1
|
Meng Z, Li T, Li J, Ding S, Liu Y, Zhao G, Chen C, Zhao P, Zhou L. LncRNAPVT1 is Associated with Cancer-Associated Fibroblasts Proliferation Through Regulating TGF-βin Oral Squamous Cell Carcinoma. Immunol Invest 2024; 53:1250-1263. [PMID: 39189542 DOI: 10.1080/08820139.2024.2395874] [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] [Indexed: 08/28/2024]
Abstract
INTRODUCTION Human oral squamous cell carcinoma (OSCC) is the most common type of oral cancer and has a poor survival rate. Cell-cell communication between OSCC cells and cancer-associated fibroblasts (CAFs) plays important roles in OSCC progression. We previously demonstrated that CAFs promote OSCC cell migration and invasion. However, how OSCC cells influence CAFs proliferation is unknown. METHODS Knockdown of PVT1 was confirmed using lentivirus infection technique. CAFs in tissues were identified by staining the cells with α-SMA using immunohistochemical technique. CCK-8 assay was used to evaluate cell proliferation. The mRNA level of a gene was measured by qRT-PCR. Secreted TGF-β were detected using ELISA assay. RESULTS We found that knockdown of the long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) was associated with a low density of CAFs in xenograft tumors in mice; further analysis revealed that PVT1 in OSCC cells induced CAF proliferation through transforming growth factor (TGF)-β. DISCUSSION Our results demonstrate that lncRNA PVT1 in tumor cells participates in CAF development in OSCC by regulating TGF-β. This study revealed a new mechanism by which PVT1 regulates OSCC progression and PVT1 is a potential therapeutic target in OSCC.
Collapse
Affiliation(s)
- Zhen Meng
- Biomedical Laboratory, Medical School of Liaocheng University, Liaocheng, Shandong Province, P.R. China
| | - Tongjuan Li
- Department of Stomatology, Anqiu Municipal Hospital, Weifang, Shandong Province, P.R. China
| | - Jun Li
- Precision Biomedical Laboratory of Liaocheng, Liaocheng People's Hospital, Medical School of Liaocheng University, Liaocheng, Shandong Province, P.R. China
| | - Shuxin Ding
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Weifang Medicial University, Weifang, Shandong Province, P.R. China
| | - Yujiao Liu
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
| | - Guoli Zhao
- Department of Pathology, Liaocheng Tumor Hospital, Liaocheng, Shandong Province, P.R. China
| | - Cheng Chen
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
| | - Peng Zhao
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
| | - Longxun Zhou
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, P.R. China
| |
Collapse
|
2
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [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: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
3
|
Cui J, Yang Z, Ma R, He W, Tao H, Li Y, Zhao Y. Placenta-targeted Treatment Strategies for Preeclampsia and Fetal Growth Restriction: An Opportunity and Major Challenge. Stem Cell Rev Rep 2024; 20:1501-1511. [PMID: 38814409 PMCID: PMC11319408 DOI: 10.1007/s12015-024-10739-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] [Accepted: 05/17/2024] [Indexed: 05/31/2024]
Abstract
The placenta plays a crucial role in maintaining normal pregnancy. The failure of spiral artery remodeling (SAR) is a key factor leading to placental ischemia and poor perfusion which is strongly associated with obstetric diseases, including preeclampsia (PE) and fetal growth restriction (FGR). Existing interventions for PE and FGR are limited and termination of pregnancy is inevitable when the maternal or fetus condition deteriorates. Considering the safety of the mother and fetus, treatments that may penetrate the placental barrier and harm the fetus are not accepted. Developing targeted treatment strategies for these conditions is urgent and necessary. With the proven efficacy of targeted therapy in treating conditions such as endometrial cancer and trophoblastic tumors, research on placental dysfunction continues to deepen. This article reviews the studies on placenta-targeted treatment and drug delivery strategies, summarizes the characteristics proposes corresponding improvement measures in targeted treatment, provides solutions for existing problems, and makes suggestions for future studies.
Collapse
Affiliation(s)
- Jianjian Cui
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zejun Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Ruilin Ma
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Wencong He
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Hui Tao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Ya'nan Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yin Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China.
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China.
| |
Collapse
|
4
|
Park J, Park SA, Kim YS, Kim D, Shin S, Lee SH, Jeun SS, Chung YJ, Ahn S. Intratumoral IL-12 delivery via mesenchymal stem cells combined with PD-1 blockade leads to long-term antitumor immunity in a mouse glioblastoma model. Biomed Pharmacother 2024; 173:115790. [PMID: 38431436 DOI: 10.1016/j.biopha.2023.115790] [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: 08/09/2023] [Revised: 10/18/2023] [Accepted: 10/26/2023] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Although PD-1 blockade is effective for treating several types of cancer, the efficacy of this agent in glioblastoma is largely limited. To overcome non-responders and the immunosuppressive tumor microenvironment, combinational immunotherapeutic strategies with anti-PD-1 need to be considered. Here, we developed IL-12-secreting mesenchymal stem cells (MSC_IL-12) with glioblastoma tropism and evaluated the therapeutic effects of anti-PD-1, MSC_IL-12, and their combination against glioblastoma. METHODS Therapeutic responses were evaluated using an immunocompetent mouse orthotopic model. Tumor-infiltrating lymphocytes (TILs) were analyzed using immunofluorescent imaging. Single-cell transcriptome was obtained from mouse brains after treatments. RESULTS Anti-PD-1 and MSC_IL-12 showed complete tumor remission in 25.0% (4/16) and 23.1% (3/13) of glioblastoma-implanted mice, respectively, and their combination yielded synergistic antitumor efficacy indicated by 50.0% (6/12) of complete tumor remission. Analyses of TILs revealed that anti-PD-1 increased CD8+ T cells, while MSC_IL-12 led to infiltration of CD4+ T cells and NK cells. Both therapies reduced frequencies of Tregs. All these aspects observed in each monotherapy group were superimposed in the combination group. Notably, no tumor growth was observed upon rechallenge in cured mice, indicating long-term immunity against glioblastoma provoked by the therapies. Single-cell RNA-seq data confirmed these results and revealed that the combined treatment led to immune-favorable tumor microenvironment-CD4+, CD8+ T cells, effector memory T cells, and activated microglia were increased, whereas exhausted T cells, Tregs, and M2 polarized microglia were reduced. CONCLUSION Anti-PD-1 and MSC_IL-12 monotherapies show long-term therapeutic responses, and their combination further enhances antitumor efficacy against glioblastoma via inducing immune-favorable tumor microenvironment.
Collapse
Affiliation(s)
- Junseong Park
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Soon A Park
- Department of Bio medicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Neurosurgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon-Seob Kim
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dokyeong Kim
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Bio medicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sun Shin
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sug Hyung Lee
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yeun-Jun Chung
- Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Bio medicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Stephen Ahn
- Department of Neurosurgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Liu X, Gao R, Wu Q, Li G, Xu X, Li W, Liu P, Wang X, Cai J, Li M, Wang Z. ITGA7 loss drives the differentiation of adipose-derived mesenchymal stem cells to cancer-associated fibroblasts. Mol Carcinog 2024; 63:479-493. [PMID: 38174862 DOI: 10.1002/mc.23665] [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: 08/11/2023] [Revised: 11/09/2023] [Accepted: 11/21/2023] [Indexed: 01/05/2024]
Abstract
Cancer-associated fibroblasts (CAFs) represent a major cellular component of the tumor (pre-)metastatic niche and play an essential role in omental dissemination of ovarian cancer. The omentum is rich in adipose, and adipose-derived mesenchymal stem cells (ADSCs) have been identified as a source of CAFs. However, the molecular events driving the phenotype shift of ADSCs remain largely unexplored. In this research, we focus on integrins, transmembrane receptors that have been widely involved in cellular plasticity. We found that integrin α7 (ITGA7) was the only member of the integrin family that positively correlated with both overall survival and progression-free survival in ovarian cancer through GEPIA2. The immunohistochemistry signal of ITGA7 was apparent in the tumor stroma, and a lower omental ITGA7 level was associated with metastasis. Primary ADSCs were isolated from the omentum of patients with ovarian cancer and identified by cellular morphology, biomarkers, and multilineage differentiation. The conditional medium of ovarian cancer cells induced ITGA7 expression decrease and phenotypic changes in ADSCs. Downregulation of ITGA7 in primary omental ADSCs led to decrease in stemness properties and emerge of characteristic morphology and biomarkers of CAFs. Moreover, the conditioned medium of ADSCs with ITGA7 depletion exhibited enhanced abilities to improve the migration and invasion of ovarian cancer cells in vitro. Overall, these findings indicate that loss of ITGA7 may induce the differentiation of ADSCs to CAFs that contribute to a tumor-supportive niche.
Collapse
Affiliation(s)
- Xiaoli Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rui Gao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiulei Wu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guoqing Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaohan Xu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenhan Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pan Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoman Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Li
- Obstetrics and Gynecology Department, Center for Reproductive Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zehua Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
6
|
Xu K, Li D, Qian J, Zhang Y, Zhang M, Zhou H, Hou X, Jiang J, Zhang Z, Sun H, Shi G, Dai H, Liu H. Single-cell disulfidptosis regulator patterns guide intercellular communication of tumor microenvironment that contribute to kidney renal clear cell carcinoma progression and immunotherapy. Front Immunol 2024; 15:1288240. [PMID: 38292868 PMCID: PMC10824999 DOI: 10.3389/fimmu.2024.1288240] [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: 09/04/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
Background Disulfidptosis, an emerging type of programmed cell death, plays a pivotal role in various cancer types, notably impacting the progression of kidney renal clear cell carcinoma (KIRC) through the tumor microenvironment (TME). However, the specific involvement of disulfidptosis within the TME remains elusive. Methods Analyzing 41,784 single cells obtained from seven samples of KIRC through single-cell RNA sequencing (scRNA-seq), this study employed nonnegative matrix factorization (NMF) to assess 24 disulfidptosis regulators. Pseudotime analysis, intercellular communication mapping, determination of transcription factor activities (TFs), and metabolic profiling of the TME subgroup in KIRC were conducted using Monocle, CellChat, SCENIC, and scMetabolism. Additionally, public cohorts were utilized to predict prognosis and immune responses within the TME subgroup of KIRC. Results Through NMF clustering and differential expression marker genes, fibroblasts, macrophages, monocytes, T cells, and B cells were categorized into four to six distinct subgroups. Furthermore, this investigation revealed the correlation between disulfidptosis regulatory factors and the biological traits, as well as the pseudotime trajectories of TME subgroups. Notably, disulfidptosis-mediated TME subgroups (DSTN+CD4T-C1 and FLNA+CD4T-C2) demonstrated significant prognostic value and immune responses in patients with KIRC. Multiple immunohistochemistry (mIHC) assays identified marker expression within both cell clusters. Moreover, CellChat analysis unveiled diverse and extensive interactions between disulfidptosis-mediated TME subgroups and tumor epithelial cells, highlighting the TNFSF12-TNFRSF12A ligand-receptor pair as mediators between DSTN+CD4T-C1, FLNA+CD4T-C2, and epithelial cells. Conclusion Our study sheds light on the role of disulfidptosis-mediated intercellular communication in regulating the biological characteristics of the TME. These findings offer valuable insights for patients with KIRC, potentially guiding personalized immunotherapy approaches.
Collapse
Affiliation(s)
- Kangjie Xu
- Central Laboratory Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Dongling Li
- Nephrology Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Jinke Qian
- Urology Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Yanhua Zhang
- Obstetrics and Gynecology Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Minglei Zhang
- Oncology Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Hai Zhou
- Central Laboratory Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Xuefeng Hou
- Central Laboratory Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Jian Jiang
- Central Laboratory Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Zihang Zhang
- Pathology Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Hang Sun
- Urology Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Guodong Shi
- Medical Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| | - Hua Dai
- Yangzhou University Clinical Medical College, Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yancheng, Jiangsu, China
| | - Hui Liu
- Urology Department, Binhai County People’s Hospital, Yancheng, Jiangsu, China
| |
Collapse
|
7
|
Park JY, Park JY, Jeong YG, Park JH, Park YH, Kim SH, Khang D. Pancreatic Tumor-Targeting Stemsome Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2300934. [PMID: 37114740 DOI: 10.1002/adma.202300934] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/18/2023] [Indexed: 06/13/2023]
Abstract
Owing to the intrinsic ability of stem cells to target the tumor environment, stem-cell-membrane-functionalized nanocarriers can target and load active anticancer drugs. In this work, a strategy that focuses on stem cells that self-target pancreatic cancer cells is developed. In particular, malignant deep tumors such as pancreatic cancer cells, one of the intractable tumors that currently have no successful clinical strategy, are available for targeting and destruction. By gaining the targeting ability of stem cells against pancreatic tumor cells, stem cell membranes can encapsulate nano-polylactide-co-glycolide loaded with doxorubicin to target and reduce deep pancreatic tumor tissues. Considering the lack of known target proteins on pancreatic tumor cells, the suggested platform technology can be utilized for targeting any malignant tumors in which surface target receptors are unavailable.
