1
|
Rakhmatullina AR, Zolotykh MA, Filina YV, Mingaleeva RN, Sagdeeva AR, Boulygina EA, Gafurbaeva DU, Bulatov ER, Rizvanov AA, Miftakhova RR. Development of a novel prostate Cancer-Stroma Sphere (CSS) model for In Vitro tumor microenvironment studies. Transl Oncol 2024; 44:101930. [PMID: 38520912 PMCID: PMC10981155 DOI: 10.1016/j.tranon.2024.101930] [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: 01/10/2024] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024] Open
Abstract
Tumor employs non-cancerous cells to gain beneficial features that promote growth and survival of cancer cells. Despite intensive research in the area of tumor microenvironment, there is still a lack of reliable and reproducible in vitro model for tumor and tumor-microenvironment cell interaction studies. Herein we report the successful development of a heterogeneous cancer-stroma sphere (CSS) model composed of prostate adenocarcinoma PC3 cells and immortalized mesenchymal stem cells (MSC). The CSS model demonstrated a structured spatial layout of the cells, with stromal cells concentrated at the center of the spheres and tumor cells located on the periphery. Significant increase in the levels of VEGFA, IL-10, and IL1a has been detected in the conditioned media of CSS as compared to PC3 spheres. Single cell RNA sequencing data revealed that VEGFA was secreted by MSC cells within heterogeneous spheroids. Enhanced expression of extracellular membrane (ECM) proteins was also shown for CSS-derived MSCs. Furthermore, we demonstrated that the multicellular architecture altered cancer cell response to chemotherapeutic agents: the inhibition of sphere formation by topotecan was 74.92 ± 4.56 % for PC3 spheres and 45.95 ± 7.84 % for CSS spheres (p < 0.01), docetaxel showed 37,51± 20,88 % and 15,67± 14,08 % inhibition, respectively (p < 0.05). Thus, CSS present an effective in vitro model for examining the extracellular matrix composition and cell-to-cell interactions within the tumor, as well as for evaluating the antitumor activity of drugs.
Collapse
Affiliation(s)
- Aigul R Rakhmatullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Maria A Zolotykh
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Yulia V Filina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Rimma N Mingaleeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Aisylu R Sagdeeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Eugenia A Boulygina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Dina U Gafurbaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Emil R Bulatov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; Division of Medical and Biological Sciences, Academy of Sciences of the Republic of Tatarstan, 420013, Kazan, Russia; I.K. Akhunbaev Kyrgyz state medical academy, 720020, Bishkek, Kyrgyzstan
| | - Regina R Miftakhova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia.
| |
Collapse
|
2
|
Li Y, Li J, Zhou L, Wang Z, Jin L, Cao J, Xie H, Wang L. Aberrant activation of TGF-β/ROCK1 enhances stemness during prostatic stromal hyperplasia. Cell Commun Signal 2024; 22:257. [PMID: 38711089 PMCID: PMC11071275 DOI: 10.1186/s12964-024-01644-4] [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: 03/10/2024] [Accepted: 04/29/2024] [Indexed: 05/08/2024] Open
Abstract
Benign prostatic hyperplasia (BPH) is a multifactorial disease in which abnormal growth factor activation and embryonic reawakening are considered important factors. Here we demonstrated that the aberrant activation of transforming growth factor β (TGF-β)/Rho kinase 1 (ROCK1) increased the stemness of BPH tissue by recruiting mesenchymal stem cells (MSCs), indicating the important role of embryonic reawakening in BPH. When TGF-β/ROCK1 is abnormally activated, MSCs are recruited and differentiate into fibroblasts/myofibroblasts, leading to prostate stromal hyperplasia. Further research showed that inhibition of ROCK1 activation suppressed MSC migration and their potential for stromal differentiation. Collectively, our findings suggest that abnormal activation of TGF-β/ROCK1 regulates stem cell lineage specificity, and the small molecule inhibitor GSK269962A could target ROCK1 and may be a potential treatment for BPH.
Collapse
Affiliation(s)
- Youyou Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Jiaren Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Liang Zhou
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Zhenxing Wang
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ling Jin
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jia Cao
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Hui Xie
- Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Long Wang
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
3
|
Saadh MJ, Alhuthali HM, Gonzales Aníbal O, Asenjo-Alarcón JA, Younus DG, Alhili A, Adhab ZH, Alsalmi O, Gharib AF, Pecho RDC, Akhavan-Sigari R. Mesenchymal stem cells and their extracellular vesicles in urological cancers: Prostate, bladder, and kidney. Cell Biol Int 2024; 48:3-19. [PMID: 37947445 DOI: 10.1002/cbin.12098] [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: 07/19/2023] [Revised: 08/31/2023] [Accepted: 10/12/2023] [Indexed: 11/12/2023]
Abstract
Mesenchymal stem cells (MSCs) are recognized for their remarkable ability to differentiate into multiple cell types. They are also known to possess properties that can fight cancer, leading to attempts to modify MSCs for use in anticancer treatments. However, MSCs have also been found to participate in pathways that promote tumor growth. Many studies have been conducted to explore the potential of MSCs for clinical applications, but the results have been inconclusive, possibly due to the diverse nature of MSC populations. Furthermore, the conflicting roles of MSCs in inhibiting tumors and promoting tumor growth hinder their adaptation to anticancer therapies. Antitumorigenic and protumorigenic properties of MSCs in urological cancers such as bladder, prostate, and renal are not as well established, and data comparing them are still limited. MSCs hold significant promise as a vehicle for delivering anticancer agents and suicide genes to tumors. Presently, numerous studies have concentrated on the products derived from MSCs, such as extracellular vesicles (EVs), as a form of cell-free therapy. This work aimed to review and discuss the current knowledge of MSCs and their EVs in urological cancer therapy.
Collapse
Affiliation(s)
| | - Hayaa M Alhuthali
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | | | | | | | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | - Ohud Alsalmi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Amal F Gharib
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | | | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
4
|
Saxena N, Chakraborty S, Dutta S, Bhardwaj G, Karnik N, Shetty O, Jadhav S, Zafar H, Sen S. Stiffness-dependent MSC homing and differentiation into CAFs - implications for breast cancer invasion. J Cell Sci 2024; 137:jcs261145. [PMID: 38108421 DOI: 10.1242/jcs.261145] [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: 03/09/2023] [Accepted: 12/03/2023] [Indexed: 12/19/2023] Open
Abstract
Cellular heterogeneity and extracellular matrix (ECM) stiffening have been shown to be drivers of breast cancer invasiveness. Here, we examine how stiffness-dependent crosstalk between cancer cells and mesenchymal stem cells (MSCs) within an evolving tumor microenvironment regulates cancer invasion. By analyzing previously published single-cell RNA sequencing datasets, we establish the existence of a subpopulation of cells in primary tumors, secondary sites and circulatory tumor cell clusters of highly aggressive triple-negative breast cancer (TNBC) that co-express MSC and cancer-associated fibroblast (CAF) markers. By using hydrogels with stiffnesses of 0.5, 2 and 5 kPa to mimic different stages of ECM stiffening, we show that conditioned medium from MDA-MB-231 TNBC cells cultured on 2 kPa gels, which mimic the pre-metastatic stroma, drives efficient MSC chemotaxis and induces stable differentiation of MSC-derived CAFs in a TGFβ (TGFB1)- and contractility-dependent manner. In addition to enhancing cancer cell proliferation, MSC-derived CAFs on 2 kPa gels maximally boost local invasion and confer resistance to flow-induced shear stresses. Collectively, our results suggest that homing of MSCs at the pre-metastatic stage and their differentiation into CAFs actively drives breast cancer invasion and metastasis in TNBC.
Collapse
Affiliation(s)
- Neha Saxena
- Department of Chemical Engineering, IIT Bombay,Mumbai 400076, India
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Soura Chakraborty
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
| | - Sarbajeet Dutta
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Garvit Bhardwaj
- Department of Electrical Engineering, IIT Kanpur, Kanpur 208016, India
| | - Nupur Karnik
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Omshree Shetty
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Sameer Jadhav
- Department of Chemical Engineering, IIT Bombay,Mumbai 400076, India
| | - Hamim Zafar
- Department of Biological Sciences and Bioengineering, IIT Kanpur, Kanpur 208016, India
- Department of Computer Science and Engineering, IIT Kanpur, Kanpur 208016, India
- Mehta Family Centre for Engineering in Medicine , IIT Kanpur, Kanpur 208016, India
| | - Shamik Sen
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| |
Collapse
|
5
|
Rivera-Cruz CM, Kumar S, Figueiredo ML. Poly I:C-priming of adipose-derived mesenchymal stromal cells promotes a pro-tumorigenic phenotype in an immunocompetent mouse model of prostate cancer. Front Cell Dev Biol 2023; 11:1145421. [PMID: 38078010 PMCID: PMC10703370 DOI: 10.3389/fcell.2023.1145421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/02/2023] [Indexed: 02/12/2024] Open
Abstract
Introduction: Mesenchymal stromal cells (MSC) are envisioned as a potential cellular vehicle for targeted cancer therapies due to their tumor tropism and immune permissiveness. An obstacle in their use is the duality in their interactions within tumors, rendering them pro-tumorigenic or anti-tumorigenic, in a context dependent manner. MSC preconditioning, or priming, has been proposed as a strategy for directing the effector properties of MSC at tumor sites. Methods: We primed human MSC derived from adipose tissues (ASC), a clinically advantageous MSC source, utilizing toll-like receptor agonists. Subsequently, we explored the consequences in tumor progression and transcriptome upon the interaction of tumor cells with primed or unprimed ASC in an in vivo model of prostate cancer, the second most common cancer and second leading cause of cancer related death in men in the USA. Results and discussion: In the studied model, poly I:C-primed ASC were found to significantly accelerate tumor growth progression. And while unprimed and LPS-primed ASC did not exert a significant effect on tumor growth at the macroscopic level, gene expression analyses suggested that all treatments promoted distinct modulatory effects in the tumor microenvironment, including altered modulation of angiogenesis, and immune response processes. However, the effects resulting from the collective interaction across these processes must be sufficiently skewed in a pro-tumorigenic or anti-tumorigenic direction for evidence of tumor progression modulation to be detectable at the macroscopic level. Our study highlights potential MSC-tumor microenvironment interactions that may be leveraged and should be considered in the development of cancer therapeutics utilizing MSC.
Collapse
Affiliation(s)
| | | | - Marxa L. Figueiredo
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
6
|
Wieder R. Fibroblasts as Turned Agents in Cancer Progression. Cancers (Basel) 2023; 15:2014. [PMID: 37046676 PMCID: PMC10093070 DOI: 10.3390/cancers15072014] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Differentiated epithelial cells reside in the homeostatic microenvironment of the native organ stroma. The stroma supports their normal function, their G0 differentiated state, and their expansion/contraction through the various stages of the life cycle and physiologic functions of the host. When malignant transformation begins, the microenvironment tries to suppress and eliminate the transformed cells, while cancer cells, in turn, try to resist these suppressive efforts. The tumor microenvironment encompasses a large variety of cell types recruited by the tumor to perform different functions, among which fibroblasts are the most abundant. The dynamics of the mutual relationship change as the sides undertake an epic battle for control of the other. In the process, the cancer "wounds" the microenvironment through a variety of mechanisms and attracts distant mesenchymal stem cells to change their function from one attempting to suppress the cancer, to one that supports its growth, survival, and metastasis. Analogous reciprocal interactions occur as well between disseminated cancer cells and the metastatic microenvironment, where the microenvironment attempts to eliminate cancer cells or suppress their proliferation. However, the altered microenvironmental cells acquire novel characteristics that support malignant progression. Investigations have attempted to use these traits as targets of novel therapeutic approaches.
