351
|
Bussard KM, Mutkus L, Stumpf K, Gomez-Manzano C, Marini FC. Tumor-associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res 2016; 18:84. [PMID: 27515302 PMCID: PMC4982339 DOI: 10.1186/s13058-016-0740-2] [Citation(s) in RCA: 556] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment is a heterogeneous population of cells consisting of the tumor bulk plus supporting cells. It is becoming increasingly evident that these supporting cells are recruited by cancer cells from nearby endogenous host stroma and promote events such as tumor angiogenesis, proliferation, invasion, and metastasis, as well as mediate mechanisms of therapeutic resistance. In addition, recruited stromal cells range in type and include vascular endothelial cells, pericytes, adipocytes, fibroblasts, and bone-marrow mesenchymal stromal cells. During normal wound healing and inflammatory processes, local stromal cells change their phenotype to become that of reactive stroma. Under certain conditions, however, tumor cells can co-opt these reactive stromal cells and further transition them into tumor-associated stromal cells (TASCs). These TASCs express higher levels of proteins, including alpha-smooth muscle actin, fibroblast activating protein, and matrix metalloproteinases, compared with their normal, non-reactive counterparts. TASCs are also known to secrete many pro-tumorigenic factors, including IL-6, IL-8, stromal-derived factor-1 alpha, vascular endothelial growth factor, tenascin-C, and matrix metalloproteinases, among others, which recruit additional tumor and pro-tumorigenic cells to the developing microenvironment. Here, we review the current literature pertaining to the origins of recruited host stroma, contributions toward tumor progression, tumor-associated stromal cells, and mechanisms of crosstalk between endogenous host stroma and tumor cells.
Collapse
Affiliation(s)
- Karen M Bussard
- Department of Cancer Biology, Wake Forest Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Lysette Mutkus
- Department of Regenerative Medicine, Wake Forest University, Winston-Salem, NC, 27157, USA
| | - Kristina Stumpf
- Department of Regenerative Medicine, Wake Forest University, Winston-Salem, NC, 27157, USA
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Frank C Marini
- Department of Cancer Biology, Wake Forest Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA. .,Department of Regenerative Medicine, Wake Forest University, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
352
|
Mezawa Y, Orimo A. The roles of tumor- and metastasis-promoting carcinoma-associated fibroblasts in human carcinomas. Cell Tissue Res 2016; 365:675-89. [PMID: 27506216 DOI: 10.1007/s00441-016-2471-1] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/04/2016] [Indexed: 12/11/2022]
Abstract
Carcinoma-associated fibroblasts (CAFs) constitute a substantial proportion of the non-neoplastic mesenchymal cell compartment in various human tumors. These fibroblasts are phenotypically converted from their progenitors via interactions with nearby cancer cells during the course of tumor progression. The resulting CAFs, in turn, support the growth and progression of carcinoma cells. These fibroblasts have a major influence on the hallmarks of carcinoma and promote tumor malignancy through the secretion of tumor-promoting growth factors, cytokines and exosomes, as well as through the remodeling of the extracellular matrix. Coevolution of CAFs and carcinoma cells during tumorigenesis is therefore essential for progression into fully malignant tumors. Recent studies have revealed the molecular mechanisms underlying CAF functions, especially in tumor invasion, metastasis and drug resistance and have highlighted the significant heterogeneity among these cells. In this review, we summarize the impacts of recently identified roles of tumor-promoting CAFs and discuss the therapeutic implications of targeting the heterotypic interactions of these fibroblasts with carcinoma cells. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Yoshihiro Mezawa
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, 113-8412, Japan
| | - Akira Orimo
- Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, 113-8412, Japan.
| |
Collapse
|
353
|
Valcz G, Galamb O, Krenács T, Spisák S, Kalmár A, Patai ÁV, Wichmann B, Dede K, Tulassay Z, Molnár B. Exosomes in colorectal carcinoma formation: ALIX under the magnifying glass. Mod Pathol 2016; 29:928-38. [PMID: 27150162 DOI: 10.1038/modpathol.2016.72] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 02/07/2023]
Abstract
Exosomes are small membrane vesicles that have important roles in transporting a great variety of bioactive molecules between epithelial compartment and their microenvironment during tumor formation including colorectal adenoma-carcinoma sequence. We tested the mRNA expression of the top 25 exosome-related markers based on ExoCharta database in healthy (n=49), adenoma (n=49) and colorectal carcinoma (n=49) patients using Affymetrix HGU133 Plus2.0 microarrays. Most related genes showed significantly elevated expression including PGK1, PKM, ANXA5, ENO1, HSP90AB1 and MSN during adenoma-carcinoma sequence. Surprisingly, the expression of ALIX (ALG 2-interacting protein X), involved in multivesicular body (MVB) and exosome formation, was significantly reduced in normal vs adenoma (P=5.02 × 10(-13)) and in normal vs colorectal carcinoma comparisons (P=1.51 × 10(-10)). ALIX also showed significant reduction (P<0.05) at the in situ protein level in the epithelial compartment of adenoma (n=35) and colorectal carcinoma (n=37) patients compared with 27 healthy individuals. Furthermore, significantly reduced ALIX protein levels were accompanied by their gradual transition from diffuse cytoplasmic expression to granular signals, which fell into the 0.6-2 μm diameter size range of MVBs. These ALIX-positive particles were seen in the tumor nests, including tumor-stroma border, which suggest their exosome function. MVB-like structures were also detected in tumor microenvironment including α-smooth muscle actin-positive stromal cells, budding off cancer cells in the tumor front as well as in cancer cells entrapped within lymphoid vessels. In conclusion, we determined the top aberrantly expressed exosome-associated markers and revealed the transition of diffuse ALIX protein signals into a MVB-like pattern during adenoma-carcinoma sequence. These tumor-associated particles seen both in the carcinoma and the surrounding microenvironment can potentially mediate epithelial-stromal interactions involved in the regulation of tumor growth, metastatic invasion and therapy response.
