51
|
Li M, Guo H, Wang Q, Chen K, Marko K, Tian X, Yang Y. Pancreatic stellate cells derived exosomal miR-5703 promotes pancreatic cancer by downregulating CMTM4 and activating PI3K/Akt pathway. Cancer Lett 2020; 490:20-30. [PMID: 32585413 DOI: 10.1016/j.canlet.2020.06.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/03/2020] [Accepted: 06/07/2020] [Indexed: 12/22/2022]
Abstract
Exosomes play important role in tumor microenvironment, and mediate the crosstalk between pancreatic cancer (PC) cells and matrix components including pancreatic stellate cells (PSCs) to regulate pancreatic cancer progression. Here we isolated primary PSCs from PC patients, and demonstrated that PSC-derived exosomes could be internalized by PC cells to promote cell proliferation. Furthermore, we identified that miR-5703 in the exosomes acted as a driver of cell proliferation and its inhibitor suppressed the function of exosomes to promote PC cell proliferation. miR-5703 directly bound to the 3'UTR of CMTM4 and downregulated its expression. CMTM4 knockdown promoted PC cell proliferation, while overexpression of CMTM4 suppressed PC cell proliferation both in vivo and in vitro. Mechanistically, we revealed that CMTM4 suppressed PI3K/Akt pathway via downregulating PAK4. In conclusion, our results suggest that PSC-derived exosomal miR-5703 could target CMTM4 in PC cells and promote cell proliferation due to PAK4 activated PI3K/Akt pathway.
Collapse
Affiliation(s)
- Mingzhe Li
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China.
| | - Huahu Guo
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Qi Wang
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China.
| | - Kai Chen
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China.
| | - Kornmann Marko
- Clinic of General, Visceral and Transplantation Surgery, University of Ulm, Ulm, 89081, Germany.
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China.
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
52
|
Sun W, Ren Y, Lu Z, Zhao X. The potential roles of exosomes in pancreatic cancer initiation and metastasis. Mol Cancer 2020; 19:135. [PMID: 32878635 PMCID: PMC7466807 DOI: 10.1186/s12943-020-01255-w] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PaCa) is an insidious and highly metastatic malignancy, with a 5-year survival rate of less than 5%. So far, the pathogenesis and progression mechanisms of PaCa have been poorly characterized. Exosomes correspond to a class of extracellular nanovesicles, produced by a broad range of human somatic and cancerous cells. These particular nanovesicles are mainly composed by proteins, genetic substances and lipids, which mediate signal transduction and material transport. A large number of studies have indicated that exosomes may play decisive roles in the occurrence and metastatic progression of PaCa. This article summarizes the specific functions of exosomes and their underlying molecular mechanisms in mediating the initiation and metastatic capability of PaCa.
Collapse
Affiliation(s)
- Wei Sun
- Department of Radiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Ying Ren
- Department of Radiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Zaiming Lu
- Department of Radiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Xiangxuan Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
53
|
Nannan L, Oudart JB, Monboisse JC, Ramont L, Brassart-Pasco S, Brassart B. Extracellular Vesicle-Dependent Cross-Talk in Cancer-Focus on Pancreatic Cancer. Front Oncol 2020; 10:1456. [PMID: 32974169 PMCID: PMC7466446 DOI: 10.3389/fonc.2020.01456] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/09/2020] [Indexed: 01/10/2023] Open
Abstract
Extracellular vesicles (EVs) like exosomes and shed microvesicles are generated by many different cells. However, among all the cells, cancer cells are now recognized to secrete more EVs than healthy cells. Tumor-derived EVs can be isolated from biofluids such as blood, urine, ascitic fluid, and saliva. Their numerous components (nucleic acids, proteins, and lipids) possess many pleiotropic functions involved in cancer progression. The tumor-derived EVs generated under the influence of tumor microenvironment play distant roles and promote cellular communication by directly interacting with different cells. Moreover, they modulate extracellular matrix remodeling and tumor progression. Tumor-derived EVs are involved in pre-metastatic niche formation, dependent on the EV-associated protein receptors, and in cancer chemoresistance as they transfer drug-resistance-related genes to recipient cells. Recent advances in preclinical and clinical fields suggest their potential use as biomarkers for diagnosis and prognosis as well as for drug delivery in cancer. In this Review, we discuss EV characteristics and pro-tumor capacities, and highlight the future crucial impact of tumor-derived EVs in pancreatic cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Lise Nannan
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.,Biomedical MRI Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jean-Baptiste Oudart
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Jean Claude Monboisse
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Laurent Ramont
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.,CHU Reims, Service de Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Sylvie Brassart-Pasco
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
| | - Bertrand Brassart
- Université de Reims Champagne Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.,CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
| |
Collapse
|
54
|
Moeng S, Son SW, Lee JS, Lee HY, Kim TH, Choi SY, Kuh HJ, Park JK. Extracellular Vesicles (EVs) and Pancreatic Cancer: From the Role of EVs to the Interference with EV-Mediated Reciprocal Communication. Biomedicines 2020; 8:biomedicines8080267. [PMID: 32756339 PMCID: PMC7459718 DOI: 10.3390/biomedicines8080267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/25/2020] [Accepted: 08/01/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is malignant and the seventh leading cause of cancer-related deaths worldwide. However, chemotherapy and radiotherapy are—at most—moderately effective, indicating the need for new and different kinds of therapies to manage this disease. It has been proposed that the biologic properties of pancreatic cancer cells are finely tuned by the dynamic microenvironment, which includes extracellular matrix, cancer-associated cells, and diverse immune cells. Accumulating evidence has demonstrated that extracellular vesicles (EVs) play an essential role in communication between heterogeneous subpopulations of cells by transmitting multiplex biomolecules. EV-mediated cell–cell communication ultimately contributes to several aspects of pancreatic cancer, such as growth, angiogenesis, metastasis and therapeutic resistance. In this review, we discuss the role of extracellular vesicles and their cargo molecules in pancreatic cancer. We also present the feasibility of the inhibition of extracellular biosynthesis and their itinerary (release and uptake) for a new attractive therapeutic strategy against pancreatic cancer.
Collapse
Affiliation(s)
- Sokviseth Moeng
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Seung Wan Son
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Jong Sun Lee
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Han Yeoung Lee
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Tae Hee Kim
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
| | - Hyo Jeong Kuh
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Jong Kook Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (S.M.); (S.W.S.); (J.S.L.); (H.Y.L.); (T.H.K.); (S.Y.C.)
- Correspondence: ; Tel.: +82-33-248-2114
| |
Collapse
|
55
|
Takeda Y, Kobayashi S, Kitakaze M, Yamada D, Akita H, Asai A, Konno M, Arai T, Kitagawa T, Ofusa K, Yabumoto M, Hirotsu T, Vecchione A, Taniguchi M, Doki Y, Eguchi H, Ishii H. Immuno-Surgical Management of Pancreatic Cancer with Analysis of Cancer Exosomes. Cells 2020; 9:cells9071645. [PMID: 32659892 PMCID: PMC7408222 DOI: 10.3390/cells9071645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/05/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023] Open
Abstract
Exosomes (EXs), a type of extracellular vesicles secreted from various cells and especially cancer cells, mesenchymal cells, macrophages and other cells in the tumor microenvironment (TME), are involved in biologically malignant behaviors of cancers. Recent studies have revealed that EXs contain microRNAs on their inside and express proteins and glycolipids on their outsides, every component of which plays a role in the transmission of genetic and/or epigenetic information in cell-to-cell communications. It is also known that miRNAs are involved in the signal transduction. Thus, EXs may be useful for monitoring the TME of tumor tissues and the invasion and metastasis, processes that are associated with patient survival. Because several solid tumors secrete immune checkpoint proteins, including programmed cell death-ligand 1, the EX-mediated mechanisms are suggested to be potent targets for monitoring patients. Therefore, a companion therapeutic approach against cancer metastasis to distant organs is proposed when surgical removal of the primary tumor is performed. However, EXs and immune checkpoint mechanisms in pancreatic cancer are not fully understood, we provide an update on the recent advances in this field and evidence that EXs will be useful for maximizing patient benefit in precision medicine.
