1
|
Dunbar KJ, Efe G, Cunningham K, Esquea E, Navaridas R, Rustgi AK. Regulation of metastatic organotropism. Trends Cancer 2025; 11:216-231. [PMID: 39732596 PMCID: PMC11903188 DOI: 10.1016/j.trecan.2024.11.012] [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: 09/29/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 12/30/2024]
Abstract
Metastasis is responsible for most cancer-related deaths. Different cancers have their own preferential sites of metastases, a phenomenon termed metastatic organotropism. The mechanisms underlying organotropism are multifactorial and include the generation of a pre-metastatic niche (PMN), metastatic homing, colonization, dormancy, and metastatic outgrowth. Historically, studies of metastatic organotropism have been limited by a lack of models allowing direct comparison of cells exhibiting different patterns of tropism. However, new innovative models and large-scale sequencing efforts have propelled organotropism research. Herein, we summarize the recent discoveries in metastatic organotropism regulation, focusing on lung, liver, brain, and bone tropism. We discuss how emerging technologies are continuing to improve our ability to model and, hopefully, predict and treat organotropism.
Collapse
Affiliation(s)
- Karen J Dunbar
- Herbert Irving Comprehensive Cancer Center, New York, NY, 10032, USA.
| | - Gizem Efe
- Herbert Irving Comprehensive Cancer Center, New York, NY, 10032, USA; Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Katherine Cunningham
- Herbert Irving Comprehensive Cancer Center, New York, NY, 10032, USA; Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Emily Esquea
- Herbert Irving Comprehensive Cancer Center, New York, NY, 10032, USA; Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Raul Navaridas
- Herbert Irving Comprehensive Cancer Center, New York, NY, 10032, USA; Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Anil K Rustgi
- Herbert Irving Comprehensive Cancer Center, New York, NY, 10032, USA; Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, 10032, USA; Division of Digestive and Liver Diseases, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
2
|
Fonseca P, Cui W, Struyf N, Tong L, Chaurasiya A, Casagrande F, Zhao H, Fernando D, Chen X, Tobin NP, Seashore-Ludlow B, Lundqvist A, Hartman J, Göndör A, Östling P, Holmgren L. A phenotypic screening approach to target p60AmotL2-expressing invasive cancer cells. J Exp Clin Cancer Res 2024; 43:107. [PMID: 38594748 PMCID: PMC11003180 DOI: 10.1186/s13046-024-03031-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Tumor cells have the ability to invade and form small clusters that protrude into adjacent tissues, a phenomenon that is frequently observed at the periphery of a tumor as it expands into healthy tissues. The presence of these clusters is linked to poor prognosis and has proven challenging to treat using conventional therapies. We previously reported that p60AmotL2 expression is localized to invasive colon and breast cancer cells. In vitro, p60AmotL2 promotes epithelial cell invasion by negatively impacting E-cadherin/AmotL2-related mechanotransduction. METHODS Using epithelial cells transfected with inducible p60AmotL2, we employed a phenotypic drug screening approach to find compounds that specifically target invasive cells. The phenotypic screen was performed by treating cells for 72 h with a library of compounds with known antitumor activities in a dose-dependent manner. After assessing cell viability using CellTiter-Glo, drug sensitivity scores for each compound were calculated. Candidate hit compounds with a higher drug sensitivity score for p60AmotL2-expressing cells were then validated on lung and colon cell models, both in 2D and in 3D, and on colon cancer patient-derived organoids. Nascent RNA sequencing was performed after BET inhibition to analyse BET-dependent pathways in p60AmotL2-expressing cells. RESULTS We identified 60 compounds that selectively targeted p60AmotL2-expressing cells. Intriguingly, these compounds were classified into two major categories: Epidermal Growth Factor Receptor (EGFR) inhibitors and Bromodomain and Extra-Terminal motif (BET) inhibitors. The latter consistently demonstrated antitumor activity in human cancer cell models, as well as in organoids derived from colon cancer patients. BET inhibition led to a shift towards the upregulation of pro-apoptotic pathways specifically in p60AmotL2-expressing cells. CONCLUSIONS BET inhibitors specifically target p60AmotL2-expressing invasive cancer cells, likely by exploiting differences in chromatin accessibility, leading to cell death. Additionally, our findings support the use of this phenotypic strategy to discover novel compounds that can exploit vulnerabilities and specifically target invasive cancer cells.