Collapse
Affiliation(s)
- Gábor Valcz
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Orsolya Galamb
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Tibor Krenács
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University and MTA-SE Tumor Progression Research Group, Budapest, Hungary
| | - Sándor Spisák
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexandra Kalmár
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Árpád V Patai
- 2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Barna Wichmann
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary
| | - Kristóf Dede
- Department of General Surgery and Surgical Oncology, Uzsoki Teaching Hospital, Budapest, Hungary
| | - Zsolt Tulassay
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary.,2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Béla Molnár
- Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary.,2nd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
354
|
Rovira J, Diekmann F, Campistol JM, Ramírez-Bajo MJ. Therapeutic application of extracellular vesicles in acute and chronic renal injury. Nefrologia 2016; 37:126-137. [PMID: 27462016 DOI: 10.1016/j.nefro.2016.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/14/2016] [Accepted: 04/28/2016] [Indexed: 12/31/2022] Open
Abstract
A new cell-to-cell communication system was discovered in the 1990s, which involves the release of vesicles into the extracellular space. These vesicles shuttle bioactive particles, including proteins, mRNA, miRNA, metabolites, etc. This particular communication has been conserved throughout evolution, which explains why most cell types are capable of producing vesicles. Extracellular vesicles (EVs) are involved in the regulation of different physiological processes, as well as in the development and progression of several diseases. EVs have been widely studied over recent years, especially those produced by embryonic and adult stem cells, blood cells, immune system and nervous system cells, as well as tumour cells. EV analysis from bodily fluids has been used as a diagnostic tool for cancer and recently for different renal diseases. However, this review analyses the importance of EVs generated by stem cells, their function and possible clinical application in renal diseases and kidney transplantation.
Collapse
Affiliation(s)
- Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Centre de Recerca Biomèdica CELLEX, Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, España; Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España
| | - Fritz Diekmann
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Centre de Recerca Biomèdica CELLEX, Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, España; Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España; Departamento de Nefrología y Trasplante Renal, Institut Clínic de Nefrologia i Urologia (ICNU), Hospital Clínic, Barcelona, España.
| | - Josep M Campistol
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Centre de Recerca Biomèdica CELLEX, Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, España; Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, España; Departamento de Nefrología y Trasplante Renal, Institut Clínic de Nefrologia i Urologia (ICNU), Hospital Clínic, Barcelona, España
| | - María José Ramírez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Centre de Recerca Biomèdica CELLEX, Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, España
| |
Collapse
|
355
|
Metabolic reprogramming of bone marrow stromal cells by leukemic extracellular vesicles in acute lymphoblastic leukemia. Blood 2016; 128:453-6. [DOI: 10.1182/blood-2015-12-688051] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
356
|
Liu CM, Hsieh CL, Shen CN, Lin CC, Shigemura K, Sung SY. Exosomes from the tumor microenvironment as reciprocal regulators that enhance prostate cancer progression. Int J Urol 2016; 23:734-44. [PMID: 27397852 DOI: 10.1111/iju.13145] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/22/2016] [Indexed: 12/21/2022]
Abstract
Distant organ metastasis of prostate cancer is a puzzle, and various theories have successively arisen to explain the mechanism of lethal cancer progression. While perhaps agreeable to many cancer biologists, the very statement of "seed and soil" proposed by Stephan Paget in 1881 is arguably still the major statement for organ-specific cancer metastasis. Since recent studies showed important correlations of regulation of cancer cells and the microenvironment, exosomes from cancer and stromal cells seem to create another important niche for metastasis. Stromal cells pretreated with exosomes from metastatic cancer cells increase the potential of change stromal cells. The poorly metastatic cancer cells could also enhance malignancy through transfer of proteins, microribonucleic acid and messenger ribonucleic acid to recipient cancer cells. Herein, we reviewed extracellular exosomes as a factor involved in cross-talk between stromal and prostate cancer epithelial cells.
Collapse
Affiliation(s)
- Che-Ming Liu
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan.,The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Cheng-Chieh Lin
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Katsumi Shigemura
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shian-Ying Sung
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
357
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
358
|
Wendler F, Stamp GW, Giamas G. Tumor-Stromal Cell Communication: Small Vesicles Signal Big Changes. Trends Cancer 2016; 2:326-329. [PMID: 28741534 DOI: 10.1016/j.trecan.2016.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/16/2016] [Accepted: 05/24/2016] [Indexed: 12/16/2022]
Abstract
Reciprocal interactions between malignant and stromal cells create a local microenvironment that fosters tumor growth. Extracellular vesicles (EVs) such as exosomes, microvesicles, and large oncosomes are involved in tumor-stroma communication by shuttling signaling cargo and other molecules. Here we discuss how EVs released by cancer or stromal cells impact the proliferation, differentiation, and metabolism of tumors.
Collapse
Affiliation(s)
- Franz Wendler
- University of Sussex, School of Life Sciences, John Maynard Smith Building, Falmer, Brighton BN1 9QG, UK.
| | - Gordon W Stamp
- Division of Diabetes, Endocrinology, and Metabolism, Imperial College Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Georgios Giamas
- University of Sussex, School of Life Sciences, John Maynard Smith Building, Falmer, Brighton BN1 9QG, UK.
| |
Collapse
|
359
|
Prakash J. Cancer-Associated Fibroblasts: Perspectives in Cancer Therapy. Trends Cancer 2016; 2:277-279. [DOI: 10.1016/j.trecan.2016.04.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/23/2016] [Accepted: 04/15/2016] [Indexed: 01/08/2023]
|
360
|
Syn N, Wang L, Sethi G, Thiery JP, Goh BC. Exosome-Mediated Metastasis: From Epithelial-Mesenchymal Transition to Escape from Immunosurveillance. Trends Pharmacol Sci 2016; 37:606-617. [PMID: 27157716 DOI: 10.1016/j.tips.2016.04.006] [Citation(s) in RCA: 374] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/08/2016] [Accepted: 04/11/2016] [Indexed: 01/02/2023]
Abstract
Exosomes are extracellular signalosomes that facilitate eukaryotic intercellular communication under a wide range of normal physiological contexts. In malignancies, this regulatory circuit is co-opted to promote cancer cell survival and outgrowth. Tumour-derived exosomes (TDEs) carry a pro-EMT (epithelial-mesenchymal transition) programme including transforming growth factor beta (TGFβ), caveolin-1, hypoxia-inducible factor 1 alpha (HIF1α), and β-catenin that enhances the invasive and migratory capabilities of recipient cells, and contributes to stromal remodelling and premetastatic niche formation. The integrin expression patterns on TDEs appear to dictate their preferential uptake by organ-specific cells, implying a crucial role of this pathway in organotropic metastasis. Through the expression of immunomodulatory molecules such as CD39 and CD73, TDEs modify the immune contexture of the tumour microenvironment, which could have implications for immunotherapy. Hence, targeting TDE dysregulation pathways, such as the heparanase/syndecan-1 axis, could represent novel therapeutic strategies in the quest to conquer cancer.
Collapse
Affiliation(s)
- Nicholas Syn
- Cancer Science Institute of Singapore, Centre for Translational Medicine, National University of Singapore, 14 Medical Drive, #12-01, Singapore 117599, Singapore; Department of Haematology-Oncology, National University Cancer Institute, 1E Kent Ridge Road, NUHS Tower Block, Level 7, Singapore 119228, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, Centre for Translational Medicine, National University of Singapore, 14 Medical Drive, #12-01, Singapore 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Jean-Paul Thiery
- Cancer Science Institute of Singapore, Centre for Translational Medicine, National University of Singapore, 14 Medical Drive, #12-01, Singapore 117599, Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; UMR 7057 Matter and Complex Systems University Paris Denis Diderot, Paris, France; Comprehensive Cancer Center Institut Gustave Roussy, Villejuif, France
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, Centre for Translational Medicine, National University of Singapore, 14 Medical Drive, #12-01, Singapore 117599, Singapore; Department of Haematology-Oncology, National University Cancer Institute, 1E Kent Ridge Road, NUHS Tower Block, Level 7, Singapore 119228, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| |
Collapse
|
361
|
Extracellular vesicles released by CD40/IL-4-stimulated CLL cells confer altered functional properties to CD4+ T cells. Blood 2016; 128:542-52. [PMID: 27118451 DOI: 10.1182/blood-2015-11-682377] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 04/20/2016] [Indexed: 01/02/2023] Open
Abstract
The complex interplay between cancer cells, stromal cells, and immune cells in the tumor microenvironment (TME) regulates tumorigenesis and provides emerging targets for immunotherapies. Crosstalk between CD4(+) T cells and proliferating chronic lymphocytic leukemia (CLL) tumor B cells occurs within lymphoid tissue pseudofollicles, and investigating these interactions is essential to understand both disease pathogenesis and the effects of immunotherapy. Tumor-derived extracellular vesicle (EV) shedding is emerging as an important mode of intercellular communication in the TME. In order to characterize tumor EVs released in response to T-cell-derived TME signals, we performed microRNA (miRNA [miR]) profiling of EVs released from CLL cells stimulated with CD40 and interleukin-4 (IL-4). Our results reveal an enrichment of specific cellular miRNAs including miR-363 within EVs derived from CD40/IL-4-stimulated CLL cells compared with parental cell miRNA content and control EVs from unstimulated CLL cells. We demonstrate that autologous patient CD4(+) T cells internalize CLL-EVs containing miR-363 that targets the immunomodulatory molecule CD69. We further reveal that autologous CD4(+) T cells that are exposed to EVs from CD40/IL-4-stimulated CLL cells exhibit enhanced migration, immunological synapse signaling, and interactions with tumor cells. Knockdown of miR-363 in CLL cells prior to CD40/IL-4 stimulation prevented the ability of CLL-EVs to induce increased synapse signaling and confer altered functional properties to CD4(+) T cells. Taken together, these data reveal a novel role for CLL-EVs in modifying T-cell function that highlights unanticipated complexity of intercellular communication that may have implications for bidirectional CD4(+) T-cell:tumor interactions within the TME.
