1
|
Romoli J, Chiodelli P, Signoroni PB, Vertua E, Ferrari C, Giuzzi E, Paini A, Scalvini E, Papait A, Stefani FR, Silini AR, Parolini O. Modeling Stromal Cells Inside the Tumor Microenvironment of Ovarian Cancer: In Vitro Generation of Cancer-Associated Fibroblast-Like Cells and Their Impact in a 3D Model. MedComm (Beijing) 2025; 6:e70172. [PMID: 40255916 PMCID: PMC12006666 DOI: 10.1002/mco2.70172] [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: 04/24/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 04/22/2025] Open
Abstract
The tumor microenvironment (TME) is the combination of cells and factors that promotes tumor progression, and cancer-associated fibroblasts (CAFs) are a key component within TME. CAF originates from various stromal cells and is activated by factors such as transforming growth factor-beta (TGF-β) secreted by tumor cells, favoring chemoresistance and metastasis. Recent publications have underlined plasticity and heterogeneity and their strong contribution to the reactive stroma within the TME. Our study aimed to replicate the TME's structure by creating a 3D in vitro model of ovarian cancer (OC). By incorporating diverse tumor and stromal cells, we simulated a physiologically relevant environment for studying CAF-like cell behavior within tumor spheroids in a context-dependent manner. CAF-like cells were generated by exposing human dermal fibroblasts to OC cell line conditioned media in the presence or absence of TGF-β. Herein, we found that different stimuli induce the generation of heterogeneous populations of CAF-like cells. Notably, we observed the ability of CAF-like cells to shape the intratumoral architecture and to contribute to functional changes in tumor cell behavior. This study highlights the importance of precise assessment of CAF for potential therapeutic interventions and further provides a reliable model for investigating novel therapeutic targets in OC.
Collapse
Affiliation(s)
- Jacopo Romoli
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
| | - Paola Chiodelli
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
| | | | - Elsa Vertua
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Clarissa Ferrari
- Research and Clinical Trials UnitFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Elisabetta Giuzzi
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Alice Paini
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Elisa Scalvini
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Andrea Papait
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCSRomeItaly
| | | | | | - Ornella Parolini
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCSRomeItaly
| |
Collapse
|
2
|
Guo J, Wang K, Sun Q, Liu J, Zheng J. Targeting B4GALT3 in BMSCs-EVs for Therapeutic Control of HCC via NF-κB pathway inhibition. Cell Biol Toxicol 2025; 41:67. [PMID: 40186771 PMCID: PMC11972216 DOI: 10.1007/s10565-025-10013-x] [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: 11/06/2024] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Examining the communications in the tumor microenvironment (TME) specific to hepatocellular carcinoma (HCC), this exploration looks into the role played by beta-1,4-Galactosyltransferase III (B4GALT3) in bone marrow mesenchymal stromal cell-derived extracellular vesicles (BMSCs-EVs) regarding the NF-κB pathway and the triggering of cancer-associated fibroblasts (CAF). Through a multidisciplinary approach combining transcriptome sequencing, bioinformatic analysis, and various experimental models, the involvement of B4GALT3 in regulating CAF activity by modulating NF-κB signaling was brought to light in our study. The outcomes suggest that targeting B4GALT3 could impede HCC cell migration and invasion, promote apoptosis, and dampen tumor progression and metastasis, offering novel insights into potential therapeutic strategies for combating HCC.
Collapse
Affiliation(s)
- Juncheng Guo
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Kaiqiong Wang
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Qigang Sun
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Jun Liu
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China
| | - Jinfang Zheng
- Department of Hepatobiliary Surgery, Hainan General Hospital, No.19 Xinhua Road, Xiuying District, Haikou, 570311, Hainan Province, China.