Collapse
Affiliation(s)
- Robert Wieder
- Rutgers New Jersey Medical School and the Cancer Institute of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
7
|
Early Cell Cultures from Prostate Cancer Tissue Express Tissue Specific Epithelial and Cancer Markers. Int J Mol Sci 2023; 24:ijms24032830. [PMID: 36769153 PMCID: PMC9917781 DOI: 10.3390/ijms24032830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Prostate cancer (PCa) is a widespread oncological disease that proceeds in the indolent form in most patients. However, in some cases, the indolent form can transform into aggressive metastatic incurable cancer. The most important task of PCa diagnostics is to search for early markers that can be used for predicting the transition of indolent cancer into its aggressive form. Currently, there are two effective preclinical models to study PCa pathogenesis: patients derived xenografts (PDXs) and patients derived organoids (PDOs). Both models have limitations that restrict their use in research. In this work, we investigated the ability of the primary 2D prostate cell cultures (PCCs) from PCa patients to express epithelial and cancer markers. Early PCCs were formed by epithelial cells that were progressively replaced with the fibroblast-like cells. Early PCCs contained tissue-specific stem cells that could grow in a 3D culture and form PDOs similar to those produced from the prostate tissue. Early PCCs and PDOs derived from the tissues of PCa patients expressed prostate basal and luminal epithelial markers, as well as cancer markers AMACR, TMPRSS2-ERG, and EZH2, the latter being a promising candidate to mark the transition from the indolent to aggressive PCa. We also identified various TMPRSS2-ERG fusion transcripts in PCCs and PDOs, including new chimeric variants resulting from the intra- and interchromosomal translocations. The results suggest that early PCCs derived from cancerous and normal prostate tissues sustain the phenotype of prostate cells and can be used as a preclinical model to study the pathogenesis of PCa.
Collapse
|
8
|
Maguire G. Chronic inflammation induced by microneedling and the use of bone marrow stem cell cytokines. J Tissue Viability 2022; 31:687-692. [DOI: 10.1016/j.jtv.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 08/07/2022] [Accepted: 08/16/2022] [Indexed: 10/14/2022]
|
9
|
Rahimi Tesiye M, Abrishami Kia Z, Rajabi-Maham H. Mesenchymal stem cells and prostate cancer: A concise review of therapeutic potentials and biological aspects. Stem Cell Res 2022; 63:102864. [PMID: 35878578 DOI: 10.1016/j.scr.2022.102864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/09/2022] Open
Abstract
Prostate cancer (PCa) arises from a cancer stem or progenitor cell with homogenous characteristics, especially among the aging men population. Over the past decade, the increasing PCa incidence has led to significant changes in both disease diagnosis and treatment. Recently, the therapeutic aspects of stem cells in many cancers, including PCa, have been debatable. The new generation of PCa studies seek to present definitive treatments with reduced therapeutic side effects. Since discovering unique properties of stem cells in modulating immunity, selective migration to inflammatory regions, and secretion of various growth factors, they have been a promising therapeutic target. The existing properties of stem cell therapy bring new opportunities for cancer inhibition: transferring chemotherapeutics, activating prodrugs, affecting the expression of genes involved in cancer, genetically modifying the production of anti-cancer compounds, proteins, and/or deriving extracellular vesicles (EVs) containing therapeutic agents from stem cells. However, their dual properties in carcinogenicity as well as their ability to inhibit cancer result in particular limitations studying them after administration. A clear understanding of the interaction between MSCs and the prostate cancer microenvironment will provide crucial information in revealing the precise applications and new practical protocols for clinical use of these cells..
Collapse
Affiliation(s)
- Maryam Rahimi Tesiye
- Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Zahra Abrishami Kia
- Faculty of Physical Education and Sport Sciences, University of Mazandaran, Babolsar, Iran.
| | - Hassan Rajabi-Maham
- Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
10
|
Frisbie L, Buckanovich RJ, Coffman L. Carcinoma Associated Mesenchymal Stem/Stromal Cells - Architects of the Pro-tumorigenic tumor microenvironment. Stem Cells 2022; 40:705-715. [PMID: 35583414 PMCID: PMC9406606 DOI: 10.1093/stmcls/sxac036] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 05/12/2022] [Indexed: 11/13/2022]
Abstract
The interaction between tumor cells and non-malignant hosts cells within the tumor microenvironment (TME) is critical to the pathophysiology of cancer. These non-malignant host cells, consisting of a variety of stromal, immune and endothelial cells, engage in a complex bidirectional crosstalk with the malignant tumor cells. Mesenchymal stem/stromal cells (MSCs) are one of these host cells, and they play a critical role in directing the formation and function of the entire TME. These MSCs are epigenetically reprogrammed by cancer cells to assume a strongly pro-tumorigenic phenotype and are referred to as carcinoma-associated mesenchymal stem/stromal cells (CA-MSCs). Studies over the last decade demonstrate that CA-MSCs not only directly interact with cancer cells to promote tumor growth and metastasis, but also orchestrate the formation of the TME. CA-MSCs can differentiate into virtually all stromal sub-lineages present in the TME, including pro-tumorigenic cancer associated fibroblasts (CAF), myofibroblasts, and adipocytes. CA-MSCs and the CAFs they produce, secrete much of the extracellular matrix in the TME. Furthermore, CA-MSC secreted factors promote angiogenesis, and recruit immunosuppressive myeloid cells effectively driving tumor immune exclusion. Thus CA-MSCs impact nearly every aspect of the TME. Despite their influence on cancer biology, as CA-MSCs represent a heterogenous population without a single definitive marker, significant confusion remains regarding the origin and proper identification CA-MSCs. This review will focus on the impact of CA-MSCs on cancer progression and metastasis and the ongoing work on CA-MSC identification, nomenclature and mechanism of action.
Collapse
Affiliation(s)
- Len Frisbie
- Department of Integrative Systems Biology, University of Pittsburgh, Pittsburgh, PA
| | - Ronald J Buckanovich
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA.,Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee Women's Research Institute, University of Pittsburgh, Pittsburgh, PA
| | - Lan Coffman
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA.,Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee Women's Research Institute, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
11
|
Li C, Zhao H, Cheng L, Wang B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci 2021; 11:187. [PMID: 34727974 PMCID: PMC8561357 DOI: 10.1186/s13578-021-00698-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/12/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cell (MSC)-based therapeutics is already available for treatment of a range of diseases or medical conditions. Autologous or allogeneic MSCs obtained from self or donors have their own advantages and disadvantages in their medical practice. Therapeutic benefits of using autologous vs. allogeneic MSCs are inconclusive. Transplanted MSCs within the body interact with their physical microenvironment or niche, physiologically or pathologically, and such cells in a newly established tissue microenvironment may be impacted by the pathological harmful environmental factors to alter their unique biological behaviors. Meanwhile, a temporary microenvironment/niche may be also altered by the resident or niche-surrounding MSCs. Therefore, the functional plasticity and heterogeneity of MSCs caused by different donors and subpopulations of MSCs may result in potential uncertainty in their safe and efficacious medical practice. Acknowledging a connection between MSCs' biology and their existing microenvironment, donor-controlled clinical practice for the long-term therapeutic benefit is suggested to further consider minimizing MSCs potential harm for MSC-based individual therapies. In this review, we summarize the advantages and disadvantages of autologous vs. allogeneic MSCs in their therapeutic applications. Among other issues, we highlight the importance of better understanding of the various microenvironments that may affect the properties of niche-surrounding MSCs and discuss the clinical applications of MSCs within different contexts for treatment of different diseases including cardiomyopathy, lupus and lupus nephritis, diabetes and diabetic complications, bone and cartilage repair, cancer and tissue fibrosis.
Collapse
Affiliation(s)
- Chenghai Li
- Stem Cell Program of Clinical Research Center, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| | - Hua Zhao
- Institute of Reproductive Medicine, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Linna Cheng
- Institute of Hematology, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China
| | - Bin Wang
- Department of Neurosurgery, People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, China.
| |
Collapse
|
12
|
Cohen L, Livney YD, Assaraf YG. Targeted nanomedicine modalities for prostate cancer treatment. Drug Resist Updat 2021; 56:100762. [PMID: 33857756 DOI: 10.1016/j.drup.2021.100762] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/17/2022]
Abstract
Prostate cancer (PC) is the second most common cause of death amongst men in the USA. Therapy of PC has been transformed in the past decade by introducing novel therapeutics, advanced functional imaging and diagnostic approaches, next generation sequencing, as well as improved application of existing therapies in localized PC. Treatment of PC at the different stages of the disease may include surgery, androgen deprivation therapy (ADT), chemotherapy and radiation therapy. However, although ADT has proven efficacious in PC treatment, its effectiveness may be temporary, as these tumors frequently develop molecular mechanisms of therapy resistance, which allow them to survive and proliferate even under conditions of testosterone deprivation, inhibition of androgen receptor signaling, or cytotoxic drug treatment. Importantly, ADT was found to induce key alterations which frequently result in the formation of metastatic tumors displaying a therapy refractory phenotype. Hence, to overcome these serious therapeutic impediments, novel PC cell-targeted therapeutic strategies are being developed. These include diverse platforms enabling specific enhanced antitumor drug uptake and increased intracellular accumulation. Studies have shown that these novel treatment modalities lead to enhanced antitumor activity and diminished systemic toxicity due to the use of selective targeting and decreased drug doses. The underlying mechanism of targeting and internalization is based upon the interaction between a selective ligand, conjugated to a drug-loaded nanoparticle or directly to an anti-cancer drug, and a specific plasma membrane biomarker, uniquely overexpressed on the surface of PC cells. Another targeted therapeutic approach is the delivery of unique anti-oncogenic signaling pathway-based therapeutic drugs, which are selectively cytotoxic to PC cells. The current paper reviews PC targeted modalities reported in the past 6 years, and discusses both the advantages and limitations of the various targeted treatment strategies.
Collapse
Affiliation(s)
- Lital Cohen
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Yoav D Livney
- The Laboratory of Biopolymers for Food and Health, Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion - Israel Institute of Technology, Haifa, 3200003, Israel.
| |
Collapse
|
13
|
McGuire JJ, Frieling JS, Lo CH, Li T, Muhammad A, Lawrence HR, Lawrence NJ, Cook LM, Lynch CC. Mesenchymal stem cell-derived interleukin-28 drives the selection of apoptosis resistant bone metastatic prostate cancer. Nat Commun 2021; 12:723. [PMID: 33526787 PMCID: PMC7851397 DOI: 10.1038/s41467-021-20962-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/06/2021] [Indexed: 01/12/2023] Open
Abstract
Bone metastatic prostate cancer (PCa) promotes mesenchymal stem cell (MSC) recruitment and their differentiation into osteoblasts. However, the effects of bone-marrow derived MSCs on PCa cells are less explored. Here, we report MSC-derived interleukin-28 (IL-28) triggers prostate cancer cell apoptosis via IL-28 receptor alpha (IL-28Rα)-STAT1 signaling. However, chronic exposure to MSCs drives the selection of prostate cancer cells that are resistant to IL-28-induced apoptosis and therapeutics such as docetaxel. Further, MSC-selected/IL-28-resistant prostate cancer cells grow at accelerated rates in bone. Acquired resistance to apoptosis is PCa cell intrinsic, and is associated with a shift in IL-28Rα signaling via STAT1 to STAT3. Notably, STAT3 ablation or inhibition impairs MSC-selected prostate cancer cell growth and survival. Thus, bone marrow MSCs drive the emergence of therapy-resistant bone metastatic prostate cancer yet this can be disabled by targeting STAT3.