Collapse
Affiliation(s)
- Pedro Fonseca
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Weiyingqi Cui
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Nona Struyf
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavägen 23a, 171 65, Stockholm, Sweden
| | - Le Tong
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Ayushi Chaurasiya
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Felipe Casagrande
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Honglei Zhao
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Dinura Fernando
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Xinsong Chen
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Nicholas P Tobin
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Brinton Seashore-Ludlow
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavägen 23a, 171 65, Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
| | - Anita Göndör
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
- Department of Clinical Molecular Biology, University of Oslo, Akershus Universitetssykehus, 1478, Lørenskog, Oslo, Norway
| | - Päivi Östling
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavägen 23a, 171 65, Stockholm, Sweden
| | - Lars Holmgren
- Department of Oncology and Pathology, Karolinska Institutet, U2, Bioclinicum J6:20, Solnavägen 30, 171 64, Solna, Stockholm, Sweden.
| |
Collapse
|
3
|
Wu X, Xu Z, Li W, Lu Y, Pu J. HIF‑1α and RACGAP1 promote the progression of hepatocellular carcinoma in a mutually regulatory way. Mol Med Rep 2023; 28:218. [PMID: 37772389 PMCID: PMC10568255 DOI: 10.3892/mmr.2023.13105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023] Open
Abstract
Hypoxia, a condition characterized by low oxygen levels, serves an important role in the progression of hepatocellular carcinoma (HCC). However, the precise molecular mechanisms underlying hypoxia‑induced HCC progression are yet to be fully elucidated. The present study assessed the involvement of two key factors, hypoxia‑inducible factor‑1α (HIF‑1α) and Rac GTPase activating protein 1 (RACGAP1), in HCC development under hypoxic conditions. HIF‑1α and RACGAP1 genes were overexpressed and knocked down in Hep3B and Huh7 cells using lentiviral transduction and the levels of HIF‑1α and RACGAP1 in the cells were assessed using quantitative PCR, western blotting and immunofluorescence. Co‑immunoprecipitation experiments were performed to evaluate the interaction between HIF‑1α and RACGAP1. Subsequently, the proliferation, apoptosis, migration and invasion of Hep3B and Huh7 cells were assessed using the Cell Counting Kit‑8 assay, flow cytometry, Transwell assay and migration experiments. The expression levels of HIF‑1α and RACGAP1 in normal and HCC tumor samples were analyzed utilizing the Gene Expression Profiling Interactive Analysis database. Furthermore, correlations between HIF‑1α/RACGAP1 gene expression levels and patient survival outcomes were evaluated using the Kaplan‑Meier plotter. Knockdown of HIF‑1α resulted in a significant decrease in RACGAP1 expression, whilst overexpression of HIF‑1α resulted in a significant increase in RACGAP1 expression. Moreover, overexpression and knockdown of RACGAP1 had the same effect on HIF‑1α expression. Additionally, it was demonstrated that HIF‑1α and RACGAP1 interacted directly within a complex. Overexpression of HIF‑1α or RACGAP1 significantly increased proliferation, invasion and migration, and significantly decreased the proportion of apoptotic Hep3B and Huh7 cells. Conversely, knockdown of HIF‑1α or RACGAP1 significantly decreased proliferation, invasion and migration, and significantly increased the proportion of apoptotic Hep3B and Huh7 cells. In addition, the combined knockdown or overexpression of HIF‑1α and RACGAP1 had a more pronounced effect on HCC cell migration compared with knockdown of HIF‑1α alone. Furthermore, there was a significant positive correlation between the expression levels of HIF‑1α and RACGAP1 in HCC tissues and patients with HCC and upregulation of both HIF‑1α and RACGAP1 demonstrated a lower overall survival probability. In conclusion, HIF‑1α and RACGAP1 may synergistically contribute to the development of HCC, highlighting their potential as valuable targets for HCC therapy.