Collapse
|
362
|
Li X, Wang S, Zhu R, Li H, Han Q, Zhao RC. Lung tumor exosomes induce a pro-inflammatory phenotype in mesenchymal stem cells via NFκB-TLR signaling pathway. J Hematol Oncol 2016; 9:42. [PMID: 27090786 PMCID: PMC4836087 DOI: 10.1186/s13045-016-0269-y] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023] Open
Abstract
Background In tumor microenvironment, a continuous cross-talk between cancer cells and other cellular components is required to sustain tumor progression. Accumulating evidence suggests that exosomes, a novel way of cell communication, play an important role in such cross-talk. Exosomes could facilitate the direct intercellular transfer of proteins, lipids, and miRNA/mRNA/DNAs between cells. Since mesenchymal stem cells (MSCs) can be attracted to tumor sites and become an important component of the tumor microenvironment, there is an urgent need to reveal the effect of tumor exosomes on MSCs and to further explore the underlying molecular mechanisms. Methods Exosomes were harvested from lung cancer cell line A549 and added to MSCs. Secretion of inflammation-associated cytokines in exosome-treated MSCs were analyzed by RT-PCR and ELISA. The growth-promoting effect of exosome-treated MSCs on lung tumor cells was evaluated by in vivo mouse xenograft model. Signaling pathway involved in exosomes-treated MSCs was detected by PCR array of human toll-like receptor signaling pathway, RT-PCR, and Western blot. Results Data showed that lung tumor cell A549-derived exosomes could induce a pro-inflammatory phenotype in MSCs named P-MSCs, which have significantly elevated secretion of IL-6, IL-8, and MCP-1. P-MSCs possess a greatly enhanced ability in promoting lung tumor growth in mouse xenograft model. Analysis of the signaling pathways in P-MSCs revealed a fast triggering of NF-κB. Genetic ablation of Toll-like receptor 2 (TLR2) by siRNA and TLR2-neutralizing antibody could block NF-κB activation by exosomes. We further found that Hsp70 present on the surface of lung tumor exosomes contributed to the induction of P-MSCs by A549 exosomes. Conclusions Our studies suggest a novel mechanism by which lung tumor cell-derived exosomes induce pro-inflammatory activity of MSCs which in turn get tumor supportive characteristics. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0269-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoxia Li
- Center of Excellence in Tissue Engineering, Key Laboratory of Beijing, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shihua Wang
- Center of Excellence in Tissue Engineering, Key Laboratory of Beijing, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Rongjia Zhu
- Center of Excellence in Tissue Engineering, Key Laboratory of Beijing, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Hongling Li
- Center of Excellence in Tissue Engineering, Key Laboratory of Beijing, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qin Han
- Center of Excellence in Tissue Engineering, Key Laboratory of Beijing, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| | - Robert Chunhua Zhao
- Center of Excellence in Tissue Engineering, Key Laboratory of Beijing, Institute of Basic Medical Sciences and School of Basic Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China. .,Center of Translational medicine Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
363
|
Belov L, Matic KJ, Hallal S, Best OG, Mulligan SP, Christopherson RI. Extensive surface protein profiles of extracellular vesicles from cancer cells may provide diagnostic signatures from blood samples. J Extracell Vesicles 2016; 5:25355. [PMID: 27086589 PMCID: PMC4834364 DOI: 10.3402/jev.v5.25355] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 02/25/2016] [Accepted: 03/15/2016] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EV) are membranous particles (30–1,000 nm in diameter) secreted by cells. Important biological functions have been attributed to 2 subsets of EV, the exosomes (bud from endosomal membranes) and the microvesicles (MV; bud from plasma membranes). Since both types of particles contain surface proteins derived from their cell of origin, their detection in blood may enable diagnosis and prognosis of disease. We have used an antibody microarray (DotScan) to compare the surface protein profiles of live cancer cells with those of their EV, based on their binding patterns to immobilized antibodies. Initially, EV derived from the cancer cell lines, LIM1215 (colorectal cancer) and MEC1 (B-cell chronic lymphocytic leukaemia; CLL), were used for assay optimization. Biotinylated antibodies specific for EpCAM (CD326) and CD19, respectively, were used to detect captured particles by enhanced chemiluminescence. Subsequently, this approach was used to profile CD19+ EV from the plasma of CLL patients. These EV expressed a subset (~40%) of the proteins detected on CLL cells from the same patients: moderate or high levels of CD5, CD19, CD31, CD44, CD55, CD62L, CD82, HLA-A,B,C, HLA-DR; low levels of CD21, CD49c, CD63. None of these proteins was detected on EV from the plasma of age- and gender-matched healthy individuals.
Collapse
Affiliation(s)
- Larissa Belov
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia;
| | - Kieran J Matic
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Susannah Hallal
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - O Giles Best
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Stephen P Mulligan
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | | |
Collapse
|
364
|
Wang J, De Veirman K, Faict S, Frassanito MA, Ribatti D, Vacca A, Menu E. Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression. J Pathol 2016; 239:162-73. [PMID: 26956697 DOI: 10.1002/path.4712] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/03/2016] [Accepted: 02/29/2016] [Indexed: 12/21/2022]
Abstract
Multiple myeloma (MM) pathogenesis and progression largely rely on the cells and extracellular factors in the bone marrow (BM) microenvironment. Compelling studies have identified tumour exosomes as key regulators in the maintenance and education of the BM microenvironment by targeting stromal cells, immune cells, and vascular cells. However, the role of MM exosomes in the modification of the BM microenvironment and MM progression remains unclear. Here, we explored the functions of MM exosomes in angiogenesis and immunosuppression in vitro and in vivo. Murine MM exosomes carrying multiple angiogenesis-related proteins enhanced angiogenesis and directly promoted endothelial cell growth. Several pathways such as signal transducer and activator of transcription 3 (STAT3), c-Jun N-terminal kinase, and p53 were modulated by the exosomes in endothelial and BM stromal cells. These exosomes promoted the growth of myeloid-derived suppressor cells (MDSCs) in naive mice through activation of the STAT3 pathway and changed their subsets to similar phenotypes to those seen in MM-bearing mice. Moreover, MM exosomes up-regulated inducible nitric oxide synthase and enhanced the immunosuppressive capacity of BM MDSCs in vivo. Our data show that MM exosomes modulate the BM microenvironment through enhancement of angiogenesis and immunosuppression, which will further facilitate MM progression. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jinheng Wang
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Sylvia Faict
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Maria Antonia Frassanito
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.,National Cancer Institute 'Giovanni Paolo II', Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| |
Collapse
|
365
|
Hall J, Prabhakar S, Balaj L, Lai CP, Cerione RA, Breakefield XO. Delivery of Therapeutic Proteins via Extracellular Vesicles: Review and Potential Treatments for Parkinson's Disease, Glioma, and Schwannoma. Cell Mol Neurobiol 2016; 36:417-27. [PMID: 27017608 PMCID: PMC4860146 DOI: 10.1007/s10571-015-0309-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 11/20/2015] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles present an attractive delivery vehicle for therapeutic proteins. They intrinsically contain many proteins which can provide information to other cells. Advantages include reduced immune reactivity, especially if derived from the same host, stability in biologic fluids, and ability to target uptake. Those from mesenchymal stem cells appear to be intrinsically therapeutic, while those from cancer cells promote tumor progression. Therapeutic proteins can be loaded into vesicles by overexpression in the donor cell, with oligomerization and membrane sequences increasing their loading. Examples of protein delivery for therapeutic benefit in pre-clinical models include delivery of: catalase for Parkinson's disease to reduce oxidative stress and thus help neurons to survive; prodrug activating enzymes which can convert a prodrug which crosses the blood-brain barrier into a toxic chemotherapeutic drug for schwannomas and gliomas; and the apoptosis-inducing enzyme, caspase-1 under a Schwann cell specific promoter for schwannoma. This therapeutic delivery strategy is novel and being explored for a number of diseases.
Collapse
Affiliation(s)
- Justin Hall
- Departments of Chemistry and Chemical Biology and Molecular Medicine, Cornell University, Ithaca, NY, 14853, USA
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, 02114, USA
| | - Shilpa Prabhakar
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, 02114, USA
| | - Leonora Balaj
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, 02114, USA
| | - Charles P Lai
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, 02114, USA
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Richard A Cerione
- Departments of Chemistry and Chemical Biology and Molecular Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Xandra O Breakefield
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, 02114, USA.
- Molecular Neurogenetics Unit, Massachusetts General Hospital-East, 13th Street, Building 149, Charlestown, MA, 02129, USA.
| |
Collapse
|
366
|
Abstract
Understanding of the lymphoma tumor microenvironment is poised to expand in the era of next-generation sequencing studies of the tumor cells themselves. Successful therapies of the future will rely on deeper appreciation of the interactions between elements of the microenvironment. Although the phenotypic, cytogenetic, and molecular characterization of tumor cells in lymphomas has progressed faster than most other solid organ tumors, concrete advancements in understanding the lymphoma microenvironment have been fewer. This article explores the composition of the lymphoma tumor microenvironment; its role in immune surveillance, evasion, and drug resistance; and its potential role in the development of targeted therapies.
Collapse
Affiliation(s)
- Mina L Xu
- Department of Pathology & Laboratory Medicine, Yale University School of Medicine, 310 Cedar Street, PO Box 208023, New Haven, CT 06520-8023, USA.
| | - Yuri Fedoriw
- University of North Carolina School of Medicine, Department of Pathology and Laboratory Medicine, NC Cancer Hospital C3162-D, 101 Manning Drive, Chapel Hill, NC 27599, USA
| |
Collapse
|
367
|
Abstract
B-cell chronic lymphocytic leukemia (CLL) is the most common adult human leukemia. Although, the molecular alterations leading to CLL onset and progression are still under investigation (specifically, the interplay and exact role of oncogenes and tumor suppressors in CLL pathogenesis). MicroRNAs are small non-coding RNAs that regulate gene expression and are expressed in a tissue specific manner. Deregulation of microRNAs can alter expression levels of genes involved in the development and/or progression of tumors. In CLL, microRNAs can function as oncogenes or tumor suppressors. Here, we review the most recent findings on the role of microRNAs in the onset/progression of CLL, and how this knowledge can be used to identify new biomarkers and targets to treat this leukemia.
Collapse
Affiliation(s)
- Veronica Balatti
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Mario Acunzo
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Yuri Pekarky
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Carlo M Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Comprehensive Cancer Center, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
368
|
Tumor microenvironment (TME)-driven immune suppression in B cell malignancy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:471-482. [DOI: 10.1016/j.bbamcr.2015.11.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/27/2015] [Accepted: 11/04/2015] [Indexed: 12/29/2022]
|
369
|
Choi MY, Kashyap MK, Kumar D. The chronic lymphocytic leukemia microenvironment: Beyond the B-cell receptor. Best Pract Res Clin Haematol 2016; 29:40-53. [PMID: 27742071 DOI: 10.1016/j.beha.2016.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 07/03/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023]
Abstract
Malignant B cells accumulate in the peripheral blood, bone marrow, and lymphoid organs of patients with chronic lymphocytic leukemia (CLL). In the tissue compartments, CLL shape a protective microenvironment by coopting normal elements. The efficacy of drugs that target these interactions further underscores their importance in the pathogenesis of CLL. While the B cell receptor (BCR) pathway clearly plays a central role in the CLL microenvironment, there is also rationale to evaluate agents that inhibit other aspects or modulate the immune cells in the microenvironment. Here we review the main cellular components, soluble factors, and signaling pathways of the CLL microenvironment, and highlight recent clinical advances. As the BCR pathway is reviewed elsewhere, we focus on other aspects of the microenvironment.