Collapse
Affiliation(s)
- Jeremy J McGuire
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jeremy S Frieling
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Chen Hao Lo
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Tao Li
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ayaz Muhammad
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Harshani R Lawrence
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Nicholas J Lawrence
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Conor C Lynch
- Tumor Biology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
14
|
Chi C, Lao Y, Ahmed AHR, Benoy EC, Li C, Dereli‐Korkut Z, Fu BM, Leong KW, Wang S. High-Throughput Tumor-on-a-Chip Platform to Study Tumor-Stroma Interactions and Drug Pharmacokinetics. Adv Healthc Mater 2020; 9:e2000880. [PMID: 32965088 DOI: 10.1002/adhm.202000880] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/07/2020] [Indexed: 12/12/2022]
Abstract
Drug screening in oncology, especially for triple-negative breast cancer (TNBC), has high demand but remains unsatisfactory. Currently available models are either nonrepresentative of the complex tumor microenvironment or only suitable for low throughput screening, resulting in a low-yield success for drug development. To tackle these issues, the L-TumorChip system is developed in this study. It is a three-layered microfluidic tumor-on-a-chip platform integrating tumor microvasculature and tumor-stromal microenvironment with high throughput screening capability. Its layered and modular design is readily scalable through simple integration of multiple units. Here, L-TumorChip is validated with a TNBC model. The L-TumorChip system emulates certain tumor-stroma complexities and tumor-endothelium interactions, including TNBC invasion through the leaky microvasculature and angiogenesis. Additionally, with this L-TumorChip, the influence of different stromal cells, including normal fibroblasts, mesenchymal stem cells, and cancer-associated fibroblasts (CAF), on cancer cell growth as well as the stromal effects on drug responses to doxorubicin treatment is investigated. The presence of CAF delays drug pharmacokinetics, while apoptotic responses indicated by caspase-3 activities are higher in coculture with normal fibroblasts. Collectively, the L-TumorChip system represents a translational high-throughput screening toolkit that enables drug screening with a scenario closer to the in vivo conditions. This potential use may therefore facilitate development of new cancer drugs.
Collapse
Affiliation(s)
- Chun‐Wei Chi
- Department of Biomedical Engineering CUNY‐ The City College of New York New York NY 10031 USA
| | - Yeh‐Hsing Lao
- Department of Biomedical Engineering Columbia University New York NY 10027 USA
| | - A. H. Rezwanuddin Ahmed
- Department of Biomedical Engineering CUNY‐ The City College of New York New York NY 10031 USA
| | - Elizabeth C. Benoy
- Department of Biomedical Engineering CUNY‐ The City College of New York New York NY 10031 USA
| | - Chenghai Li
- Department of Biomedical Engineering CUNY‐ The City College of New York New York NY 10031 USA
| | - Zeynep Dereli‐Korkut
- Department of Biomedical Engineering CUNY‐ The City College of New York New York NY 10031 USA
| | - Bingmei M. Fu
- Department of Biomedical Engineering CUNY‐ The City College of New York New York NY 10031 USA
| | - Kam W. Leong
- Department of Biomedical Engineering Columbia University New York NY 10027 USA
| | - Sihong Wang
- Department of Biomedical Engineering CUNY‐ The City College of New York New York NY 10031 USA
| |
Collapse
|
15
|
Interaction of cancer cells with mesenchymal stem cells: implications in metastatic progression. J Indian Inst Sci 2020. [DOI: 10.1007/s41745-020-00182-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
16
|
Aloperine in combination with therapeutic adenoviral vector synergistically suppressed the growth of non-small cell lung cancer. J Cancer Res Clin Oncol 2020; 146:861-874. [DOI: 10.1007/s00432-020-03157-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/13/2020] [Indexed: 12/25/2022]
|
17
|
Atiya H, Frisbie L, Pressimone C, Coffman L. Mesenchymal Stem Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1234:31-42. [PMID: 32040853 DOI: 10.1007/978-3-030-37184-5_3] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The interactions between tumor cells and the non-malignant stromal and immune cells that make up the tumor microenvironment (TME) are critical to the pathophysiology of cancer. Mesenchymal stem cells (MSCs) are multipotent stromal stem cells found within most cancers and play a critical role influencing the formation and function of the TME. MSCs have been reported to support tumor growth through a variety of mechanisms including (i) differentiation into other pro-tumorigenic stromal components, (ii) suppression of the immune response, (iii) promotion of angiogenesis, (iv) enhancement of an epithelial-mesenchymal transition (EMT), (v) enrichment of cancer stem-like cells (CSC), (vi) increase in tumor cell survival, and (vii) promotion of tumor metastasis. In contrast, MSCs have also been reported to have antitumorigenic functions including (i) enhancement of the immune response, (ii) inhibition of angiogenesis, (iii) regulation of cellular signaling, and (iv) induction of tumor cell apoptosis. Although literature supporting both arguments exists, most studies point to MSCs acting in a cancer supporting role within the confines of the TME. Tumor-suppressive effects are observed when MSCs are used in higher ratios to tumor cells. Additionally, MSC function appears to be tissue type dependent and may rely on cancer education to reprogram a naïve MSC with antitumor effects into a cancer-educated or cancer-associated MSC (CA-MSC) which develops pro-tumorigenic function. Further work is required to delineate the complex crosstalk between MSCs and other components of the TME to accurately assess the impact of MSCs on cancer initiation, growth, and spread.
Collapse
Affiliation(s)
- Huda Atiya
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leonard Frisbie
- Division of Hematology/Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Lan Coffman
- Division of Hematology/Oncology, Division of Gynecologic Oncology, Department of Medicine, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Lokeshwar SD, Patel P, Shah SM, Ramasamy R. A Systematic Review of Human Trials Using Stem Cell Therapy for Erectile Dysfunction. Sex Med Rev 2020; 8:122-130. [DOI: 10.1016/j.sxmr.2019.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/19/2019] [Accepted: 08/24/2019] [Indexed: 01/08/2023]
|
19
|
Rahmatizadeh F, Gholizadeh-Ghaleh Aziz S, Khodadadi K, Lale Ataei M, Ebrahimie E, Soleimani Rad J, Pashaiasl M. Bidirectional and Opposite Effects of Naïve Mesenchymal Stem Cells on Tumor Growth and Progression. Adv Pharm Bull 2019; 9:539-558. [PMID: 31857958 PMCID: PMC6912184 DOI: 10.15171/apb.2019.063] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/31/2019] [Accepted: 08/13/2019] [Indexed: 12/16/2022] Open
Abstract
Cancer has long been considered as a heterogeneous population of uncontrolled proliferation of
different transformed cell types. The recent findings concerning tumorigeneses have highlighted
the fact that tumors can progress through tight relationships among tumor cells, cellular, and
non-cellular components which are present within tumor tissues. In recent years, studies have
shown that mesenchymal stem cells (MSCs) are essential components of non-tumor cells within
the tumor tissues that can strongly affect tumor development. Several forms of MSCs have been
identified within tumor stroma. Naïve (innate) mesenchymal stem cells (N-MSCs) derived from
different sources are mostly recruited into the tumor stroma. N-MSCs exert dual and divergent
effects on tumor growth through different conditions and factors such as toll-like receptor
priming (TLR-priming), which is the primary underlying causes of opposite effects. Moreover,
MSCs also have the contrary effects by various molecular mechanisms relying on direct cellto-
cell connections and indirect communications through the autocrine, paracrine routes, and
tumor microenvironment (TME).
Overall, cell-based therapies will hold great promise to provide novel anticancer treatments.
However, the application of intact MSCs in cancer treatment can theoretically cause adverse
clinical outcomes. It is essential that to extensively analysis the effective factors and conditions
in which underlying mechanisms are adopted by MSCs when encounter with cancer.
The aim is to review the cellular and molecular mechanisms underlying the dual effects of
MSCs followed by the importance of polarization of MSCs through priming of TLRs.
Collapse
Affiliation(s)
- Faramarz Rahmatizadeh
- Department of Molecular Medicine, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Khodadad Khodadadi
- Murdoch Children's Research Institute, Royal Children's Hospital, The University of Melbourne, Melbourne, Australia
| | - Maryam Lale Ataei
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esmaeil Ebrahimie
- Adelaide Medical School, University of Adelaide, Adelaide, Australia.,School of Animal and Veterinary Sciences, University of Adelaide, Adelaide, Australia
| | - Jafar Soleimani Rad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
| | - Maryam Pashaiasl
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran.,Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Brennen WN, Isaacs JT. Mesenchymal stem cells and the embryonic reawakening theory of BPH. Nat Rev Urol 2019; 15:703-715. [PMID: 30214054 DOI: 10.1038/s41585-018-0087-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The prostate is the only organ in a man that continues to grow with age. John McNeal proposed, 40 years ago, that this BPH is characterized by an age-related reinitiation of benign neoplastic growth selectively in developmentally abortive distal ducts within the prostate transition-periurethral zone (TPZ), owing to a reawakening of inductive stroma selectively within these zones. An innovative variant of this hypothesis is that, owing to its location, the TPZ is continuously exposed to urinary components and/or autoantigens, which produces an inflammatory TPZ microenvironment that promotes recruitment of bone marrow-derived mesenchymal stem cells (MSCs) and generates a paracrine-inductive stroma that reinitiates benign neoplastic nodular growth. In support of this hypothesis, MSCs infiltrate human BPH tissue and have the ability to stimulate epithelial stem cell growth. These results provide a framework for defining both the aetiology of BPH in ageing men and insights into new therapeutic approaches.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA.
| | - John T Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA. .,Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Bozkurt C, Karaöz E, Adaklı Aksoy B, Aydoğdu S, Fışgın T. The Use of Allogeneic Mesenchymal Stem Cells in Childhood Steroid-Resistant Acute Graft-Versus-Host Disease: A Retrospective Study of a Single-Center Experience. Turk J Haematol 2019; 36:186-192. [PMID: 31208159 PMCID: PMC6682770 DOI: 10.4274/tjh.galenos.2019.2019.0090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Objective: Steroid-resistant acute graft-versus-host disease (srAGVHD) is the most important cause of morbidity and mortality after allogeneic stem cell transplantation. There are several treatment methods available, including mesenchymal stem cell (MSC) application. The aim of this study was to evaluate the results of MSC therapy performed in children with srAGVHD. Materials and Methods: MSC therapy was used in our center between November 2014 and December 2017 for 22 patients who developed srAGVHD. The patients were retrospectively evaluated in terms of treatment response and survival. Results: After application of MSCs, complete response was obtained in 45.5% of the subjects, partial response was obtained in 13.6%, and no response was obtained in 40.9%. We found that 45.5% of the patients were alive and 54.5% had died and our treatment results were similar to those in the literature. Response to MSC treatment was found to be the only prognostic marker affecting mortality. Conclusion: MSC application is a treatment method that can be used safely together with other treatment methods in srAGVHD, a condition that has a high mortality rate. There are almost no acute side effects. There are also no serious long-term side effects in the literature. Prospective randomized studies are required to obtain high-quality data.
Collapse
Affiliation(s)
- Ceyhun Bozkurt
- İstinye University Faculty of Medicine, Department of Pediatrics, İstanbul, Turkey,Altınbaş University Faculty of Medicine, Bahçelievler Medical Park Hospital Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Erdal Karaöz
- İstinye University Faculty of Medicine, Department of Histology-Embryology, İstanbul, Turkey,İstinye University Faculty of Medicine, Stem Cell and Tissue Engineering Research and Application Center, İstanbul, Turkey,Liv Hospital, Regenerative Medicine, Stem Cell Production Center, İstanbul, Turkey
| | - Başak Adaklı Aksoy
- İstinye University Faculty of Medicine, Department of Pediatrics, İstanbul, Turkey,Altınbaş University Faculty of Medicine, Bahçelievler Medical Park Hospital Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Selime Aydoğdu
- Altınbaş University Faculty of Medicine, Bahçelievler Medical Park Hospital Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| | - Tunç Fışgın
- Altınbaş University Faculty of Medicine, Bahçelievler Medical Park Hospital Pediatric Bone Marrow Transplantation Unit, İstanbul, Turkey
| |
Collapse
|
22
|
Abstract
Cancers are not composed merely of cancer cells alone; instead, they are complex 'ecosystems' comprising many different cell types and noncellular factors. The tumour stroma is a critical component of the tumour microenvironment, where it has crucial roles in tumour initiation, progression, and metastasis. Most anticancer therapies target cancer cells specifically, but the tumour stroma can promote the resistance of cancer cells to such therapies, eventually resulting in fatal disease. Therefore, novel treatment strategies should combine anticancer and antistromal agents. Herein, we provide an overview of the advances in understanding the complex cancer cell-tumour stroma interactions and discuss how this knowledge can result in more effective therapeutic strategies, which might ultimately improve patient outcomes.