Collapse
Affiliation(s)
- Yu Takeda
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Masatoshi Kitakaze
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Ayumu Asai
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Artificial Intelligence Research Center, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan;
| | - Masamitsu Konno
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Takahiro Arai
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Unitech Co., Ltd., Kashiwa 277-0005, Japan
| | - Toru Kitagawa
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
- Kyowa-kai Medical Corporation, Osaka 540-0008, Japan
| | - Ken Ofusa
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Prophoenix Division, Food and Life-Science Laboratory, Idea Consultants, Inc., Osaka-city, Osaka 559-8519, Japan
| | - Masami Yabumoto
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
- Kinshu-kai Medical Corporation, Osaka 558-0041, Japan
| | - Takaaki Hirotsu
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Hirotsu Bio Science Inc., Tokyo 107-0062, Japan
| | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, University of Rome “Sapienza”, Santo Andrea Hospital, via di Grottarossa, 1035-00189 Rome, Italy;
| | - Masateru Taniguchi
- Artificial Intelligence Research Center, The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan;
| | - Yuichiro Doki
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Hidetoshi Eguchi
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
| | - Hideshi Ishii
- Center of Medical Innovation and Translational Research (CoMIT), Osaka University Graduate School of Medicine, Suita, Yamadaoka 2-2, Osaka 565-0871, Japan; (Y.T.); (M.K.); (A.A.); (M.K.); (T.A.); (T.K.); (K.O.); (M.Y.); (T.H.); (Y.D.); (H.E.)
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (S.K.); (D.Y.); (H.A.)
- Correspondence: ; Tel.: +81-(0)6-6210-8406 (ext. 8405); Fax: +81-(0)6-6210-8407
| |
Collapse
|
56
|
Gašić U, Ćirić I, Pejčić T, Radenković D, Djordjević V, Radulović S, Tešić Ž. Polyphenols as Possible Agents for Pancreatic Diseases. Antioxidants (Basel) 2020; 9:antiox9060547. [PMID: 32585831 PMCID: PMC7346180 DOI: 10.3390/antiox9060547] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/26/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is very aggressive and it is estimated that it kills nearly 50% of patients within the first six months. The lack of symptoms specific to this disease prevents early diagnosis and treatment. Today, gemcitabine alone or in combination with other cytostatic agents such as cisplatin (Cis), 5-fluorouracil (5-FU), irinotecan, capecitabine, or oxaliplatin (Oxa) is used in conventional therapy. Outgoing literature provides data on the use of polyphenols, biologically active compounds, in the treatment of pancreatic cancer and the prevention of acute pancreatitis. Therefore, the first part of this review gives a brief overview of the state of pancreatic disease as well as the procedures for its treatment. The second part provides a detailed overview of the research regarding the anticancer effects of both pure polyphenols and their plant extracts. The results regarding the antiproliferative, antimetastatic, as well as inhibitory effects of polyphenols against PC cell lines as well as the prevention of acute pancreatitis are presented in detail. Finally, particular emphasis is given to the polyphenolic profiles of apples, berries, cherries, sour cherries, and grapes, given the fact that these fruits are rich in polyphenols and anthocyanins. Polyphenolic profiles, the content of individual polyphenols, and their relationships are discussed. Based on this, significant data can be obtained regarding the amount of fruit that should be consumed daily to achieve a therapeutic effect.
Collapse
Affiliation(s)
- Uroš Gašić
- Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia;
| | - Ivanka Ćirić
- Innovation Center, University of Belgrade—Faculty of Chemistry, P.O. Box 51, 11158 Belgrade, Serbia;
| | - Tomislav Pejčić
- Clinic of Urology, Clinical Centre of Serbia, Pasterova 2, 11000 Belgrade, Serbia;
| | - Dejan Radenković
- University of Belgrade—Faculty of Medicine, dr Subotića 8, 11000 Belgrade, Serbia;
- First Surgical Clinic, Clinical Center of Serbia, Koste Todorovića 6, 11000 Belgrade, Serbia;
| | - Vladimir Djordjević
- First Surgical Clinic, Clinical Center of Serbia, Koste Todorovića 6, 11000 Belgrade, Serbia;
| | - Siniša Radulović
- Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia;
| | - Živoslav Tešić
- University of Belgrade—Faculty of Chemistry, Studentski trg 12–16, P.O. Box 51, 11158 Belgrade, Serbia
- Correspondence: ; Tel.: +381-113336733
| |
Collapse
|
57
|
Cui S, Lou S, Guo W, Jian S, Wu Y, Liu X, Lan X, Jia X. Prediction of MiR-21-5p in Promoting the Development of Lung Adenocarcinoma via PDZD2 Regulation. Med Sci Monit 2020; 26:e923366. [PMID: 32535612 PMCID: PMC7313425 DOI: 10.12659/msm.923366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Lung adenocarcinoma currently accounts for the highest cancer-related mortality rate worldwide. MiR-21-5p has a vital role in various types of cancers. We have analyzed the miR-21-5p expression level, prognosis, and associated molecular pathways in lung adenocarcinoma with multiple bioinformatics databases. MATERIAL AND METHODS The Cancer Genome Atlas (TCGA) database was employed to fetch the miR-21-5p expression profile in multiple tumors. We used the UALCAN platform to assess the differential regulation of the miR-21-5p in healthy tissue and lung adenocarcinoma. Also, the survival prognosis of the miR-21-5p in each stage of lung adenocarcinoma was done by the Kaplan-Meier database. The STARBASE and UALCAN databases were employed to predict the miR-21-5p target genes, and the levels of target genes and their prognostic value were analyzed. RESULTS MiR-21-5p was overexpressed in the majority of human cancers. MiR-21-5p demonstrated escalated expression in the lung adenocarcinoma tissue in contrast to the normal tissue (P<0.05). Poor prognosis was witnessed in the miR-21-5p high expression group as compared to the low expression group (hazard ratio [HR]= 1.59, P<0.05). PDZD2 was predicted as a miR-21-5p potential target. We found a negative correlation between PDZD2 and miR-21-5p (r=-0.255, P<0.05). PDZD2 was downregulated in lung adenocarcinoma (P<0.05). Overexpression of PDZD2 was associated with a better prognosis of survival in lung adenocarcinoma patients (HR=0.45, P<0.05). CONCLUSIONS MiR-21-5p exhibits the potential to act as a biomarker for the survival prognosis of lung adenocarcinoma. It might be responsible for the onset and progression of lung adenocarcinoma through PDZD2 regulation.
