1
|
Soni N, Bissa B. Exosomes, circadian rhythms, and cancer precision medicine: New frontiers. Biochimie 2024; 227:172-181. [PMID: 39032591 DOI: 10.1016/j.biochi.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/23/2024]
Abstract
"The environment shapes people's actions," a well-known proverb, strongly dictates that a change in our way of life changes our behavior. Circadian rhythms have been identified as a mechanism for maintaining homeostasis in the body, which, if disrupted by sleeping patterns, could result in significant metabolic alterations that adversely affect our health. The changes induced by circadian rhythm alter the secretion and cargo selection in exosomes which are nanovesicles important for intercellular communication. Exosomes were formerly known as "junk particles" but are now recognized as miniature copies of a cell's genetic material. Dysregulation of circadian rhythm has shown that it changes the gene expression of a cell to some extent and significantly alters the exosomal release. Meanwhile, cells secrete exosomes continuously to align the rhythmicity of the biological clock. In this study, we integrate circadian rhythms and exosomes with precision medicines to find better approaches to early diagnosis and treatment of disease.
Collapse
Affiliation(s)
- Naveen Soni
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Bhawana Bissa
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India.
| |
Collapse
|
2
|
Huang C, Zhang J, Wang H, Liang C. Exosomes That Have Different Cellular Origins Followed by the Impact They Have on Prostate Tumor Development in the Tumor Microenvironment. Cancer Rep (Hoboken) 2024; 7:e70001. [PMID: 39229670 PMCID: PMC11372288 DOI: 10.1002/cnr2.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/15/2024] [Accepted: 08/11/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the most common urinary tumor with the highest incidence rate and the second among the leading causes of death worldwide for adult males. In the worldwide cancer incidence rate, PCa is on the increase. The cancerous cells in the prostate and cells in the microenvironment surrounding the tumor communicate through signal transduction, which is crucial for the development and spread of PCa. RECENT FINDINGS Exosomes are nanoscale vesicles released into body fluids by various cells that can aid intercellular communication by releasing nucleic acids and proteins. Exosomes published by different types of cells in the tumor microenvironment can have varying impacts on the proliferation and growth of tumor cells via various signaling pathways, modes of action, and secreted cytokines. CONCLUSION The main purpose of this review is to describe the effects of different cell-derived exosomes in the tumor microenvironment of PCa on the progression of tumor cells, as well as to summarize and discuss the prospects for the application of exosomes in the treatment and diagnosis of PCa.
Collapse
Affiliation(s)
- Cong Huang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Key Laboratory of Genitourinary Diseases Anhui Province, Anhui Medical University, Hefei, China
| | - Jialong Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Key Laboratory of Genitourinary Diseases Anhui Province, Anhui Medical University, Hefei, China
| | - Hongzhi Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Key Laboratory of Genitourinary Diseases Anhui Province, Anhui Medical University, Hefei, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- Institute of Urology, Anhui Medical University, Hefei, China
- Key Laboratory of Genitourinary Diseases Anhui Province, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Yang Z, Chen JQ, Liu TJ, Chen YL, Ma ZK, Fan YZ, Wang ZX, Xu S, Wang K, Wang XY, Li L, Xie HJ. Knocking down AR promotes osteoblasts to recruit prostate cancer cells by altering exosomal circ-DHPS/miR-214-3p/CCL5 pathway. Asian J Androl 2024; 26:195-204. [PMID: 37966336 PMCID: PMC10919426 DOI: 10.4103/aja202351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/04/2023] [Indexed: 11/16/2023] Open
Abstract
Tumor-derived exosomes have been shown to play a key role in organ-specific metastasis, and the androgen receptor regulates prostate cancer (PCa) progression. It is unclear whether the androgen receptor regulates the recruitment of prostate cancer cells to the bone microenvironment, even bone metastases, through exosomes. Here, we found that exosomes isolated from PCa cells after knocking down androgen receptor (AR) or enzalutamide treatment can facilitate the migration of prostate cancer cells to osteoblasts. In addition, AR silencing or treatment with the AR antagonist enzalutamide may increase the expression of circular RNA-deoxyhypusine synthase (circ-DHPS) in PCa cells, which can be transported to osteoblasts by exosomes. Circ-DHPS acts as a competitive endogenous RNA (ceRNA) against endogenous miR-214-3p to promote C-C chemokine ligand 5 ( CCL5 ) levels in osteoblasts. Increasing the level of CCL5 in osteoblasts could recruit more PCa cells into the bone microenvironment. Thus, blocking the circ-DHPS/miR-214-3p/CCL5 signal may decrease exosome-mediated migration of prostate cancer cells to osteoblasts.
Collapse
Affiliation(s)
- Zhao Yang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Jia-Qi Chen
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Tian-Jie Liu
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yu-Le Chen
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Zhen-Kun Ma
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yi-Zeng Fan
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Zi-Xi Wang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Xin-Yang Wang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Hong-Jun Xie
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
4
|
Dey D, Ghosh S, Mirgh D, Panda SP, Jha NK, Jha SK. Role of exosomes in prostate cancer and male fertility. Drug Discov Today 2023; 28:103791. [PMID: 37777169 DOI: 10.1016/j.drudis.2023.103791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/09/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Prostate cancer (PCa) is the second most common and fifth most aggressive neoplasm among men worldwide. In the last decade, extracellular vesicle (EV) research has decoded multiple unsolved cancer-related mysteries. EVs can be classified as microvesicles, apoptotic bodies, and exosomes, among others. Exosomes play a key role in cellular signaling. Their internal cargos (nucleic acids, proteins, lipids) influence the recipient cell. In PCa, the exosome is the regulator of cancer progression. It is also a promising theranostics tool for PCa. Moreover, exosomes have strong participation in male fertility complications. This review aims to highlight the exosome theranostics signature in PCa and its association with male fertility.
Collapse
Affiliation(s)
- Dwaipayan Dey
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, Rahara, West Bengal 700118, India
| | - Srestha Ghosh
- Department of Microbiology, Lady Brabourne College, Kolkata 700017, West Bengal, India
| | - Divya Mirgh
- Johns Hopkins University, Baltimore, MD 21218, USA
| | - Siva Parsad Panda
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India.
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Department of Biotechnology, School of Applied and Life Sciences (SALS), Uttaranchal, University, Dehradun, India.
| |
Collapse
|
5
|
Soupir AC, Tian Y, Stewart PA, Nunez-Lopez YO, Manley BJ, Pellini B, Bloomer AM, Zhang J, Mo Q, Marchion DC, Liu M, Koomen JM, Siegel EM, Wang L. Detectable Lipidomes and Metabolomes by Different Plasma Exosome Isolation Methods in Healthy Controls and Patients with Advanced Prostate and Lung Cancer. Int J Mol Sci 2023; 24:1830. [PMID: 36768152 PMCID: PMC9916336 DOI: 10.3390/ijms24031830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Circulating exosomes in the blood are promising tools for biomarker discovery in cancer. Due to their heterogeneity, different isolation methods may enrich distinct exosome cargos generating different omic profiles. In this study, we evaluated the effects of plasma exosome isolation methods on detectable multi-omic profiles in patients with non-small cell lung cancer (NSCLC), castration-resistant prostate cancer (CRPC), and healthy controls, and developed an algorithm to quantify exosome enrichment. Plasma exosomes were isolated from CRPC (n = 10), NSCLC (n = 14), and healthy controls (n = 10) using three different methods: size exclusion chromatography (SEC), lectin binding, and T-cell immunoglobulin domain and mucin domain-containing protein 4 (TIM4) binding. Molecular profiles were determined by mass spectrometry of extracted exosome fractions. Enrichment analysis of uniquely detected molecules was performed for each method with MetaboAnalyst. The exosome enrichment index (EEI) scores methods based on top differential molecules between patient groups. The lipidomic analysis detected 949 lipids using exosomes from SEC, followed by 246 from lectin binding and 226 from TIM4 binding. The detectable metabolites showed SEC identifying 191 while lectin binding and TIM4 binding identified 100 and 107, respectively. When comparing uniquely detected molecules, different methods showed preferential enrichment of different sets of molecules with SEC enriching the greatest diversity. Compared to controls, SEC identified 28 lipids showing significant difference in NSCLC, while only 1 metabolite in NSCLC and 5 metabolites in CRPC were considered statistically significant (FDR < 0.1). Neither lectin-binding- nor TIM4-binding-derived exosome lipids or metabolites demonstrated significant differences between patient groups. We observed the highest EEI from SEC in lipids (NSCLC: 871.33) which was also noted in metabolites. These results support that the size exclusion method of exosome extraction implemented by SBI captures more heterogeneous exosome populations. In contrast, lectin-binding and TIM4-binding methods bind surface glycans or phosphatidylserine moieties of the exosomes. Overall, these findings suggest that specific isolation methods select subpopulations which may significantly impact cancer biomarker discovery.
Collapse
Affiliation(s)
- Alex C. Soupir
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Yijun Tian
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Paul A. Stewart
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Yury O. Nunez-Lopez
- Advent Health, Translational Research Institute for Metabolism and Diabetes, Orlando, FL 32804, USA
| | - Brandon J. Manley
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Bruna Pellini
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Amanda M. Bloomer
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jingsong Zhang
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Qianxing Mo
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Min Liu
- Proteomics & Metabolomics Core, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - John M. Koomen
- Department of Molecular Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Erin M. Siegel
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Liang Wang
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
6
|
Folkmanis K, Junk E, Merdane E, Folkmane I, Folkmanis V, Ivanovs I, Eglitis J, Jakubovskis M, Laabs S, Isajevs S, Lietuvietis V. Clinicopathological Significance of Exosomal Proteins CD9 and CD63 and DNA Mismatch Repair Proteins in Prostate Adenocarcinoma and Benign Hyperplasia. Diagnostics (Basel) 2022; 12:diagnostics12020287. [PMID: 35204378 PMCID: PMC8871402 DOI: 10.3390/diagnostics12020287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction. Recently, it has been shown that exosomal biomarkers and DNA mismatch repair proteins (MMR) could play an important role in cancer risk stratification and prognosis assessment. The gold standard for prostate carcinoma (PCa) diagnosis is biopsy and histopathological examination. Thus, the complex evaluation of exosomal and MMR proteins could be beneficial for prostate cancer risk stratification and diagnostics. The aim of the current study was to evaluate and compare the expression of exosomal proteins CD9 and CD63 and MMR proteins in the tissue of patients with prostate benign hyperplasia (BPH) and PCa. Methods. The study was retrospective. Altogether, 92 patients with PCa and 20 patients with BPH (control group) were enrolled in the study. Exosomal and MMR protein expression was analyzed by immunohistochemistry (IHC). The follow-up for each PCa patient in our study lasted till disease progression and/or a maximum of 5 years. Results. Low-grade PCa was observed in 56 patients and high-grade PCa in 36 patients. CD63 expression was significantly higher in patients with high-grade PCa compared to those with low-grade PCa. CD9 expression was significantly downregulated in PCa patients compared to the control group. MMR protein expression deficiency was observed in 10 PCa patients. MMR proteins were maintained in all cases of BPH. The study found a negative correlation between MMR protein loss and PCa ISUP grade groups. Progression-free survival (PFS) in patients with MMR deficiency was significantly shorter than in patients with maintained MMR expression. Conclusions. CD9 protein expression was downregulated in PCa, compared to BPH, while CD63 protein expression was upregulated in high-grade PCa but downregulated in low-grade PCa. CD63 protein upregulation, CD9 downregulation, and loss of MMR protein characterized the shorter PFS of high-grade PCa patients. CD9, CD63, and MMR could be the routine immunohistochemical biomarkers for the diagnosis and risk stratification of PCa.
Collapse
Affiliation(s)
- Kristofs Folkmanis
- Faculty of Medicine, University of Latvia, LV-1004 Riga, Latvia; (E.J.); (E.M.); (I.F.); (V.F.); (I.I.); (J.E.); (S.I.)
- Department of Urology, Elbe Hospital in Stade—Teaching Hospital of Hamburg-Eppendorf University Hospital, 20246 Stade, Germany;
- Correspondence: ; Tel.: +49-152-136-57241
| | - Elizabete Junk
- Faculty of Medicine, University of Latvia, LV-1004 Riga, Latvia; (E.J.); (E.M.); (I.F.); (V.F.); (I.I.); (J.E.); (S.I.)
| | - Evelina Merdane
- Faculty of Medicine, University of Latvia, LV-1004 Riga, Latvia; (E.J.); (E.M.); (I.F.); (V.F.); (I.I.); (J.E.); (S.I.)
| | - Inese Folkmane
- Faculty of Medicine, University of Latvia, LV-1004 Riga, Latvia; (E.J.); (E.M.); (I.F.); (V.F.); (I.I.); (J.E.); (S.I.)
| | - Valdis Folkmanis
- Faculty of Medicine, University of Latvia, LV-1004 Riga, Latvia; (E.J.); (E.M.); (I.F.); (V.F.); (I.I.); (J.E.); (S.I.)
| | - Igors Ivanovs
- Faculty of Medicine, University of Latvia, LV-1004 Riga, Latvia; (E.J.); (E.M.); (I.F.); (V.F.); (I.I.); (J.E.); (S.I.)
- Department of Urology, East Clinical University Hospital, LV-1007 Riga, Latvia; (M.J.); (V.L.)
| | - Janis Eglitis
- Faculty of Medicine, University of Latvia, LV-1004 Riga, Latvia; (E.J.); (E.M.); (I.F.); (V.F.); (I.I.); (J.E.); (S.I.)
- Department of Urology, East Clinical University Hospital, LV-1007 Riga, Latvia; (M.J.); (V.L.)
| | - Maris Jakubovskis
- Department of Urology, East Clinical University Hospital, LV-1007 Riga, Latvia; (M.J.); (V.L.)
- Department of Urology, Faculty of Medicine, Riga Stradins University, LV-1007 Riga, Latvia
| | - Sven Laabs
- Department of Urology, Elbe Hospital in Stade—Teaching Hospital of Hamburg-Eppendorf University Hospital, 20246 Stade, Germany;
| | - Sergejs Isajevs
- Faculty of Medicine, University of Latvia, LV-1004 Riga, Latvia; (E.J.); (E.M.); (I.F.); (V.F.); (I.I.); (J.E.); (S.I.)
- Department of Urology, East Clinical University Hospital, LV-1007 Riga, Latvia; (M.J.); (V.L.)
| | - Vilnis Lietuvietis
- Department of Urology, East Clinical University Hospital, LV-1007 Riga, Latvia; (M.J.); (V.L.)
- Department of Urology, Faculty of Medicine, Riga Stradins University, LV-1007 Riga, Latvia
| |
Collapse
|
7
|
Hosseini K, Ranjbar M, Pirpour Tazehkand A, Asgharian P, Montazersaheb S, Tarhriz V, Ghasemnejad T. Evaluation of exosomal non-coding RNAs in cancer using high-throughput sequencing. J Transl Med 2022; 20:30. [PMID: 35033106 PMCID: PMC8760667 DOI: 10.1186/s12967-022-03231-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 01/05/2022] [Indexed: 12/13/2022] Open
Abstract
Clinical oncologists need more reliable and non-invasive diagnostic and prognostic biomarkers to follow-up cancer patients. However, the existing biomarkers are often invasive and costly, emphasizing the need for the development of biomarkers to provide convenient and precise detection. Extracellular vesicles especially exosomes have recently been the focus of translational research to develop non-invasive and reliable biomarkers for several diseases such as cancers, suggesting as a valuable source of tumor markers. Exosomes are nano-sized extracellular vesicles secreted by various living cells that can be found in all body fluids including serum, urine, saliva, cerebrospinal fluid, and ascites. Different molecular and genetic contents of their origin such as nucleic acids, proteins, lipids, and glycans in a stable form make exosomes a promising approach for various cancers' diagnoses, prediction, and follow-up in a minimally invasive manner. Since exosomes are used by cancer cells for intercellular communication, they play a critical role in the disease process, highlighting the importance of their use as clinically relevant biomarkers. However, regardless of the advantages that exosome-based diagnostics have, they suffer from problems regarding their isolation, detection, and characterization of their contents. This study reviews the history and biogenesis of exosomes and discusses non-coding RNAs (ncRNAs) and their potential as tumor markers in different types of cancer, with a focus on next generation sequencing (NGS) as a detection method. Moreover, the advantages and challenges associated with exosome-based diagnostics are also presented.
Collapse
Affiliation(s)
- Kamran Hosseini
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Ranjbar
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Pirpour Tazehkand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parina Asgharian
- Department of Pharmacognosy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Tohid Ghasemnejad
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Li H, Zheng Q, Xie X, Wang J, Zhu H, Hu H, He H, Lu Q. Role of Exosomal Non-Coding RNAs in Bone-Related Diseases. Front Cell Dev Biol 2022; 9:811666. [PMID: 35004702 PMCID: PMC8733689 DOI: 10.3389/fcell.2021.811666] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Bone-related diseases seriously affect the lives of patients and carry a heavy economic burden on society. Treatment methods cannot meet the diverse clinical needs of affected patients. Exosomes participate in the occurrence and development of many diseases through intercellular communication, including bone-related diseases. Studies have shown that exosomes can take-up and “package” non-coding RNAs and “deliver” them to recipient cells, thereby regulating the function of recipient cells. The exosomal non-coding RNAs secreted by osteoblasts, osteoclasts, chondrocytes, and other cells are involved in the regulation of bone-related diseases by inhibiting osteoclasts, enhancing chondrocyte activity and promoting angiogenesis. Here, we summarize the role and therapeutic potential of exosomal non-coding RNAs in the bone-related diseases osteoporosis, osteoarthritis, and bone-fracture healing, and discuss the clinical application of exosomes in patients with bone-related diseases.
Collapse
Affiliation(s)
- Hang Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Qiyue Zheng
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinyan Xie
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China.,College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaojiao Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Haihong Zhu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Haoye Hu
- Department of Medical Genetics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hao He
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
9
|
Martens‐Uzunova ES, Kusuma GD, Crucitta S, Lim HK, Cooper C, Riches JE, Azad A, Ochiya T, Boyle GM, Southey MC, Del Re M, Lim R, Ramm GA, Jenster GW, Soekmadji C. Androgens alter the heterogeneity of small extracellular vesicles and the small RNA cargo in prostate cancer. J Extracell Vesicles 2021; 10:e12136. [PMID: 34434533 PMCID: PMC8374107 DOI: 10.1002/jev2.12136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022] Open
Abstract
Proliferation and survival of prostate cancer cells are driven by the androgen receptor (AR) upon binding to androgen steroid hormones. Manipulating the AR signalling axis is the focus for prostate cancer therapy; thus, it is crucial to understand the role of androgens and AR on extracellular vesicle (EV) secretion and cargo. In this study, we report that plasma-derived circulating vesicles consisting of CD9 and double-positive for CD9 and Prostate Specific Membrane Antigen (PSMA) are increased in patients with advanced metastatic prostate cancer, whereas double positives for CD9 and CD63 small extracellular vesicles (S-EVs) are significantly higher in patients with localised prostate cancer. Androgen manipulation by dihydrotestosterone (DHT) and the clinical antagonist enzalutamide (ENZ) altered the heterogeneity and size of CD9 positive S-EVs in AR expressing prostate cancer cells, while assessment of the total number and protein cargo of total S-EVs was unaltered across different treatment groups. Furthermore, hormone stimulation caused strong and specific effects on the small RNA cargo of S-EVs. A total of 543 small RNAs were found to be regulated by androgens including miR-19-3p and miR-361-5p. Analysis of S-EVs heterogeneity and small RNA cargo may provide clinical utility for prostate cancer and be informative to understand further the mechanism of resistance to androgen targeted therapy in castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Elena S. Martens‐Uzunova
- Department of Urology, Erasmus MC, Cancer InstituteUniversity Medical Centre RotterdamRotterdamThe Netherlands
| | - Gina D. Kusuma
- The Ritchie Centre, Hudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity Hospital of PisaPisaItaly
| | - Hong Kiat Lim
- Department of Cell and Molecular BiologyQIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Crystal Cooper
- Central Analytical Research FacilityInstitute for Future EnvironmentsQueensland University of TechnologyBrisbaneAustralia
| | - James E. Riches
- Central Analytical Research FacilityInstitute for Future EnvironmentsQueensland University of TechnologyBrisbaneAustralia
| | - Arun Azad
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneParkvilleVictoriaAustralia
- Department of Medical OncologyPeter MacCallum Cancer CentreMelbourneAustralia
| | - Takahiro Ochiya
- Institute of Medical ScienceTokyo Medical UniversityTokyoJapan
| | - Glen M. Boyle
- Department of Cell and Molecular BiologyQIMR Berghofer Medical Research InstituteBrisbaneAustralia
- School of Biomedical Sciences, Faculty of MedicineUniversity of QueenslandBrisbaneAustralia
| | - Melissa C. Southey
- Genetic Epidemiology Laboratory, Department of PathologyThe University of MelbourneMelbourneAustralia
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental MedicineUniversity Hospital of PisaPisaItaly
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityClaytonVictoriaAustralia
| | - Grant A. Ramm
- Department of Cell and Molecular BiologyQIMR Berghofer Medical Research InstituteBrisbaneAustralia
- School of Biomedical Sciences, Faculty of MedicineUniversity of QueenslandBrisbaneAustralia
| | - Guido W. Jenster
- Department of Urology, Erasmus MC, Cancer InstituteUniversity Medical Centre RotterdamRotterdamThe Netherlands
| | - Carolina Soekmadji
- Department of Cell and Molecular BiologyQIMR Berghofer Medical Research InstituteBrisbaneAustralia
- School of Biomedical Sciences, Faculty of MedicineUniversity of QueenslandBrisbaneAustralia
| |
Collapse
|
10
|
Urabe F, Patil K, Ramm GA, Ochiya T, Soekmadji C. Extracellular vesicles in the development of organ-specific metastasis. J Extracell Vesicles 2021; 10:e12125. [PMID: 34295457 PMCID: PMC8287318 DOI: 10.1002/jev2.12125] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
Distant organ metastasis, often termed as organotropic metastasis or metastatic organotropism, is a fundamental feature of malignant tumours and accounts for most cancer-related mortalities. This process is orchestrated by many complex biological interactions and processes that are mediated by a combination of anatomical, genetic, pathophysiological and biochemical factors. Recently, extracellular vesicles (EVs) are increasingly being demonstrated as critical mediators of bi-directional tumour-host cell interactions, controlling organ-specific infiltration, adaptation and colonization at the secondary site. EVs govern organotropic metastasis by modulating the pre-metastatic microenvironment through upregulation of pro-inflammatory gene expression and immunosuppressive cytokine secretion, induction of phenotype-specific differentiation and recruitment of specific stromal cell types. This review discusses EV-mediated metastatic organotropism in visceral (brain, lung, liver, and lymph node) and skeletal (bone) metastasis, and discusses how the pre-metastatic education by EVs transforms the organ into a hospitable, tumour cell-friendly milieu that supports the growth of metastatic cells. Decoding the organ-specific traits of EVs and their functions in organotropic metastasis is essential in accelerating the clinical application of EVs in cancer management.
Collapse
Affiliation(s)
- Fumihiko Urabe
- Department of UrologyThe Jikei University School of MedicineTokyoJapan
- Department of Molecular and Cellular MedicineTokyo Medical UniversityTokyoJapan
| | - Kalyani Patil
- Department of Molecular PathophysiologyTranslational Research InstituteAcademic Health SystemHamad Medical CorporationDohaQatar
| | - Grant A. Ramm
- Department of Cell and Molecular BiologyQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Takahiro Ochiya
- Department of Molecular and Cellular MedicineTokyo Medical UniversityTokyoJapan
| | - Carolina Soekmadji
- Department of Cell and Molecular BiologyQIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
11
|
Verdi J, Ketabchi N, Noorbakhsh N, Saleh M, Ebrahimi-Barough S, Seyhoun I, Kavianpour M. Development and Clinical Application of Tumor-derived Exosomes in Patients with Cancer. