science, Carl R. Woese Institute of Genomic Biology, and Cancer Center of Illinois, University of Illinois at Urbana-Champaign, 2122, Siebel Center, 201 N. Goodwin Ave., Urbana, IL, 61801, USA.
| |
Collapse
|
35
|
Beauséjour M, Boutin A, Vachon PH. Anoikis and the Human Gut Epithelium in Health and Disease. ANOIKIS 2021:95-126. [DOI: 10.1007/978-3-030-73856-3_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
36
|
Li GS, Hou W, Chen G, Yao YX, Chen XY, Zhang XG, Liang Y, Li MX, Huang ZG, Dang YW, Liang QH, Wu HY, Li RQ, Wei HY. Clinical Significance of Integrin Subunit Beta 4 in Head and Neck Squamous Cell Carcinoma. Cancer Biother Radiopharm 2020; 37:256-275. [PMID: 33179959 DOI: 10.1089/cbr.2020.3943] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background: The expression level and clinical significance of integrin subunit beta 4 (ITGB4) in head and neck squamous cell carcinoma (HNSCC) remain unclear. Materials and Methods: Expression of ITGB4 in HNSCC tissues were evaluated by calculating standard mean differences (SMDs) based on gene chips, RNA-seq, and immunohistochemistry data (n = 2330) from multiple sources. Receiver operating characteristic (ROC) curves were used to detect the ability of ITGB4 to distinguish HNSCC from non-HNSCC samples. The relationship between the expression level of ITGB4 and clinical parameters was evaluated by calculating SMDs. Results: Identical results of mRNA and protein levels indicated remarkable up-expression of ITGB4 in HNSCC tissues. Further ROC curves showed that ITGB4 could distinguish HNSCC from non-HNSCC samples. Genetic alteration analysis of ITGB4 in HNSCC indicated that overexpression of ITGB4 in HNSCC was likely not owing to genetic alteration of ITGB4. Moreover, ITGB4 overexpression level may be correlated with clinical T stage. Conclusion: ITGB4 likely plays an essential role in HNSCC occurrence based on our study and its potential diagnostic value is worthy of further exploration in the future.
Collapse
Affiliation(s)
- Guo-Sheng Li
- Department of Organic Chemistry and Medicinal Chemistry, Pharmaceutical College, Guangxi Medical University, Nanning, People's Republic of China
| | - Wei Hou
- Guangxi Key Laboratory of Thalassemia Research, Life Sciences Institute, Guangxi Medical University, Nanning, People's Republic of China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Yu-Xuan Yao
- Department of Organic Chemistry and Medicinal Chemistry, Pharmaceutical College, Guangxi Medical University, Nanning, People's Republic of China
| | - Xiao-Yi Chen
- Department of Organic Chemistry and Medicinal Chemistry, Pharmaceutical College, Guangxi Medical University, Nanning, People's Republic of China
| | - Xiao-Guohui Zhang
- Department of Organic Chemistry and Medicinal Chemistry, Pharmaceutical College, Guangxi Medical University, Nanning, People's Republic of China
| | - Yao Liang
- Department of Organic Chemistry and Medicinal Chemistry, Pharmaceutical College, Guangxi Medical University, Nanning, People's Republic of China
| | - Ming-Xuan Li
- Department of Organic Chemistry and Medicinal Chemistry, Pharmaceutical College, Guangxi Medical University, Nanning, People's Republic of China
| | - Zhi-Guang Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Yi-Wu Dang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Qing-Hua Liang
- Department of Clinical Laboratory, Guangxi Jiangbin Hospital, Nanning, People's Republic of China
| | - Hua-Yu Wu
- Department of Cell Biology and Genetics, School of Pre-Clinical Medicine, Guangxi Medical University, Nanning, People's Republic of China
| | - Rong-Qiao Li
- Department of Clinical Laboratory, Guangxi Jiangbin Hospital, Nanning, People's Republic of China
| | - Hong-Yu Wei
- Department of Organic Chemistry and Medicinal Chemistry, Pharmaceutical College, Guangxi Medical University, Nanning, People's Republic of China
| |
Collapse
|
37
|
Bladder Cancer Metastasis Induced by Chronic Everolimus Application Can Be Counteracted by Sulforaphane In Vitro. Int J Mol Sci 2020; 21:ijms21155582. [PMID: 32759798 PMCID: PMC7432076 DOI: 10.3390/ijms21155582] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/13/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic treatment with the mTOR inhibitor, everolimus, fails long-term in preventing tumor growth and dissemination in cancer patients. Thus, patients experiencing treatment resistance seek complementary measures, hoping to improve therapeutic efficacy. This study investigated metastatic characteristics of bladder carcinoma cells exposed to everolimus combined with the isothiocyanate sulforaphane (SFN), which has been shown to exert cancer inhibiting properties. RT112, UMUC3, or TCCSUP bladder carcinoma cells were exposed short- (24 h) or long-term (8 weeks) to everolimus (0.5 nM) or SFN (2.5 µM), alone or in combination. Adhesion and chemotaxis along with profiling details of CD44 receptor variants (v) and integrin α and β subtypes were evaluated. The functional impact of CD44 and integrins was explored by blocking studies and siRNA knock-down. Long-term exposure to everolimus enhanced chemotactic activity, whereas long-term exposure to SFN or the SFN-everolimus combination diminished chemotaxis. CD44v4 and v7 increased on RT112 cells following exposure to SFN or SFN-everolimus. Up-regulation of the integrins α6, αV, and β1 and down-regulation of β4 that was present with everolimus alone could be prevented by combining SFN and everolimus. Down-regulation of αV, β1, and β4 reduced chemotactic activity, whereas knock-down of CD44 correlated with enhanced chemotaxis. SFN could, therefore, inhibit resistance-related tumor dissemination during everolimus-based bladder cancer treatment.