Collapse
Affiliation(s)
- Shengjin Cui
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China (mainland)
| | - Shuang Lou
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China (mainland)
| | - Weiquan Guo
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China (mainland)
| | - Shihui Jian
- Department of Clinical Laboratory, Guangzhou Huayin Medical Inspection Center Co., Ltd., Guangzhou, Guangdong, China (mainland)
| | - Yunfeng Wu
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China (mainland)
| | - Xintong Liu
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China (mainland)
| | - Xi Lan
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China (mainland)
| | - Xingwang Jia
- Department of Clinical Laboratory, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China (mainland)
| |
Collapse
|
58
|
Ma Q, Wu H, Xiao Y, Liang Z, Liu T. Upregulation of exosomal microRNA‑21 in pancreatic stellate cells promotes pancreatic cancer cell migration and enhances Ras/ERK pathway activity. Int J Oncol 2020; 56:1025-1033. [PMID: 32319558 DOI: 10.3892/ijo.2020.4986] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 01/20/2020] [Indexed: 11/05/2022] Open
Abstract
Pancreatic stellate cells (PSCs) are typically activated in pancreatic ductal adenocarcinoma (PDAC) and release exosomes containing high levels of microRNA‑21 (miR‑21). However, the specific roles of exosomal miR‑21 in regulating the PDAC malignant phenotype remain unknown. The present study aimed to determine the effects of exosomal miR‑21 on the migratory ability of PDAC cells and explore the potential underlying molecular mechanism. Weighted gene correlation network and The Cancer Genome Atlas database analysis revealed that high miR‑21 levels were associated with a poor prognosis in patients with pancreatic adenocarcinoma, and that the Ras/ERK signaling pathway may be a potential target of miR‑21. In vitro, PDAC cells were demonstrated to internalize the PSC-derived exosome, resulting in high miR‑21 levels, which subsequently promoted cell migration, induced epithelial‑to‑mesenchymal transition (EMT) and increased matrix metalloproteinase‑2/9 activity. In addition, exosomal miR‑21 increased the levels of ERK1/2 and Akt phosphorylation in PDAC cells. Collectively, these results suggested that PSC‑derived exosomal miR‑21 may promote PDAC cell migration and EMT and enhance Ras/ERK signaling activity. Thus, miR‑21 may be a potential cause of poor prognosis in patients with pancreatic cancer and a new treatment target.
Collapse
Affiliation(s)
- Qiang Ma
- Department of Pathology, Peking Union Medical College Hospital, Beijing 100730, P. R. China
| | - Huanwen Wu
- Department of Pathology, Peking Union Medical College Hospital, Beijing 100730, P. R. China
| | - Ying Xiao
- Department of Pathology, Peking Union Medical College Hospital, Beijing 100730, P. R. China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, Beijing 100730, P. R. China
| | - Tonghua Liu
- Department of Pathology, Peking Union Medical College Hospital, Beijing 100730, P. R. China
| |
Collapse
|
59
|
Han S, Underwood P, Hughes SJ. From tumor microenvironment communicants to biomarker discovery: Selectively packaged extracellular vesicular cargoes in pancreatic cancer. Cytokine Growth Factor Rev 2020; 51:61-68. [PMID: 32005635 PMCID: PMC8711854 DOI: 10.1016/j.cytogfr.2020.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/30/2019] [Accepted: 01/06/2020] [Indexed: 02/06/2023]
Abstract
Virtually all cells release various types of vesicles into the extracellular environment. These extracellular vesicles (EVs) transport molecular cargoes, performing as communicants for information exchange both within the tumor microenvironment (TME) and to distant organs. Thus, understanding the selective packaging of EV cargoes and the mechanistic impact of those cargoes - including metabolites, lipids, proteins, and/or nucleic acids - offers an opportunity to increase our knowledge of cancer biology and identify EV cargoes that might serve as cancer biomarkers in blood, saliva, or urine samples. In this review, we collect and organize recent advances in this field with an emphasis on pancreatic cancer (pancreatic adenocarcinoma, PDAC) and the concept that cells selectively package cargo into EVs. These studies demonstrate PDAC EV cargoes signal to reprogram and remodel the TME and impact distant organs. EV cargoes identified as potential PDAC diagnostic and prognostic biomarkers are summarized.
Collapse
Affiliation(s)
- Song Han
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, United States.
| | - Patrick Underwood
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Steven J Hughes
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL 32610, United States
| |
Collapse
|
60
|
Ayala‐Mar S, Donoso‐Quezada J, Gallo‐Villanueva RC, Perez‐Gonzalez VH, González‐Valdez J. Recent advances and challenges in the recovery and purification of cellular exosomes. Electrophoresis 2019; 40:3036-3049. [PMID: 31373715 PMCID: PMC6972601 DOI: 10.1002/elps.201800526] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
Exosomes are nanovesicles secreted by most cellular types that carry important biochemical compounds throughout the body with different purposes, playing a preponderant role in cellular communication. Because of their structure, physicochemical properties and stability, recent studies are focusing in their use as nanocarriers for different therapeutic compounds for the treatment of different diseases ranging from cancer to Parkinson's disease. However, current bioseparation protocols and methodologies are selected based on the final exosome application or intended use and present both advantages and disadvantages when compared among them. In this context, this review aims to present the most important technologies available for exosome isolation while discussing their advantages and disadvantages and the possibilities of being combined with other strategies. This is critical since the development of novel exosome-based therapeutic strategies will be constrained to the effectiveness and yield of the selected downstream purification methodologies for which a thorough understanding of the available technological resources is needed.
Collapse
Affiliation(s)
- Sergio Ayala‐Mar
- Tecnologico de MonterreySchool of Engineering and Science, AvEugenio Garza Sada 2501 SurMonterreyNLMexico
| | - Javier Donoso‐Quezada
- Tecnologico de MonterreySchool of Engineering and Science, AvEugenio Garza Sada 2501 SurMonterreyNLMexico
| | | | - Victor H. Perez‐Gonzalez
- Tecnologico de MonterreySchool of Engineering and Science, AvEugenio Garza Sada 2501 SurMonterreyNLMexico
| | - José González‐Valdez
- Tecnologico de MonterreySchool of Engineering and Science, AvEugenio Garza Sada 2501 SurMonterreyNLMexico
| |
Collapse
|
61
|
Patras L, Banciu M. Intercellular Crosstalk Via Extracellular Vesicles in Tumor Milieu as Emerging Therapies for Cancer Progression. Curr Pharm Des 2019; 25:1980-2006. [DOI: 10.2174/1381612825666190701143845] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
:Increasing evidence has suggested that extracellular vesicles (EV) mediated bidirectional transfer of functional molecules (such as proteins, different types of RNA, and lipids) between cancer cells and tumor stromal cells (immune cells, endothelial cells, fibroblasts, stem cells) and strongly contributed to the reinforcement of cancer progression. Thus, intercellular EV-mediated signaling in tumor microenvironment (TME) is essential in the modulation of all processes that support and promote tumor development like immune suppression, angiogenesis, invasion and metastasis, and resistance of tumor cells to anticancer treatments.:Besides EV potential to revolutionize our understanding of the cancer cell-stromal cells crosstalk in TME, their ability to selectively transfer different cargos to recipient cells has created excitement in the field of tumortargeted delivery of specific molecules for anticancer treatments. Therefore, in tight connection with previous findings, this review brought insight into the dual role of EV in modulation of TME. Thus, on one side EV create a favorable phenotype of tumor stromal cells for tumor progression; however, as a future new class of anticancer drug delivery systems EV could re-educate the TME to overcome main supportive processes for malignancy progression.