Collapse
Affiliation(s)
- Jun-Young Park
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
| | - Jun Young Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Yong-Gyu Jeong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
| | - Joo-Hwan Park
- Division of Medical Oncology, Department of Internal Medicine, Gil Medical Center, College of Medicine, Gachon University, Incheon, 21565, South Korea
| | - Yeon Ho Park
- Department of Surgery, Gil Medical Center, College of Medicine, Gachon University, Incheon, 21565, South Korea
| | - Sang-Hyun Kim
- CMRI, Department of Pharmacology, College of Medicine, Kyungpook National University, Daegu, 41944, South Korea
| | - Dongwoo Khang
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, 21999, South Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, South Korea
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, South Korea
- Ectosome Inc., Incheon, 21999, South Korea
| |
Collapse
|
8
|
Fan M, Zhang Y, Shi H, Xiang L, Yao H, Lin R. Bone mesenchymal stem cells promote gastric cancer progression through TGF-β1/Smad2 positive feedback loop. Life Sci 2023; 323:121657. [PMID: 37019301 DOI: 10.1016/j.lfs.2023.121657] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023]
Abstract
AIMS Bone marrow-derived mesenchymal stem cells (BMSCs) have been proven to be recruited into the tumor microenvironment and contribute to gastric cancer (GC) progression, but the underlying mechanism is still unclear. The purpose of this study is to explore the exact role and potential mechanism of BMSCs in the progression of GC. MATERIALS AND METHODS Bioinformatics analyzed were used to clarify the correlation between TGF-β1 and prognosis of gastric cancer. Cell co-culture were used to explore the interaction between gastric cancer cells (GCs) and BMSCs. Quantitative real time-PCR and Western blot assay were used to detect gene and protein expression, respectively. The biological characteristics of GCs and BMSCs were detected by immunofluorescence, Transwell migration, Elisa and invasion assay. Xenograft models in nude mice were constructed to evaluate GC development in vivo. KEY FINDINGS TGF-β1 was overexpressed in GC cells and tissues, and is positively related to the poor prognosis of patients. TGF-β1 from GCs activated the Smad2 pathway in BMSCs, promoting their differentiation into carcinoma-associated fibroblasts (CAFs) and TGF-β1 expression. Concomitantly, TGF-β1 secreted by CAFs activate Smad2 signaling in GC cells, thus inducing their epithelial-mesenchymal transition (EMT) and TGF-β1 secretion. BMSCs can dramatically promote the proliferation, migration, and invasion of GCs while blocking TGF-β1/Smad2 positive feedback loop can reverse these effects. SIGNIFICANCE The TGF-β1/Smad2 positive feedback loop between GCs and BMSCs, promotes the CAFs differentiation of BMSCs and the epithelial-mesenchymal transition of GCs, resulting in the progression of GC.
Collapse
|
9
|
Toledo B, Picon-Ruiz M, Marchal JA, Perán M. Dual Role of Fibroblasts Educated by Tumour in Cancer Behavior and Therapeutic Perspectives. Int J Mol Sci 2022; 23:15576. [PMID: 36555218 PMCID: PMC9778751 DOI: 10.3390/ijms232415576] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Tumours are complex systems with dynamic interactions between tumour cells, non-tumour cells, and extracellular components that comprise the tumour microenvironment (TME). The majority of TME's cells are cancer-associated fibroblasts (CAFs), which are crucial in extracellular matrix (ECM) construction, tumour metabolism, immunology, adaptive chemoresistance, and tumour cell motility. CAF subtypes have been identified based on the expression of protein markers. CAFs may act as promoters or suppressors in tumour cells depending on a variety of factors, including cancer stage. Indeed, CAFs have been shown to promote tumour growth, survival and spread, and secretome changes, but they can also slow tumourigenesis at an early stage through mechanisms that are still poorly understood. Stromal-cancer interactions are governed by a variety of soluble factors that determine the outcome of the tumourigenic process. Cancer cells release factors that enhance the ability of fibroblasts to secrete multiple tumour-promoting chemokines, acting on malignant cells to promote proliferation, migration, and invasion. This crosstalk between CAFs and tumour cells has given new prominence to the stromal cells, from being considered as mere physical support to becoming key players in the tumour process. Here, we focus on the concept of cancer as a non-healing wound and the relevance of chronic inflammation to tumour initiation. In addition, we review CAFs heterogeneous origins and markers together with the potential therapeutic implications of CAFs "re-education" and/or targeting tumour progression inhibition.
Collapse
Affiliation(s)
- Belén Toledo
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
| | - Manuel Picon-Ruiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Instituto de Investigación Sanitaria ibs. GRANADA, Hospitales Universitarios de Granada-Universidad de Granada, E-18071 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, E-18016 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, E-23071 Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, E-18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, E-18016 Granada, Spain
| |
Collapse
|
10
|
Sharpe BP, Hayden A, Manousopoulou A, Cowie A, Walker RC, Harrington J, Izadi F, Breininger SP, Gibson J, Pickering O, Jaynes E, Kyle E, Saunders JH, Parsons SL, Ritchie AA, Clarke PA, Collier P, Mongan NP, Bates DO, Yacqub-Usman K, Garbis SD, Walters Z, Rose-Zerilli M, Grabowska AM, Underwood TJ. Phosphodiesterase type 5 inhibitors enhance chemotherapy in preclinical models of esophageal adenocarcinoma by targeting cancer-associated fibroblasts. Cell Rep Med 2022; 3:100541. [PMID: 35732148 PMCID: PMC9244979 DOI: 10.1016/j.xcrm.2022.100541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/14/2021] [Accepted: 01/28/2022] [Indexed: 12/03/2022]
Abstract
The chemotherapy resistance of esophageal adenocarcinomas (EACs) is underpinned by cancer cell extrinsic mechanisms of the tumor microenvironment (TME). We demonstrate that, by targeting the tumor-promoting functions of the predominant TME cell type, cancer-associated fibroblasts (CAFs) with phosphodiesterase type 5 inhibitors (PDE5i), we can enhance the efficacy of standard-of-care chemotherapy. In ex vivo conditions, PDE5i prevent the transdifferentiation of normal fibroblasts to CAF and abolish the tumor-promoting function of established EAC CAFs. Using shotgun proteomics and single-cell RNA-seq, we reveal PDE5i-specific regulation of pathways related to fibroblast activation and tumor promotion. Finally, we confirm the efficacy of PDE5i in combination with chemotherapy in close-to-patient and in vivo PDX-based model systems. These findings demonstrate that CAFs drive chemotherapy resistance in EACs and can be targeted by repurposing PDE5i, a safe and well-tolerated class of drug administered to millions of patients world-wide to treat erectile dysfunction.
Collapse
Affiliation(s)
- Benjamin P Sharpe
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Annette Hayden
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | | | - Andrew Cowie
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Robert C Walker
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Jack Harrington
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Fereshteh Izadi
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK; Centre for NanoHealth, Swansea University Medical School, Singleton Campus, Swansea SA2 8PP, UK
| | - Stella P Breininger
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Jane Gibson
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Oliver Pickering
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Eleanor Jaynes
- University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Ewan Kyle
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - John H Saunders
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Salford Royal NHS Foundation Trust, Salford M6 8HD, UK
| | - Simon L Parsons
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Nottingham University Hospitals NHS Trust, Hucknall Road, Nottingham NG5 1PB, UK
| | - Alison A Ritchie
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Philip A Clarke
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Pamela Collier
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nigel P Mongan
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA; Biodiscovery Institute, School of Veterinary Medicine and Science, University of Nottingham, Nottingham NG5 1PB, UK
| | - David O Bates
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Kiren Yacqub-Usman
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | | | - Zoë Walters
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Matthew Rose-Zerilli
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK
| | - Anna M Grabowska
- Ex Vivo Cancer Pharmacology Centre of Excellence, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Timothy J Underwood
- School of Cancer Sciences, Faculty of Medicine, Room CS B2, MP824, Somers Cancer Research Building, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK.
| |
Collapse
|
11
|
Fan C, Liao M, Xie L, Huang L, Lv S, Cai S, Su X, Wang Y, Wang H, Wang M, Liu Y, Wang Y, Guo H, Yang H, Liu Y, Wang T, Ma L. Single-Cell Transcriptome Integration Analysis Reveals the Correlation Between Mesenchymal Stromal Cells and Fibroblasts. Front Genet 2022; 13:798331. [PMID: 35360851 PMCID: PMC8961367 DOI: 10.3389/fgene.2022.798331] [Citation(s) in RCA: 5] [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: 10/20/2021] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Mesenchymal stromal cells (MSCs) and fibroblasts show similar morphology, surface marker expression, and proliferation, differentiation, and immunomodulatory capacities. These similarities not only blur their cell identities but also limit their application. Methods: We performed single-cell transcriptome sequencing of the human umbilical cord and foreskin MSCs (HuMSCs and FSMSCs) and extracted the single-cell transcriptome data of the bone marrow and adipose MSCs (BMSCs and ADMSCs) from the Gene Expression Omnibus (GEO) database. Then, we performed quality control, batch effect correction, integration, and clustering analysis of the integrated single-cell transcriptome data from the HuMSCs, FMSCs, BMSCs, and ADMSCs. The cell subsets were annotated based on the surface marker phenotypes for the MSCs (CD105 + , CD90 +, CD73 +, CD45 -, CD34 -, CD19 -, HLA-DRA -, and CD11b -), fibroblasts (VIM +, PECAM1 -, CD34 -, CD45 -, EPCAM -, and MYH11 -), and pericytes (CD146 +, PDGFRB +, PECAM1 -, CD34 -, and CD45 -). The expression levels of common fibroblast markers (ACTA2, FAP, PDGFRA, PDGFRB, S100A4, FN1, COL1A1, POSTN, DCN, COL1A2, FBLN2, COL1A2, DES, and CDH11) were also analyzed in all cell subsets. Finally, the gene expression profiles, differentiation status, and the enrichment status of various gene sets and regulons were compared between the cell subsets. Results: We demonstrated 15 distinct cell subsets in the integrated single-cell transcriptome sequencing data. Surface marker annotation demonstrated the MSC phenotype in 12 of the 15 cell subsets. C10 and C14 subsets demonstrated both the MSC and pericyte phenotypes. All 15 cell subsets demonstrated the fibroblast phenotype. C8, C12, and C13 subsets exclusively demonstrated the fibroblast phenotype. We identified 3,275 differentially expressed genes, 305 enriched gene sets, and 34 enriched regulons between the 15 cell subsets. The cell subsets that exclusively demonstrated the fibroblast phenotype represented less primitive and more differentiated cell types. Conclusion: Cell subsets with the MSC phenotype also demonstrated the fibroblast phenotype, but cell subsets with the fibroblast phenotype did not necessarily demonstrate the MSC phenotype, suggesting that MSCs represented a subclass of fibroblasts. We also demonstrated that the MSCs and fibroblasts represented highly heterogeneous populations with distinct cell subsets, which could be identified based on the differentially enriched gene sets and regulons that specify proliferating, differentiating, metabolic, and/or immunomodulatory functions.
Collapse
Affiliation(s)
- Chuiqin Fan
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Maochuan Liao
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Lichun Xie
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University (The Women and Children’s Medical Center of Guangzhou Medical University), Guangzhou, China
| | - Liangping Huang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Siyu Lv
- Department of Hematology and Oncology, Shenzhen Children’s Hospital of China Medical University, Shenzhen, China
| | - Siyu Cai
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xing Su
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yue Wang
- Department of Hematology and Oncology, Shenzhen Children’s Hospital of China Medical University, Shenzhen, China
| | - Hongwu Wang
- Department of Hematology and Oncology, Shenzhen Children’s Hospital of China Medical University, Shenzhen, China
| | - Manna Wang
- Department of Hematology and Oncology, Shenzhen Children’s Hospital of China Medical University, Shenzhen, China
| | - Yulin Liu
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yu Wang
- Department of Hematology and Oncology, Shenzhen Children’s Hospital of China Medical University, Shenzhen, China
| | - Huijie Guo
- Department of Hematology and Oncology, Shenzhen Children’s Hospital of China Medical University, Shenzhen, China
| | - Hanhua Yang
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University (The Women and Children’s Medical Center of Guangzhou Medical University), Guangzhou, China
| | - Yufeng Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tianyou Wang
- Department of Hematology and Oncology, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Lian Ma
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Pediatrics, The Third Affiliated Hospital of Guangzhou Medical University (The Women and Children’s Medical Center of Guangzhou Medical University), Guangzhou, China
- Department of Hematology and Oncology, Shenzhen Children’s Hospital of China Medical University, Shenzhen, China
| |
Collapse
|
12
|
Bellei B, Migliano E, Picardo M. Research update of adipose tissue-based therapies in regenerative dermatology. Stem Cell Rev Rep 2022; 18:1956-1973. [PMID: 35230644 DOI: 10.1007/s12015-022-10328-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2022] [Indexed: 12/09/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) have a spontaneous propensity to support tissue homeostasis and regeneration. Among the several sources of MSCs, adipose-derived tissue stem cells (ADSCs) have received major interest due to the higher mesenchymal stem cells concentration, ease, and safety of access. However, since a significant part of the natural capacity of ADSCs to repair damaged tissue is ascribable to their secretory activity that combines mitogenic factors, cytokines, chemokines, lipids, and extracellular matrix components, several studies focused on cell-free strategies. Furthermore, adipose cell-free derivatives are becoming more attractive especially for non-volumizing purposes, such as most dermatological conditions. However, when keratinocytes, fibroblasts, melanocytes, adipocytes, and hair follicle cells might not be locally sourced, graft of materials containing concentrated ADSCs is preferred. The usage of extracellular elements of adipose tissue aims to promote a self-autonomous regenerative microenvironment in the receiving area restoring physiological homeostasis. Hence, ADSCs or their paracrine activity are currently being studied in several dermatological settings including wound healing, skin fibrosis, burn, and aging.The present work analyzing both preclinical and clinical experiences gives an overview of the efficacy of adipose tissue-derivatives like autologous fat, the stromal vascular fraction (SVF), purified ADSCs, secretome and extracellular matrix graft in the field of regenerative medicine for the skin.