Collapse
|
23
|
Schweizer MT, Wang H, Bivalacqua TJ, Partin AW, Lim SJ, Chapman C, Abdallah R, Levy O, Bhowmick NA, Karp JM, De Marzo A, Isaacs JT, Brennen WN, Denmeade SR. A Phase I Study to Assess the Safety and Cancer-Homing Ability of Allogeneic Bone Marrow-Derived Mesenchymal Stem Cells in Men with Localized Prostate Cancer. Stem Cells Transl Med 2019; 8:441-449. [PMID: 30735000 PMCID: PMC6477003 DOI: 10.1002/sctm.18-0230] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022] Open
Abstract
Animal models show that systemically administered bone marrow‐derived mesenchymal stem cells (MSCs) home to sites of primary and metastatic prostate cancer (PC)—making them candidates to selectively deliver cytotoxic agents. To further assess this potential as a cell‐based therapeutic vehicle, a phase I study testing homing of systemically infused allogeneic MSCs preprostatectomy was conducted. The primary objective was to assess safety and feasibility and to determine if MSCs accumulate within primary PC tissue. MSCs were quantified using beads, emulsion, amplification, magnetics digital polymerase chain reaction (limit of detection: ≥0.01% MSCs) to measure allogeneic MSC DNA relative to recipient DNA. MSCs were harvested from healthy donors and expanded ex vivo using standard protocols by the Johns Hopkins Cell Therapy Laboratory. PC patients planning to undergo prostatectomy were eligible for MSC infusion. Enrolled subjects received a single intravenous infusion 4–6 days prior to prostatectomy. The first three subjects received 1 x 106 cells per kilogram (maximum 1 x 108 cells), and subsequent four patients received 2 x 106 cells per kilogram (maximum 2 x 108 cells). No dose‐limiting toxicities were observed and all patients underwent prostatectomy without delay. Pathologic assessment of prostate cores revealed ≥70% tumor involvement in cores from four subjects, with benign tissue in the others. MSCs were undetectable in all subjects, and the study was stopped early for futility. MSC infusions appear safe in PC patients. Although intended for eventual use in metastatic PC patients, in this study, MSCs did not home primary tumors in sufficient levels to warrant further development as a cell‐based therapeutic delivery strategy using standard ex vivo expansion protocols. stem cells translational medicine2019;8:441–449
Collapse
Affiliation(s)
- Michael T Schweizer
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Hao Wang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Trinity J Bivalacqua
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alan W Partin
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Su Jin Lim
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Carolyn Chapman
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rehab Abdallah
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Oren Levy
- Center for Nanomedicine and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Neil A Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jeffrey M Karp
- Center for Nanomedicine and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA.,Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Angelo De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John T Isaacs
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - W Nathaniel Brennen
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Samuel R Denmeade
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Krueger TE, Thorek DLJ, Meeker AK, Isaacs JT, Brennen WN. Tumor-infiltrating mesenchymal stem cells: Drivers of the immunosuppressive tumor microenvironment in prostate cancer? Prostate 2019; 79:320-330. [PMID: 30488530 PMCID: PMC6549513 DOI: 10.1002/pros.23738] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/17/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prostate cancer is characterized by T-cell exclusion, which is consistent with their poor responses to immunotherapy. In addition, T-cells restricted to the adjacent stroma and benign areas are characterized by anergic and immunosuppressive phenotypes. In order for immunotherapies to produce robust anti-tumor responses in prostate cancer, this exclusion barrier and immunosuppressive microenvironment must first be overcome. We have previously identified mesenchymal stem cells (MSCs) in primary and metastatic human prostate cancer tissue. METHODS An Opal Multiplex immunofluorescence assay based on CD73, CD90, and CD105 staining was used to identify triple-labeled MSCs in human prostate cancer tissue. T-cell suppression assays and flow cytometry were used to demonstrate the immunosuppressive potential of primary MSCs expanded from human bone marrow and prostate cancer tissue from independent donors. RESULTS Endogenous MSCs were confirmed to be present at sites of human prostate cancer. These prostate cancer-infiltrating MSCs suppress T-cell proliferation in a dose-dependent manner similar to their bone marrow-derived counterparts. Also similar to bone marrow-derived MSCs, prostate cancer-infiltrating MSCs upregulate expression of PD-L1 and PD-L2 on their cell surface in the presence of IFNγ and TNFα. CONCLUSION Prostate cancer-infiltrating MSCs suppress T-cell proliferation similar to canonical bone marrow-derived MSCs, which have well-documented immunosuppressive properties with numerous effects on both innate and adaptive immune system function. Thus, we hypothesize that selective depletion of MSCs infiltrating sites of prostate cancer should restore immunologic recognition and elimination of malignant cells via broad re-activation of cytotoxic pro-inflammatory pathways.
Collapse
Affiliation(s)
- Timothy E. Krueger
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel L. J. Thorek
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, Missouri
- Department of Biomedical Engineering, Washington University School of Medicine, Saint Louis, Missouri
| | - Alan K. Meeker
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland
| | - John T. Isaacs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - W. Nathaniel Brennen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
25
|
Päth G, Perakakis N, Mantzoros CS, Seufert J. Stem cells in the treatment of diabetes mellitus - Focus on mesenchymal stem cells. Metabolism 2019; 90:1-15. [PMID: 30342065 DOI: 10.1016/j.metabol.2018.10.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/25/2018] [Accepted: 10/14/2018] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus type 1 and type 2 have become a global epidemic with dramatically increasing incidences. Poorly controlled diabetes is associated with severe life-threatening complications. Beside traditional treatment with insulin and oral anti-diabetic drugs, clinicians try to improve patient's care by cell therapies using embryonic stem cells (ESC), induced pluripotent stem cells (iPSC) and adult mesenchymal stem cells (MSC). ESC display a virtually unlimited plasticity, including the differentiation into insulin producing β-cells, but they raise ethical concerns and bear, like iPSC, the risk of tumours. IPSC may further inherit somatic mutations and remaining somatic transcriptional memory upon incomplete re-programming, but allow the generation of patient/disease-specific cell lines. MSC avoid such issues but have not been successfully differentiated into β-cells. Instead, MSC and their pericyte phenotypes outside the bone marrow have been recognized to secrete numerous immunomodulatory and tissue regenerative factors. On this account, the term 'medicinal signaling cells' has been proposed to define the new conception of a 'drug store' for injured tissues and to stay with the MSC nomenclature. This review presents the biological background and the resulting clinical potential and limitations of ESC, iPSC and MSC, and summarizes the current status quo of cell therapeutic concepts and trials.
Collapse
Affiliation(s)
- Günter Päth
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.
| | - Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jochen Seufert
- Division of Endocrinology and Diabetology, Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
26
|
Yin Z, Jiang K, Li R, Dong C, Wang L. Multipotent mesenchymal stromal cells play critical roles in hepatocellular carcinoma initiation, progression and therapy. Mol Cancer 2018; 17:178. [PMID: 30593276 PMCID: PMC6309092 DOI: 10.1186/s12943-018-0926-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/16/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, with high morbidity, relapse and mortality rates. Multipotent mesenchymal stromal cells (MSCs) can be recruited to and become integral components of the HCC microenvironment and can influence tumor progression. This review discusses MSC migration to liver fibrosis and the HCC microenvironment, MSC involvement in HCC initiation and progression and the widespread application of MSCs in HCC-targeted therapy, thus clarifying the critical roles of MSCs in HCC.
Collapse
Affiliation(s)
- Zeli Yin
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China.,Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China.,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Keqiu Jiang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China.,Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China.,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Rui Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China.,Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China.,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China
| | - Chengyong Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China. .,Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China. .,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China.
| | - Liming Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027, Liaoning, China. .,Engineering Research Center for New Materials and Precision Treatment Technology of Malignant Tumors Therapy, Dalian Medical University, Dalian, 116027, Liaoning, China. .,Engineering Technology Research Center for Translational Medicine, Dalian Medical University, Dalian, 116027, Liaoning, China.
| |
Collapse
|
27
|
Moazen B, Zarrinhaghighi A, Nejatollahi F. Selection and Evaluation of Specific Single Chain Antibodies against CD90, a Marker for Mesenchymal and Cancer Stem Cells. Rep Biochem Mol Biol 2018; 7:45-51. [PMID: 30324117 PMCID: PMC6175597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/29/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND CD90, a membrane-associated glycoprotein is a marker used to identify mesenchymal stem cells (MSCs). Recent studies have introduced CD90, which induces tumorigenic activity, as a cancer stem cell (CSC) marker in various malignancies. Blocking CD90 activity with anti-CD90 monoclonal antibodies enhanced anti-tumor effects. To date, highly specific antibody single-chain variable fragments (scFvs) have been isolated against various targets and showed promising results in cancer immunotherapy. METHODS A phage antibody was produced from a scFv library using M13KO7 helper phage. The phage library was panned against a CD90 epitope. To select specific clones, PCR and DNA fingerprinting were performed and common patterns were identified. The panning results were confirmed by phage ELISA. RESULTS Of 20 clones selected after panning, 16 shared identical fingerprints. One clone from this group reacted specifically with the epitope in phage ELISA. The average absorbance of wells coated with the CD90 peptide was significantly greater than that of wells containing no peptide (p=0.03). CONCLUSION Currently, recombinant antibodies are used not only as highly specific detection tools, but due to their specific characteristics, are applied in targeted cancer therapies. The anti-CD90 scFv selected in this study has the potential to be used to detect MSCs and target CSCs and offers promising strategies for treatment of various cancers.
Collapse
Affiliation(s)
- Bahareh Moazen
- Shiraz HIV/AIDS Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Recombinant antibody laboratory, Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Zarrinhaghighi
- Shiraz HIV/AIDS Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Recombinant antibody laboratory, Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Foroogh Nejatollahi
- Shiraz HIV/AIDS Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Recombinant antibody laboratory, Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
28
|
Heterogeneous cancer-associated fibroblast population potentiates neuroendocrine differentiation and castrate resistance in a CD105-dependent manner. Oncogene 2018; 38:716-730. [PMID: 30177832 PMCID: PMC7182071 DOI: 10.1038/s41388-018-0461-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/27/2018] [Accepted: 07/24/2018] [Indexed: 11/20/2022]
Abstract
Heterogeneous prostatic carcinoma associated fibroblasts (CAF) contribute to tumor progression and resistance to androgen signaling deprivation therapy (ADT). CAF subjected to extended passaging, compared to low passage CAF, were found to lose tumor expansion potential and heterogeneity. Cell surface endoglin (CD105), known to be expressed on proliferative endothelia and mesenchymal stem cells, was diminished in high passage CAF. RNA-sequencing revealed SFRP1 to be distinctly expressed by tumor-inductive CAF, which was further demonstrated to occur in a CD105-dependent manner. Moreover, ADT resulted in further expansion of the CD105+ fibroblastic population and downstream SFRP1 in 3-dimensional cultures and patient derived xenograft tissues. In patients, CD105+ fibroblasts were found to circumscribe epithelia with neuroendocrine differentiation. CAF-derived SFRP1, driven by CD105 signaling, was necessary and sufficient to induce prostate cancer neuroendocrine differentiation in a paracrine manner. A partially humanized CD105 neutralizing antibody, TRC105, inhibited fibroblastic SFRP1 expression and epithelial neuroendocrine differentiation. In a novel synthetic lethality paradigm, we found that simultaneously targeting the epithelia and its microenvironment with ADT and TRC105, respectively, reduced castrate resistant tumor progression, in a model where either ADT or TRC105 alone had little effect.