Collapse
Affiliation(s)
- Michael Y Choi
- Moores Cancer Center, UCSD-Moores Cancer Center, La Jolla, 92093-0820, CA, USA.
| | - Manoj Kumar Kashyap
- Moores Cancer Center, UCSD-Moores Cancer Center, La Jolla, 92093-0820, CA, USA.
| | - Deepak Kumar
- Moores Cancer Center, UCSD-Moores Cancer Center, La Jolla, 92093-0820, CA, USA.
| |
Collapse
|
370
|
Wang L, Gao CJ. [Role of extracellular vesicles in hematological malignancies]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2016; 37:258-261. [PMID: 27033771 PMCID: PMC7342940 DOI: 10.3760/cma.j.issn.0253-2727.2016.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Indexed: 06/05/2023]
Affiliation(s)
| | - C J Gao
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
371
|
Jeong S, Park J, Pathania D, Castro CM, Weissleder R, Lee H. Integrated Magneto-Electrochemical Sensor for Exosome Analysis. ACS NANO 2016; 10:1802-9. [PMID: 26808216 PMCID: PMC4802494 DOI: 10.1021/acsnano.5b07584] [Citation(s) in RCA: 335] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Extracellular vesicles, including exosomes, are nanoscale membrane particles that carry molecular information on parental cells. They are being pursued as biomarkers of cancers that are difficult to detect or serially follow. Here we present a compact sensor technology for rapid, on-site exosome screening. The sensor is based on an integrated magneto-electrochemical assay: exosomes are immunomagnetically captured from patient samples and profiled through electrochemical reaction. By combining magnetic enrichment and enzymatic amplification, the approach enables (i) highly sensitive, cell-specific exosome detection and (ii) sensor miniaturization and scale-up for high-throughput measurements. As a proof-of-concept, we implemented a portable, eight-channel device and applied it to screen extracellular vesicles in plasma samples from ovarian cancer patients. The sensor allowed for the simultaneous profiling of multiple protein markers within an hour, outperforming conventional methods in assay sensitivity and speed.
Collapse
Affiliation(s)
- Sangmoo Jeong
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
| | - Jongmin Park
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
| | - Divya Pathania
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
| | - Cesar M. Castro
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
- Massachusetts General Hospital Cancer Center, Boston, MA 02114
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
- Massachusetts General Hospital Cancer Center, Boston, MA 02114
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114
- Corresponding author: H. Lee, PhD, Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114, 617-726-8226,
| |
Collapse
|
372
|
Harp D, Driss A, Mehrabi S, Chowdhury I, Xu W, Liu D, Garcia-Barrio M, Taylor RN, Gold B, Jefferson S, Sidell N, Thompson W. Exosomes derived from endometriotic stromal cells have enhanced angiogenic effects in vitro. Cell Tissue Res 2016; 365:187-96. [PMID: 26841879 PMCID: PMC4917586 DOI: 10.1007/s00441-016-2358-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/30/2015] [Indexed: 02/07/2023]
Abstract
Our objective has been to establish a pro-angiogenic role for exosomes in endometriosis and to determine whether a differential expression profile of cellular and exosomal microRNAs (miRNAs) exists in endometriosis. We performed an in vitro study of human primary endometrial stromal cells (ESCs) and human umbilical vein endothelial cells (HUVECs). We isolated and characterized exosomes from ESCs from five endometriosis patients and five phase-matched controls. Exosomes were characterized by transmission electron microscopy and NanoSight technology. MiRNA was assessed by deep sequencing and reverse transcription with quantitative polymerase chain reaction. Exosome uptake studies were achieved by means of confocal microscopy. The pro-angiogenic experiments were executed by treating HUVECs with ESC-derived exosomes. We observed differential profiles of exosomal miRNA expression between exosomes derived from endometriosis lesion cells and diseased eutopic stromal cells compared with exosomes derived from control ESCs. We also demonstrated autocrine cellular uptake of exosomes and paracrine functional angiogenic effects of exosomes on HUVECs. The results of this study support the hypothesis that exosomes derived from ESCs play autocrine/paracrine roles in the development of endometriosis, potentially modulating angiogenesis. The broader clinical implications are that Sampson’s theory of retrograde menstruation possibly encompasses the finding that exosomes work as intercellular communication modulators in endometriosis.
Collapse
Affiliation(s)
- Djana Harp
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA.
| | - Adel Driss
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA
| | - Sharifeh Mehrabi
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA
| | - Indrajit Chowdhury
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA
| | - Wei Xu
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA
| | - Dong Liu
- Cardiovascular Research Institute, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA.,Department of Physiology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA
| | - Minerva Garcia-Barrio
- Cardiovascular Research Institute, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA.,Department of Physiology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Bert Gold
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | | | - Neil Sidell
- Department of Gynecology & Obstetrics, Emory University School of Medicine, 1639 Pierce Dr., WMB 4303, Atlanta, GA 30322, USA
| | - Winston Thompson
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA.,Department of Physiology, Morehouse School of Medicine, 720 Westview Drive, SW, Atlanta, GA 30310, USA
| |
Collapse
|
373
|
Tissot T, Ujvari B, Solary E, Lassus P, Roche B, Thomas F. Do cell-autonomous and non-cell-autonomous effects drive the structure of tumor ecosystems? Biochim Biophys Acta Rev Cancer 2016; 1865:147-54. [PMID: 26845682 DOI: 10.1016/j.bbcan.2016.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/28/2016] [Accepted: 01/30/2016] [Indexed: 12/21/2022]
Abstract
By definition, a driver mutation confers a growth advantage to the cancer cell in which it occurs, while a passenger mutation does not: the former is usually considered as the engine of cancer progression, while the latter is not. Actually, the effects of a given mutation depend on the genetic background of the cell in which it appears, thus can differ in the subclones that form a tumor. In addition to cell-autonomous effects generated by the mutations, non-cell-autonomous effects shape the phenotype of a cancer cell. Here, we review the evidence that a network of biological interactions between subclones drives cancer cell adaptation and amplifies intra-tumor heterogeneity. Integrating the role of mutations in tumor ecosystems generates innovative strategies targeting the tumor ecosystem's weaknesses to improve cancer treatment.