Collapse
Affiliation(s)
- Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Emilia Migliano
- Department of Plastic and Reconstructive Surgery, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
13
|
Abouelnaga H, El-Khateeb D, Moemen Y, El-Fert A, Elgazzar M, Khalil A. Characterization of mesenchymal stem cells isolated from Wharton’s jelly of the human umbilical cord. EGYPTIAN LIVER JOURNAL 2022. [DOI: 10.1186/s43066-021-00165-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Isolation of post-partum umbilical cord Wharton’s jelly stem cells has gained attention as an alternative source of the bone marrow. Because easy isolation, lack of ethical concerns, and the presence of both embryonic and adult stem cells have made them a valuable source for use in therapeutic applications and regenerative medicine. The study utilized a modified protocol using in-house human pooled cord blood serum for isolation and expansion of the mesenchymal stem cells obtained from the human umbilical cord Wharton’s jelly. Cell proliferation and population doubling time and tri-lineage differentiation were assessed, and the expressions of mesenchymal cell surface markers CD44, CD90, CD105, and CD34 were assessed by flow cytometry and RT-PCR. The genetic stability of the isolated cells was assessed by chromosomal karyotype.
Results
The isolated cells displayed fibroblastic-like morphology and tri-lineage differentiation into adipocyte, chondrocyte, and osteocyte. The isolated cells maintained the proliferative competence with a doubling time ranged from 38 to 42h and corresponded well with the standard positive and negative molecular markers (CD44+, CD90+, CD 105+, and CD34−). Cell senescence occurred at the later passage of the cells (P15) affecting, about 25% of the population. Metaphases spread of the cells showed normal diploid karyotypes, with typical chromosomal plates indicating genetic stability of the isolated cells.
Conclusion
The primary cultures exhibited success in isolating the umbilical cord Wharton’s jelly mesenchymal stem cells, which maintained their tri-lineage differentiation potential, phenotypes and karyotype characteristics on further passage and expansion.
Collapse
|
14
|
Oishi Y, Tamura R, Yoshida K, Toda M. Histopathological Investigation of Dura-like Membrane in Vestibular Schwannomas. Brain Sci 2021; 11:brainsci11121649. [PMID: 34942951 PMCID: PMC8699540 DOI: 10.3390/brainsci11121649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 11/30/2022] Open
Abstract
The dura-like membrane (DLM) is an outermost membranous structure arising from the dura mater adjacent to the internal auditory meatus (IAM) that envelops some vestibular schwannomas (VSs). Its recognition is important for the preservation of the facial and cochlear nerves during tumor resection. This study analyzes the histopathological characteristics of the DLM. The expression of CD34 and αSMA was histopathologically analyzed in tumor and DLM tissue of 10 primary VSs with and without a DLM. Tumor volume, resection volume percentage, microvessel density (MVD), and vessel diameter were analyzed. Volumetric analysis revealed that the presence of a DLM was significantly associated with lower tumor resection volume (p < 0.05). Intratumoral vessel diameter was significantly larger in the DLM group than the non-DLM group (p < 0.01). Larger VSs showed a higher intratumoral MVD in the DLM group (p < 0.05). Multilayered αSMA-positive vessels were identified in the DLM, tumor, and border; there tended to be more of these vessels within the tumor in the DLM group compared to the non-DLM group (p = 0.08). These arteriogenic characteristics suggest that the DLM is formed as the tumor induces feeding vessels from the dura mater around the IAM.
Collapse
|
15
|
Ghollasi M, Ghasembaglou S, Rahban D, Korani M, Motallebnezhad M, Asadi M, Zarredar H, Salimi A. Prospects for Manipulation of Mesenchymal Stem Cells in Tumor Therapy: Anti-Angiogenesis Property on the Spotlight. Int J Stem Cells 2021; 14:351-365. [PMID: 34456189 PMCID: PMC8611310 DOI: 10.15283/ijsc20146] [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: 09/16/2020] [Revised: 06/01/2021] [Accepted: 06/16/2021] [Indexed: 11/10/2022] Open
Abstract
The interactions between the tumor microenvironment and the tumor cells confers a condition that accelerate or decelerate the development of tumor. Of these cells, mesenchymal stem cells (MSCs) have the potential to modulate the tumor cells. MSCs have been established with double functions, whereby contribute to a tumorigenic or anti-tumor setting. Clinical studies have indicated the potential of MSCs to be used as tool in treating the human cancer cells. One of the advantageous features of MSCs that make them as a well-suited tool for cancer therapy is the natural tumor-trophic migration potential. A key specification of the tumor development has been stablished to be angiogenesis. As a result, manipulation of angiogenesis has become an attractive approach for cancer therapy. This review article will seek to clarify the anti-angiogenesis strategy in modulating the MSCs to treat the tumor cells.
Collapse
Affiliation(s)
- Marzieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran
| | - Shahram Ghasembaglou
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Dariush Rahban
- Department of Nanomedicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Korani
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Morteza Motallebnezhad
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Ali Salimi
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Bagheri R, Bitazar R, Talebi S, Alaeddini M, Etemad-Moghadam S, Eini L. Conditioned media derived from mesenchymal stem cells induces apoptosis and decreases cell viability and proliferation in squamous carcinoma cell lines. Gene 2021; 782:145542. [PMID: 33675953 DOI: 10.1016/j.gene.2021.145542] [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: 10/20/2020] [Revised: 01/26/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022]
Abstract
Squamous cell carcinoma (SCC) is a relatively common cancer with a low survival rate, poor prognosis and no effective treatment strategy. The use of cell-free conditioned media derived from mesenchymal stem cells (CM-MSCs) has shown promising results in treating various diseases. This study aimed to evaluate the effects of CM-MSCs on proliferation and apoptosis of CAL-27 and FaDu SCC cell lines. CM derived from human bone marrow and human amniotic membrane MSCs (BM-MSCs and AM-MSCs) was used in this investigation. MTT assay demonstrated that CM-BMMSC decreased the viability of CAL-27 and FaDu cell lines, 24, 48, and 72 h after treatment. Quantitative real-time PCR indicated that mRNA expression of PCNA as a proliferative marker, and BCL-2 as an anti-apoptotic protein, decreased in both cell lines treated with CM-BMMSC. Based on the flow cytometry results, the number of positive proliferative Ki67 cells and apoptotic Annexin-V cells decreased and increased in both cell lines treated with CM-BMMSC, respectively. However, CM-AMMSC treatment had both pro-and anti-neoplastic effects in our samples and showed considerable differences between the two cell lines. Taken together, our findings demonstrated that CM-BMMSC and, to a lesser degree, CM-AMMSC decrease cell viability and proliferation and increase cell apoptosis in SCC cell lines in a time-dependent manner. However, further studies are needed, especially to evaluate the anti-tumor potential of CM-BMMSC in vivo.
Collapse
Affiliation(s)
- Rezvan Bagheri
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, P.O. Box: 14155-5583, Tehran, Iran; Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, University of Sherbrooke, 3001 North 12th Avenue, Sherbrooke, QC J1H 5N4, Canada
| | - Razieh Bitazar
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, P.O. Box: 14155-5583, Tehran, Iran
| | - Saeed Talebi
- Department of Medical Genetics and Molecular Biology, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614525, Tehran, Iran
| | - Mojgan Alaeddini
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, P.O. Box: 14155-5583, Tehran, Iran
| | - Shahroo Etemad-Moghadam
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, P.O. Box: 14155-5583, Tehran, Iran
| | - Leila Eini
- Division of Histology, Department of Basic Science, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, P.O. Box: 1477893855, Tehran, Iran.
| |
Collapse
|
17
|
Sato M, Tamura R, Morimoto Y, Oishi Y, Yoshida K, Toda M. Quiescent and Activated Fibroblasts in Lateral Ventricular Meningioma With a Dura-like Membrane. World Neurosurg 2020; 147:e215-e224. [PMID: 33316485 DOI: 10.1016/j.wneu.2020.12.025] [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: 09/18/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Lateral ventricular meningioma (LVM) is a rare entity, accounting for 0.5%-5% of all intracranial meningiomas. This type of meningioma arises from meningothelial inclusion bodies in the tela choroidea and/or mesenchymal stroma of the choroid plexus. Although not yet fully characterized, a membranous structure is frequently observed around LVMs. This study analyzed quiescent and activated fibroblast phenotypes in LVMs with focus on the relationship between tumor growth and development of the membranous structure. METHODS This retrospective study analyzed 9 LVM cases for which gross total removal was achieved. Expression of the ependymal cell marker (Forkhead Box J1 [FoxJ1]) was histopathologically evaluated. The distribution of quiescent and activated fibroblasts was also analyzed using anti-fibroblast-specific protein-1 (FSP1)/S100A4 antibody and anti-α-smooth muscle actin (αSMA) antibody, respectively. The control group was 5 cases with primary convexity meningioma for which Simpson grade I removal was achieved. RESULTS Small LVMs (≤30 mm) were covered by a FoxJ1-positive(+) ependymal cell monolayer; no αSMA(+) cells were detected in the tumor; and a thick membrane capsule was not observed. None of the convexity meningiomas showed FoxJ1(+) cells. Large LVMs (>30 mm) had thick membrane capsules without an ependymal cell monolayer, which resembled dura mater. The FSP1/S100A4(+) and αSM(+) cells were clearly concentrated in the peripheral area just below the thick dura mater-like membrane capsules. CONCLUSIONS This study found an association between activated fibroblasts and dura mater-like membrane capsules in LVMs. The characteristics of membranous structure in LVMs may differ depending on tumor size.
Collapse
Affiliation(s)
- Mizuto Sato
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yukina Morimoto
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yumiko Oishi
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
18
|
Dai L, Song K, Di W. Adipocytes: active facilitators in epithelial ovarian cancer progression? J Ovarian Res 2020; 13:115. [PMID: 32967712 PMCID: PMC7513299 DOI: 10.1186/s13048-020-00718-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
There is growing evidence that adipocytes play important roles in the progression of multiple cancers. Moreover, in obesity, adipocytes alter their original functions and contribute to the metabolic and inflammatory changes of adipose tissue microenvironment, which can further enhance tumor development. At present, the roles of adipocytes in the pathogenesis of epithelial ovarian cancer (EOC) are far from being fully elucidated. Herein, we summarized the recent advances in understanding the roles of adipocytes in EOC progression. Adipocytes, close neighbors of EOC tissue, promote EOC growth, invasion, metastasis and angiogenesis through adipokine secretion, metabolic remodeling and immune microenvironment modulation. Moreover, adipocytes are important therapeutic targets and may work as useful anticancer drug delivery depot for EOC treatment. Furthermore, adipocytes also act as a therapeutic obstacle for their involvement in EOC treatment resistance. Hence, better characterization of the adipocytes in EOC microenvironment and the crosstalk between adipocytes and EOC cells may provide insights into EOC progression and suggest novel therapeutic opportunities.
Collapse
Affiliation(s)
- Lan Dai
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China. .,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Keqi Song
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wen Di
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China. .,Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China. .,State Key Laboratory of Oncogene and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
19
|
Castro-Oropeza R, Vazquez-Santillan K, Díaz-Gastelum C, Melendez-Zajgla J, Zampedri C, Ferat-Osorio E, Rodríguez-González A, Arriaga-Pizano L, Maldonado V. Adipose-derived mesenchymal stem cells promote the malignant phenotype of cervical cancer. Sci Rep 2020; 10:14205. [PMID: 32848147 PMCID: PMC7450089 DOI: 10.1038/s41598-020-69907-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 05/27/2020] [Indexed: 12/24/2022] Open
Abstract
Epidemiological studies indicate that obesity negatively affects the progression and treatment of cervical-uterine cancer. Recent evidence shows that a subpopulation of adipose-derived stem cells can alter cancer properties. In the present project, we described for the first time the impact of adipose-derived stem cells over the malignant behavior of cervical cancer cells. The transcriptome of cancer cells cultured in the presence of stem cells was analyzed using RNA-seq. Changes in gene expression were validated using digital-PCR. Bioinformatics tools were used to identify the main transduction pathways disrupted in cancer cells due to the presence of stem cells. In vitro and in vivo assays were conducted to validate cellular and molecular processes altered in cervical cancer cells owing to stem cells. Our results show that the expression of 95 RNAs was altered in cancer cells as a result of adipose-derived stem cells. Experimental assays indicate that stem cells provoke an increment in migration, invasion, angiogenesis, and tumorigenesis of cancer cells; however, no alterations were found in proliferation. Bioinformatics and experimental analyses demonstrated that the NF-kappa B signaling pathway is enriched in cancer cells due to the influence of adipose-derived stem cells. Interestingly, the tumor cells shift their epithelial to a mesenchymal morphology, which was reflected by the increased expression of specific mesenchymal markers. In addition, stem cells also promote a stemness phenotype in the cervical cancer cells. In conclusion, our results suggest that adipose-derived stem cells induce cervical cancer cells to acquire malignant features where NF-kappa B plays a key role.
Collapse
Affiliation(s)
- Rosario Castro-Oropeza
- Epigenetics Laboratories, National Institute of Genomic Medicine (INMEGEN), 14610, Mexico City, Mexico
| | - Karla Vazquez-Santillan
- Epigenetics Laboratories, National Institute of Genomic Medicine (INMEGEN), 14610, Mexico City, Mexico
| | - Claudia Díaz-Gastelum
- Epigenetics Laboratories, National Institute of Genomic Medicine (INMEGEN), 14610, Mexico City, Mexico
| | - Jorge Melendez-Zajgla
- Functional Genomics Laboratories, National Institute of Genomic Medicine (INMEGEN), 14610, Mexico City, Mexico
| | - Cecilia Zampedri
- Functional Genomics Laboratories, National Institute of Genomic Medicine (INMEGEN), 14610, Mexico City, Mexico
| | - Eduardo Ferat-Osorio
- Gastrosurgery Service, UMAE, National Medical Center "Siglo XXI", Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Arturo Rodríguez-González
- Gastrosurgery Service, UMAE, National Medical Center "Siglo XXI", Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Lourdes Arriaga-Pizano
- Medical Research Unit on Immunochemistry, National Medical Center "Siglo XXI", Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Vilma Maldonado
- Epigenetics Laboratories, National Institute of Genomic Medicine (INMEGEN), 14610, Mexico City, Mexico.
| |
Collapse
|
20
|
Al-kharboosh R, ReFaey K, Lara-Velazquez M, Grewal SS, Imitola J, Quiñones-Hinojosa A. Inflammatory Mediators in Glioma Microenvironment Play a Dual Role in Gliomagenesis and Mesenchymal Stem Cell Homing: Implication for Cellular Therapy. Mayo Clin Proc Innov Qual Outcomes 2020; 4:443-459. [PMID: 32793872 PMCID: PMC7411162 DOI: 10.1016/j.mayocpiqo.2020.04.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma is the most aggressive malignant primary brain tumor, with a dismal prognosis and a devastating overall survival. Despite aggressive surgical resection and adjuvant treatment, average survival remains approximately 14.6 months. The brain tumor microenvironment is heterogeneous, comprising multiple populations of tumor, stromal, and immune cells. Tumor cells evade the immune system by suppressing several immune functions to enable survival. Gliomas release immunosuppressive and tumor-supportive soluble factors into the microenvironment, leading to accelerated cancer proliferation, invasion, and immune escape. Mesenchymal stem cells (MSCs) isolated from bone marrow, adipose tissue, or umbilical cord are a promising tool for cell-based therapies. One crucial mechanism mediating the therapeutic outcomes often seen in MSC application is their tropism to sites of injury. Furthermore, MSCs interact with host immune cells to regulate the inflammatory response, and data points to the possibility of using MSCs to achieve immunomodulation in solid tumors. Interleukin 1β, interleukin 6, tumor necrosis factor α, transforming growth factor β, and stromal cell-derived factor 1 are notably up-regulated in glioblastoma and dually promote immune and MSC trafficking. Mesenchymal stem cells have widely been regarded as hypoimmunogenic, enabling this cell-based administration across major histocompatibility barriers. In this review, we will highlight (1) the bidirectional communication of glioma cells and tumor-associated immune cells, (2) the inflammatory mediators enabling leukocytes and transplantable MSC migration, and (3) review preclinical and human clinical trials using MSCs as delivery vehicles. Mesenchymal stem cells possess innate abilities to migrate great distances, cross the blood-brain barrier, and communicate with surrounding cells, all of which make them desirable "Trojan horses" for brain cancer therapy.
Collapse
Key Words
- 5-FC, 5-fluorocytosine
- AMSC, adipose tissue–derived mesenchymal stem cell
- BBB, blood-brain barrier
- BMSC, bone marrow–derived mesenchymal stem cell
- CED, convection-enhanced delivery
- DC, dendritic cell
- EGFRvIII, EGFR variant III
- GBM, glioblastoma
- GSC, glioma stem cell
- IFN, interferon
- IL, interleukin
- MDSC, myeloid-derived suppressor cell
- MHC, major histocompatibility complex
- MSC, mesenchymal stem cell
- NSC, neural stem cell
- TAM, tumor-associated macrophage
- TGF, transforming growth factor
- TNF, tumor necrosis factor
- UC-MSC, umbilical cord MSC
Collapse
Affiliation(s)
- Rawan Al-kharboosh
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL
- Mayo Clinic College of Medicine and Science, Mayo Clinic Graduate School of Biomedical Sciences (Neuroscience Track), Regenerative Sciences Training Program, Mayo Clinic, Rochester, MN
| | - Karim ReFaey
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL
| | - Montserrat Lara-Velazquez
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL
- Plan of Combined Studies in Medicine (MD/PhD), National Autonomous University of Mexico, Mexico City
| | | | - Jaime Imitola
- Department of Neurology Research, Division of Multiple Sclerosis and Translational Neuroimmunology, UConn School of Medicine, Farmington, CT
| | | |
Collapse
|
21
|
Han P, Cao P, Hu S, Kong K, Deng Y, Zhao B, Li F. Esophageal Microenvironment: From Precursor Microenvironment to Premetastatic Niche. Cancer Manag Res 2020; 12:5857-5879. [PMID: 32765088 PMCID: PMC7371556 DOI: 10.2147/cmar.s258215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/29/2020] [Indexed: 12/15/2022] Open
Abstract
Esophageal cancer (EC) is the sixth most deadly cancer, and its incidence is still increasing year by year. Although the researches on the molecular mechanisms of EC have been widely carried out and incremental progress has been made, its overall survival rate is still low. There is cumulative evidence showing that the esophageal microenvironment plays a vital role in the development of EC. In precancerous lesions of the esophagus, high-risk environmental factors can promote the development of precancerous lesions by inducing the production of inflammatory factors and the recruitment of immune cells. In the tumor microenvironment, tumor-promoting cells can inhibit anti-tumor immunity and promote tumor progression through a variety of pathways, such as bone marrow-derived suppressor cells (MDSCs), tumor-associated fibroblasts (CAFs), and regulatory T cells (Tregs). The formation of extracellular hypoxia and acidic microenvironment and the change of extracellular matrix stiffness are also important factors affecting tumor progression and metastasis. Simultaneously, primary tumor-derived cytokines and bone marrow-derived immune cells can also promote the formation of pre-metastasis niche of EC lymph nodes, which are beneficial to EC lymph node metastasis. Further research on the specific mechanism of these processes in the occurrence, development, and metastasis of each EC subtype will support us to grasp the overall pre-cancerous prevention, targeted treatment, and metastatic assessment of EC.
Collapse
Affiliation(s)
- Peng Han
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Peng Cao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Shan Hu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Kangle Kong
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Yu Deng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Bo Zhao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Fan Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| |
Collapse
|
22
|
Yeung TL, Sheng J, Leung CS, Li F, Kim J, Ho SY, Matzuk MM, Lu KH, Wong STC, Mok SC. Systematic Identification of Druggable Epithelial-Stromal Crosstalk Signaling Networks in Ovarian Cancer. J Natl Cancer Inst 2020; 111:272-282. [PMID: 29860390 PMCID: PMC6410941 DOI: 10.1093/jnci/djy097] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 04/04/2018] [Accepted: 04/27/2018] [Indexed: 12/28/2022] Open
Abstract
Background Bulk tumor tissue samples are used for generating gene expression profiles in most research studies, making it difficult to decipher the stroma–cancer crosstalk networks. In the present study, we describe the use of microdissected transcriptome profiles for the identification of cancer–stroma crosstalk networks with prognostic value, which presents a unique opportunity for developing new treatment strategies for ovarian cancer. Methods Transcriptome profiles from microdissected ovarian cancer–associated fibroblasts (CAFs) and ovarian cancer cells from patients with high-grade serous ovarian cancer (n = 70) were used as input data for the computational systems biology program CCCExplorer to uncover crosstalk networks between various cell types within the tumor microenvironment. The crosstalk analysis results were subsequently used for discovery of new indications for old drugs in ovarian cancer by computational ranking of candidate agents. Survival analysis was performed on ovarian tumor–bearing Dicer/Pten double-knockout mice treated with calcitriol, a US Food and Drug Administration–approved agent that suppresses the Smad signaling cascade, or vehicle control (9–11 mice per group). All statistical tests were two-sided. Results Activation of TGF-β-dependent and TGF-β-independent Smad signaling was identified in a particular subtype of CAFs and was associated with poor patient survival (patients with higher levels of Smad-regulated gene expression by CAFs: median overall survival = 15 months, 95% confidence interval [CI] = 12.7 to 17.3 months; vs patients with lower levels of Smad-regulated gene expression: median overall survival = 26 months, 95% CI = 15.9 to 36.1 months, P = .02). In addition, the activated Smad signaling identified in CAFs was found to be targeted by repositioning calcitriol. Calcitriol suppressed Smad signaling in CAFs, inhibited tumor progression in mice, and prolonged the median survival duration of ovarian cancer–bearing mice from 36 to 48 weeks (P = .04). Conclusions Our findings suggest the feasibility of using novel multicellular systems biology modeling to identify and repurpose known drugs targeting cancer–stroma crosstalk networks, potentially leading to faster and more effective cures for cancers.
Collapse
Affiliation(s)
- Tsz-Lun Yeung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jianting Sheng
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center, Weill Cornell Medicine, Houston, TX.,Center for Modeling Cancer Development, Houston Methodist Cancer Center, Houston, TX
| | - Cecilia S Leung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Fuhai Li
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH
| | - Jaeyeon Kim
- Department of Biochemistry and Molecular Biology, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN
| | - Samuel Y Ho
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Martin M Matzuk
- Department of Pathology and Immunology and Center for Drug Discovery, Baylor College of Medicine, Houston, TX
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center, Weill Cornell Medicine, Houston, TX.,Center for Modeling Cancer Development, Houston Methodist Cancer Center, Houston, TX
| | - Samuel C Mok
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX
| |
Collapse
|
23
|
Schröder C, Khatri R, Petry SF, Linn T. Class I and II Histone Deacetylase Inhibitor LBH589 Promotes Endocrine Differentiation in Bone Marrow Derived Human Mesenchymal Stem Cells and Suppresses Uncontrolled Proliferation. Exp Clin Endocrinol Diabetes 2020; 129:357-364. [PMID: 32052390 DOI: 10.1055/a-1103-1900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells are useful tools employed in clinical and preclinical medicine. Their beneficial potential in especially degenerative as well as autoimmune diseases is a constant focus of research. Regarding diabetes mellitus, transplantation of stem cells is seen as a possible therapeutic approach to overcome the loss of endocrine pancreatic cells. It was reported that co-transplantation of mesenchymal stem cells with pancreatic islet cells improves function and survival of the graft. However, these multipotent progenitors may be able to form tumors, especially under immunosuppressed conditions. Histone deacetylase inhibitors might offer the potential to overcome this issue. These small molecules can induce cell differentiation and control proliferation. Their potential to control lineage development of stem cells has been distinctly demonstrated in the treatment of cancer, mainly in hematopoietic neoplasias.In this study, we demonstrate that human bone marrow-derived mesenchymal stem cells exhibit low carcinogenic potential in an immunosuppressed condition in vivo. Further, the effect of histone deacetylase inhibitors LBH589, MS-275, and MGCD0103 was examined after normalizing histone deacetylase activities in culture. Interestingly, transcripts of insulin gene enhancer protein and paired-box-gene 6, two markers of pancreatic endocrine differentiation were constitutively expressed in the cell line. The broad spectrum inhibitor of class I and class II histone deacetylases LBH589 upregulated the expression of these transcription factors in a significant way, whereas addition of selective class I histone deacetylase inhibitors MS-275 and MGCD0103 did not result in significant changes in gene expression.In conclusion, we deliver evidence that a combined class I and II histone deacetylase inhibition is able to modulate the transcripts of differentiation markers of mesenchymal stem cells. The treatment holds the capability to facilitate endocrine differentiation in future approaches to replace endocrine cells by stem cell therapy.
Collapse
Affiliation(s)
- Christoph Schröder
- Clinical Research Unit, Centre of Internal Medicine, Justus Liebig University Giessen, Germany.,Medizinische Hochschule Hannover, Hannover, Germany
| | - Rahul Khatri
- Clinical Research Unit, Centre of Internal Medicine, Justus Liebig University Giessen, Germany
| | | | - Thomas Linn
- Clinical Research Unit, Centre of Internal Medicine, Justus Liebig University Giessen, Germany
| |
Collapse
|
24
|
Álvarez MV, Gutiérrez LM, Auzmendi J, Correa A, Lazarowski A, Bolontrade MF. Acquisition of stem associated-features on metastatic osteosarcoma cells and their functional effects on mesenchymal stem cells. Biochim Biophys Acta Gen Subj 2020; 1864:129522. [PMID: 31945406 DOI: 10.1016/j.bbagen.2020.129522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 12/20/2019] [Accepted: 01/10/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND Osteosarcoma (OS) is the most frequent malignant bone tumor, affecting predominantly children and young adults. Metastases are a major clinical challenge in OS. In this context, 20% of OS patients are diagnosed with metastatic OS, but near 80% of all OS patients could present non-detectable micrometastases at the moment of diagnosis. METHODS Osteogenic differentiation; doxorubicin exclusion assay; fluorescence microscopy; RT-qPCR; proteomic analysis. RESULTS Our results suggest that metastatic OS cells possess a diminished osteoblastic differentiation potential with a gain of metastatic traits like the capacity to modify intracellular localization of chemodrugs and higher levels of expression of stemness-related genes. On the opposite hand, non-metastatic OS cells possess bone-associated traits like higher osteoblastic differentiation and also an osteoblastic-inducer secretome. OS cells also differ in the nature of their interaction with mesenchymal stem cells (MSCs), with opposites impacts on MSCs phenotype and behavior. CONCLUSIONS All this suggests that a major trait acquired by metastatic cells is a switch into a stem-like state that could favor its survival in the pulmonary niche, opening new possibilities for personalized chemotherapeutic schemes. GENERAL SIGNIFICANCE Our work provides new insights regarding differences among metastatic and non-metastatic OS cells, with particular emphasis on differentiation potential, multidrug resistance and interaction with MSCs.
Collapse
Affiliation(s)
- Matías Valenzuela Álvarez
- Remodeling processes and cellular niches laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET- Hospital Italiano Buenos Aires (HIBA), Instituto Universitario del Hospital Italiano (IUHI), C1199ACL Buenos Aires, Argentina
| | - Luciana M Gutiérrez
- Remodeling processes and cellular niches laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET- Hospital Italiano Buenos Aires (HIBA), Instituto Universitario del Hospital Italiano (IUHI), C1199ACL Buenos Aires, Argentina
| | - Jerónimo Auzmendi
- INFIBIOC, Clinical Biochemistry Department, School of Pharmacy and Biochemistry (FFyB), University of Buenos Aires (UBA), C1113AAD Buenos Aires, Argentina
| | | | - Alberto Lazarowski
- INFIBIOC, Clinical Biochemistry Department, School of Pharmacy and Biochemistry (FFyB), University of Buenos Aires (UBA), C1113AAD Buenos Aires, Argentina
| | - Marcela F Bolontrade
- Remodeling processes and cellular niches laboratory, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), CONICET- Hospital Italiano Buenos Aires (HIBA), Instituto Universitario del Hospital Italiano (IUHI), C1199ACL Buenos Aires, Argentina.
| |
Collapse
|
25
|
Tan HX, Xiao ZG, Huang T, Fang ZX, Liu Y, Huang ZC. CXCR4/TGF-β1 mediated self-differentiation of human mesenchymal stem cells to carcinoma-associated fibroblasts and promoted colorectal carcinoma development. Cancer Biol Ther 2019; 21:248-257. [PMID: 31818187 DOI: 10.1080/15384047.2019.1685156] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background: Tumor microenvironment (TME) is a crucial part of tumor hallmarks. Mesenchymal stem cells (MSCs), important components of TME, are the main source of Carcinoma-associated fibroblasts (CAFs), but the mechanism of transformation regulation is still unclear. Transforming growth factor-β1 (TGF-β1), chemokine Stromal cell-derived factor-1 (SDF-1) and its endogenous receptor CXCR4 may play important roles during this process.Methods: Co-culture technique was used to explore the effects of MSCs on the proliferation, migration and invasion of colorectal carcinoma (CRC) cells and how they induced MSCs to differentiate into CAFs. The expression of α-SMA, Vimentin, S100A4 and FAP were detected as CAFs markers. Inhibitors AMD3100 and cyclophosphamide (Cy) were pre-treated in MSCs to verify the functions of CXCR4/TGF-β1. Finally, the xenograft models in nude mice were generated to further verify this process in vivo.Results: MSCs promoted the CRCs proliferation, invasion and migration, and induced SDF-1 expression and secretion, which dramatically up-regulated CXCR4 and TGF-β1 expression in MSCs. The levels of CAFs markers elevated in MSCs, indicating CAFs differentiation occurred in MSCs. AMD3100 and Cy treatment significantly blocked this differentiation process of MSCs by suppressing CXCR4 expression and TGF-β1 secretion. In vivo xenograft experiments also demonstrated that MSCs promoted differentiation into CAFs through CXCR4/TGF-β1 signaling in either primary tumor tissues or hepatic metastatic tissues of CRC.Conclusion: Our studies have revealed the essential role of CXCR4/TGF-β1 axis playing in the transformation of tumor microenvironment by mediating MSCs differentiation into CAFs, promoting CRCs growth and metastasis.
Collapse
Affiliation(s)
- Hao-Xiang Tan
- Department of General Surgery, Hunan Province People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China.,Department of General Surgery, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, P.R. China
| | - Zhi-Gang Xiao
- Department of General Surgery, Hunan Province People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| | - Tao Huang
- Department of General Surgery, Hunan Province People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| | - Zhi-Xue Fang
- Department of General Surgery, Hunan Province People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| | - Yu Liu
- Department of General Surgery, Hunan Province People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| | - Zhong-Cheng Huang
- Department of General Surgery, Hunan Province People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, P.R. China
| |
Collapse
|
26
|
Gyukity-Sebestyén E, Harmati M, Dobra G, Németh IB, Mihály J, Zvara Á, Hunyadi-Gulyás É, Katona R, Nagy I, Horváth P, Bálind Á, Szkalisity Á, Kovács M, Pankotai T, Borsos B, Erdélyi M, Szegletes Z, Veréb ZJ, Buzás EI, Kemény L, Bíró T, Buzás K. Melanoma-Derived Exosomes Induce PD-1 Overexpression and Tumor Progression via Mesenchymal Stem Cell Oncogenic Reprogramming. Front Immunol 2019; 10:2459. [PMID: 31681332 PMCID: PMC6813737 DOI: 10.3389/fimmu.2019.02459] [Citation(s) in RCA: 40] [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: 03/21/2019] [Accepted: 10/01/2019] [Indexed: 01/31/2023] Open
Abstract
Recently, it has been described that programmed cell death protein 1 (PD-1) overexpressing melanoma cells are highly aggressive. However, until now it has not been defined which factors lead to the generation of PD-1 overexpressing subpopulations. Here, we present that melanoma-derived exosomes, conveying oncogenic molecular reprogramming, induce the formation of a melanoma-like, PD-1 overexpressing cell population (mMSCPD-1+) from naïve mesenchymal stem cells (MSCs). Exosomes and mMSCPD-1+ cells induce tumor progression and expression of oncogenic factors in vivo. Finally, we revealed a characteristic, tumorigenic signaling network combining the upregulated molecules (e.g., PD-1, MET, RAF1, BCL2, MTOR) and their upstream exosomal regulating proteins and miRNAs. Our study highlights the complexity of exosomal communication during tumor progression and contributes to the detailed understanding of metastatic processes.
Collapse
Affiliation(s)
- Edina Gyukity-Sebestyén
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
- Doctoral School of Interdisciplinary Sciences, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Mária Harmati
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
- Doctoral School of Interdisciplinary Sciences, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Gabriella Dobra
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
- Doctoral School of Interdisciplinary Sciences, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - István B. Németh
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Johanna Mihály
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ágnes Zvara
- Laboratory of Functional Genomics, Institute of Genetics, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
| | - Éva Hunyadi-Gulyás
- Laboratory of Proteomics Research, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
| | - Róbert Katona
- Artificial Chromosome and Stem Cell Research Laboratory, Institute of Genetics, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
| | - István Nagy
- Sequencing Platform, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
| | - Péter Horváth
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
| | - Árpád Bálind
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
| | - Ábel Szkalisity
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
| | - Mária Kovács
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tibor Pankotai
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Barbara Borsos
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Miklós Erdélyi
- Advanced Optical Imaging Group, Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Zsolt Szegletes
- Atomic Force Microscope Laboratory, Institute of Biophysics, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
| | - Zoltán J. Veréb
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Edit I. Buzás
- MTA-SE Immuno-proteogenomics Extracellular Vesicle Research Group, Department of Genetics, Cell- and Immunobiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Tamás Bíró
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Krisztina Buzás
- Laboratory of Microscopic Image Analysis and Machine Learning, Institute of Biochemistry, Biological Research Centre of Hungarian Academy of Sciences, Szeged, Hungary
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, Szeged, Hungary
| |
Collapse
|
27
|
Javan MR, Khosrojerdi A, Moazzeni SM. New Insights Into Implementation of Mesenchymal Stem Cells in Cancer Therapy: Prospects for Anti-angiogenesis Treatment. Front Oncol 2019; 9:840. [PMID: 31555593 PMCID: PMC6722482 DOI: 10.3389/fonc.2019.00840] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment interacts with tumor cells, establishing an atmosphere to contribute or suppress the tumor development. Among the cells which play a role in the tumor microenvironment, mesenchymal stem cells (MSCs) have been demonstrated to possess the ability to orchestrate the fate of tumor cells, drawing the attention to the field. MSCs have been considered as cells with double-bladed effects, implicating either tumorigenic or anti-tumor activity. On the other side, the promising potential of MSCs in treating human cancer cells has been observed from the clinical studies. Among the beneficial characteristics of MSCs is the natural tumor-trophic migration ability, providing facility for drug delivery and, therefore, targeted treatment to detach tumor and metastatic cells. Moreover, these cells have been the target of engineering approaches, due to their easily implemented traits, in order to obtain the desired expression of anti-angiogenic, anti-proliferative, and pro-apoptotic properties, according to the tumor type. Tumor angiogenesis is the key characteristic of tumor progression and metastasis. Manipulation of angiogenesis has become an attractive approach for cancer therapy since the introduction of the first angiogenesis inhibitor, namely bevacizumab, for metastatic colorectal cancer therapy. This review tries to conclude the approaches, with focus on anti-angiogenesis approach, in implementing the MSCs to combat against tumor cell progression.
Collapse
Affiliation(s)
- Mohammad Reza Javan
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Arezou Khosrojerdi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mohammad Moazzeni
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
28
|
Aldinucci D, Borghese C, Casagrande N. Formation of the Immunosuppressive Microenvironment of Classic Hodgkin Lymphoma and Therapeutic Approaches to Counter It. Int J Mol Sci 2019; 20:ijms20102416. [PMID: 31096713 PMCID: PMC6566335 DOI: 10.3390/ijms20102416] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022] Open
Abstract
Classic Hodgkin lymphoma (cHL) is characterized by a few tumor cells surrounded by a protective, immunosuppressive tumor microenvironment composed of normal cells that are an active part of the disease. Hodgkin and Reed-Sternberg (HRS) cells evade the immune system through a variety of different mechanisms. They evade antitumor effector T cells and natural killer cells and promote T cell exhaustion. Using cytokines and extracellular vesicles, they recruit normal cells, induce their proliferation and "educate" (i.e. reprogram) them to become immunosuppressive and protumorigenic. Therefore, alternative treatment strategies are being developed to target not only tumor cells but also the tumor microenvironment. Here we summarize current knowledge on the ability of HRS cells to build their microenvironment and to educate normal cells to become immunosuppressive. We also describe therapeutic strategies to counteract formation of the tumor microenvironment and related processes leading to T cell exhaustion and repolarization of immunosuppressive tumor-associated macrophages.
Collapse
Affiliation(s)
- Donatella Aldinucci
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| | - Cinzia Borghese
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| | - Naike Casagrande
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| |
Collapse
|
29
|
Raz Y, Cohen N, Shani O, Bell RE, Novitskiy SV, Abramovitz L, Levy C, Milyavsky M, Leider-Trejo L, Moses HL, Grisaru D, Erez N. Bone marrow-derived fibroblasts are a functionally distinct stromal cell population in breast cancer. J Exp Med 2018; 215:3075-3093. [PMID: 30470719 PMCID: PMC6279405 DOI: 10.1084/jem.20180818] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/05/2018] [Accepted: 10/23/2018] [Indexed: 12/21/2022] Open
Abstract
Raz et al. demonstrate that the expression of PDGFRα distinguishes two functional CAF populations in breast tumors and lung metastases and identify a subpopulation of CAFs that are specifically recruited to the tumor microenvironment from mesenchymal stromal cells in the BM. Cancer-associated fibroblasts (CAFs) are highly prominent in breast tumors, but their functional heterogeneity and origin are still largely unresolved. We report that bone marrow (BM)–derived mesenchymal stromal cells (MSCs) are recruited to primary breast tumors and to lung metastases and differentiate to a distinct subpopulation of CAFs. We show that BM-derived CAFs are functionally important for tumor growth and enhance angiogenesis via up-regulation of Clusterin. Using newly generated transgenic mice and adoptive BM transplantations, we demonstrate that BM-derived fibroblasts are a substantial source of CAFs in the tumor microenvironment. Unlike resident CAFs, BM-derived CAFs do not express PDGFRα, and their recruitment resulted in a decrease in the percentage of PDGFRα-expressing CAFs. Strikingly, decrease in PDGFRα in breast cancer patients was associated with worse prognosis, suggesting that BM-derived CAFs may have deleterious effects on survival. Therefore, PDGFRα expression distinguishes two functionally unique CAF populations in breast tumors and metastases and may have important implications for patient stratification and precision therapeutics.