Collapse
|
29
|
Krueger TEG, Thorek DLJ, Denmeade SR, Isaacs JT, Brennen WN. Concise Review: Mesenchymal Stem Cell-Based Drug Delivery: The Good, the Bad, the Ugly, and the Promise. Stem Cells Transl Med 2018; 7:651-663. [PMID: 30070053 PMCID: PMC6127224 DOI: 10.1002/sctm.18-0024] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/15/2018] [Accepted: 05/30/2018] [Indexed: 12/12/2022] Open
Abstract
The development of mesenchymal stem cells (MSCs) as cell‐based drug delivery vectors for numerous clinical indications, including cancer, has significant promise. However, a considerable challenge for effective translation of these approaches is the limited tumor tropism and broad biodistribution observed using conventional MSCs, which raises concerns for toxicity to nontarget peripheral tissues (i.e., the bad). Consequently, there are a variety of synthetic engineering platforms in active development to improve tumor‐selective targeting via increased homing efficiency and/or specificity of drug activation, some of which are already being evaluated clinically (i.e., the good). Unfortunately, the lack of robust quantification and widespread adoption of standardized methodologies with high sensitivity and resolution has made accurate comparisons across studies difficult, which has significantly impeded progress (i.e., the ugly). Herein, we provide a concise review of active and passive MSC homing mechanisms and biodistribution postinfusion; in addition to in vivo cell tracking methodologies and strategies to enhance tumor targeting with a focus on MSC‐based drug delivery strategies for cancer therapy. Stem Cells Translational Medicine2018;1–13
Collapse
Affiliation(s)
- Timothy E G Krueger
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel L J Thorek
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins, Baltimore, Maryland, USA
| | - Samuel R Denmeade
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins, Baltimore, Maryland, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John T Isaacs
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins, Baltimore, Maryland, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - W Nathaniel Brennen
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center (SKCCC) at Johns Hopkins, Baltimore, Maryland, USA
| |
Collapse
|
30
|
Vlaeminck-Guillem V. Extracellular Vesicles in Prostate Cancer Carcinogenesis, Diagnosis, and Management. Front Oncol 2018; 8:222. [PMID: 29951375 PMCID: PMC6008571 DOI: 10.3389/fonc.2018.00222] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/29/2018] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), especially exosomes, are now well recognized as major ways by which cancer cells interact with each other and stromal cells. The meaningful messages transmitted by the EVs are carried by all components of the EVs, i.e., the membrane lipids and the cargo (DNAs, RNAs, microRNAs, long non-coding RNAs, proteins). They are clearly part of the armed arsenal by which cancer cells obtain and share more and more advantages to grow and conquer new spaces. Identification of these messages offers a significant opportunity to better understand how a cancer occurs and then develops both locally and distantly. But it also provides a powerful means by which cancer progression can be detected and monitored. In the last few years, significant research efforts have been made to precisely identify how the EV trafficking is modified in cancer cells as compared to normal cells and how this trafficking is altered during cancer progression. Prostate cancer has not escaped this trend. The aim of this review is to describe the results obtained when assessing the meaningful content of prostate cancer- and stromal-derived EVs in terms of a better comprehension of the cellular and molecular mechanisms underlying prostate cancer occurrence and development. This review also deals with the use of EVs as powerful tools to diagnose non-indolent prostate cancer as early as possible and to accurately define, in a personalized approach, its present and potential aggressiveness, its response to treatment (androgen deprivation, chemotherapy, radiation, surgery), and the overall patients’ prognosis.
Collapse
Affiliation(s)
- Virginie Vlaeminck-Guillem
- Medical Unit of Molecular Oncology and Transfer, Department of Biochemistry and Molecular Biology, Centre Hospitalier Lyon-Sud, Hospices Civils of Lyon, Pierre-Bénite, France.,Cancer Research Centre of Lyon, U1052 INSERM, CNRS 5286, Claude Bernard University Lyon 1, Léon Bérard Centre, Lyon, France
| |
Collapse
|
31
|
Brennen WN, Zhang B, Kulac I, Kisteman LN, Antony L, Wang H, Meeker AK, De Marzo AM, Garraway IP, Denmeade SR, Isaacs JT. Mesenchymal stem cell infiltration during neoplastic transformation of the human prostate. Oncotarget 2018; 8:46710-46727. [PMID: 28493842 PMCID: PMC5564518 DOI: 10.18632/oncotarget.17362] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 04/01/2017] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal Stem Cells (MSCs) have been identified in prostate cancer, raising the critical question of their physical and temporal source. Therefore, MSCs were quantified and characterized in benign and malignant prostate tissue representing different disease states and a wide range of age groups from fetal development through adult death using analytical and functional methodologies. In contrast to lineage-restricted Mesenchymal Progenitor Cells (MPCs) found in normal prostate tissue, MSCs with tri-lineage differentiation potential (adipogenesis, osteogenesis, and chondrogenesis) are identified in prostate tissue from a subset of men with prostate cancer, consistent with an influx of more stem-like progenitors (i.e. MSCs) from the bone marrow. Additionally, prostate tissue from a subset of these patients is highly enriched in MSCs, suggesting their enumeration may have prognostic value for identifying men with aggressive disease. This influx is an ongoing process continuing throughout disease progression as documented by the presence of MSCs in metastatic lesions from multiple organ sites harvested at the time of death in metastatic castration-resistant prostate cancer (mCRPC) patients. This infiltration of MSCs from systemic circulation provides the rationale for their use as a cell-based vector to deliver therapeutic agents.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Baohui Zhang
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ibrahim Kulac
- Department of Pathology at the SKCCC at Johns Hopkins, Baltimore, MD, USA
| | - L Nelleke Kisteman
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Lizamma Antony
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Hao Wang
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Alan K Meeker
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Department of Pathology at the SKCCC at Johns Hopkins, Baltimore, MD, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Angelo M De Marzo
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Department of Pathology at the SKCCC at Johns Hopkins, Baltimore, MD, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Isla P Garraway
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Samuel R Denmeade
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John T Isaacs
- Department of Oncology at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.,Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
32
|
Lugini L, Valtieri M, Federici C, Cecchetti S, Meschini S, Condello M, Signore M, Fais S. Exosomes from human colorectal cancer induce a tumor-like behavior in colonic mesenchymal stromal cells. Oncotarget 2018; 7:50086-50098. [PMID: 27418137 PMCID: PMC5226570 DOI: 10.18632/oncotarget.10574] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/29/2016] [Indexed: 12/30/2022] Open
Abstract
Background Cancer cells, including colorectal cancer ones (CRC), release high amounts of nanovesicles (exosomes), delivering biochemical messages for paracrine or systemic crosstalk. Mesenchymal stromal cells (MSCs) have been shown to play contradicting roles in tumor progression. Results CRC exosomes induce in cMSCs: i) atypical morphology, higher proliferation, migration and invasion; ii) formation of spheroids; iii) an acidic extracellular environment associated with iv) a plasma membrane redistribution of vacuolar H+-ATPase and increased expression of CEA. Colon cancer derived MSCs, which were isolated from tumor masses, produce umbilicated spheroids, a future frequently observed in the inner core of rapidly growing tumors and recapitulate the changes observed in normal colonic MSCs exposed to CRC exosomes. Materials and Methods Tissue specific colonic (c)MSCs were exposed to primary or metastatic CRC exosomes and analysed by light and electron microscopy, proliferation in 2D and 3D cultures, migration and invasion assays, Western blot and confocal microscopy for vacuolar H+-ATPase expression. Conclusions CRC exosomes are able to induce morphological and functional changes in colonic MSCs, which may favour tumor growth and its malignant progression. Our results suggest that exosomes are actively involved in cancer progression and that inhibiting tumor exosome release may represent a way to interfere with cancer.
Collapse
Affiliation(s)
- Luana Lugini
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Valtieri
- Department of Ematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Cristina Federici
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Serena Cecchetti
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Stefania Meschini
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Condello
- Department of Technology and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Michele Signore
- Department of Ematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Fais
- Department of Therapeutic Research and Medicine Evaluation, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
33
|
Assessing angiogenic responses induced by primary human prostate stromal cells in a three-dimensional fibrin matrix assay. Oncotarget 2018; 7:71298-71308. [PMID: 27542256 PMCID: PMC5342079 DOI: 10.18632/oncotarget.11347] [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: 06/01/2016] [Accepted: 08/10/2016] [Indexed: 01/08/2023] Open
Abstract
Accurate modeling of angiogenesis in vitro is essential for guiding the preclinical development of novel anti-angiogenic agents and treatment strategies. The formation of new blood vessels is a multifactorial and multi-stage process dependent upon paracrine factors produced by stromal cells in the local microenvironment. Mesenchymal stem cells (MSCs) are multipotent cells in adults that can be recruited to sites of inflammation and tissue damage where they aid in wound healing through regenerative, trophic, and immunomodulatory properties. Primary stromal cultures derived from human bone marrow, normal prostate, or prostate cancer tissue are highly enriched in MSCs and stromal progenitors. Using conditioned media from these primary cultures, a robust pro-angiogenic response was observed in a physiologically-relevant three-dimensional fibrin matrix assay. To evaluate the utility of this assay, the allosteric HDAC4 inhibitor tasquinimod and the anti-VEGF monoclonal antibody bevacizumab were used as model compounds with distinct mechanisms of action. While both agents had a profound inhibitory effect on endothelial sprouting, only bevacizumab induced significant regression of established vessels. Additionally, the pro-angiogenic properties of MSCs derived from prostate cancer patients provides further evidence that selective targeting of this population may be of therapeutic benefit.
Collapse
|
34
|
Trivanović D, Krstić J, Jauković A, Bugarski D, Santibanez JF. Mesenchymal stromal cell engagement in cancer cell epithelial to mesenchymal transition. Dev Dyn 2017; 247:359-367. [PMID: 28850772 DOI: 10.1002/dvdy.24583] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 07/04/2017] [Accepted: 08/10/2017] [Indexed: 12/14/2022] Open
Abstract
Due to coexistence of stromal and epithelial tumor cells, their dynamic interactions have been widely recognized as significant cellular components to the tumor tissue integrity. Initiation and outcome of epithelial to mesenchymal transition (EMT) in tumor cells are dependent on their interaction with adjacent or recruited mesenchymal stromal cells (MSCs). A plethora of mechanisms are involved in MSCs-controlled employment of the developmental processes of EMT that contribute to loss of epithelial cell phenotype and acquisition of stemness, invasiveness and chemoresistance of tumor cells. Interplay of MSCs with tumor cells, including interchange of soluble biomolecules, plasma membrane structures, cytoplasmic content, and organelles, is established through cell-cell contact and/or by means of paracrine signaling. The main focus of this review is to summarize knowledge about involvement of MSCs in cancer cell EMT. Understanding the underlying cellular and molecular mechanism involved in the interplay between MSCs and cancer EMT is essential for development of effective therapy approaches, which in combination with current treatments may improve the control of tumor progression. Developmental Dynamics 247:359-367, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Drenka Trivanović
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Jelena Krstić
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Aleksandra Jauković
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Diana Bugarski
- Laboratory for Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Juan F Santibanez
- Group for Molecular Oncology, Institute for Medical Research, University of Belgrade, Belgrade, Republic of Serbia
| |
Collapse
|
35
|
Tao X, Xu L, Yin L, Han X, Qi Y, Xu Y, Song S, Zhao Y, Peng J. Dioscin induces prostate cancer cell apoptosis through activation of estrogen receptor-β. Cell Death Dis 2017; 8:e2989. [PMID: 28796245 PMCID: PMC5596577 DOI: 10.1038/cddis.2017.391] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 06/22/2017] [Accepted: 07/10/2017] [Indexed: 12/31/2022]
Abstract
Recent researches have shown that estrogen receptor-β (ERβ) activator may be a potent anticancer agent for prostate cancer (PCa), and our previous study also indicated that dioscin can upregulate the expression of ERβ in MC3T3-E1 cell. In the present work, the activity and mechanism of dioscin, a natural product, against PCa were investigated. The results showed that dioscin markedly inhibited cell viability, colony formation, motility and induced apoptosis in PC3 cells. Moreover, dioscin disrupted the formation of PC3 cell-derived mammospheres and reduced aldehyde dehydrogenase (ALDH) level and the CD133+/CD44+ cells, indicating that dioscin had a potent inhibitory activity on prostate cancer stem cells (PCSCs). In vivo results also showed that dioscin significantly suppressed the tumor growth of PC3 cell xenografts in nude mice. Furthermore, mechanism investigation showed that dioscin markedly upregulated ERβ expression level, subsequently increased prolyl hydroxylase 2 level, decreased the levels of hypoxia-inducible factor-1α, vascular endothelial growth factor A and BMI-1, and thus induced cell apoptosis by regulating the expression levels of caspase-3 and Bcl-2 family proteins. In addition, transfection experiment of ERβ-siRNA further indicated that diosicn showed excellent activity against PCa in vitro and in vivo by increasing ERβ expression level. The co-immunoprecipitation (Co-IP) results further suggested that dioscin promoted the interaction of c-ABL and ERβ, but did not change c-ABL expression. Moreover, the molecular docking assay showed that dioscin processed powerful affinity toward to ERβ mainly through the strong hydrogen bonding and hydrophobic effects, and the actions of dioscin on ERβ activation and tumor cells inhibition were significantly weakened in the mutational (Phe-336, Phe-468) PC3 cells. Collectively, these findings proved that dioscin exerted efficient anti-PCa activity via activation of ERβ, which should be developed as an efficient candidate in clinical for treating this cancer in the future.