| |
Collapse
|
3
|
Zhang S, Cao G, Shen S, Wu Y, Tan X, Jiang X. CAF-derived miR-642a-3p supports migration, invasion, and EMT of hepatocellular carcinoma cells by targeting SERPINE1. PeerJ 2024; 12:e18428. [PMID: 39544420 PMCID: PMC11562775 DOI: 10.7717/peerj.18428] [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: 06/07/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024] Open
Abstract
Background Cancer-associated fibroblasts (CAFs) and hepatocellular carcinoma (HCC) cells interact to promote HCC progression, but the underlying mechanisms remain unclear. Serpin family E member 1 (SERPINE1) has conflicting roles in HCC, and microRNAs (miRNAs) are known to regulate tumor progression through intercellular communication. Therefore, we investigated the potential involvement of miRNA/SERPINE1 axis in crosstalk between CAFs and HCC cells. Methods In this study, candidate miRNAs targeting SERPINE1 3' UTR were predicted using multiple miRNA databases. The miRNAs and SERPINE1 mRNA expression in Huh7 cells was assessed after co-culture with CAFs using RT-qPCR. Huh7 cell proliferation and invasion were detected after SERPINE1 siRNA. The functions of the CAF-derived miR-642a-3p/SERPINE1 axis in HCC cells were examined using CCK-8, wound healing, transwell assays, western blot, and dual-luciferase reporter assays. Moreover, a orthotopic xenograft model was used to investigate the contribution of miR-642a-3p knockdown in HCC. Results SERPINE1 mRNA expression decreased, while miR-642a-3p expression increased in Huh7 cells co-cultured with CAFs. SERPINE1 knockdown enhanced Huh7 cell proliferation and invasion as well as miR-642a-3p expression. miR-642a-3p overexpression promoted migration, invasion, and epithelial-mesenchymal transition (EMT) in Huh7 cells by targeting SERPINE1, while miR-642a-3p knockdown yielded the opposite effect. Rescue experiments confirmed that SERPINE1 knockdown attenuated the inhibitory effects of miR-642a-3p knockdown on migration, invasion, and EMT in Huh7 cells. Importantly, miR-642a-3p knockdown suppressed growth and EMT in orthotopic liver tumors. Conclusion CAF-derived miR-642a-3p/SERPINE1 axis facilitated migration, invasion, and EMT in the HCC cells, suggesting miR-642a-3p/SERPINE1 axis can be a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Pharmacy, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Gang Cao
- Office of the Dean, Nantong Maternal and Child Health Care Hospital, Nantong, China
| | - Shuijie Shen
- Department of Science and Education, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Yu Wu
- Department of Science and Education, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Xiying Tan
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyan Jiang
- Department of Pharmacy, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| |
Collapse
|
4
|
Hu S, Zhang C, Ma Q, Li M, Yu X, Zhang H, Lv S, Shi Y, He X. Unveiling the multifaceted roles of microRNAs in extracellular vesicles derived from mesenchymal stem cells: implications in tumor progression and therapeutic interventions. Front Pharmacol 2024; 15:1438177. [PMID: 39161894 PMCID: PMC11330784 DOI: 10.3389/fphar.2024.1438177] [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: 05/25/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) have the capacity to migrate to tumor sites in vivo and transmit paracrine signals by secreting extracellular vesicles (EVs) to regulate tumor biological behaviors. MSC-derived EVs (MSC-EVs) have similar tumor tropism and pro- or anti-tumorigenesis as their parental cells and exhibit superior properties in drug delivery. MSC-EVs can transfer microRNAs (miRNAs) to tumor cells, thereby manipulating multiple key cancer-related pathways, and further playing a vital role in the tumor growth, metastasis, drug resistance and other aspects. In addition, tumor cells can also influence the behaviors of MSCs in the tumor microenvironment (TME), orchestrating this regulatory process via miRNAs in EVs (EV-miRNAs). Clarifying the specific mechanism by which MSC-derived EV-miRNAs regulate tumor progression, as well as investigating the roles of EV-miRNAs in the TME will contribute to their applications in tumor pharmacotherapy. This article mainly reviews the multifaceted roles and mechanism of miRNAs in MSC-EVs affecting tumor progression, the crosstalk between MSCs and tumor cells caused by EV-miRNAs in the TME. Eventually, the clinical applications of miRNAs in MSC-EVs in tumor therapeutics are illustrated.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shuang Lv
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yingai Shi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xu He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Gładyś A, Mazurski A, Czekaj P. Potential Consequences of the Use of Adipose-Derived Stem Cells in the Treatment of Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:7806. [PMID: 39063048 PMCID: PMC11277008 DOI: 10.3390/ijms25147806] [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: 06/12/2024] [Revised: 07/07/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks as the most prevalent of primary liver cancers and stands as the third leading cause of cancer-related deaths. Early-stage HCC can be effectively managed with available treatment modalities ranging from invasive techniques, such as liver resection and thermoablation, to systemic therapies primarily employing tyrosine kinase inhibitors. Unfortunately, these interventions take a significant toll on the body, either through physical trauma or the adverse effects of pharmacotherapy. Consequently, there is an understandable drive to develop novel HCC therapies. Adipose-derived stem cells (ADSCs) are a promising therapeutic tool. Their facile extraction process, coupled with the distinctive immunomodulatory capabilities of their secretome, make them an intriguing subject for investigation in both oncology and regenerative medicine. The factors they produce are both enzymes affecting the extracellular matrix (specifically, metalloproteinases and their inhibitors) as well as cytokines and growth factors affecting cell proliferation and invasiveness. So far, the interactions observed with various cancer cell types have not led to clear conclusions. The evidence shows both inhibitory and stimulatory effects on tumor growth. Notably, these effects appear to be dependent on the tumor type, prompting speculation regarding their potential inhibitory impact on HCC. This review briefly synthesizes findings from preclinical and clinical studies examining the effects of ADSCs on cancers, with a specific focus on HCC, and emphasizes the need for further research.
Collapse
Affiliation(s)
- Aleksandra Gładyś
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, 40-752 Katowice, Poland;
| | - Adam Mazurski
- Students Scientific Society, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, 40-752 Katowice, Poland;
| | - Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, 40-752 Katowice, Poland;
| |
Collapse
|
6
|
Salah RA, El-Derby AM, El-Gammal Z, Wadie B, Ahmed SM, Elshenawy SE, Magdy S, Salah A, Gabr M, Mohamed I, El-Badri N. Hepatocellular carcinoma patients serum modulates the regenerative capacities of adipose mesenchymal stromal cells. Heliyon 2024; 10:e24794. [PMID: 38333871 PMCID: PMC10850426 DOI: 10.1016/j.heliyon.2024.e24794] [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: 04/22/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent cancers causing the highest mortality rate worldwide. Treatment options of surgery, radiation, cytotoxic drugs and liver transplantation suffer significant side effects and a high frequency of relapse. Stem cell therapy has been proposed as a new effective therapy, however, controversial reports are emerging on the role of mesenchymal stem cells in cancer. In this work, we aimed to assess the regenerative capacities of adipose mesenchymal stem cells when exposed to serum from HCC patients, by assessing the effect of the sera on modulating the regenerative capacities of h-AMSCs and the cancer properties in HCC cells. This will pave the way for maximizing the efficacy of MSCs in cancer therapy. Our data show that HCC serum-treated hA-MSCs suffered oncogene-induced senescence as shown by their altered morphology and ameliorated proliferation and differentiation. The cells were enlarged with small irregular nuclei, swollen rough endoplasmic reticulum cisternae, and aging lysosomes typified by dark residual bodies. HCC serum-treated Huh-7 cancer cells on the other hand displayed higher tumor aggressiveness as depicted by altered morphology, increased cellular proliferation and migration, and decreased percentage of early and late apoptotic cells. Our findings provide evidence that exposure of hA-MSCs to the serum of HCC patients decreases their regenerative capacities and should be considered when employed as a potential therapy in HCC patients.
Collapse
Affiliation(s)
- Radwa Ayman Salah
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Azza M. El-Derby
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Zaynab El-Gammal
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
- Stem Cells and Regenerative Medicine Department, Egypt Center for Research and Regenerative Medicine (ECRRM), Giza, 12578, Egypt
| | - Bishoy Wadie
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Sara M. Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Shimaa E. Elshenawy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
- Stem Cells and Regenerative Medicine Department, Egypt Center for Research and Regenerative Medicine (ECRRM), Giza, 12578, Egypt
| | - Shireen Magdy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Ayman Salah
- Department of Hepatogastroenterology, Kasr El-Aini Cairo University, Cairo, Egypt
| | - Mahmoud Gabr
- Urology and Nephrology Center, Mansoura, 35516, Egypt
| | - Ihab Mohamed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, 12578, Egypt
| |
Collapse
|
7
|
Akkız H. Emerging Role of Cancer-Associated Fibroblasts in Progression and Treatment of Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:3941. [PMID: 36835352 PMCID: PMC9964606 DOI: 10.3390/ijms24043941] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 02/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent cancers worldwide and the fourth leading cause of cancer-related death globally. Tumor cells recruit and remodel various types of stromal and inflammatory cells to form a tumor microenvironment (TME), which encompasses cellular and molecular entities, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), immune cells, myeloid-derived suppressor cells (MDSCs), immune checkpoint molecules and cytokines that promote cancer cell growth, as well as their drug resistance. HCC usually arises in the context of cirrhosis, which is always associated with an enrichment of activated fibroblasts that are owed to chronic inflammation. CAFs are a major component of the TME, providing physical support in it and secreting various proteins, such as extracellular matrices (ECMs), hepatocyte growth factor (HGF), insulin-like growth factor 1/2 (ILGF1/2) and cytokines that can modulate tumor growth and survival. As such, CAF-derived signaling may increase the pool of resistant cells, thus reducing the duration of clinical responses and increasing the degree of heterogeneity within tumors. Although CAFs are often implicated to be associated with tumor growth, metastasis and drug resistance, several studies have reported that CAFs have significant phenotypic and functional heterogeneity, and some CAFs display antitumor and drug-sensitizing properties. Multiple studies have highlighted the relevance of crosstalk between HCC cells, CAFs and other stromal cells in influence of HCC progression. Although basic and clinical studies partially revealed the emerging roles of CAFs in immunotherapy resistance and immune evasion, a better understanding of the unique functions of CAFs in HCC progression will contribute to development of more effective molecular-targeted drugs. In this review article, molecular mechanisms involved in crosstalk between CAFs, HCC cells and other stromal cells, as well as the effects of CAFs on HCC-cell growth, metastasis, drug resistance and clinical outcomes, are comprehensively discussed.
Collapse
Affiliation(s)
- Hikmet Akkız
- Department of Gastroenterology and Hepatology, The University of Bahçeşehir, Istanbul 34425, Turkey
| |
Collapse
|
8
|
Ying F, Chan MSM, Lee TKW. Cancer-Associated Fibroblasts in Hepatocellular Carcinoma and Cholangiocarcinoma. Cell Mol Gastroenterol Hepatol 2023; 15:985-999. [PMID: 36708970 PMCID: PMC10040968 DOI: 10.1016/j.jcmgh.2023.01.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
Primary liver cancer (PLC) includes hepatocellular carcinoma and intrahepatic cholangiocarcinoma and is the sixth most common cancer worldwide with poor prognosis. PLC is characterized by an abundant stromal reaction in which cancer-associated fibroblasts (CAFs) are one of the major stromal components. Solid evidence has demonstrated the crucial role of CAFs in tumor progression, and CAF abundance is often correlated with poor clinical outcomes. Although CAFs are regarded as an attractive and promising target for PLC treatment, a poor understanding of CAF origins and heterogeneity and a lack of specific CAF markers are the major hurdles to efficient CAF-specific therapy. In this review, we examine recent advances in the understanding of CAF diversity in the context of biomarkers, subtypes, and functions in PLC. The regulatory roles of CAFs in extracellular matrix remodeling, metastasis, cancer stemness, and therapeutic resistance are summarized. With an increasing link between CAF abundance and reduced antitumor immune responses, we provide updated knowledge on the crosstalk between CAFs and immune cells within the tumor microenvironment, which leads to immune resistance. In addition, we present current CAF-targeted therapies and describe some future perspectives. A better understanding of CAF biology will shed light on a novel therapeutic strategy against PLC.
Collapse
Affiliation(s)
- Fan Ying
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong
| | - Mandy Sze Man Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong
| | - Terence Kin Wah Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hong Kong.
| |
Collapse
|
9
|
De Falco E, Bordin A, Menna C, Dhori X, Picchio V, Cozzolino C, De Marinis E, Floris E, Maria Giorgiano N, Rosa P, Angelo Rendina E, Ibrahim M, Calogero A. Remote Adipose Tissue-Derived Stromal Cells of Patients with Lung Adenocarcinoma Generate a Similar Malignant Microenvironment of the Lung Stromal Counterpart. JOURNAL OF ONCOLOGY 2023; 2023:1011063. [PMID: 36733673 PMCID: PMC9889152 DOI: 10.1155/2023/1011063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/25/2022] [Accepted: 12/25/2022] [Indexed: 01/26/2023]
Abstract
Cancer alters both local and distant tissue by influencing the microenvironment. In this regard, the interplay with the stromal fraction is considered critical as this latter can either foster or hamper the progression of the disease. Accordingly, the modality by which tumors may alter distant niches of stromal cells is still unclear, especially at early stages. In this short report, we attempt to better understand the biology of this cross-talk. In our "autologous stromal experimental setting," we found that remote adipose tissue-derived mesenchymal stem cells (mediastinal AMSC) obtained from patients with lung adenocarcinoma sustain proliferation and clonogenic ability of A549 and human primary lung adenocarcinoma cells similarly to the autologous stromal lung counterpart (LMSC). This effect is not observed in lung benign diseases such as the hamartochondroma. This finding was validated by conditioning benign AMSC with supernatants from LAC for up to 21 days. The new reconditioned media of the stromal fraction so obtained, was able to increase cell proliferation of A549 cells at 14 and 21 days similar to that derived from AMSC of patients with lung adenocarcinoma. The secretome generated by remote AMSC revealed overlapping to the corresponding malignant microenvironment of the autologous local LMSC. Among the plethora of 80 soluble factors analyzed by arrays, a small pool of 5 upregulated molecules including IL1-β, IL-3, MCP-1, TNF-α, and EGF, was commonly shared by both malignant-like autologous A- and L-MSC derived microenvironments vs those benign. The bioinformatics analysis revealed that these proteins were strictly and functionally interconnected to lung fibrosis and proinflammation and that miR-126, 101, 486, and let-7-g were their main targets. Accordingly, we found that in lung cancer tissues and blood samples from the same set of patients here employed, miR-126 and miR-486 displayed the highest expression levels in tissue and blood, respectively. When the miR-126-3p was silenced in A549 treated with AMSC-derived conditioned media from patients with lung adenocarcinoma, cell proliferation decreased compared to control media.
Collapse
Affiliation(s)
- Elena De Falco
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Antonella Bordin
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| | - Cecilia Menna
- Department of Thoracic Surgery, University “Sapienza”, S. Andrea Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Xhulio Dhori
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| | - Vittorio Picchio
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| | - Claudia Cozzolino
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| | - Elisabetta De Marinis
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| | - Erica Floris
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| | - Noemi Maria Giorgiano
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| | - Paolo Rosa
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| | - Erino Angelo Rendina
- Department of Thoracic Surgery, University “Sapienza”, S. Andrea Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Mohsen Ibrahim
- Department of Thoracic Surgery, University “Sapienza”, S. Andrea Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Antonella Calogero
- Department of Medical-Surgical Science and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome “Sapienza”, C.so della Repubblica 79 04100, Latina, Italy
| |
Collapse
|
10
|
Stem Cells for Cancer Therapy: Translating the Uncertainties and Possibilities of Stem Cell Properties into Opportunities for Effective Cancer Therapy. Int J Mol Sci 2023; 24:ijms24021012. [PMID: 36674525 PMCID: PMC9864033 DOI: 10.3390/ijms24021012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Cancer recurrence and drug resistance following treatment, as well as metastatic forms of cancer, are trends that are commonly encountered in cancer management. Amidst the growing popularity of personalized medicine and targeted therapy as effective cancer treatment, studies involving the use of stem cells in cancer therapy are gaining ground as promising translational treatment options that are actively pursued by researchers due to their unique tumor-homing activities and anti-cancer properties. Therefore, this review will highlight cancer interactions with commonly studied stem cell types, namely, mesenchymal stroma/stem cells (MSC), induced pluripotent stem cells (iPSC), iPSC-derived MSC (iMSC), and cancer stem cells (CSC). A particular focus will be on the effects of paracrine signaling activities and exosomal miRNA interaction released by MSC and iMSCs within the tumor microenvironment (TME) along with their therapeutic potential as anti-cancer delivery agents. Similarly, the role of exosomal miRNA released by CSCs will be further discussed in the context of its role in cancer recurrence and metastatic spread, which leads to a better understanding of how such exosomal miRNA could be used as potential forms of non-cell-based cancer therapy.
Collapse
|
11
|
Peng H, Zhu E, Zhang Y. Advances of cancer-associated fibroblasts in liver cancer. Biomark Res 2022; 10:59. [PMID: 35971182 PMCID: PMC9380339 DOI: 10.1186/s40364-022-00406-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/01/2022] [Indexed: 12/24/2022] Open
Abstract
Liver cancer is one of the most common malignant tumors worldwide, it is ranked sixth in incidence and fourth in mortality. According to the distinct origin of malignant tumor cells, liver cancer is mainly divided into hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). Since most cases are diagnosed at an advanced stage, the prognosis of liver cancer is poor. Tumor growth depends on the dynamic interaction of various cellular components in the tumor microenvironment (TME). As the most abundant components of tumor stroma, cancer-associated fibroblasts (CAFs) have been involved in the progression of liver cancer. The interplay between CAFs and tumor cells, immune cells, or vascular endothelial cells in the TME through direct cell-to-cell contact or indirect paracrine interaction, affects the initiation and development of tumors. Additionally, CAFs are not a homogeneous cell population in liver cancer. Recently, single-cell sequencing technology has been used to help better understand the diversity of CAFs in liver cancer. In this review, we mainly update the knowledge of CAFs both in HCC and CCA, including their cell origins, chemoresistance, tumor stemness induction, tumor immune microenvironment formation, and the role of tumor cells on CAFs. Understanding the context-dependent role of different CAFs subsets provides new strategies for precise liver cancer treatment.
Collapse
Affiliation(s)
- Hao Peng
- Medical School, Southeast University, Nanjing, 210009, China
| | - Erwei Zhu
- The Second People's Hospital of Lianyungang (The Oncology Hospital of Lianyungang), Lianyungang, 222006, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210009, China.
| |
Collapse
|