Collapse
Affiliation(s)
- Yael Raz
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Obstetrics and Gynecology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Noam Cohen
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ophir Shani
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rachel E Bell
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sergey V Novitskiy
- Department of Cancer Biology, Vanderbilt University School of Medicine and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, TN
| | - Lilach Abramovitz
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Carmit Levy
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michael Milyavsky
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Leonor Leider-Trejo
- Department of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Harold L Moses
- Department of Cancer Biology, Vanderbilt University School of Medicine and Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, TN
| | - Dan Grisaru
- Department of Obstetrics and Gynecology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Neta Erez
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
30
|
Fu X, Xie F, Gong F, Yang Z, Lv X, Li X, Jiao H, Wang Q, Liu X, Yan L, Xiao R. Suppression of PTBP1 signaling is responsible for mesenchymal stem cell induced invasion of low malignancy cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1552-1565. [PMID: 30327198 DOI: 10.1016/j.bbamcr.2018.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/03/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022]
Abstract
Mesenchymal stem cells (MSCs) hold great promise as attractive vehicles to deliver therapeutic agents against cancer, while the cross-talk between MSCs and cancer cells remains controversial. Here in an indirect co-culture system we observed that MSCs induced the malignancy transformation of low malignancy cancer cells HT29 and MCF7, whereas MSCs were reprogrammed by high malignancy cancer cells HCT116 and MDA-MB-231 without exerting an obvious influence on them. We further demonstrated that the RNA-binding protein polypyrimidine tract-binding protein 1 (PTBP1) was suppressed in low malignancy cancer cells co-cultured with MSCs. Moreover, shRNA mediated silencing of PTBP1 could promote the invasiveness of HT29 cells while over-expression of PTBP1 attenuate the MSC-induced invasion of HT29 cells. Our results suggested that differential effects of MSCs on the invasion of cancer cells partially corresponded to PTBP1 expression in cancer cells and the maintenance of biological characteristics in MSCs, which insight could provide a theoretical basis for evaluating the safety of MSC application and PTBP1 targeting in cancer treatment.
Collapse
Affiliation(s)
- Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Fangnan Xie
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Fuxing Gong
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Xiaoyan Lv
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Xintian Li
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Hu Jiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Ba-Da-Chu Road, Beijing 100144, PR China.
| |
Collapse
|
31
|
Calvo-Asensio I, Dillon ET, Lowndes NF, Ceredig R. The Transcription Factor Hif-1 Enhances the Radio-Resistance of Mouse MSCs. Front Physiol 2018; 9:439. [PMID: 29755367 PMCID: PMC5932323 DOI: 10.3389/fphys.2018.00439] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/06/2018] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent progenitors supporting bone marrow hematopoiesis. MSCs have an efficient DNA damage response (DDR) and are consequently relatively radio-resistant cells. Therefore, MSCs are key to hematopoietic reconstitution following total body irradiation (TBI) and bone marrow transplantation (BMT). The bone marrow niche is hypoxic and via the heterodimeric transcription factor Hypoxia-inducible factor-1 (Hif-1), hypoxia enhances the DDR. Using gene knock-down, we have previously shown that the Hif-1α subunit of Hif-1 is involved in mouse MSC radio-resistance, however its exact mechanism of action remains unknown. In order to dissect the involvement of Hif-1α in the DDR, we used CRISPR/Cas9 technology to generate a stable mutant of the mouse MSC cell line MS5 lacking Hif-1α expression. Herein, we show that it is the whole Hif-1 transcription factor, and not only the Hif-1α subunit, that modulates the DDR of mouse MSCs. This effect is dependent upon the presence of a Hif-1α protein capable of binding to both DNA and its heterodimeric partner Arnt (Hif-1β). Detailed transcriptomic and proteomic analysis of Hif1a KO MS5 cells leads us to conclude that Hif-1α may be acting indirectly on the DNA repair process. These findings have important implications for the modulation of MSC radio-resistance in the context of BMT and cancer.
Collapse
Affiliation(s)
- Irene Calvo-Asensio
- Regenerative Medicine Institute, School of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland.,Genome Stability Laboratory, Centre for Chromosome Biology, National University of Ireland, Galway, Ireland
| | - Eugène T Dillon
- Proteome Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Noel F Lowndes
- Genome Stability Laboratory, Centre for Chromosome Biology, National University of Ireland, Galway, Ireland
| | - Rhodri Ceredig
- Regenerative Medicine Institute, School of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| |
Collapse
|
32
|
Byun JS, Park S, Caban A, Jones A, Gardner K. Linking Race, Cancer Outcomes, and Tissue Repair. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:317-328. [PMID: 29137950 PMCID: PMC5785534 DOI: 10.1016/j.ajpath.2017.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 02/07/2023]
Abstract
The burden of cancer in the United States is unevenly spread across its different populations, with stark differences in both disease prevalence and outcome on the basis of race and ethnicity. Although a large portion of these differences can be explained by a variety of sociobehavioral and socioeconomic factors, even after these exposures are taken into consideration, considerable disparities persist. In this review, we explore a conceptual framework of biological theories and unifying concepts, based on an evolutionary perspective, that may help better define common guiding principles for exploration of underlying causes of cancer health disparities. The ultimate goal of this conceptual perspective is to outline approaches that may aid in establishing integrated pathway and processes analyses to provide useful insights to guide the development of future interventions. These interventions will improve outcome, increase prevention, and ultimately eliminate all disparities.
Collapse
Affiliation(s)
- Jung S Byun
- National Institute on Minority Health and Health Disparities, Bethesda, Maryland
| | - Samson Park
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Ambar Caban
- National Institute on Minority Health and Health Disparities, Bethesda, Maryland
| | - Alana Jones
- National Institute on Minority Health and Health Disparities, Bethesda, Maryland
| | - Kevin Gardner
- National Institute on Minority Health and Health Disparities, Bethesda, Maryland; Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
33
|
Devetzi M, Goulielmaki M, Khoury N, Spandidos DA, Sotiropoulou G, Christodoulou I, Zoumpourlis V. Genetically‑modified stem cells in treatment of human diseases: Tissue kallikrein (KLK1)‑based targeted therapy (Review). Int J Mol Med 2018; 41:1177-1186. [PMID: 29328364 PMCID: PMC5819898 DOI: 10.3892/ijmm.2018.3361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
The tissue kallikrein-kinin system (KKS) is an endogenous multiprotein metabolic cascade which is implicated in the homeostasis of the cardiovascular, renal and central nervous system. Human tissue kallikrein (KLK1) is a serine protease, component of the KKS that has been demonstrated to exert pleiotropic beneficial effects in protection from tissue injury through its anti-inflammatory, anti-apoptotic, anti-fibrotic and anti-oxidative actions. Mesenchymal stem cells (MSCs) or endothelial progenitor cells (EPCs) constitute populations of well-characterized, readily obtainable multipotent cells with special immunomodulatory, migratory and paracrine properties rendering them appealing potential therapeutics in experimental animal models of various diseases. Genetic modification enhances their inherent properties. MSCs or EPCs are competent cellular vehicles for drug and/or gene delivery in the targeted treatment of diseases. KLK1 gene delivery using adenoviral vectors or KLK1 protein infusion into injured tissues of animal models has provided particularly encouraging results in attenuating or reversing myocardial, renal and cerebrovascular ischemic phenotype and tissue damage, thus paving the way for the administration of genetically modified MSCs or EPCs with the human tissue KLK1 gene. Engraftment of KLK1-modified MSCs and/or KLK1-modified EPCs resulted in advanced beneficial outcome regarding heart and kidney protection and recovery from ischemic insults. Collectively, findings from pre-clinical studies raise the possibility that tissue KLK1 may be a novel future therapeutic target in the treatment of a wide range of cardiovascular, cerebrovascular and renal disorders.
Collapse
Affiliation(s)
- Marina Devetzi
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Goulielmaki
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Nicolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Ioannis Christodoulou
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| |
Collapse
|
34
|
Giornelli GH, Mandó P. A Theoretical View of Ovarian Cancer Relapse. EUROPEAN MEDICAL JOURNAL 2017. [DOI: 10.33590/emj/10312894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is a disease that almost invariably relapses even after optimal primary cytoreductive surgery and standard first-line platinum-based chemotherapy. After recurrence, progressions occur at shorter intervals in the natural history of the disease. However, the biologic and cellular events underlying recurrence and progression (maintenance phase) are yet to be completely understood. Ovarian adenocarcinoma, like any other tissue, after reduction of the cell population (cytoreduction) either by surgery, chemotherapy, radiotherapy, or targeted therapies induced cell-death, tends to its own renewal through cancer stem cells (CSC). CSC remain quiescent most of their lives and then ‘wake up’, generating a proliferative progeny that differentiates as they become different clones of daughter cells. What defines them is their ‘self-renewal’ potential, thus perpetuating the disease with higher tumour volume relapses in which CSC increase in number.
We propose a theory of how recurrence/relapse occurs in which CSC play a key role in the genesis of relapse. These self-renewing CSC can generate a proliferative progeny and this population is sensitive to chemotherapy, anti-angiogenic agents, and PARP inhibitors, which so far have only increased the disease/relapse free survival (‘maintenance phase’). In OC it seems we are not addressing the ‘root’ of recurrence/relapse. As with any theory, this is based on both proven facts and suggested hypotheses, which may serve as investigation drivers towards finally making a substantial improvement in OC management.
Collapse
Affiliation(s)
- Gonzalo H. Giornelli
- Genito Urinary Oncology Department, Instituto Alexander Fleming, Buenos Aires, Argentina
| | - Pablo Mandó
- Genito Urinary Oncology Department, Instituto Alexander Fleming, Buenos Aires, Argentina
| |
Collapse
|
35
|
Cheng W, Ge Q, Wan L, Wang X, Chen X, Wu X. A method to establish a mouse model of bone marrow microenvironment injury. Exp Anim 2017. [PMID: 28626156 PMCID: PMC5682345 DOI: 10.1538/expanim.16-0116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A normal bone marrow microenvironment plays a very important role in the normal
functioning of hematopoietic stem cells. Once disturbed, this microenvironment can become
favorable for the occurrence of blood disorders, cancers, and other diseases. Therefore,
further studies on the bone marrow microenvironment should be performed to reveal
regulatory and stem cell fate determination mechanisms and promote the development of bone
marrow transplantation, tissue repair and regenerative medicine, and other fields. A small
animal model for further research is also urgently needed. In this study, an electric
shock device was designed to elicit a femur bone marrow microenvironment injury in mice. A
wire was inserted into the distal femur but not into the proximal femur, and the bone
marrow microenvironment was evidently damaged by application of 100 ± 10 V for 1.5 ± 0.5
min ; mortality, however, was low in the mice. Gross observation, hematoxylin and eosin
staining, immunohistochemistry, bright-field microscopy, and micro-CT scanning were also
conducted. A large number of new blood capillaries and sinusoids appeared in the injured
distal femur after 2 weeks. The capillaries in the injured femur disappeared after 4
weeks, and mature blood vessels were scattered throughout the injured area. Red blood
cells disappeared, and the cellular structure and trabecular bone were better than those
observed 2 weeks previously. Thus, we developed a simply operated, accurate, reliable, and
easily controlled small animal model as a good technical platform to examine angiogenesis
and segmentation damage in the bone marrow microenvironment.