Collapse
Affiliation(s)
- Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Shasha Song
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yanyan Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| |
Collapse
|
36
|
Chemaly ER, Troncone L, Lebeche D. SERCA control of cell death and survival. Cell Calcium 2017; 69:46-61. [PMID: 28747251 DOI: 10.1016/j.ceca.2017.07.001] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/03/2017] [Accepted: 07/03/2017] [Indexed: 12/31/2022]
Abstract
Intracellular calcium (Ca2+) is a critical coordinator of various aspects of cellular physiology. It is increasingly apparent that changes in cellular Ca2+ dynamics contribute to the regulation of normal and pathological signal transduction that controls cell growth and survival. Aberrant perturbations in Ca2+ homeostasis have been implicated in a range of pathological conditions, such as cardiovascular diseases, diabetes, tumorigenesis and steatosis hepatitis. Intracellular Ca2+ concentrations are therefore tightly regulated by a number of Ca2+ handling enzymes, proteins, channels and transporters located in the plasma membrane and in Ca2+ storage organelles, which work in concert to fine tune a temporally and spatially precise Ca2+ signal. Chief amongst them is the sarco/endoplasmic reticulum (SR/ER) Ca2+ ATPase pump (SERCA) which actively re-accumulates released Ca2+ back into the SR/ER, therefore maintaining Ca2+ homeostasis. There are at least 14 different SERCA isoforms encoded by three ATP2A1-3 genes whose expressions are species- and tissue-specific. Altered SERCA expression and activity results in cellular malignancy and induction of ER stress and ER stress-associated apoptosis. The role of SERCA misregulation in the control of apoptosis in various cell types and disease setting with prospective therapeutic implications is the focus of this review. Ca2+ is a double edge sword for both life as well as death, and current experimental evidence supports a model in which Ca2+ homeostasis and SERCA activity represent a nodal point that controls cell survival. Pharmacological or genetic targeting of this axis constitutes an incredible therapeutic potential to treat different diseases sharing similar biological disorders.
Collapse
Affiliation(s)
- Elie R Chemaly
- Division of Nephrology and Hypertension, Department of Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Luca Troncone
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Djamel Lebeche
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Diabetes, Obesity and Metabolism Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Graduate School of Biological Sciences, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA.
| |
Collapse
|
37
|
Dalla Pozza E, Forciniti S, Palmieri M, Dando I. Secreted molecules inducing epithelial-to-mesenchymal transition in cancer development. Semin Cell Dev Biol 2017; 78:62-72. [PMID: 28673679 DOI: 10.1016/j.semcdb.2017.06.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/27/2017] [Accepted: 06/29/2017] [Indexed: 02/08/2023]
Abstract
The epithelial-mesenchymal transition (EMT) is a biologic process that allows a polarized epithelial cell to undergo multiple biochemical changes that enable it to assume a mesenchymal cell phenotype. EMT is involved in embryo development, wound healing, tissue regeneration, organ fibrosis and has also been proposed as the critical mechanism for the acquisition of malignant phenotypes by epithelial cancer cells. These cells have been shown to acquire a mesenchymal phenotype when localized at the invasive front of primary tumours increasing aggressiveness, invasiveness, metastatic potential and resistance to chemotherapy. There is now increasing evidence demonstrating that a crucial role in the development of this process is played by factors secreted by cells of the tumour microenvironment or by the tumour cells themselves. This review summarises the current knowledge of EMT induction in cancer by paracrine or autocrine mechanisms, by exosomes or free proteins and miRNAs.
Collapse
Affiliation(s)
- Elisa Dalla Pozza
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Stefania Forciniti
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| | - Marta Palmieri
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy.
| | - Ilaria Dando
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Verona, Italy
| |
Collapse
|
38
|
Endaya B, Guan SP, Newman JP, Huynh H, Sia KC, Chong ST, Kok CYL, Chung AYF, Liu BB, Hui KM, Lam PYP. Human mesenchymal stem cells preferentially migrate toward highly oncogenic human hepatocellular carcinoma cells with activated EpCAM signaling. Oncotarget 2017; 8:54629-54639. [PMID: 28903370 PMCID: PMC5589609 DOI: 10.18632/oncotarget.17633] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 02/27/2017] [Indexed: 01/16/2023] Open
Abstract
The epithelial cell adhesion molecule (EpCAM) is a type I transmembrane glycoprotein that is regarded as one of the markers for tumor initiating cells (TIC) in human hepatocellular carcinoma (HCC). Much work has been directed towards targeting these TICs as a mean of placing these master regulators of cell proliferation and drug resistance under control. Human bone marrow-derived mesenchymal stem cells are known to exhibit an innate property of tumor tropism. However, the possible relationship between MSC and TIC is not well understood. In this study, we show that MSC migration to HCC can be effectively inhibited by TACE and γ-secretase inhibitors that stop the activation of EpCAM signaling event. Silencing of EpCAM expression through siRNA and antibody approaches also resulted in impaired MSC migration. By contrast, increase levels of EpICD proteins in HCC cells and HCC mouse xenografts resulted in enhanced MSC migration. Taken together, these findings show that MSC is drawn to the more oncogenic population of HCC, and could potentially serve as a cell-based carrier of therapeutic genes to target EpICD-enriched hepatic tumor cells.
Collapse
Affiliation(s)
- Berwini Endaya
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore City, Singapore.,Griffith Health Institute, Griffith University, Southport, Australia
| | - Shou P Guan
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore City, Singapore
| | - Jennifer P Newman
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore City, Singapore
| | - Hung Huynh
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore City, Singapore
| | - Kian C Sia
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore City, Singapore
| | - Siao T Chong
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore City, Singapore
| | - Catherine Y L Kok
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore City, Singapore
| | - Alexander Y F Chung
- Department of General Surgery, Singapore General Hospital, Singapore City, Singapore
| | - Bin B Liu
- Liver Cancer Institute of Fudan University, Shanghai, China
| | - Kam M Hui
- Department of Biochemistry, National University of Singapore, Singapore City, Singapore.,Institute of Molecular and Cell Biology, ASTAR, Singapore City, Singapore.,Cancer and Stem Cells Biology Program, Duke-NUS Graduate Medical School, Singapore City, Singapore
| | - Paula Y P Lam
- Division of Cellular and Molecular Research, National Cancer Centre, Singapore City, Singapore.,Department of Physiology, National University of Singapore, Singapore City, Singapore.,Cancer and Stem Cells Biology Program, Duke-NUS Graduate Medical School, Singapore City, Singapore
| |
Collapse
|
39
|
Sharpe B, Alghezi DA, Cattermole C, Beresford M, Bowen R, Mitchard J, Chalmers AD. A subset of high Gleason grade prostate carcinomas contain a large burden of prostate cancer syndecan-1 positive stromal cells. Prostate 2017; 77:1312-1324. [PMID: 28744948 DOI: 10.1002/pros.23391] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 06/26/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND There is a pressing need to identify prognostic and predictive biomarkers for prostate cancer to aid treatment decisions in both early and advanced disease settings. Syndecan-1, a heparan sulfate proteoglycan, has been previously identified as a potential prognostic biomarker by multiple studies at the tissue and serum level. However, other studies have questioned its utility. METHODS Anti-Syndecan-1 immunohistochemistry was carried out on 157 prostate tissue samples (including cancerous, adjacent normal tissue, and non-diseased prostate) from three independent cohorts of patients. A population of Syndecan-1 positive stromal cells was identified and the number and morphological parameters of these cells quantified. The identity of the Syndecan-1-positive stromal cells was assessed by multiplex immunofluorescence using a range of common cell lineage markers. Finally, the burden of Syndecan-1 positive stromal cells was tested for association with clinical parameters. RESULTS We identified a previously unreported cell type which is marked by Syndecan-1 expression and is found in the stroma of prostate tumors and adjacent normal tissue but not in non-diseased prostate. We call these cells Prostate Cancer Syndecan-1 Positive (PCSP) cells. Immunofluorescence analysis revealed that the PCSP cell population did not co-stain with markers of common prostate epithelial, stromal, or immune cell populations. However, morphological analysis revealed that PCSP cells are often elongated and displayed prominent lamellipodia, suggesting they are an unidentified migratory cell population. Analysis of clinical parameters showed that PCSP cells were found with a frequency of 20-35% of all tumors evaluated, but were not present in non-diseased normal tissue. Interestingly, a subset of primary Gleason 5 prostate tumors had a high burden of PCSP cells. CONCLUSIONS The current study identifies PCSP cells as a novel, potentially migratory cell type, which is marked by Syndecan-1 expression and is found in the stroma of prostate carcinomas, adjacent normal tissue, but not in non-diseased prostate. A subset of poor prognosis high Gleason grade 5 tumors had a particularly high PCSP cell burden, suggesting an association between this unidentified cell type and tumor aggressiveness.
Collapse
Affiliation(s)
- Benjamin Sharpe
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Dhafer A Alghezi
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Thi Qar University, Dhi Qar, Iraq
| | - Claire Cattermole
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Mark Beresford
- Department of Oncology, Royal United Hospital, Bath, United Kingdom
| | - Rebecca Bowen
- Department of Oncology, Royal United Hospital, Bath, United Kingdom
| | - John Mitchard
- Department of Cellular Pathology, Royal United Hospital, Bath, United Kingdom
| | - Andrew D Chalmers
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| |
Collapse
|
40
|
Ridge SM, Sullivan FJ, Glynn SA. Mesenchymal stem cells: key players in cancer progression. Mol Cancer 2017; 16:31. [PMID: 28148268 PMCID: PMC5286812 DOI: 10.1186/s12943-017-0597-8] [Citation(s) in RCA: 362] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/19/2017] [Indexed: 02/08/2023] Open
Abstract
Tumour progression is dependent on the interaction between tumour cells and cells of the surrounding microenvironment. The tumour is a dynamic milieu consisting of various cell types such as endothelial cells, fibroblasts, cells of the immune system and mesenchymal stem cells (MSCs). MSCs are multipotent stromal cells that are known to reside in various areas such as the bone marrow, fat and dental pulp. MSCs have been found to migrate towards inflammatory sites and studies have shown that they also migrate towards and incorporate into the tumour. The key question is how they interact there. MSCs may interact with tumour cells through paracrine signalling. On the other hand, MSCs have the capacity to differentiate to various cell types such as osteocytes, chondrocytes and adipocytes and it is possible that MSCs differentiate at the site of the tumour. More recently it has been shown that cross-talk between tumour cells and MSCs has been shown to increase metastatic potential and promote epithelial-to-mesenchymal transition. This review will focus on the role of MSCs in tumour development at various stages of progression from growth of the primary tumour to the establishment of distant metastasis.