Collapse
Affiliation(s)
- Xianjian Wu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Zuoming Xu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Wenchuan Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yuan Lu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Jian Pu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| |
Collapse
|
4
|
Carranza-Rosales P, Valencia-Mercado D, Esquivel-Hernández O, González-Geroniz MI, Bañuelos-García JI, Castruita-Ávila AL, Sánchez-Prieto MA, Viveros-Valdez E, Morán-Martínez J, Balderas-Rentería I, Guzmán-Delgado NE, Carranza-Torres IE. Breast Cancer Tissue Explants: An Approach to Develop Personalized Therapy in Public Health Services. J Pers Med 2023; 13:1521. [PMID: 37888132 PMCID: PMC10608341 DOI: 10.3390/jpm13101521] [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: 09/23/2023] [Revised: 10/18/2023] [Accepted: 10/21/2023] [Indexed: 10/28/2023] Open
Abstract
Breast cancer is one of the main causes of death worldwide. Lately, there is great interest in developing methods that assess individual sensitivity and/or resistance of tumors to antineoplastics to provide personalized therapy for patients. In this study we used organotypic culture of human breast tumor slices to predict the experimental effect of antineoplastics on the viability of tumoral tissue. Samples of breast tumor were taken from 27 patients with clinically advanced breast cancer; slices were obtained and incubated separately for 48 h with paclitaxel, docetaxel, epirubicin, 5-fluorouracil, cyclophosphamide, and cell culture media (control). We determined an experimental tumor sensitivity/resistance (S/R) profile by evaluating tissue viability using the Alamar Blue® metabolic test, and by structural viability (histopathological analyses, necrosis, and inflammation). These parameters were related to immunohistochemical expression of the estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. The predominant histological type found was infiltrating ductal carcinoma (85.2%), followed by lobular carcinoma (7.4%) and mixed carcinoma (7.4%). Experimental drug resistance was related to positive hormone receptor status in 83% of samples treated with cyclophosphamide (p = 0.027). Results suggest that the tumor S/R profile can help to predict personalized therapy or optimize chemotherapeutic treatments in breast cancer.
Collapse
Affiliation(s)
- Pilar Carranza-Rosales
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Calle Jesús Dionisio González # 501, Col. Independencia, Monterrey 64720, NL, Mexico;
| | - Daniel Valencia-Mercado
- Unidad Médica de Alta Especialidad, Hospital de Ginecología y Obstetricia No. 23, Instituto Mexicano del Seguro Social, Avenida Constitución y Félix U, Gómez s/n, Colonia Centro, Monterrey 64000, NL, Mexico; (D.V.-M.); (O.E.-H.); (M.I.G.-G.); (J.I.B.-G.)
| | - Olga Esquivel-Hernández
- Unidad Médica de Alta Especialidad, Hospital de Ginecología y Obstetricia No. 23, Instituto Mexicano del Seguro Social, Avenida Constitución y Félix U, Gómez s/n, Colonia Centro, Monterrey 64000, NL, Mexico; (D.V.-M.); (O.E.-H.); (M.I.G.-G.); (J.I.B.-G.)
| | - Manuel Ismael González-Geroniz
- Unidad Médica de Alta Especialidad, Hospital de Ginecología y Obstetricia No. 23, Instituto Mexicano del Seguro Social, Avenida Constitución y Félix U, Gómez s/n, Colonia Centro, Monterrey 64000, NL, Mexico; (D.V.-M.); (O.E.-H.); (M.I.G.-G.); (J.I.B.-G.)
| | - José Inocente Bañuelos-García
- Unidad Médica de Alta Especialidad, Hospital de Ginecología y Obstetricia No. 23, Instituto Mexicano del Seguro Social, Avenida Constitución y Félix U, Gómez s/n, Colonia Centro, Monterrey 64000, NL, Mexico; (D.V.-M.); (O.E.-H.); (M.I.G.-G.); (J.I.B.-G.)
| | - Ana Lilia Castruita-Ávila
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 25, Instituto Mexicano del Seguro Social, Av Fidel Velázquez s/n, Mitras Nte., Monterrey 64180, NL, Mexico; (A.L.C.-Á.); (M.A.S.-P.)
| | - Mario Alberto Sánchez-Prieto
- Unidad Médica de Alta Especialidad, Hospital de Especialidades No. 25, Instituto Mexicano del Seguro Social, Av Fidel Velázquez s/n, Mitras Nte., Monterrey 64180, NL, Mexico; (A.L.C.-Á.); (M.A.S.-P.)