Collapse
Affiliation(s)
- Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
62
|
Jiang R, Huang H, Lian Z, Hu Z, Lloyd RS, Fang D, Li Y, Xian H, Yuan J, Sha Y, Wang S, Hu D. Exosomal miR-221 derived from hydroquinone-transformed malignant human bronchial epithelial cells is involved in cell viability of recipient cells. J Appl Toxicol 2019; 40:224-233. [PMID: 31468561 DOI: 10.1002/jat.3898] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/01/2019] [Accepted: 08/01/2019] [Indexed: 12/16/2022]
Abstract
miR-221, an oncogenic microRNA, can promote cell proliferation and is highly expressed in various types of tumors. However, the role of exosomal miR-221 in benzene-caused carcinogenesis remains elusive. Our study was designed to investigate whether exosomes secreted by the hydroquinone (HQ; an active metabolite of benzene)-transformed malignant cells can transmit miR-221 to normal recipient cells and its possible effects on cell viability. Our investigation revealed that expression levels of miR-221 were significantly increased in HQ-transformed malignant cells relative to normal controls. Furthermore, exposure of control cells to exosomes that were derived from HQ-transformed malignant cells increased miR-221 levels and promoted their proliferation. Analyses of the biological potency of exosomes derived from HQ-transformed malignant cells in which miR-221 levels were decreased using an inhibitor, showed that both miR-221 levels and proliferation of recipient cells were decreased, but still were higher than those of normal 16HBE cells. Our study indicates that exosomal miR-221 derived from HQ-transformed malignant human bronchial epithelial cells is involved in the proliferation of recipient cells.
Collapse
Affiliation(s)
- Ran Jiang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Haoyu Huang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhenwei Lian
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zuqing Hu
- School of Medicine, Jiamusi University, Jiamusi, China
| | - R Stephen Lloyd
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon
| | - Daokui Fang
- Department of Environmental Health, Center for Disease Control and Prevention of Shenzhen City, Shenzhen, China
| | - Yanfeng Li
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hongyi Xian
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen, China
| | - Yan Sha
- Shenzhen Prevention and Treatment Center for Occupational Disease, Institute of Occupational Disease, Shenzhen, China
| | - Sanming Wang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, SAR, China
| | - Dalin Hu
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
63
|
Guo XY, Xiao F, Li J, Zhou YN, Zhang WJ, Sun B, Wang G. Exosomes and pancreatic diseases: status, challenges, and hopes. Int J Biol Sci 2019; 15:1846-1860. [PMID: 31523187 PMCID: PMC6743302 DOI: 10.7150/ijbs.35823] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic disease, including pathologies such as acute pancreatitis (AP), chronic pancreatitis (CP), and pancreatic cancer (PC), is a complicated and dangerous clinical condition involving the disruption of exocrine or endocrine function. PC has one of the highest mortality rates among cancers due to insufficient diagnosis in early stages. Furthermore, efficient treatment options for the disease etiologies of AP and CP are lacking. Thus, the identification of new therapeutic targets and reliable biomarkers is required. As essential couriers in intercellular communication, exosomes have recently been confirmed to play an important role in pancreatic disease, but the specific underlying mechanisms are unknown. Herein, we summarize the current knowledge of exosomes in pancreatic disease with respect to diagnosis, molecular mechanisms, and treatment, proposing new ideas for the study of pancreatic disease.
Collapse
Affiliation(s)
- Xiao-Yu Guo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fan Xiao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jie Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yi-Nan Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wang-Jun Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
64
|
Batista IA, Melo SA. Exosomes and the Future of Immunotherapy in Pancreatic Cancer. Int J Mol Sci 2019; 20:ijms20030567. [PMID: 30699928 PMCID: PMC6387297 DOI: 10.3390/ijms20030567] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/25/2019] [Indexed: 12/26/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease, associated with a late diagnosis and a five-year survival rate of 8%. Currently available treatments fall short in improving the survival and quality of life of PDAC patients. The only possible curative option is still the surgical resection of the tumor. Exosomes are extracellular vesicles secreted by cells that transport proteins, lipids, and nucleic acids to other cells, triggering phenotypic changes in the recipient cells. Tumor cells often secrete increased amounts of exosomes. Tumor exosomes are now accepted as important players in the remodeling of PDAC tumor stroma, particularly in the establishment of an immunosuppressive microenvironment. This has sparked the interest in their usefulness as mediators of immunomodulatory effects for the treatment of PDAC. In fact, exosomes are now under study to understand their potential as nanocarriers to stimulate an immune response against cancer. This review highlights the latest findings regarding the function of exosomes in tumor-driven immunomodulation, and the challenges and advantages associated with the use of these vesicles to potentiate immunotherapy in PDAC.
Collapse
Affiliation(s)
- Ines A Batista
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal (i3S), 4200-135 Porto, Portugal.
- Institute of Molecular Pathology & Immunology of the University of Porto (IPATIMUP), 4200-135 Porto, Portugal.
| | - Sonia A Melo
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal (i3S), 4200-135 Porto, Portugal.
- Institute of Molecular Pathology & Immunology of the University of Porto (IPATIMUP), 4200-135 Porto, Portugal.
- Medical Faculty of the University of Porto (FMUP), 4200-319 Porto, Portugal.
| |
Collapse
|
65
|
Chen G, Hu M, Qu X, Wang K, Qu Y. MicroRNA‑584 directly targets CCND1 and inhibits cell proliferation and invasion in pancreatic cancer. Mol Med Rep 2018; 19:719-726. [PMID: 30431107 DOI: 10.3892/mmr.2018.9651] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 10/16/2018] [Indexed: 11/06/2022] Open
Abstract
Multiple previous studies have demonstrated that the dysregulation of microRNAs (miRNAs) is implicated in the occurrence and development of pancreatic cancer. Therefore, a further characterisation of deregulated miRNAs in pancreatic cancer may provide novel insight into the oncogenesis and progression of pancreatic cancer, which may facilitate the identification of effective therapeutic targets for treating patients with this disease. In the present study, reverse transcription‑quantitative polymerase chain reaction analysis demonstrated that the expression level of miRNA‑584‑5p (miR‑584) was significantly decreased in pancreatic cancer tissues and cell lines. It was demonstrated that restoration of miR‑584 expression significantly suppressed the proliferative and invasive ability of pancreatic cancer cells. Bioinformatics analysis predicted that cyclin D1 (CCND1) was a putative target of miR‑584. Subsequent experiments demonstrated that CCND1 was a direct target gene of miR‑584 in pancreatic cancer cells. Furthermore, the inhibition of CCND1 mimicked the suppressive effect of miR‑584 overexpression in pancreatic cancer cells. The restoration of CCND1 expression significantly abolished the inhibitory effects of miR‑584 overexpression on pancreatic cancer cells. Collectively, the present results demonstrated that miR‑584 inhibited the development of pancreatic cancer by directly targeting CCND1, suggesting that this miRNA may represent a potential therapeutic target for this fatal disease.
Collapse
Affiliation(s)
- Gang Chen
- Department of Gastroenterology, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Ming Hu
- Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Xiusheng Qu
- Department of Radiochemotherapy, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Kaifeng Wang
- Department of Vascular Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| | - Yikun Qu
- Department of General Surgery, First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang 154003, P.R. China
| |
Collapse
|
66
|
Qian L, Yu S, Chen Z, Meng Z, Huang S, Wang P. Functions and clinical implications of exosomes in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2018; 1871:75-84. [PMID: 30419313 DOI: 10.1016/j.bbcan.2018.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/31/2018] [Accepted: 11/04/2018] [Indexed: 01/18/2023]
Abstract
Pancreatic cancer is one of the most aggressive human malignancies and is associated with a dismal prognosis, which can be contributed to its atypical symptoms, metastatic propensity, and significant chemoresistance. Emerging evidence shows that pancreatic cancer cell-derived exosomes (PEXs) play critical roles in tumorigenesis and tumor development, as they are involved in drug resistance, immune evasion and metabolic reprograming, and distant metastasis of pancreatic cancer. Their numerous differentially expressed and functional contents make PEXs promising screening tools and therapeutic targets, which require further exploration. In this review, we focus on the functions of PEX contents and their clinical implications in pancreatic cancer.