Collapse
|
38
|
Ye Y, Zhang R, Feng H. Fibronectin promotes tumor cells growth and drugs resistance through a CDC42-YAP-dependent signaling pathway in colorectal cancer. Cell Biol Int 2020; 44:1840-1849. [PMID: 32437085 DOI: 10.1002/cbin.11390] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/17/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022]
Abstract
Fibronectin (FN) is a high-molecular-weight glycoprotein of the extracellular matrix (ECM) that binds to membrane-spanning receptor proteins or other elements in ECM. The expression of FN could be involved in the cancer cells proliferation or migration, and the molecular mechanisms responsible for FN induced protumor signals begin to be elucidated. Here, we report that the elevated expression of FN was observed in those chemoresistant tumor tissues from patients with colorectal cancer. Consistently, FN culture significantly strengthened the proliferation of colorectal cancer cells, induced the colorectal tumor sustained growth and drug resistance in vitro and in vivo. In mechanism, FN could bind to integrin αvβ1, resulting the downstream cell division cycle 42/yes-associated protein 1 (CDC42/YAP-1) signaling pathway activation. The activation of CDC42/YAP-1 signal induces the upregulation of transcription factor SOX2, causing the sustained growth and drugs resistance in colorectal cancer. Blockade of integrin αvβ1 significantly suppressed the colorectal cancer growth and drugs resistance development in vitro and in vivo, which provides a new target for clinical colorectal cancer treatment.
Collapse
Affiliation(s)
- Yu Ye
- Department of General Surgery, Zhejiang Chinese Medicine and Western Medicine Integrated Hospital/Hangzhou Red Cross Hospital, Hangzhou, China
| | - Ruifeng Zhang
- Department of Gastrointestinal Surgery, Shanxian Dongda Hospital, Heze, China
| | - Haiyang Feng
- Department of Colorectal Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China.,Department of Colorectal Surgery, Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
39
|
Kotelevets L, Chastre E. Rac1 Signaling: From Intestinal Homeostasis to Colorectal Cancer Metastasis. Cancers (Basel) 2020; 12:cancers12030665. [PMID: 32178475 PMCID: PMC7140047 DOI: 10.3390/cancers12030665] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 12/14/2022] Open
Abstract
The small GTPase Rac1 has been implicated in a variety of dynamic cell biological processes, including cell proliferation, cell survival, cell-cell contacts, epithelial mesenchymal transition (EMT), cell motility, and invasiveness. These processes are orchestrated through the fine tuning of Rac1 activity by upstream cell surface receptors and effectors that regulate the cycling Rac1-GDP (off state)/Rac1-GTP (on state), but also through the tuning of Rac1 accumulation, activity, and subcellular localization by post translational modifications or recruitment into molecular scaffolds. Another level of regulation involves Rac1 transcripts stability and splicing. Downstream, Rac1 initiates a series of signaling networks, including regulatory complex of actin cytoskeleton remodeling, activation of protein kinases (PAKs, MAPKs) and transcription factors (NFkB, Wnt/β-catenin/TCF, STAT3, Snail), production of reactive oxygen species (NADPH oxidase holoenzymes, mitochondrial ROS). Thus, this GTPase, its regulators, and effector systems might be involved at different steps of the neoplastic progression from dysplasia to the metastatic cascade. After briefly placing Rac1 and its effector systems in the more general context of intestinal homeostasis and in wound healing after intestinal injury, the present review mainly focuses on the several levels of Rac1 signaling pathway dysregulation in colorectal carcinogenesis, their biological significance, and their clinical impact.
Collapse
Affiliation(s)
- Larissa Kotelevets
- Institut National de la Santé et de la Recherche Médicale, UMR S 938, Centre de Recherche Saint-Antoine, 75012 Paris, France
- Sorbonne Université, Hôpital Saint-Antoine, Site Bâtiment Kourilsky, 75012 Paris, France
- Correspondence: (L.K.); (E.C.)
| | - Eric Chastre
- Institut National de la Santé et de la Recherche Médicale, UMR S 938, Centre de Recherche Saint-Antoine, 75012 Paris, France
- Sorbonne Université, Hôpital Saint-Antoine, Site Bâtiment Kourilsky, 75012 Paris, France
- Correspondence: (L.K.); (E.C.)
| |
Collapse
|
40
|
Beaulieu JF. Colorectal Cancer Research: Basic, Preclinical, and Clinical Approaches. Cancers (Basel) 2020; 12:cancers12020416. [PMID: 32053872 PMCID: PMC7072293 DOI: 10.3390/cancers12020416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/23/2022] Open
Affiliation(s)
- Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; ; Tel.: +1-819-821-8000 (ext. 75269)
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|