Collapse
Affiliation(s)
- Sarah M Ridge
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, Costello Road, Galway, Ireland.,Prostate Cancer Institute, School of Medicine, Costello Road, Galway, Ireland
| | - Francis J Sullivan
- Prostate Cancer Institute, School of Medicine, Costello Road, Galway, Ireland
| | - Sharon A Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, Costello Road, Galway, Ireland. .,Prostate Cancer Institute, School of Medicine, Costello Road, Galway, Ireland.
| |
Collapse
|
41
|
Saulite L, Dapkute D, Pleiko K, Popena I, Steponkiene S, Rotomskis R, Riekstina U. Nano-engineered skin mesenchymal stem cells: potential vehicles for tumour-targeted quantum-dot delivery. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2017; 8:1218-1230. [PMID: 28685122 PMCID: PMC5480321 DOI: 10.3762/bjnano.8.123] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/10/2017] [Indexed: 05/20/2023]
Abstract
Nanotechnology-based drug design offers new possibilities for the use of nanoparticles in imaging and targeted therapy of tumours. Due to their tumour-homing ability, nano-engineered mesenchymal stem cells (MSCs) could be utilized as vectors to deliver diagnostic and therapeutic nanoparticles into a tumour. In the present study, uptake and functional effects of carboxyl-coated quantum dots QD655 were studied in human skin MSCs. The effect of QD on MSCs was examined using a cell viability assay, Ki67 expression analysis, and tri-lineage differentiation assay. The optimal conditions for QD uptake in MSCs were determined using flow cytometry. The QD uptake route in MSCs was examined via fluorescence imaging using endocytosis inhibitors for the micropinocytosis, phagocytosis, lipid-raft, clathrin- and caveolin-dependent endocytosis pathways. These data showed that QDs were efficiently accumulated in the cytoplasm of MSCs after incubation for 6 h. The main uptake route of QDs in skin MSCs was clathrin-mediated endocytosis. QDs were mainly localized in early endosomes after 6 h as well as in late endosomes and lysosomes after 24 h. QDs in concentrations ranging from 0.5 to 64 nM had no effect on cell viability and proliferation. The expression of MSC markers, CD73 and CD90, and hematopoietic markers, CD34 and CD45, as well as the ability to differentiate into adipocytes, chondrocytes, and osteocytes, were not altered in the presence of QDs. We observed a decrease in the QD signal from labelled MSCs over time that could partly reflect QD excretion. Altogether, these data suggest that QD-labelled MSCs could be used for targeted drug delivery studies.
Collapse
Affiliation(s)
- Liga Saulite
- Faculty of Medicine, University of Latvia, Raina blvd. 19, LV-1586, Riga, Latvia
| | - Dominyka Dapkute
- Biomedical Physics Laboratory, National Cancer Institute, P. Baublio Street 3b, LT-08406 Vilnius, Lithuania
- Life Science Center, Vilnius University, Sauletekio al. 7, LT-10257, Vilnius, Lithuania
| | - Karlis Pleiko
- Faculty of Medicine, University of Latvia, Raina blvd. 19, LV-1586, Riga, Latvia
| | - Ineta Popena
- Faculty of Medicine, University of Latvia, Raina blvd. 19, LV-1586, Riga, Latvia
| | - Simona Steponkiene
- Biomedical Physics Laboratory, National Cancer Institute, P. Baublio Street 3b, LT-08406 Vilnius, Lithuania
| | - Ricardas Rotomskis
- Biomedical Physics Laboratory, National Cancer Institute, P. Baublio Street 3b, LT-08406 Vilnius, Lithuania
- Laser research center, Vilnius University, Sauletekio al. 9, corp. 3, LT-10222, Vilnius, Lithuania
| | - Una Riekstina
- Faculty of Medicine, University of Latvia, Raina blvd. 19, LV-1586, Riga, Latvia
| |
Collapse
|
42
|
Effect of PI3K/Akt Signaling Pathway on the Process of Prostate Cancer Metastasis to Bone. Cell Biochem Biophys 2016; 72:171-7. [PMID: 27040945 DOI: 10.1007/s12013-014-0433-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We sought to study the effects of PI3K/Akt pathway and its downstream substrate NF-κB on prostate cancer bone metastatic process. Expression level of active p-Akt in PC3 cells was upregulated by transient expression with constitutively active plasmid CA-Akt or, alternatively, suppressed by dominant negative construct DN-Akt. NF-κB activity was determined by luciferase reporter assays. mRNA and protein expressions of receptor activator of NF-κB ligand (RANKL), parathyroid hormone-related protein (PTHrP), and bone morphogenetic protein 2 (BMP-2) were evaluated using RT-PCR and Western blotting. The effect of cross-talk between PC3 and SaOS2 cells on cell proliferation was analyzed using a co-culture system. Stimulation of p-Akt promoted NF-κB activity, and led to an increase in mRNA and protein expressions of RANKL, PTHrP, and BMP-2 in PC3 PCa cells through NF-κB. Co-culturing PC3 and SaOS2 cells significantly increased the expression of p-Akt and the activity of NF-κB, and promoted proliferation of both PC3 and SaOS2 cells. Increasing expression levels of p-Akt by transfection with CA-Akt led to further increase in cells proliferation, whereas NF-κB inhibitor PDTC partially blocked this effect. PI3K/Akt pathway stimulates the expressions of RANKL, PTHrP, and BMP-2 partly through NF-κB, suggesting its importance for bone metastasis of prostate carcinoma. Interaction of prostate cancer cells with bone cells has a stimulatory effect on cell proliferation.
Collapse
|
43
|
Wang L, Xie L, Tintani F, Xie H, Li C, Cui Z, Wan M, Zu X, Qi L, Cao X. Aberrant Transforming Growth Factor-β Activation Recruits Mesenchymal Stem Cells During Prostatic Hyperplasia. Stem Cells Transl Med 2016; 6:394-404. [PMID: 28191756 PMCID: PMC5442798 DOI: 10.5966/sctm.2015-0411] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 07/28/2016] [Indexed: 02/05/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is the overgrowth of prostate tissues with high prevalence in older men. BPH pathogenesis is not completely understood, but it is believed to be a result of de novo overgrowth of prostatic stroma. In this study, we show that aberrant activation of transforming growth factor‐β (TGF‐β) mobilizes mesenchymal/stromal stem cells (MSCs) in circulating blood, which are recruited for the prostatic stromal hyperplasia. Elevated levels of active TGF‐β were observed in both a phenylephrine‐induced prostatic hyperplasia mouse model and human BPH tissues. Nestin lineage tracing revealed that 39.6% ± 6.3% of fibroblasts and 73.3% ± 4.2% smooth muscle cells were derived from nestin+ cells in Nestin‐Cre, Rosa26‐YFPflox/+mice. Nestin+ MSCs were increased in the prostatic hyperplasia mice. Our parabiosis experiment demonstrate that nestin+ MSCs were mobilized and recruited to the prostatic stroma of wild‐type mice and gave rise to the fibroblasts. Moreover, injection of a TGF‐β neutralizing antibody (1D11) inhibits mobilization of MSCs, their recruitment to the prostatic stroma and hyperplasia. Importantly, knockout of TβRII in nestin+ cell lineage ameliorated stromal hyperplasia. Thus, elevated levels of TGF‐β‐induced mobilization and recruitment of MSCs to the reactive stroma resulting in overgrowth of prostate tissues in BPH and, thus, inhibition of TGF‐β activity could be a potential therapy for BPH. Stem Cells Translational Medicine2017;6:394–404
Collapse
Affiliation(s)
- Long Wang
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Liang Xie
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Francis Tintani
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hui Xie
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Changjun Li
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zhuang Cui
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mei Wan
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Lin Qi
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xu Cao
- Department of Orthopedic Surgery and Institute of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
44
|
Shearer JJ, Wold EA, Umbaugh CS, Lichti CF, Nilsson CL, Figueiredo ML. Inorganic Arsenic-Related Changes in the Stromal Tumor Microenvironment in a Prostate Cancer Cell-Conditioned Media Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:1009-15. [PMID: 26588813 PMCID: PMC4937864 DOI: 10.1289/ehp.1510090] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 11/12/2015] [Indexed: 05/18/2023]
Abstract
BACKGROUND The tumor microenvironment plays an important role in the progression of cancer by mediating stromal-epithelial paracrine signaling, which can aberrantly modulate cellular proliferation and tumorigenesis. Exposure to environmental toxicants, such as inorganic arsenic (iAs), has also been implicated in the progression of prostate cancer. OBJECTIVE The role of iAs exposure in stromal signaling in the tumor microenvironment has been largely unexplored. Our objective was to elucidate molecular mechanisms of iAs-induced changes to stromal signaling by an enriched prostate tumor microenvironment cell population, adipose-derived mesenchymal stem/stromal cells (ASCs). RESULTS ASC-conditioned media (CM) collected after 1 week of iAs exposure increased prostate cancer cell viability, whereas CM from ASCs that received no iAs exposure decreased cell viability. Cytokine array analysis suggested changes to cytokine signaling associated with iAs exposure. Subsequent proteomic analysis suggested a concentration-dependent alteration to the HMOX1/THBS1/TGFβ signaling pathway by iAs. These results were validated by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blotting, confirming a concentration-dependent increase in HMOX1 and a decrease in THBS1 expression in ASC following iAs exposure. Subsequently, we used a TGFβ pathway reporter construct to confirm a decrease in stromal TGFβ signaling in ASC following iAs exposure. CONCLUSIONS Our results suggest a concentration-dependent alteration of stromal signaling: specifically, attenuation of stromal-mediated TGFβ signaling following exposure to iAs. Our results indicate iAs may enhance prostate cancer cell viability through a previously unreported stromal-based mechanism. These findings indicate that the stroma may mediate the effects of iAs in tumor progression, which may have future therapeutic implications. CITATION Shearer JJ, Wold EA, Umbaugh CS, Lichti CF, Nilsson CL, Figueiredo ML. 2016. Inorganic arsenic-related changes in the stromal tumor microenvironment in a prostate cancer cell-conditioned media model. Environ Health Perspect 124:1009-1015; http://dx.doi.org/10.1289/ehp.1510090.
Collapse
Affiliation(s)
- Joseph J. Shearer
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Eric A. Wold
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Charles S. Umbaugh
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Cheryl F. Lichti
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Carol L. Nilsson
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Marxa L. Figueiredo
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
45
|
Brennen WN, Kisteman LN, Isaacs JT. Rapid selection of mesenchymal stem and progenitor cells in primary prostate stromal cultures. Prostate 2016; 76:552-64. [PMID: 26732992 PMCID: PMC4856028 DOI: 10.1002/pros.23145] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/15/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Carcinoma-associated fibroblasts (CAFs) are a dominant component of the tumor microenvironment with pro-tumorigenic properties. Despite this knowledge, their physiologic origins remain poorly understood. Mesenchymal stem cells (MSCs) can be recruited from the bone marrow to areas of tissue damage and inflammation, including prostate cancer. MSCs can generate and have many overlapping properties with CAFs in preclinical models. METHODS Multiparameter flow cytometry and multipotent differentiation assays used to define MSCs in primary prostate stromal cultures derived from young (<25 yrs) organ donors and prostate cancer patients compared with bone marrow-derived stromal cultures. Population doubling times, population doublings, cell size, and differentiation potential determined under multiple culture conditions, including normoxia, hypoxia, and a variety of media. TGF-β measured by ELISA. RESULTS MSCs and stromal progenitors are not only present in normal and malignant prostate tissue, but are quickly selected for in primary stromal cultures derived from these tissues; becoming the dominant population within just a few passages. Growth potential inversely associated with TGF-β concentrations. All conditions generated populations with an average cell diameter >15 µm. All cultures tested had the ability to undergo osteogenic and chondrogenic differentiation, but unlike bone marrow-derived MSCs, primary stromal cultures derived from normal prostate tissue lack adipogenic differentiation potential. In contrast, a subset of stromal cultures derived from prostate cancer patients retain the ability to differentiate into adipocytes; a property that is significantly suppressed under hypoxic conditions in both bone marrow- and prostate-derived MSCs. CONCLUSIONS Primary prostate stromal cultures are highly enriched in cells with an MSC or stromal progenitor phenotype. The use of primary cultures such as these to study CAFs raises interesting implications when considering their overlapping properties. The lack of adipogenesis in stromal cultures derived from normal prostates suggests they have a lineage-restricted progenitor phenotype. The retention of adipogenic differentiation in cultures from a subset of prostate cancer patients suggests the active recruitment of less committed progenitors or MSCs from the bone marrow as a function of disease progression. This recruitment can potentially be exploited for prognostic purposes or a cell-based platform for the systemic delivery of cytotoxic agents to sites of prostate cancer.