Collapse
Affiliation(s)
- Ling Qian
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 2000332, China
| | - Shulin Yu
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 2000332, China
| | - Zhen Chen
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 2000332, China
| | - Zhiqiang Meng
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 2000332, China
| | - Shenglin Huang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 2000332, China; Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 2000332, China.
| | - Peng Wang
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 2000332, China.
| |
Collapse
|
67
|
Zhou J, Li X, Wu X, Zhang T, Zhu Q, Wang X, Wang H, Wang K, Lin Y, Wang X. Exosomes Released from Tumor-Associated Macrophages Transfer miRNAs That Induce a Treg/Th17 Cell Imbalance in Epithelial Ovarian Cancer. Cancer Immunol Res 2018; 6:1578-1592. [PMID: 30396909 DOI: 10.1158/2326-6066.cir-17-0479] [Citation(s) in RCA: 279] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/22/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022]
Abstract
The immune microenvironment is crucial for epithelial ovarian cancer (EOC) progression and consists of tumor-associated macrophages (TAM) and T lymphocytes, such as regulatory T cells (Treg) and T helper 17 (Th17) cells. In this study, the Treg/Th17 ratio was significantly higher in EOC in situ and in metastatic peritoneal tissues than in benign ovarian tumors and benign peritoneum. The Treg/Th17 ratio was associated with histologic grade and was an independent prognostic factor for overall survival of EOC patients. On the basis of microarray analysis of exosomes derived from TAMs, we identified miRNAs enriched in the exosomes, including miR-29a-3p and miR-21-5p. When the two miRNA mimics were transfected into CD4+ T cells, they directly suppressed STAT3 and regulated Treg/Th17 cells, inducing an imbalance, and they had a synergistic effect on STAT3 inhibition. Taken together, these results indicate that exosomes mediate the interaction between TAMs and T cells, generating an immune-suppressive microenvironment that facilitates EOC progression and metastasis. These findings suggest that targeting these exosomes or their associated miRNAs might pave the way for the development of novel treatments for EOC.
Collapse
Affiliation(s)
- Jieru Zhou
- Department of Obstetrics and Gynaecology, XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoduan Li
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoli Wu
- Department of Obstetrics and Gynaecology, XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Zhang
- Center for Reproductive Medicine, Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Qinyi Zhu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xinjing Wang
- Department of Obstetrics and Gynaecology, XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Husheng Wang
- Department of Obstetrics and Gynaecology, XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yingying Lin
- Department of Neurosurgery, RenJi Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynaecology, XinHua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
68
|
Samandari M, Julia MG, Rice A, Chronopoulos A, Del Rio Hernandez AE. Liquid biopsies for management of pancreatic cancer. Transl Res 2018; 201:98-127. [PMID: 30118658 DOI: 10.1016/j.trsl.2018.07.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/17/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is one of the main causes of cancer-related deaths worldwide. It is asymptomatic at an early stage, and most diagnosis occurs when the disease is already at a late stage, by which time the tumor is nonresectable. In order to increase the overall survival of patients with pancreatic cancer, as well as to decrease the cancer burden, it is necessary to perform early diagnosis, prognosis stratifications and cancer monitoring using accurate, minimally invasive, and cost-effective methods. Liquid biopsies seek to detect tumor-associated biomarkers in a variety of extractable body fluids and can help to monitor treatment response and disease progression, and even predict patient outcome. In patients with pancreatic cancer, tumor-derived materials, primarily circulating tumor DNA, circulating tumor cells and exosomes, are being studied for inclusion in the management of the disease. This review focuses on describing the biology of these biomarkers, methods for their enrichment and detection, as well as their potential for clinical application. Moreover, we discuss the future direction of liquid biopsies and introduce how they can be exploited toward point of care personalized medicine for the management of pancreatic cancer.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - María Gil Julia
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Antonios Chronopoulos
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Armando E Del Rio Hernandez
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom.
| |
Collapse
|
69
|
Sun Q, Zhang B, Hu Q, Qin Y, Xu W, Liu W, Yu X, Xu J. The impact of cancer-associated fibroblasts on major hallmarks of pancreatic cancer. Am J Cancer Res 2018; 8:5072-5087. [PMID: 30429887 PMCID: PMC6217060 DOI: 10.7150/thno.26546] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 09/04/2018] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) constitutes one of the most challenging lethal tumors and has a very poor prognosis. In addition to cancer cells, the tumor microenvironment created by a repertoire of resident and recruited cells and the extracellular matrix also contribute to the acquisition of hallmarks of cancer. Among these factors, cancer-associated fibroblasts (CAFs) are critical components of the tumor microenvironment. CAFs originate from the activation of resident fibroblasts and pancreatic stellate cells, the differentiation of bone marrow-derived mesenchymal stem cells and epithelial-to-mesenchymal transition. CAFs acquire an activated phenotype via various cytokines and promote tumor proliferation and growth, accelerate invasion and metastasis, induce angiogenesis, promote inflammation and immune destruction, regulate tumor metabolism, and induce chemoresistance; these factors contribute to the acquisition of major hallmarks of PDAC. Therefore, an improved understanding of the impact of CAFs on the major hallmarks of PDAC will highlight the diagnostic and therapeutic values of these targeted cells.
Collapse
|
70
|
Kanat O, Ertas H. Shattering the castle walls: Anti-stromal therapy for pancreatic cancer. World J Gastrointest Oncol 2018; 10:202-210. [PMID: 30147846 PMCID: PMC6107476 DOI: 10.4251/wjgo.v10.i8.202] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/19/2018] [Accepted: 06/27/2018] [Indexed: 02/05/2023] Open
Abstract
Despite the availability of potent chemotherapy regimens, such as 5-fluorouracil, folinic acid, irinotecan, and oxaliplatin (FOLFIRINOX) and nab-paclitaxel plus gemcitabine, treatment outcomes in metastatic pancreatic cancer (PC) remain unsatisfactory. The presence of an abundant fibrous stroma in PC is considered a crucial factor for its unfavorable condition. Apparently, stroma acts as a physical barrier to restrict intratumoral cytotoxic drug penetration and creates a hypoxic environment that reduces the efficacy of radiotherapy. In addition, stroma plays a vital supportive role in the development and progression of PC, which has prompted researchers to assess the potential benefits of agents targeting several cellular (e.g., stellate cells) and acellular (e.g., hyaluronan) elements of the stroma. This study aims to briefly review the primary structural properties of PC stroma and its interaction with cancer cells and summarize the current status of anti-stromal therapies in the management of metastatic PC.