| | - Ezequiel Viveros-Valdez
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, San Nicolás de los Garza 66450, NL, Mexico;
| | - Javier Morán-Martínez
- Departamento de Biología Celular y Ultraestructura, Facultad de Medicina, Universidad Autónoma de Coahuila, Av. Morelos 900-Oriente, Primera de Cobián Centro, Torreón 27000, CH, Mexico;
| | - Isaías Balderas-Rentería
- Facultad de Ciencias Químicas, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, San Nicolás de los Garza 66450, NL, Mexico;
| | - Nancy Elena Guzmán-Delgado
- Unidad Médica de Alta Especialidad, Hospital de Cardiología No. 34, Instituto Mexicano del Seguro Social, Av. Lincoln S/N, Col. Valle Verde 2do. Sector, Monterrey 64360, NL, Mexico
| | - Irma Edith Carranza-Torres
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, Calle Jesús Dionisio González # 501, Col. Independencia, Monterrey 64720, NL, Mexico;
- Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Av. Pedro de Alba s/n, San Nicolás de los Garza 66450, NL, Mexico;
| |
Collapse
|
5
|
Pape J, Micalet A, Alsheikh W, Ezbakh N, Virjee RI, Al Hosni R, Moeendarbary E, Cheema U. Biophysical Parameters Can Induce Epithelial-to-Mesenchymal Phenotypic and Genotypic Changes in HT-29 Cells: A Preliminary Study. Int J Mol Sci 2023; 24:3956. [PMID: 36835368 PMCID: PMC9962772 DOI: 10.3390/ijms24043956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) in cancer is the process described where cancer epithelial cells acquire mesenchymal properties which can lead to enhanced invasiveness. Three-dimensional cancer models often lack the relevant and biomimetic microenvironment parameters appropriate to the native tumour microenvironment thought to drive EMT. In this study, HT-29 epithelial colorectal cells were cultivated in different oxygen and collagen concentrations to investigate how these biophysical parameters influenced invasion patterns and EMT. Colorectal HT-29 cells were grown in physiological hypoxia (5% O2) and normoxia (21% O2) in 2D, 3D soft (60 Pa), and 3D stiff (4 kPa) collagen matrices. Physiological hypoxia was sufficient to trigger expression of markers of EMT in the HT-29 cells in 2D by day 7. This is in contrast to a control breast cancer cell line, MDA-MB-231, which expresses a mesenchymal phenotype regardless of the oxygen concentration. In 3D, HT-29 cells invaded more extensively in a stiff matrix environment with corresponding increases in the invasive genes MMP2 and RAE1. This demonstrates that the physiological environment can directly impact HT-29 cells in terms of EMT marker expression and invasion, compared to an established cell line, MDA-MB-231, which has already undergone EMT. This study highlights the importance of the biophysical microenvironment to cancer epithelial cells and how these factors can direct cell behaviour. In particular, that stiffness of the 3D matrix drives greater invasion in HT-29 cells regardless of hypoxia. It is also pertinent that some cell lines (already having undergone EMT) are not as sensitive to the biophysical features of their microenvironment.
Collapse
Affiliation(s)
- Judith Pape
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| | - Auxtine Micalet
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
- Department of Mechanical Engineering, University College London, Gower Street, London WC1E 6BT, UK
| | - Wissal Alsheikh
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| | - Nadia Ezbakh
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| | - Rania-Iman Virjee
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| | - Rawiya Al Hosni
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, Gower Street, London WC1E 6BT, UK
| | - Umber Cheema
- Centre for 3D Models of Health and Disease, Department of Targeted Intervention, Division of Surgery and Interventional Science, University College London, Charles Bell House, 43-45 Foley Street, London W1W 7TS, UK
| |
Collapse
|
6
|
Ahmadi M, Mahmoodi M, Shoaran M, Nazari-Khanamiri F, Rezaie J. Harnessing Normal and Engineered Mesenchymal Stem Cells Derived Exosomes for Cancer Therapy: Opportunity and Challenges. Int J Mol Sci 2022; 23:ijms232213974. [PMID: 36430452 PMCID: PMC9699149 DOI: 10.3390/ijms232213974] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