Collapse
Affiliation(s)
- W. Nathaniel Brennen
- Correspondence to: W. Nathaniel Brennen, Department of Oncology, Prostate Cancer Program, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, 1650 Orleans St., CRB-I, Rm 1M87, Baltimore, MD 21287.
| | | | | |
Collapse
|
46
|
Strand DW, Aaron L, Henry G, Franco OE, Hayward SW. Isolation and analysis of discreet human prostate cellular populations. Differentiation 2016; 91:139-51. [PMID: 26546040 PMCID: PMC4854811 DOI: 10.1016/j.diff.2015.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 10/27/2015] [Indexed: 02/03/2023]
Abstract
The use of lineage tracing in transgenic mouse models has revealed an abundance of subcellular phenotypes responsible for maintaining prostate homeostasis. The ability to use fresh human tissues to examine the hypotheses generated by these mouse experiments has been greatly enhanced by technical advances in tissue processing, flow cytometry and cell culture. We describe in detail the optimization of protocols for each of these areas to facilitate research on solving human prostate diseases through the analysis of human tissue.
Collapse
Affiliation(s)
- Douglas W Strand
- Department of Urology, UT Southwestern University Medical Center, Dallas, TX, USA
| | - LaTayia Aaron
- Department of Cancer Biology, Meharry Medical College, Nashville, TN, USA
| | - Gervaise Henry
- Department of Urology, UT Southwestern University Medical Center, Dallas, TX, USA
| | - Omar E Franco
- Department of Surgery, NorthShore University Health System, Research Institute, Evanston, IL, USA
| | - Simon W Hayward
- Department of Surgery, NorthShore University Health System, Research Institute, Evanston, IL, USA.
| |
Collapse
|
47
|
Levy O, Brennen WN, Han E, Rosen DM, Musabeyezu J, Safaee H, Ranganath S, Ngai J, Heinelt M, Milton Y, Wang H, Bhagchandani SH, Joshi N, Bhowmick N, Denmeade SR, Isaacs JT, Karp JM. A prodrug-doped cellular Trojan Horse for the potential treatment of prostate cancer. Biomaterials 2016; 91:140-150. [PMID: 27019026 DOI: 10.1016/j.biomaterials.2016.03.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/21/2016] [Accepted: 03/15/2016] [Indexed: 01/10/2023]
Abstract
Despite considerable advances in prostate cancer research, there is a major need for a systemic delivery platform that efficiently targets anti-cancer drugs to sites of disseminated prostate cancer while minimizing host toxicity. In this proof-of-principle study, human mesenchymal stem cells (MSCs) were loaded with poly(lactic-co-glycolic acid) (PLGA) microparticles (MPs) that encapsulate the macromolecule G114, a thapsigargin-based prostate specific antigen (PSA)-activated prodrug. G114-particles (∼950 nm in size) were internalized by MSCs, followed by the release of G114 as an intact prodrug from loaded cells. Moreover, G114 released from G114 MP-loaded MSCs selectively induced death of the PSA-secreting PCa cell line, LNCaP. Finally, G114 MP-loaded MSCs inhibited tumor growth when used in proof-of-concept co-inoculation studies with CWR22 PCa xenografts, suggesting that cell-based delivery of G114 did not compromise the potency of this pro-drug in-vitro or in-vivo. This study demonstrates a potentially promising approach to assemble a cell-based drug delivery platform, which inhibits cancer growth in-vivo without the need of genetic engineering. We envision that upon achieving efficient homing of systemically infused MSCs to cancer sites, this MSC-based platform may be developed into an effective, systemic 'Trojan Horse' therapy for targeted delivery of therapeutic agents to sites of metastatic PCa.
Collapse
Affiliation(s)
- Oren Levy
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - W Nathaniel Brennen
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, United States
| | - Edward Han
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - David Marc Rosen
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, United States
| | - Juliet Musabeyezu
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Helia Safaee
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Sudhir Ranganath
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Jessica Ngai
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Martina Heinelt
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Yuka Milton
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Hao Wang
- Department of Oncology, Division of Biostatistics at the Sidney Kimmel Comprehensive Cancer Center, United States
| | - Sachin H Bhagchandani
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Nitin Joshi
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States
| | - Neil Bhowmick
- The Samuel Oschin Comprehensive Cancer Institute at the Cedars-Sinai Medical Center, United States
| | - Samuel R Denmeade
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, United States.
| | - John T Isaacs
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, United States.
| | - Jeffrey M Karp
- Division of Biomedical Engineering, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - MIT Division of Health Sciences and Technology, United States.
| |
Collapse
|
48
|
Chowdhury R, Webber JP, Gurney M, Mason MD, Tabi Z, Clayton A. Cancer exosomes trigger mesenchymal stem cell differentiation into pro-angiogenic and pro-invasive myofibroblasts. Oncotarget 2015; 6:715-31. [PMID: 25596732 PMCID: PMC4359250 DOI: 10.18632/oncotarget.2711] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 11/11/2014] [Indexed: 12/25/2022] Open
Abstract
Stromal fibroblasts become altered in response to solid cancers, to exhibit myofibroblastic characteristics, with disease promoting influence. Infiltrating mesenchymal stem cells (MSC) may contribute towards these changes, but the factors secreted by cancer cells that impact MSC differentiation are poorly understood. We investigated the role of nano-metre sized vesicles (exosomes), secreted by prostate cancer cells, on the differentiation of bone-marrow MSC (BM-MSC), and the subsequent functional consequences of such changes. Purified exosomes impaired classical adipogenic differentiation, skewing differentiation towards alpha-smooth muscle actin (αSMA) positive myofibroblastic cells. A single exosomes treatment generated myofibroblasts secreting high levels of VEGF-A, HGF and matrix regulating factors (MMP-1, −3 and −13). Differentiated MSC had pro-angiogenic functions and enhanced tumour proliferation and invasivity assessed in a 3D co-culture model. Differentiation was dependent on exosomal-TGFβ, but soluble TGFβ at matched dose could not generate the same phenotype. Exosomes present in the cancer cell secretome were the principal factors driving this phenotype. Prostate cancer exosomes dominantly dictate a programme of MSC differentiation generating myofibroblasts with functional properties consistent with disease promotion.
Collapse
Affiliation(s)
- Ridwana Chowdhury
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Jason P Webber
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.,Cardiff Institute for Tissue Engineering and Repair, Cardiff University
| | - Mark Gurney
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Malcolm D Mason
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Zsuzsanna Tabi
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL
| | - Aled Clayton
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.Cardiff Institute for Tissue Engineering and Repair, Cardiff University.,Institute of Cancer and Genetics, School of Medicine, Cardiff University, Velindre Cancer Centre, Whitchurch, Cardiff, United Kingdom, CF14 2TL.Cardiff Institute for Tissue Engineering and Repair, Cardiff University
| |
Collapse
|
49
|
Yasmin-Karim S, King MR, Messing EM, Lee YF. E-selectin ligand-1 controls circulating prostate cancer cell rolling/adhesion and metastasis. Oncotarget 2015; 5:12097-110. [PMID: 25301730 PMCID: PMC4322988 DOI: 10.18632/oncotarget.2503] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 09/16/2014] [Indexed: 12/18/2022] Open
Abstract
Circulating prostate cancer (PCa) cells preferentially roll and adhere on bone marrow vascular endothelial cells, where abundant E-selectin and stromal cell-derived factor 1 (SDF-1) are expressed, subsequently initiating a cascade of activation events that eventually lead to the development of metastases. To elucidate the roles of circulating PCa cells' rolling and adhesion behaviors in cancer metastases, we applied a dynamic cylindrical flow-based microchannel device that is coated with E-selectin and SDF-1, mimicking capillary endothelium. Using this device we captured a small fraction of rolling PCa cells. These rolling cells display higher static adhesion ability, more aggressive cancer phenotypes and stem-like properties. Importantly, mice received rolling PCa cells, but not floating PCa cells, developed cancer metastases. Genes coding for E-selectin ligands and genes associated with cancer stem cells and metastasis were elevated in rolling PCa cells. Knock down of E-selectin ligand 1(ESL-1), significantly impaired PCa cells' rolling capacity and reduced cancer aggressiveness. Moreover, ESL-1 activates RAS and MAP kinase signal cascade, consequently inducing the downstream targets. In summary, circulating PCa cells' rolling capacity contributes to PCa metastasis, and that is in part controlled by ESL-1.
Collapse
Affiliation(s)
- Sayeda Yasmin-Karim
- Departments of Urology and Pathology and Laboratory Medicine, and Chemical Engineering, University of Rochester, Rochester, NY 14642
| | - Michael R King
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Edward M Messing
- Departments of Urology and Pathology and Laboratory Medicine, and Chemical Engineering, University of Rochester, Rochester, NY 14642
| | - Yi-Fen Lee
- Departments of Urology and Pathology and Laboratory Medicine, and Chemical Engineering, University of Rochester, Rochester, NY 14642
| |
Collapse
|
50
|
Montico F, Kido LA, San Martin R, Rowley DR, Cagnon VHA. Reactive stroma in the prostate during late life: The role of microvasculature and antiangiogenic therapy influences. Prostate 2015; 75:1643-61. [PMID: 26184673 DOI: 10.1002/pros.23045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/02/2015] [Indexed: 11/06/2022]
Abstract
BACKGROUND Prostate cancer is associated to a reactive stroma microenvironment characterized by angiogenic processes that are favorable for tumor progression. Senescence has been identified as a predisposing factor for prostate malignancies. In turn, the relationships between aging, reactive stroma, and the mechanisms that induce this phenotype are largely unknown. Thus, we investigated the occurrence of reactive stroma in the mouse prostate during advanced age as well as the effects of antiangiogenic and androgen ablation therapies on reactive stroma recruitment. METHODS Male mice (52-week-old FVB) were treated with two classes of angiogenesis inhibitors: direct (TNP-470; 15 mg/kg; s.c.) and/or indirect (SU5416; 6 mg/kg; i.p.). Androgen ablation was carried out by finasteride administration (20 mg/kg; s.c.), alone or in association to both inhibitors. The Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) model was used as a paradigm of cancer-associated reactive stroma. The dorsolateral prostate was collected for α-actin (αSMA), vimentin (VIM), and transforming growth factor-beta (TGF-β) immunohistochemical and Western blotting analyses as well as for CD34/αSMA and CD34/VIM colocalization. RESULTS Senescence was associated with increased αSMA, VIM, and TGF-β expression as well as with the recruitment of CD34/αSMA and CD34/VIM dual-positive fibroblasts. These observations were similar to those verified in TRAMP mice. Antiangiogenic treatment promoted the recovery of senescence-associated stromal changes. Hormonal ablation, despite having led to impaired CD34/αSMA and CD34/VIM dual-positive cell recruitment, did not result in decreased stimulus to reactive stroma development, due to enhanced TGF-β expression in relation to the aged controls. CONCLUSIONS Reactive stroma develops in the prostate of non-transgenic mice as a result of aging. The periacinar microvasculature is a candidate source for the recruitment of reactive stroma-associated cells, which may be derived either from perivascular-resident mesenchymal stem cells (MSCs) or from an endothelial-to-mesenchymal transition (EndMT) process. Thus, antiangiogenic therapy is a promising approach for preventing age-associated prostate malignancies by means of its negative interference in the development of reactive stroma phenotype from the vascular wall.
Collapse
Affiliation(s)
- Fabio Montico
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Larissa Akemi Kido
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Rebeca San Martin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - David R Rowley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Valéria H A Cagnon
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|