Collapse
Affiliation(s)
- Ozkan Kanat
- Department of Medical Oncology, Faculty of Medicine, Uludag University, Bursa 16059, Turkey
| | - Hulya Ertas
- Department of Medical Oncology, Faculty of Medicine, Uludag University, Bursa 16059, Turkey
| |
Collapse
|
71
|
Yu LL, Zhu J, Liu JX, Jiang F, Ni WK, Qu LS, Ni RZ, Lu CH, Xiao MB. A Comparison of Traditional and Novel Methods for the Separation of Exosomes from Human Samples. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3634563. [PMID: 30148165 PMCID: PMC6083592 DOI: 10.1155/2018/3634563] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/17/2018] [Indexed: 01/11/2023]
Abstract
Exosomes are discrete populations of small (40-200 nm in diameter) membranous vesicles that are released into the extracellular space by most cell types, eventually accumulating in the circulation. As molecular messengers, exosomes exert a broad array of vital physiologic functions by transporting information between different cell types. Because of these functional properties, they may have potential as biomarker sources for prognostic and diagnostic disease. Recent research has found that exosomes have potential to be utilized as drug delivery agents for therapeutic targets. However, basic researches on exosomes and researches on their therapeutic potential both require the existence of effective and rapid methods for their separation from human samples. In the current absence of a standardized method, there are several methods available for the separation of exosomes, but very few studies have previously compared the efficiency and suitability of these different methods. This review summarized and compared the available traditional and novel methods for the extraction of exosomes from human samples and considered their advantages and disadvantages for use in clinical laboratories and point-of-care settings.
Collapse
Affiliation(s)
- Li-Li Yu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Jing Zhu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Jin-Xia Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Feng Jiang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Wen-Kai Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Li-Shuai Qu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Run-Zhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Cui-Hua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Ming-Bing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
72
|
Abstract
Pancreatic cancers with poor prognosis are highly malignant, readily metastatic and of immune tolerance, mainly due to delayed detection. The metastatic progression and immune tolerance of pancreatic cancer is greatly attributed to the intercellular communication. However, exosomes are deemed to be the most important tool of intercellular communicators. Thus, we present a review of pancreatic cancer and exosomes in this article. We intensively summarize the progress of early pancreatic cancer and the relationship of the proliferation, progression and metastasis of pancreatic cancer and pancreatic cancer-derived exosomes, and propose new ideas of the study of pancreatic cancer.
Collapse
Affiliation(s)
- Chengfei Zhao
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350108, Fujian, China.,Department of Pharmacy, Pharmacy and Medical Technology School, Putian University, Putian 351100, Fujian, China.,Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350108, Fujian, China
| | - Feng Gao
- Department of Pathology, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China.,Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350108, Fujian, China
| | - Shaohuang Weng
- Department of Pharmaceutical Analysis, Faculty of Pharmacy, Fujian Medical University, Fuzhou 350108, Fujian, China
| | - Qicai Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian, China
| |
Collapse
|
73
|
Xue R, Jia K, Wang J, Yang L, Wang Y, Gao L, Hao J. A Rising Star in Pancreatic Diseases: Pancreatic Stellate Cells. Front Physiol 2018; 9:754. [PMID: 29967585 PMCID: PMC6015921 DOI: 10.3389/fphys.2018.00754] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/29/2018] [Indexed: 12/17/2022] Open
Abstract
Pancreatic stellate cell (PSC) is a type of pluripotent cell located between pancreatic lobules and the surrounding area of acinars. When activated, PSC can be transformed into myofibroblast-like cell. A number of evidences suggest that activated PSC is the main source of the accumulation of extracellular matrix (ECM) protein under the pathological conditions, which lead to pancreatic fibrosis in chronic pancreatitis and pancreatic cancer. Recent studies have found that PSC also plays an important role in the endocrine cell function, islet fibrosis and diabetes. In order to provide new strategies for the treatment of pancreatic diseases, this paper systematically summarizes the recent researches about the biological behaviors of PSC, including its stem/progenitor cell characteristics, secreted exosomes, cellular senescence, epithelial mesenchymal transformation (EMT), energy metabolism and direct mechanical reprogramming.
Collapse
Affiliation(s)
- Ran Xue
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Kai Jia
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jianxin Wang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lixin Yang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yanbin Wang
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lingyun Gao
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jianyu Hao
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
74
|
Armstrong EA, Beal EW, Chakedis J, Paredes AZ, Moris D, Pawlik TM, Schmidt CR, Dillhoff ME. Exosomes in Pancreatic Cancer: from Early Detection to Treatment. J Gastrointest Surg 2018; 22:737-750. [PMID: 29423813 DOI: 10.1007/s11605-018-3693-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/12/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Pancreatic cancer (PC) remains one of the most fatal forms of cancer worldwide with incidence nearly equal to mortality. This is often attributed to the fact that diagnosis is often not made until later disease stages when treatment proves difficult. Efforts have been made to reduce the mortality of PC through improvements in early screening techniques and treatments of late-stage disease. Exosomes, small extracellular vesicles involved in cellular communication, have shown promise in helping understand PC disease biology. METHODS In this review, we discuss current studies of the role of exosomes in PC physiology, and their potential use as diagnostic and treatment tools. RESULTS Exosomes have a role in diagnosing pancreatic cancer and in understanding tumor biology including migration, proliferation, chemoresistance, immunosuppression, cachexia and diabetes, and have a potential role in therapy for pancreatic cancer. CONCLUSIONS Exosomal analysis is beneficial in demonstrating mechanisms behind PC growth and metastasis, immunosuppression, drug resistance, and paraneoplastic conditions. Furthermore, the use of exosomes can be beneficial in detecting early-stage PC and exosomes have potential applications as therapeutic targets.
Collapse
Affiliation(s)
- Emily A Armstrong
- The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Eliza W Beal
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA.
| | - Jeffery Chakedis
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Anghela Z Paredes
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Demetrios Moris
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Carl R Schmidt
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| | - Mary E Dillhoff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 320 W 10th Ave. M256 Starling Loving Hall, Columbus, OH, 43210, USA
| |
Collapse
|
75
|
Yan Y, Fu G, Ming L. Role of exosomes in pancreatic cancer. Oncol Lett 2018; 15:7479-7488. [PMID: 29731898 PMCID: PMC5920881 DOI: 10.3892/ol.2018.8348] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/20/2017] [Indexed: 01/10/2023] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies. Exosomes, which are released by multiple cell types, such as cancer cells, contain functional biomolecules (including proteins, nucleic acids and lipids) that can be horizontally delivered to recipient cells. Exosomes act as the most prominent mediator of intercellular communication and can regulate, instruct and re-educate their surrounding microenvironment and target specific organs. The present review performed an extensive search of multiple databases from 2005 to April 23 2017, for eligible literature relating to exosomes and their role in pancreatic cancer. With a focus on the latest findings for pancreatic cancer exosomes, their role in tumorigenesis was summarized, as well as their aggressive behaviors and their contribution to immunosuppression and therapy resistance in pancreatic cancer. In addition, the potential function of exosomes as novel diagnostic biomarkers is briefly discussed. Finally, we propose potential clinical applications for exosomes in pancreatic cancer.
Collapse
Affiliation(s)
- Yunmeng Yan
- Department of Clinical Laboratory and Key Clinical Laboratory of The Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Guangzhen Fu
- Department of Clinical Laboratory and Key Clinical Laboratory of The Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Liang Ming
- Department of Clinical Laboratory and Key Clinical Laboratory of The Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
76
|
Fu Y, Liu S, Zeng S, Shen H. The critical roles of activated stellate cells-mediated paracrine signaling, metabolism and onco-immunology in pancreatic ductal adenocarcinoma. Mol Cancer 2018; 17:62. [PMID: 29458370 PMCID: PMC5817854 DOI: 10.1186/s12943-018-0815-z] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/12/2018] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignant diseases worldwide. It is refractory to conventional treatments, and consequently has a documented 5-year survival rate as low as 7%. Increasing evidence indicates that activated pancreatic stellate cells (PSCs), one of the stromal components in tumor microenvironment (TME), play a crucial part in the desmoplasia, carcinogenesis, aggressiveness, metastasis associated with PDAC. Despite the current understanding of PSCs as a "partner in crime" to PDAC, detailed regulatory roles of PSCs and related microenvironment remain obscure. In addition to multiple paracrine signaling pathways, recent research has confirmed that PSCs-mediated tumor microenvironment may influence behaviors of PDAC via diverse mechanisms, such as rewiring metabolic networks, suppressing immune responses. These new activities are closely linked with treatment and prognosis of PDAC. In this review, we discuss the recent advances regarding new functions of activated PSCs, including PSCs-cancer cells interaction, mechanisms involved in immunosuppressive regulation, and metabolic reprogramming. It's clear that these updated experimental or clinical studies of PSCs may provide a promising approach for PDAC treatment in the near future.