Collapse
Affiliation(s)
- Wenzhe Cheng
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Quanhu Ge
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Longfei Wan
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Xiaoyi Wang
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| | - Xueling Chen
- Department of Immunology, School of Medicine, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Xiangwei Wu
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China.,Laboratory of Translational Medicine, School of Medicine, Shihezi University, Shihezi, Xinjiang 832008, P.R. China
| |
Collapse
|
36
|
Caron JM, Ames JJ, Contois L, Liebes L, Friesel R, Muggia F, Vary CPH, Oxburgh L, Brooks PC. Inhibition of Ovarian Tumor Growth by Targeting the HU177 Cryptic Collagen Epitope. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 186:1649-61. [PMID: 27216148 DOI: 10.1016/j.ajpath.2016.01.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/22/2015] [Accepted: 01/19/2016] [Indexed: 12/17/2022]
Abstract
Evidence suggests that stromal cells play critical roles in tumor growth. Uncovering new mechanisms that control stromal cell behavior and their accumulation within tumors may lead to development of more effective treatments. We provide evidence that the HU177 cryptic collagen epitope is selectively generated within human ovarian carcinomas and this collagen epitope plays a role in SKOV-3 ovarian tumor growth in vivo. The ability of the HU177 epitope to regulate SKOV-3 tumor growth depends in part on its ability to modulate stromal cell behavior because targeting this epitope inhibited angiogenesis and, surprisingly, the accumulation of α-smooth muscle actin-expressing stromal cells. Integrin α10β1 can serve as a receptor for the HU177 epitope in α-smooth muscle actin-expressing stromal cells and subsequently regulates Erk-dependent migration. These findings are consistent with a mechanism by which the generation of the HU177 collagen epitope provides a previously unrecognized α10β1 ligand that selectively governs angiogenesis and the accumulation of stromal cells, which in turn secrete protumorigenic factors that contribute to ovarian tumor growth. Our findings provide a new mechanistic understanding into the roles by which the HU177 epitope regulates ovarian tumor growth and provide new insight into the clinical results from a phase 1 human clinical study of the monoclonal antibody D93/TRC093 in patients with advanced malignant tumors.
Collapse
Affiliation(s)
- Jennifer M Caron
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Jacquelyn J Ames
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Liangru Contois
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Leonard Liebes
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Robert Friesel
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Franco Muggia
- New York University Langone Medical Center, Division of Hematology and Medical Oncology, New York, New York
| | - Calvin P H Vary
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Leif Oxburgh
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Peter C Brooks
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine.
| |
Collapse
|
37
|
You D, Jang MJ, Kim BH, Choi KR, Lee C, Song G, Shin HC, Jeong IG, Suh N, Kim YM, Ahn TY, Kim CS. Bone marrow–derived mesenchymal stromal cell therapy in a rat model of cavernous nerve injury: Preclinical study for approval. Cytotherapy 2016; 18:870-80. [DOI: 10.1016/j.jcyt.2016.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 04/27/2016] [Accepted: 04/29/2016] [Indexed: 10/21/2022]
|
38
|
Murgai M, Giles A, Kaplan R. Physiological, Tumor, and Metastatic Niches: Opportunities and Challenges for Targeting the Tumor Microenvironment. Crit Rev Oncog 2016; 20:301-14. [PMID: 26349421 DOI: 10.1615/critrevoncog.2015013668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The primary tumor niche and the related but distinct premetastatic/metastatic niche comprise a number of essential players, including immune cells, stromal cells, and extracellular matrix. The cross-talk between these components is key to tumor progression. Many of these cell types and signaling pathways in the tumor microenvironment also are found in physiological and stem cell niches, such as the bone marrow, colonic crypt, and skin bulge. Here they play tightly regulated roles in wound healing and tissue homeostasis. Understanding the similarities and differences between these distinct niches may better inform our ability to therapeutically target the tumor microenvironment. In this review we discuss a number of tumor and metastatic niche components as they relate to stem cell niches and highlight potential therapeutic strategies in pediatric cancers.
Collapse
Affiliation(s)
- Meera Murgai
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Amber Giles
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Rosandra Kaplan
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
39
|
Hagenhoff A, Bruns CJ, Zhao Y, von Lüttichau I, Niess H, Spitzweg C, Nelson PJ. Harnessing mesenchymal stem cell homing as an anticancer therapy. Expert Opin Biol Ther 2016; 16:1079-92. [PMID: 27270211 DOI: 10.1080/14712598.2016.1196179] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs) are non-hematopoietic progenitor cells that have been exploited as vehicles for cell-based cancer therapy. The general approach is based on the innate potential of adoptively applied MSC to undergo facilitated recruitment to malignant tissue. MSC from different tissue sources have been engineered using a variety of therapy genes that have shown efficacy in solid tumor models. AREAS COVERED In this review we will focus on the current developments of MSC-based gene therapy, in particular the diverse approaches that have been used for MSCs-targeted tumor therapy. We also discuss some outstanding issues and general prospects for their clinical application. EXPERT OPINION The use of modified mesenchymal stem cells as therapy vehicles for the treatment of solid tumors has progressed to the first generation of clinical trials, but the general field is still in its infancy. There are many questions that need to be addressed if this very complex therapy approach is widely applied in clinical settings. More must be understood about the mechanisms underlying tumor tropism and we need to identify the optimal source of the cells used. Outstanding issues also include the therapy transgenes used, and which tumor types represent viable targets for this therapy.
Collapse
Affiliation(s)
- Anna Hagenhoff
- a Department of Pediatrics and Pediatric Oncology Center, Klinikum rechts der Isar , Technical University , Munich , Germany
| | - Christiane J Bruns
- b Department of Surgery , Otto-von-Guericke University , Magdeburg , Germany
| | - Yue Zhao
- b Department of Surgery , Otto-von-Guericke University , Magdeburg , Germany
| | - Irene von Lüttichau
- a Department of Pediatrics and Pediatric Oncology Center, Klinikum rechts der Isar , Technical University , Munich , Germany
| | - Hanno Niess
- c Department of General, Visceral, Transplantation, Vascular and Thoracic Surgery , University of Munich , Munich , Germany
| | - Christine Spitzweg
- d Department of Internal Medicine II , University of Munich , Munich , Germany
| | - Peter J Nelson
- e Clinical Biochemistry Group, Medizinische Klinik und Poliklinik IV , University of Munich , Munich , Germany
| |
Collapse
|
40
|
Mesenchymal stem cells generate pericytes to promote tumor recurrence via vasculogenesis after stereotactic body radiation therapy. Cancer Lett 2016; 375:349-359. [DOI: 10.1016/j.canlet.2016.02.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 01/30/2016] [Accepted: 02/17/2016] [Indexed: 01/16/2023]
|
41
|
Zhang J, Sun D, Fu Q, Cao Q, Zhang H, Zhang K. Bone mesenchymal stem cells differentiate into myofibroblasts in the tumor microenvironment. Oncol Lett 2016; 12:644-650. [PMID: 27347194 DOI: 10.3892/ol.2016.4645] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 02/19/2016] [Indexed: 01/23/2023] Open
Abstract
The aim of the present study was to investigate the tropism of mesenchymal stem cells (MSCs) to the tumor microenvironment, and to evaluate the feasibility of bone marrow mesenchymal stem cells differentiating into myofibroblasts in vitro. A total of 1 ml bone marrow was extracted from the greater trochanter of one male New Zealand rabbit, and MSCs were obtained by density gradient centrifugation and cultured routinely. The surface markers were analyzed by flow cytometry. A VX2 tumor was aseptically excised from another male New Zealand rabbit and primary cultured. The tropism of MSCs for 30% and 50% VX2 conditioned medium was determined by using Transwell migration assays. MSCs were incubated in 30% VX2 conditioned medium for 7 or 14 days. The messenger (m)RNA levels and protein expression of α-smooth muscle actin (α-SMA) and vimentin were measured by reverse transcription-polymerase chain reaction and western blotting. MSCs were observed to have a spindle shape. The cultured MSCs were cluster of differentiation (CD)44+, CD105+, CD106+ and CD34-. VX2 cells demonstrated a spindle or polygon shape. In the Transwell assay, it was observed that the migrated cells appeared more frequently in the 30% VX2 conditioned medium group compared with the other groups when microscopically examined, which was additionally confirmed by the results of a colorimetric assay. The mRNA levels and protein expression of α-SMA and vimentin significantly increased in the test group compared with the control group at 7 days (P<0.01), and further increased in the test group at 14 days (P<0.01). The results of the present study demonstrated that MSCs have tropism for the tumor microenvironment and furthermore, may differentiate into myofibroblasts in the tumor microenvironment in vitro. The present study suggested that MSCs may migrate to the tumor and subsequently differentiate into myofibroblasts due to the tumor microenvironment, which may lead to promotion of the growth of the tumor. The present study additionally suggested that MSCs may be the precursors of tumor/carcinoma-associated myofibroblasts.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Dingqi Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Qiang Fu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Qingwei Cao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hui Zhang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Keqin Zhang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
42
|
Maria ATJ, Maumus M, Le Quellec A, Jorgensen C, Noël D, Guilpain P. Adipose-Derived Mesenchymal Stem Cells in Autoimmune Disorders: State of the Art and Perspectives for Systemic Sclerosis. Clin Rev Allergy Immunol 2016; 52:234-259. [DOI: 10.1007/s12016-016-8552-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Vartanian A, Karshieva S, Dombrovsky V, Belyavsky A. Melanoma educates mesenchymal stromal cells towards vasculogenic mimicry. Oncol Lett 2016; 11:4264-4268. [PMID: 27313776 DOI: 10.3892/ol.2016.4523] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 04/08/2016] [Indexed: 01/05/2023] Open
Abstract
Accumulating evidence suggests that mesenchymal stromal cells (MSCs) are recruited to the tumor, and promote tumor development and growth. The present study was performed to investigate the communication between aggressive melanoma and MSCs in vasculogenic mimicry (VM). Normal human MSCs plated on Matrigel were unable to form capillary-like structures (CLSs). By contrast, MSCs co-cultured with aggressive melanoma cell lines, namely, Mel Cher, Mel Kor and Mel P, generated CLSs. Significantly, MSCs co-cultured with poorly aggressive melanoma cells, namely, Mel Me, failed to form CLSs. To identify factors responsible for VM, the effects of vascular endothelial growth factor A (VEGFA), pro-epidermal growth factor, basic fibroblast growth factor and stromal cell-derived factor 1α on the formation of CLSs by MSCs were tested. VM was induced by the addition of VEGFA, whereas other cytokines were inefficient. To confirm the hypothesis that aggressive tumor cells can increase the vasculogenic ability of MSCs, a standard B16/F10 mouse melanoma test system was used. MSCs isolated from the adipose tissues of C57BL/6 mice with melanoma formed a vascular-like network on Matrigel, whereas MSCs from healthy mice failed to form such structures. This study provides the first direct evidence that melanoma tumors educate MSCs to engage in VM. The education may occur distantly. These findings offer promise for novel therapeutic directions in the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Amalia Vartanian
- Department of Experimental Diagnosis and Biotherapy of Tumors, Blokhin Russian Cancer Research Center, Moscow 115478, Russia
| | - Saida Karshieva
- Laboratory of Stem and Progenitor Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Vladislav Dombrovsky
- Department of Experimental Diagnosis and Biotherapy of Tumors, Blokhin Russian Cancer Research Center, Moscow 115478, Russia
| | - Alexander Belyavsky
- Laboratory of Stem and Progenitor Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| |
Collapse
|
44
|
DUBON MARIAJOSE, PARK KISOOK. The mechanisms of substance P-mediated migration of bone marrow-derived mesenchymal stem cell-like ST2 cells. Int J Mol Med 2016; 37:1105-11. [DOI: 10.3892/ijmm.2016.2496] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 02/17/2016] [Indexed: 11/06/2022] Open
|
45
|
Onion D, Argent RH, Reece-Smith AM, Craze ML, Pineda RG, Clarke PA, Ratan HL, Parsons SL, Lobo DN, Duffy JP, Atherton JC, McKenzie AJ, Kumari R, King P, Hall BM, Grabowska AM. 3-Dimensional Patient-Derived Lung Cancer Assays Reveal Resistance to Standards-of-Care Promoted by Stromal Cells but Sensitivity to Histone Deacetylase Inhibitors. Mol Cancer Ther 2016; 15:753-63. [PMID: 26873730 DOI: 10.1158/1535-7163.mct-15-0598] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/10/2015] [Indexed: 11/16/2022]
Abstract
There is a growing recognition that current preclinical models do not reflect the tumor microenvironment in cellular, biological, and biophysical content and this may have a profound effect on drug efficacy testing, especially in the era of molecular-targeted agents. Here, we describe a method to directly embed low-passage patient tumor-derived tissue into basement membrane extract, ensuring a low proportion of cell death to anoikis and growth complementation by coculture with patient-derived cancer-associated fibroblasts (CAF). A range of solid tumors proved amenable to growth and pharmacologic testing in this 3D assay. A study of 30 early-stage non-small cell lung cancer (NSCLC) specimens revealed high levels of de novo resistance to a large range of standard-of-care agents, while histone deacetylase (HDAC) inhibitors and their combination with antineoplastic drugs displayed high levels of efficacy. Increased resistance was seen in the presence of patient-derived CAFs for many agents, highlighting the utility of the assay for tumor microenvironment-educated drug testing. Standard-of-care agents showed similar responses in the 3D ex vivo and patient-matched in vivo models validating the 3D-Tumor Growth Assay (3D-TGA) as a high-throughput screen for close-to-patient tumors using significantly reduced animal numbers. Mol Cancer Ther; 15(4); 753-63. ©2016 AACR.
Collapse
Affiliation(s)
- David Onion
- Ex Vivo Cancer Pharmacology Centre of Excellence, Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom. University of Nottingham Flow Cytometry Facility, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Richard H Argent
- Ex Vivo Cancer Pharmacology Centre of Excellence, Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Alexander M Reece-Smith
- Ex Vivo Cancer Pharmacology Centre of Excellence, Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Madeleine L Craze
- Ex Vivo Cancer Pharmacology Centre of Excellence, Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Robert G Pineda
- Ex Vivo Cancer Pharmacology Centre of Excellence, Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Philip A Clarke
- Ex Vivo Cancer Pharmacology Centre of Excellence, Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Hari L Ratan
- Department of Urology, Nottingham University NHS Trust, QMC, Nottingham, United Kingdom
| | - Simon L Parsons
- Nottingham University NHS Trust, City Hospital Campus, Nottingham, United Kingdom
| | - Dileep N Lobo
- Nottingham Digestive Diseases National Institute of Health Research Biomedical Research Unit, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - John P Duffy
- Department of Thoracic Surgery, Nottingham University NHS Trust, City Hospital Campus, Nottingham United Kingdom
| | - John C Atherton
- Nottingham Digestive Diseases National Institute of Health Research Biomedical Research Unit, University of Nottingham and Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | | | - Rajendra Kumari
- Crown Bioscience UK Ltd, Hillcrest, Loughborough, United Kingdom
| | - Peter King
- Janssen Research and Development, Spring House, Pennsylvania
| | - Brett M Hall
- Janssen Research and Development, Spring House, Pennsylvania
| | - Anna M Grabowska
- Ex Vivo Cancer Pharmacology Centre of Excellence, Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, United Kingdom.
| |
Collapse
|
46
|
Takiguchi G, Nishita M, Kurita K, Kakeji Y, Minami Y. Wnt5a-Ror2 signaling in mesenchymal stem cells promotes proliferation of gastric cancer cells by activating CXCL16-CXCR6 axis. Cancer Sci 2016; 107:290-7. [PMID: 26708384 PMCID: PMC4814243 DOI: 10.1111/cas.12871] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/14/2015] [Accepted: 12/21/2015] [Indexed: 12/27/2022] Open
Abstract
Wnt5a‐Ror2 signaling has been shown to play important roles in promoting aggressiveness of various cancer cells in a cell‐autonomous manner. However, little is known about its function in cancer‐associated stromal cells, including mesenchymal stem cells (MSCs). Thus, we examined the role of Wnt5a‐Ror2 signaling in bone marrow‐derived MSCs in regulating proliferation of undifferentiated gastric cancer cells. Coculture of a gastric cancer cell line, MKN45, with MSCs either directly or indirectly promotes proliferation of MKN45 cells, and suppressed expression of Ror2 in MSCs prior to coculture inhibits enhanced proliferation of MKN45 cells. In addition, conditioned media from MSCs, treated with control siRNA, but not siRNAs against Ror2, can enhance proliferation of MKN45 cells. Interestingly, it was found that expression of CXCL16 in MSCs is augmented by Wnt5a‐Ror2 signaling, and that recombinant chemokine (C‐X‐C motif) ligand (CXCL)16 protein can enhance proliferation of MKN45 cells in the absence of MSCs. In fact, suppressed expression of CXCL16 in MSCs or an addition of a neutralizing antibody against CXCL16 fails to promote proliferation of MKN45 cells in either direct or indirect coculture with MSCs. Importantly, we show that MKN45 cells express chemokine (C‐X‐C motif) receptor (CXCR)6, a receptor for CXCL16, and that suppressed expression of CXCR6 in MKN45 cells results in a failure of its enhanced proliferation in either direct or indirect coculture with MSCs. These findings indicate that Wnt5a‐Ror2 signaling enhances expression of CXCL16 in MSCs and, as a result, enhanced secretion of CXCL16 from MSCs might act on CXCR6 expressed on MKN45, leading to the promotion of its proliferation.
Collapse
Affiliation(s)
- Gosuke Takiguchi
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan.,Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Michiru Nishita
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Kana Kurita
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yoshihiro Kakeji
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| |
Collapse
|
47
|
Yi T, Kim SN, Lee HJ, Kim J, Cho YK, Shin DH, Tak SJ, Moon SH, Kang JE, Ji IM, Lim HJ, Lee DS, Jeon MS, Song SU. Manufacture of Clinical-Grade Human Clonal Mesenchymal Stem Cell Products from Single Colony Forming Unit-Derived Colonies Based on the Subfractionation Culturing Method. Tissue Eng Part C Methods 2015; 21:1251-62. [PMID: 26421757 DOI: 10.1089/ten.tec.2015.0017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Stem cell products derived from mesenchymal stem cells (MSCs) have been widely used in clinical trials, and a few products have been already commercialized. However, the therapeutic effects of clinical-grade MSCs are still controversial owing to mixed results from recent clinical trials. A potential solution to overcome this hurdle may be to use clonal stem cells as the starting cell material to increase the homogeneity of the final stem cell products. We have previously developed an alternative isolation and culture protocol for establishing a population of clonal MSCs (cMSCs) from single colony forming unit (CFU)-derived colonies. In this study, we established a good manufacturing practice (GMP)-compatible procedure for the clinical-grade production of human bone marrow-derived cMSCs based on the subfractionation culturing method. We optimized the culture procedures to expand and obtain a clonal population of final MSC products from single CFU-derived colonies in a GMP facility. The characterization results of the final cMSC products met our preset criteria. Animal toxicity tests were performed in a good laboratory practice facility, and showed no toxicity or tumor formation in vivo. These tests include single injection toxicity, multiple injection toxicity, biodistribution analysis, and tumorigenicity tests in vivo. No chromosomal abnormalities were detected by in situ karyotyping using oligo-fluorescence in situ hydridization (oligo-FISH), providing evidence of genetic stability of the clinical-grade cMSC products. The manufacture and quality control results indicated that our GMP methodology could produce sufficient clonal population of MSC products from a small amount of bone marrow aspirate to treat a number of patients.
Collapse
Affiliation(s)
- TacGhee Yi
- 1 Translational Research Center, Inha University School of Medicine , Incheon, Republic of Korea.,2 Inha Research Institute for Medical Science, Inha University School of Medicine , Incheon, Republic of Korea.,3 SCM Lifescience Co., Ltd. , Incheon, Republic of Korea
| | - Si-na Kim
- 4 Drug Development Program, Department of Biomedical Science, Inha University School of Medicine , Incheon, Republic of Korea
| | - Hyun-Joo Lee
- 4 Drug Development Program, Department of Biomedical Science, Inha University School of Medicine , Incheon, Republic of Korea
| | - Junghee Kim
- 4 Drug Development Program, Department of Biomedical Science, Inha University School of Medicine , Incheon, Republic of Korea
| | - Yun-Kyoung Cho
- 3 SCM Lifescience Co., Ltd. , Incheon, Republic of Korea
| | - Dong-Hee Shin
- 1 Translational Research Center, Inha University School of Medicine , Incheon, Republic of Korea.,2 Inha Research Institute for Medical Science, Inha University School of Medicine , Incheon, Republic of Korea
| | - Sun-Ji Tak
- 1 Translational Research Center, Inha University School of Medicine , Incheon, Republic of Korea
| | - Sun-Hwa Moon
- 3 SCM Lifescience Co., Ltd. , Incheon, Republic of Korea
| | - Ji-Eun Kang
- 3 SCM Lifescience Co., Ltd. , Incheon, Republic of Korea
| | - In-Mi Ji
- 3 SCM Lifescience Co., Ltd. , Incheon, Republic of Korea
| | - Huyn-Ja Lim
- 3 SCM Lifescience Co., Ltd. , Incheon, Republic of Korea
| | - Dong-Soon Lee
- 5 Department of Pathology, Seoul National University School of Medicine , Seoul, Republic of Korea
| | - Myung-Shin Jeon
- 1 Translational Research Center, Inha University School of Medicine , Incheon, Republic of Korea
| | - Sun U Song
- 1 Translational Research Center, Inha University School of Medicine , Incheon, Republic of Korea.,3 SCM Lifescience Co., Ltd. , Incheon, Republic of Korea
| |
Collapse
|
48
|
Bonuccelli G, Avnet S, Grisendi G, Salerno M, Granchi D, Dominici M, Kusuzaki K, Baldini N. Role of mesenchymal stem cells in osteosarcoma and metabolic reprogramming of tumor cells. Oncotarget 2015; 5:7575-88. [PMID: 25277190 PMCID: PMC4202145 DOI: 10.18632/oncotarget.2243] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The tumor microenvironment plays an important role in cancer progression. Here, we focused on the role of reactive mesenchymal stem cells (MSC) in osteosarcoma (OS), and used human adipose MSC and a panel of OS cell lines (Saos-2, HOS, and 143B) to investigate the mutual effect of normal-cancer cell metabolic programming. Our results showed that MSC are driven by oxidative stress induced by OS cells to undergo Warburg metabolism, with increased lactate production. Therefore, we analyzed the expression of lactate monocarboxylate transporters. By real time PCR and immunofluorescence, in MSC we detected the expression of MCT-4, the transporter for lactate efflux, whereas MCT-1, responsible for lactate uptake, was expressed in OS cells. In agreement, silencing of MCT-1 by siRNA significantly affected the ATP production in OS cancer cells. Thus, cancer cells directly increase their mitochondrial biogenesis using this energy-rich metabolite that is abundantly provided by MSC as an effect of the altered microenvironmental conditions induced by OS cells. We also showed that lactate produced by MSC promotes the migratory ability of OS cells. These data provide novel information to be exploited for cancer therapies targeting the mutual metabolic reprogramming of cancer cells and their stroma.
Collapse
Affiliation(s)
- Gloria Bonuccelli
- Department of Biomedical and Neuromotion Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Sofia Avnet
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Giulia Grisendi
- Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Manuela Salerno
- Department of Biomedical and Neuromotion Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Donatella Granchi
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children and Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Katsuyuki Kusuzaki
- Department of Molecular Cell Physiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kyoto, Japan
| | - Nicola Baldini
- Department of Biomedical and Neuromotion Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy. Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|
49
|
Expression of α-Smooth Muscle Actin Determines the Fate of Mesenchymal Stromal Cells. Stem Cell Reports 2015; 4:1016-30. [PMID: 26028530 PMCID: PMC4471834 DOI: 10.1016/j.stemcr.2015.05.004] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 05/04/2015] [Accepted: 05/04/2015] [Indexed: 02/07/2023] Open
Abstract
Pro-fibrotic microenvironments of scars and tumors characterized by increased stiffness stimulate mesenchymal stromal cells (MSCs) to express α-smooth muscle actin (α-SMA). We investigated whether incorporation of α-SMA into contractile stress fibers regulates human MSC fate. Sorted α-SMA-positive MSCs exhibited high contractile activity, low clonogenicity, and differentiation potential limited to osteogenesis. Knockdown of α-SMA was sufficient to restore clonogenicity and adipogenesis in MSCs. Conversely, α-SMA overexpression induced YAP translocation to the nucleus and reduced the high clonogenicity and adipogenic potential of α-SMA-negative MSCs. Inhibition of YAP rescued the decreased adipogenic differentiation potential induced by α-SMA, establishing a mechanistic link between matrix stiffness, α-SMA, YAP, and MSC differentiation. Consistent with in vitro findings, nuclear localization of YAP was positively correlated in α-SMA expressing stromal cells of adiposarcoma and osteosarcoma. We propose that α-SMA mediated contraction plays a critical role in mechanically regulating MSC fate by controlling YAP/TAZ activation. The α-SMA-positive myofibroblast fraction of human MSCs exhibits low clonogenicity Formation of α-SMA stress fibers enhances nuclear translocation of YAP/TAZ in MSCs α-SMA knockdown favors adipogenesis, while overexpression promotes osteogenesis α-SMA-mediated lineage choice of MSCs is YAP dependent
Collapse
|
50
|
Effect of Metformin on Viability, Morphology, and Ultrastructure of Mouse Bone Marrow-Derived Multipotent Mesenchymal Stromal Cells and Balb/3T3 Embryonic Fibroblast Cell Line. BIOMED RESEARCH INTERNATIONAL 2015; 2015:769402. [PMID: 26064951 PMCID: PMC4430655 DOI: 10.1155/2015/769402] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/27/2015] [Accepted: 04/15/2015] [Indexed: 12/27/2022]
Abstract
Metformin, a popular drug used to treat diabetes, has recently gained attention as a potentially useful therapeutic agent for treating cancer. In our research metformin was added to in vitro cultures of bone marrow-derived multipotent mesenchymal stromal cells (BMSCs) and Balb/3T3 fibroblast at concentration of 1 mM, 5 mM, and 10 mM. Obtained results indicated that metformin negatively affected proliferation activity of investigated cells. The drug triggered the formation of autophagosomes and apoptotic bodies in all tested cultures. Additionally, we focused on determination of expression of genes involved in insulin-like growth factor 2 (IGF2) signaling pathway. The most striking finding was that the mRNA level of IGF2 was constant in both BMSCs and Balb/3T3. Further, the analysis of IGF2 concentration in cell supernatants showed that it decreased in BMSC cultures after 5 and 10 mM metformin treatments. In case of Balb/3T3 the concentration of IGF2 in culture supernatants decreased after 1 and 5 mM and increased after 10 mM of metformin. Our results suggest that metformin influences the cytophysiology of somatic cells in a dose- and time-dependent manner causing inhibition of proliferation and abnormalities of their morphology and ultrastructure.
Collapse
|