There remains a vital necessity for new therapeutic approaches to combat metastatic cancers, which cause globally over 8 million deaths per year. Mesenchymal stem cells (MSCs) display aptitude as new therapeutic choices for cancer treatment. Exosomes, the most important mediator of MSCs, regulate tumor progression. The potential of harnessing exosomes from MSCs (MSCs-Exo) in cancer therapy is now being documented. MSCs-Exo can promote tumor progression by affecting tumor growth, metastasis, immunity, angiogenesis, and drug resistance. However, contradictory evidence has suggested that MSCs-Exo suppress tumors through several mechanisms. Therefore, the exact association between MSCs-Exo and tumors remains controversial. Accordingly, the applications of MSCs-Exo as novel drug delivery systems and standalone therapeutics are being extensively explored. In addition, engineering MSCs-Exo for targeting tumor cells has opened a new avenue for improving the efficiency of antitumor therapy. However, effective implementation in the clinical trials will need the establishment of standards for MSCs-Exo isolation and characterization as well as loading and engineering methods. The studies outlined in this review highlight the pivotal roles of MSCs-Exo in tumor progression and the promising potential of MSCs-Exo as therapeutic drug delivery vehicles for cancer treatment.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5665665811, Iran
| | - Monireh Mahmoodi
- Department of Biology, Faculty of Science, Arak University, Arak 3815688349, Iran
| | - Maryam Shoaran
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz 5665665811, Iran
| | - Fereshteh Nazari-Khanamiri
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia 5714783734, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia 5714783734, Iran
- Correspondence: ; Tel.: +98-9148548503; Fax: +98-4432222010
| |
Collapse
|
7
|
He L, Deng C. Recent advances in organotypic tissue slice cultures for anticancer drug development. Int J Biol Sci 2022; 18:5885-5896. [PMID: 36263166 PMCID: PMC9576528 DOI: 10.7150/ijbs.78997] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 01/12/2023] Open
Abstract
Organotypic tissue slice culture is established from animal or patient tissues and cultivated in an in vitro ecosystem. This technique has made countless contributions to anticancer drug development due to the vast number of advantages, such as the preservation of the cell repertoire and immune components, identification of invasive ability of tumors, toxicity determination of compounds, quick assessment of therapeutic efficacy, and high predictive performance of drug responses. Importantly, it serves as a reliable tool to stratify therapeutic responders from nonresponders and select the optimal standard-of-care treatment regimens for personalized medicine, which is expected to become a potent platform and even the gold standard for anticancer drug screening of individualization in the near future.
Collapse
Affiliation(s)
- Lin He
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China
| | - Chuxia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China.,Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Macau SAR, China.,MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China.,✉ Corresponding author: Chu-Xia Deng, Ph.D. Dean and Chair Professor, E12, Room 4041, Faculty of Health Sciences, University of Macau, Macau SAR, China. Phone: (853) 8822 4997; Fax: 8822 2314; E-mail:
| |
Collapse
|
8
|
Zhang Y, Bao J, Gong X, Shi W, Liu T, Wang X. Transcriptomics and metabolomics revealed the molecular mechanism of the toxic effect of mancozeb on liver of mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:114003. [PMID: 36007320 DOI: 10.1016/j.ecoenv.2022.114003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Mancozeb (MCZ), a broad-spectrum fungicide, has been widely used in crops (tomatoes and potatoes) in the past few decades, resulting in its bioaccumulation in the food web. However, the mechanism of MCZ on liver injury has not been reported yet. This study combined transcriptomics and metabolomics to explore the potential mechanism of MCZ on liver injury. MCZ group was given 100 mg/kg MCZ every day, and the C group was given 0.2 mL of deionized water every day. One hundred mg/kg MCZ led to unclear hepatocyte structure and hemorrhagic inflammatory cell infiltration. Transcriptomics and metabolomics analyses showed that the MCZ group resulted in 326 differentially expressed genes (DEGs) and 179 differential metabolites. Joint analysis showed that DEGs and differential metabolites were mainly enriched in the adenosine monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway. We found that MCZ could increase the content of reactive oxygen species (ROS) and reduce the activities of superoxide dismutase (SOD) and catalase (CAT). The contents of DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B) in the liver decreased significantly, and the state of DNA methylation was significantly higher than the control (C) group (p < 0.05). Our results suggest that AMPK and mitogen‑activated protein kinase (MAPK) signaling pathways play an important role in MCZ-induced liver injury and are the key mechanisms for understanding the hepatotoxicity of MCZ.
Collapse
Affiliation(s)
- Yan Zhang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Jialu Bao
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China
| | - Xincheng Gong
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071001, China
| | - Wanyu Shi
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071001, China
| | - Tao Liu
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071001, China
| | - Xiaodan Wang
- College of Traditional Chinese Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071001, China.
| |
Collapse
|
9
|
Rezaie J, Ahmadi M, Ravanbakhsh R, Mojarad B, Mahbubfam S, Shaban SA, Shadi K, Berenjabad NJ, Etemadi T. Tumor-derived extracellular vesicles: The metastatic organotropism drivers. Life Sci 2022; 289:120216. [PMID: 34890589 DOI: 10.1016/j.lfs.2021.120216] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023]
Abstract
The continuous growing, spreading, and metastasis of tumor cells depend on intercellular communication within cells resident in a tissue environment. Such communication is mediated through the secretion of particles from tumor cells and resident cells known as extracellular vesicles (EVs) within a microenvironment. EVs are a heterogeneous population of membranous vesicles released from tumor cells that transfer many types of active biomolecules to recipient cells and induce physiologic and phenotypic alterations in the tissue environment. Spreading the 'seeds' of metastasis needs the EVs that qualify the 'soil' at distant sites to promote the progress of arriving tumor cells. Growing evidence indicates that EVs have vital roles in tumorigenesis, including pre-metastatic niche formation and organotropic metastasis. These EVs mediate organotropic metastasis by modifying the pre-metastatic microenvironment through different pathways including induction of phenotypic alternation and differentiation of cells, enrolment of distinct supportive stromal cells, up-regulation of the expression of pro-inflammatory genes, and induction of immunosuppressive status. However, instead of pre-metastatic niche formation, evidence suggests that EVs may mediate reawakening of dormant niches. Findings regarding EVs function in tumor metastasis have led to growing interests in the interdisciplinary significance of EVs, including targeted therapy, cell-free therapy, drug-delivery system, and diagnostic biomarker. In this review, we discuss EVs-mediated pre-metastatic niche formation and organotropic metastasis in visceral such as lung, liver, brain, lymph node, and bone with a focus on associated signaling, causing visceral environment hospitable for metastatic cells. Furthermore, we present an overview of the possible therapeutic application of EVs in cancer management.
Collapse
Affiliation(s)
- Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Mahdi Ahmadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Ravanbakhsh
- Department of Aquatic Biotechnology, Artemia and Aquaculture Research Institute, Urmia University, Urmia, Iran
| | - Behnam Mojarad
- Biology Department, Faculty of Sciences, Urmia University, Urmia, Iran
| | - Shadi Mahbubfam
- Biology Department, Faculty of Sciences, Urmia University, Urmia, Iran
| | | | - Kosar Shadi
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Tahereh Etemadi
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| |
Collapse
|
10
|
Supadmanaba IGP, Comandatore A, Morelli L, Giovannetti E, Lagerweij T. Organotypic-liver slide culture systems to explore the role of extracellular vesicles in pancreatic cancer metastatic behavior and guide new therapeutic approaches. Expert Opin Drug Metab Toxicol 2021; 17:937-946. [PMID: 33945374 DOI: 10.1080/17425255.2021.1925646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/30/2021] [Indexed: 02/08/2023]
Abstract
Introduction: Recent studies suggested that extracellular vesicles (EVs) play a role both in the metastatic niche formation and in the progression of several tumors, including pancreatic cancer. In particular, the effects of EVs on metastasis should be studied in model systems that take into account both the tumor cells and the metastatic site/tumor microenvironment. Studies with labeled EVs or EV-secreting cells in ex vivo models will reflect the physiological and pathological functions of EVs. The organotypic-tissue slide culture systems can fulfill such a role.Areas covered: This review provides an overview of available organotypic-culture slide systems. We specifically focus on the assay system of liver culture-slides in combination with pancreatic tumors, which can be modulated to test the efficacy of new therapeutic approaches.Expert opinion: The intercellular exchange of EVs has emerged as a biologically relevant phenomenon to drive cancer metastasis. However, further models need to be developed to better elucidate the functional roles of EVs. The use of novel organotypic slide culture systems provides the opportunity to explore the role of EVs in the metastatic behavior of pancreatic cancer, decreasing the use of costly and cumbersome organoid or animal models.
Collapse
Affiliation(s)
- I Gede Putu Supadmanaba
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Biochemistry Department, Faculty of Medicine, Universitas Udayana, Denpasar, Bali, Indonesia
| | - Annalisa Comandatore
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Luca Morelli
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per La Scienza, Pisa, Italy
| | - Tonny Lagerweij
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|