Collapse
Affiliation(s)
- Yaojie Fu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Shanshan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
77
|
Qiu J, Yang G, Feng M, Zheng S, Cao Z, You L, Zheng L, Zhang T, Zhao Y. Extracellular vesicles as mediators of the progression and chemoresistance of pancreatic cancer and their potential clinical applications. Mol Cancer 2018; 17:2. [PMID: 29304816 PMCID: PMC5756395 DOI: 10.1186/s12943-017-0755-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 12/27/2017] [Indexed: 12/17/2022] Open
Abstract
Pancreatic cancer is one of the most lethal cancers worldwide due to its insidious symptoms, early metastasis, and chemoresistance. Hence, the underlying mechanisms contributing to pancreatic cancer progression require further exploration. Based on accumulating evidence, extracellular vesicles, including exosomes and microvesicles, play a crucial role in pancreatic cancer progression and chemoresistance. Furthermore, they also possess the potential to be promising biomarkers, therapy targets and tools for treating pancreatic cancer. Therefore, in-depth studies on the role of extracellular vesicles in pancreatic cancer are meaningful. In this review, we focus on the regulatory effects of extracellular vesicles on pancreatic cancer progression, metastasis, cancer-related immunity and chemoresistance, particularly their potential roles as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Mengyu Feng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Suli Zheng
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China. .,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, 100730, China.
| |
Collapse
|
78
|
Han S, Gonzalo DH, Feely M, Rinaldi C, Belsare S, Zhai H, Kalra K, Gerber MH, Forsmark CE, Hughes SJ. Stroma-derived extracellular vesicles deliver tumor-suppressive miRNAs to pancreatic cancer cells. Oncotarget 2017; 9:5764-5777. [PMID: 29464032 PMCID: PMC5814172 DOI: 10.18632/oncotarget.23532] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/28/2017] [Indexed: 01/18/2023] Open
Abstract
The biology of tumor-associated stroma (TAS) in pancreatic ductal adenocarcinoma (PDAC) is not well understood. The paradoxical observation that stroma-depletion strategies lead to progression of PDAC reinforced the need to critically evaluate the functional contribution of TAS in the initiation and progression of PDAC. PDAC and TAS cells are unique in their expression of specific miRNAs, and this specific miRNA expression pattern alters host to tumor microenvironment interactions. Using primary human pancreatic TAS cells and primary xenograft PDAC cells co-culture, we provide evidence of miRNA trafficking and exchanging between TAS and PDAC cells, in a two-way, cell-contact independent fashion, via extracellular vesicles (EVs) transportation. Selective packaging of miRNAs into EVs led to enrichment of stromal specific miR-145 in EVs secreted by TAS cells. Exosomes, but not microvesicles, derived from human TAS cells demonstrated a tumor suppressive role by inducing PDAC cell apoptosis. This effect was mitigated by anti-miR-145 sequences. Our data suggest that TAS-derived miRNAs are delivered to adjacent PDAC cells via exosomes and suppress tumor cell growth. These data highlight that TAS cells secrete exosomes carrying tumor suppressive genetic materials, a possible anti-tumor capacity. Future work of the development of patient-derived exosomes could have therapeutic implications for unresectable PDAC.
Collapse
Affiliation(s)
- Song Han
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - David H Gonzalo
- Department of Pathology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Michael Feely
- Department of Pathology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Carlos Rinaldi
- Department of Biomedical Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | - Sayali Belsare
- Department of Biomedical Engineering, University of Florida College of Medicine, Gainesville, FL, USA
| | | | | | - Michael H Gerber
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christopher E Forsmark
- Division of Gastroenterology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Steven J Hughes
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, USA
| |
Collapse
|
79
|
Fibrosis-related miRNAs as serum biomarkers for pancreatic ductal adenocarcinoma. Oncotarget 2017; 9:4451-4460. [PMID: 29435115 PMCID: PMC5796986 DOI: 10.18632/oncotarget.23377] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 11/29/2017] [Indexed: 12/17/2022] Open
Abstract
We investigated whether serum microRNAs (miRNAs) could be diagnostic or prognostic markers in pancreatic ductal adenocarcinoma (PDAC). We first identified miRNAs showing altered expression in human pancreatic stellate cells (hPSCs) co-cultured with PDAC cells (Panc-1 and BxPC-3) as compared to hPSCs cultured alone. Among the miRNAs with altered expression, let-7d exhibited reduced expression in an in silico analysis of The Cancer Genome Atlas data. Inhibition of let-7d resulted in enhanced expression of fibrosis-related genes. We extracted serum miRNA from 45 PDAC patients and 42 healthy controls and quantified expression let-7d using digital PCR. Based on the level of let-7d expression, we were able to distinguish between PDAC patients and controls. Additionally, reduced let-7d expression correlated with poor overall survival. Thus, fibrosis-related miRNAs may be serum biomarkers for PDAC.
Collapse
|
80
|
Barteneva NS, Baiken Y, Fasler-Kan E, Alibek K, Wang S, Maltsev N, Ponomarev ED, Sautbayeva Z, Kauanova S, Moore A, Beglinger C, Vorobjev IA. Extracellular vesicles in gastrointestinal cancer in conjunction with microbiota: On the border of Kingdoms. Biochim Biophys Acta Rev Cancer 2017; 1868:372-393. [DOI: 10.1016/j.bbcan.2017.06.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/26/2017] [Accepted: 06/26/2017] [Indexed: 12/16/2022]
|
81
|
Masamune A, Yoshida N, Hamada S, Takikawa T, Nabeshima T, Shimosegawa T. Exosomes derived from pancreatic cancer cells induce activation and profibrogenic activities in pancreatic stellate cells. Biochem Biophys Res Commun 2017; 495:71-77. [PMID: 29111329 DOI: 10.1016/j.bbrc.2017.10.141] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/26/2017] [Indexed: 12/21/2022]
Abstract
Pancreatic cancer cells (PCCs) interact with pancreatic stellate cells (PSCs), which play a pivotal role in pancreatic fibrogenesis, to develop the cancer-conditioned tumor microenvironment. Exosomes are membrane-enclosed nanovesicles, and have been increasingly recognized as important mediators of cell-to-cell communications. The aim of this study was to clarify the effects of PCC-derived exosomes on cell functions in PSCs. Exosomes were isolated from the conditioned medium of Panc-1 and SUIT-2 PCCs. Human primary PSCs were treated with PCC-derived exosomes. PCC-derived exosomes stimulated the proliferation, migration, activation of ERK and Akt, the mRNA expression of α-smooth muscle actin (ACTA2) and fibrosis-related genes, and procollagen type I C-peptide production in PSCs. Ingenuity pathway analysis of the microarray data identified transforming growth factor β1 and tumor necrosis factor as top upstream regulators. PCCs increased the expression of miR-1246 and miR-1290, abundantly contained in PCC-derived exosomes, in PSCs. Overexpression of miR-1290 induced the expression of ACTA2 and fibrosis-related genes in PSCs. In conclusion, PCC-derived exosomes stimulate activation and profibrogenic activities in PSCs. Exosome-mediated interactions between PSCs and PCCs might play a role in the development of the tumor microenvironment.
Collapse
Affiliation(s)
- Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Naoki Yoshida
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shin Hamada
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Takikawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tatsuhide Nabeshima
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
82
|
Rani A, O'Shea A, Ianov L, Cohen RA, Woods AJ, Foster TC. miRNA in Circulating Microvesicles as Biomarkers for Age-Related Cognitive Decline. Front Aging Neurosci 2017; 9:323. [PMID: 29046635 PMCID: PMC5632661 DOI: 10.3389/fnagi.2017.00323] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 09/20/2017] [Indexed: 11/13/2022] Open
Abstract
Community dwelling older individuals from the North Florida region were examined for health status and a comprehensive neuropsychological battery, including the Montreal Cognitive Assessment (MoCA), was performed on each participant. A subpopulation (58 females and 39 males) met the criteria for age (60–89) and no evidence of mild cognitive impairment, with a MoCA score ≥23. Despite the stringent criteria for participation, MoCA scores were negatively correlated within the limited age range. Extracellular microvesicles were isolated from the plasma and samples were found to be positive for the exosome marker CD63, with an enrichment of particles within the size range for exosomes. miRNA was extracted and examined using next generation sequencing with a stringent criterion (average of ≥10 counts per million reads) resulting in 117 miRNA for subsequent analysis. Characterization of expression confirmed pervious work concerning the relative abundance and overall pattern of expression of miRNA in plasma. Correlation analysis indicated that most of the miRNAs (74 miRNAs) were positively correlated with age (p <0.01). Multiple regression was employed to identify the relationship of miRNA expression and MoCA score, accounting for age. MoCA scores were negatively correlated with 13 miRNAs. The pattern of expression for cognition-related miRNA did not match that previously described for Alzheimer’s disease. Enrichment analysis was employed to identify miRNA–gene interactions to reveal possible links to brain function.
Collapse
Affiliation(s)
- Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Andrew O'Shea
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States.,Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Lara Ianov
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Genetics and Genomics Program, Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Ronald A Cohen
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States.,Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Adam J Woods
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States.,Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, United States
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.,Genetics and Genomics Program, Genetics Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
83
|
Abstract
PURPOSE OF REVIEW Pancreatic stellate cells (PSCs) play an integral role in the pathogenesis of pancreatitis and pancreatic cancer. With the developing knowledge of this important cell type, we are at the cusp of developing effective therapies for the above diseases based upon targeting the PSC and modulating its function. RECENT FINDINGS The major themes of the recent PSC literature include: PSC interactions with the extracellular matrix and other stromal components; intracellular calcium physiology as drivers of mechanical interactions and necrosis; the relationship between proinflammatory, protumoural, angiogenic, and metabolic pathways in pancreatic necrosis, fibrosis, and carcinogenesis; and targeting of the stroma for antitumoural and antifibrotic effects. SUMMARY Traditionally, there have been few treatment options for pancreatitis and pancreatic cancer. The elucidation of the wide-ranging functions of PSCs provide an opportunity for treatments based on stromal reprogramming.
Collapse
|
84
|
Liu Q, Liao Q, Zhao Y. Chemotherapy and tumor microenvironment of pancreatic cancer. Cancer Cell Int 2017; 17:68. [PMID: 28694739 PMCID: PMC5498917 DOI: 10.1186/s12935-017-0437-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is an extremely dismal malignance. Chemotherapy has been widely applied to treat this intractable tumor. It has exclusive tumor microenvironment (TME), characterized by dense desmoplasia and profound infiltrations of immunosuppressive cells. Interactions between stromal cells and cancer cells play vital roles to affect the biological behaviors of pancreatic cancer. Targeting the stromal components of pancreatic cancer has shown promising results. In addition to the direct toxic effects of chemotherapeutic drugs on cancer cells, they can also remodel the TME, eventually affecting their efficacy. Herein, we reviewed the following four aspects; (1) clinical landmark advances of chemotherapy in pancreatic cancer, since 2000; (2) interactions and mechanisms between stromal cells and pancreatic cancer cells; (3) remodeling effects and mechanisms of chemotherapy on TME; (4) targeting stromal components in pancreatic cancer.
Collapse
Affiliation(s)
- Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| |
Collapse
|
85
|
Makler A, Narayanan R. Mining Exosomal Genes for Pancreatic Cancer Targets. Cancer Genomics Proteomics 2017; 14:161-172. [PMID: 28446531 PMCID: PMC5420817 DOI: 10.21873/cgp.20028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/03/2017] [Accepted: 04/05/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Exosomes, cell-derived vesicles encompassing lipids, DNA, proteins coding genes and noncoding RNAs (ncRNAs) are present in diverse body fluids. They offer novel biomarker and drug therapy potential for diverse diseases, including cancer. MATERIALS AND METHODS Using gene ontology, exosomal genes database and GeneCards metadata analysis tools, a database of cancer-associated protein coding genes and ncRNAs (n=2,777) was established. Variant analysis, expression profiling and pathway mapping were used to identify putative pancreatic cancer exosomal gene candidates. RESULTS Five hundred and seventy-five protein-coding genes, 26 RNA genes and one pseudogene directly associated with pancreatic cancer were identified in the study. Nine open reading frames (ORFs) encompassing enzymes, apoptosis and transcriptional regulators, and secreted factors and five cDNAs (enzymes) emerged from the analysis. Among the ncRNA class, 26 microRNAs (miRs), one pseudogene, one long noncoding RNA (LNC) and one antisense gene were identified. Furthermore, 22 exosome-associated protein-coding targets (a cytokine, enzymes, membrane glycoproteins, receptors, and a transporter) emerged as putative leads for pancreatic cancer therapy. Seven of these protein-coding targets are FDA-approved and the drugs-based on these could provide repurposing opportunities for pancreatic cancer. CONCLUSION The database of exosomal genes established in this study provides a framework for developing novel biomarkers and drug therapy targets for pancreatic cancer.
Collapse
Affiliation(s)
- Amy Makler
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, U.S.A
| | - Ramaswamy Narayanan
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, U.S.A.
| |
Collapse
|
86
|
Exosome-Mediated Intercellular Communication Between Stellate Cells and Cancer Cells in Pancreatic Ductal Adenocarcinoma. Pancreas 2017; 46:1-4. [PMID: 27977625 PMCID: PMC5171232 DOI: 10.1097/mpa.0000000000000686] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has long being viewed as a consequence of recurrent or unresolved inflammation, associated with fibrogenesis and extensive cross-communication between different cell types within the tumor microenvironment. A plethora of cytokines, chemokines and DAMPs such as TNF-α, IL-6, MCP-1, ATP and HMGB1, produced in response to pancreas damage from injury fuel inflammatory and fibrogenic responses. These and other factors contribute to the intercellular interplay within the complex milieu of the tumor microenvironment. However, precisely which factors represent appropriate or inappropriate signals, which cell type(s) produces the factors, and the detailed understanding of the sequence of events by which regeneration and malignant transformation are orchestrated have not yet been unraveled. Clearly, further understanding of cause-and effect relationships between the diverse cell populations and the factors they release into the microenvironment is urgently needed.
Collapse
|