Collapse
Affiliation(s)
- Tazzio Tissot
- CREEC/MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France.
| | - Beata Ujvari
- Centre for Integrative Ecology, School of Life and Environmental Sciences, Deakin University, Waurn Ponds, Australia
| | - Eric Solary
- INSERM U1170, Gustave Roussy, 94805 Villejuif, France; University Paris-Saclay, Faculty of Medicine, 94270 Le Kremlin-Bicêtre, France
| | - Patrice Lassus
- CNRS, UMR 5535, Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Montpellier, France
| | - Benjamin Roche
- CREEC/MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France; Unité mixte internationale de Modélisation Mathématique et Informatique des Systèmes Complexes (UMI IRD/UPMC UMMISCO), 32 Avenue Henri Varagnat, 93143 Bondy Cedex, France
| | - Frédéric Thomas
- CREEC/MIVEGEC, UMR IRD/CNRS/UM 5290, 911 Avenue Agropolis, BP 64501, 34394 Montpellier Cedex 5, France
| |
Collapse
|
374
|
Esparza-Baquer A, Labiano I, Bujanda L, Perugorria MJ, Banales JM. MicroRNAs in cholangiopathies: Potential diagnostic and therapeutic tools. Clin Res Hepatol Gastroenterol 2016; 40:15-27. [PMID: 26774196 DOI: 10.1016/j.clinre.2015.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/23/2015] [Accepted: 10/02/2015] [Indexed: 02/07/2023]
Abstract
Cholangiopathies are the group of diseases targeting the bile duct epithelial cells (i.e. cholangiocytes). These disorders arise from different etiologies and represent a current diagnostic, prognostic and therapeutic challenge. Different molecular mechanisms participate in the development and progression of each type of biliary disease. However, microRNA deregulation is a common central event occurring in all of them that plays a key role in their pathogenesis. MicroRNAs are highly stable small non-coding RNAs present in cells, extracellular microvesicles and biofluids, representing valuable diagnostic tools and potential targets for therapy. In the following sections, the most novel and significant discoveries in this field are summarized and their potential clinical value is highlighted.
Collapse
Affiliation(s)
- Aitor Esparza-Baquer
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastián, Spain
| | - Ibone Labiano
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastián, Spain
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastián, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain
| | - María J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastián, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesús M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastián, Spain; National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
375
|
Pocsfalvi G, Stanly C, Fiume I, Vékey K. Chromatography and its hyphenation to mass spectrometry for extracellular vesicle analysis. J Chromatogr A 2016; 1439:26-41. [PMID: 26830636 DOI: 10.1016/j.chroma.2016.01.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs), such as exosomes, microvesicles and apoptotic bodies are released by cells, both under physiological and pathological conditions. EVs can participate in a novel type of intercellular communication and deliver cargo of nucleic acids, proteins and lipids near or to distant host cells. EV research is proceeding at a fast pace; now they start to appear as promising therapeutic targets, diagnostic tools and drug delivery systems. Isolation and analysis of EVs are prerequisites for understanding their biological roles and for their clinical exploitation. In this process chromatography and mass spectrometry (MS)-based strategies are rapidly gaining importance; and are reviewed in the present communication. Isolation and purification of EVs is mostly performed by ultracentrifugation at present. Chromatography-based strategies are gaining ground, among which affinity and size exclusion chromatography (SEC) are particularly strong contenders. Their major advantages are the relative simplicity, robustness and throughput. Affinity chromatography has the added advantage of separating EV subtypes based on molecular recognition of EV surface motifs. SEC has the advantage that isolated EVs may retain their biological activity. EVs are typically isolated in small amounts, therefore high sensitivity is required for their analysis. Study of the molecular content of EVs (all compounds beside nucleic acids) is predominantly based on liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis. The chromatographic separation is mostly performed by reverse phase, nanoscale, ultra high performance LC technique. The MS analysis relying typically on nano-electrospray ionization MS/MS provides high sensitivity, selectivity and resolution, so that thousand(s) of proteins can be detected/identified/quantified in a EV sample. Beside protein identification, quantitation and characterization of protein post-translational modifications (PTMs), like glycosylation and phosphorylation are becoming feasible and increasingly important. Along with conventional LC-MS/MS, other chromatographic approaches hyphenated to MS are gaining importance for EV characterization. Hydrophilic interaction LC is used to characterize PTMs; LC-inductively coupled plasma/MS to identify metal containing molecules; while gas chromatography-MS to analyze some lipids and metabolites.
Collapse
Affiliation(s)
- Gabriella Pocsfalvi
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy.
| | - Christopher Stanly
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy
| | - Immacolata Fiume
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy
| | - Károly Vékey
- Mass Spectrometry Proteomics Group, Institute of Organic Chemistry, Research Centre for Natural Sciences of the Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
376
|
Sorokina T, Shipounova I, Bigildeev A, Petinati N, Drize N, Turkina A, Chelysheva E, Shukhov O, Kuzmina L, Parovichnikova E, Savchenko V. The ability of multipotent mesenchymal stromal cells from the bone marrow of patients with leukemia to maintain normal hematopoietic progenitor cells. Eur J Haematol 2016; 97:245-52. [PMID: 26643284 DOI: 10.1111/ejh.12713] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND The development of leukemia impairs normal hematopoiesis and marrow stromal microenvironment. The aim of the investigation was to study the ability of multipotent mesenchymal stromal cells (MSCs) derived from the bone marrow of patients with leukemia to maintain normal hematopoietic progenitor cells. METHODS MSCs were obtained from the bone marrow of 14 patients with acute lymphoblastic (ALL), 25 with myeloid (AML), and 15 with chronic myeloid (CML) leukemia. As a control, MSCs from 22 healthy donors were used. The incidence of cobblestone area forming cells (CAFC 7-8 d) in the bone marrow of healthy donor cultivated on the supportive layer of patients MSCs was measured. RESULTS The ability of MSCs from AML and ALL patients at the moment of diagnosis to maintain normal CAFC was significantly decreased when compared to donors. After chemotherapy, the restoration of ALL patients' MSCs functions was slower than that of AML. CML MSCs maintained CAFC better than donors' at the moment of diagnosis and this ability increased with treatment. CONCLUSIONS The ability of patients' MSCs to maintain normal hematopoietic progenitor cells was shown to change in comparison with MSCs from healthy donors and depended on nosology. During treatment, the functional capacity of patients' MSCs had been partially restored.
Collapse
Affiliation(s)
| | | | | | | | - Nina Drize
- National Research Center for Hematology, Moscow, Russia
| | - Anna Turkina
- National Research Center for Hematology, Moscow, Russia
| | | | - Oleg Shukhov
- National Research Center for Hematology, Moscow, Russia
| | | | | | | |
Collapse
|
377
|
Abstract
In this issue of Blood, Paggetti et al present novel findings that chronic lymphocytic leukemia (CLL)-derived exosomes and their molecular cargo are actively transferred to stromal cells that reside in the lymphoid tumor microenvironment (TME), promoting the reprogramming of these cells into cancer-associated fibroblasts (CAFs).
Collapse
|
378
|
Taylor MA. Circulating MicroRNAs as Biomarkers and Mediators of Cell-Cell Communication in Cancer. Biomedicines 2015; 3:270-281. [PMID: 28536412 PMCID: PMC5344225 DOI: 10.3390/biomedicines3040270] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/03/2015] [Accepted: 11/05/2015] [Indexed: 12/21/2022] Open
Abstract
The realization of personalized medicine for cancer will rely not only on the development of new therapies, but on biomarkers that direct these therapies to the right patient. MicroRNA expression profiles in the primary tumor have been shown to differ between cancer patients and healthy individuals, suggesting they might make useful biomarkers. However, examination of microRNA expression in the primary tumor requires an invasive biopsy procedure. More recently, microRNAs have been shown to be released from the primary tumor into the circulation where they can be utilized as non-invasive biomarkers to diagnose patients, predict prognosis, or indicate therapeutic response. This review provides an overview of the current use of circulating microRNAs as biomarkers as well as recent findings on their role in regulating cell signaling interactions in the tumor microenvironment.
Collapse
Affiliation(s)
- Molly A Taylor
- AstraZeneca, R&D Oncology iMed, Room 33F83/7 Mereside, Alderley Park, Macclesfield SK10 4TG, UK.
| |
Collapse
|
379
|
Exosome-mediated microenvironment dysregulation in leukemia. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:464-470. [PMID: 26384870 DOI: 10.1016/j.bbamcr.2015.09.017] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/05/2015] [Accepted: 09/11/2015] [Indexed: 12/23/2022]
Abstract
The hematopoietic stem cell (HSC) niche is composed of a complex set of stromal support cells that maintain HSCs and promote normal hematopoiesis. We now know that molecular changes within the hematopoietic niche contribute to leukemia development. Leukemia cells often reorganize the hematopoietic niche to promote and support their own survival and growth. Here we will summarize recent works that decipher the normal hematopoietic niche cellular components and describe how the leukemia-transformed niche contributes to hematological malignances. Finally, we will discuss recent publications that highlight a possible role for exosomes in the leukemia-induced niche reorganization. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
|
380
|
Gandellini P, Andriani F, Merlino G, D'Aiuto F, Roz L, Callari M. Complexity in the tumour microenvironment: Cancer associated fibroblast gene expression patterns identify both common and unique features of tumour-stroma crosstalk across cancer types. Semin Cancer Biol 2015; 35:96-106. [PMID: 26320408 DOI: 10.1016/j.semcancer.2015.08.008] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/17/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022]
Abstract
Cancer is a complex disease, driven by the accumulation of several somatic aberrations but fostered by a two-way interaction between tumour cells and the surrounding microenvironment. Cancer associated fibroblasts (CAFs) represent one of the major players in tumour-stroma crosstalk. Recent in vitro and in vivo studies, often conducted by employing high throughput approaches, have started unravelling the key pathways involved in their functional effects. This review focus on open challenges in the study of CAF properties and function, highlighting at the same time the existence of common mechanisms as well as peculiarities in different cancer types (breast, prostate and lung cancer). Although still limited by current experimental models, which are unable to deal with the full level of complexity of the tumour microenvironment, a better understanding of these mechanisms may enable the identification of new biomarkers and therapeutic targets, to improve current strategies for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Paolo Gandellini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Andriani
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giuseppe Merlino
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca D'Aiuto
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Roz
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maurizio Callari
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|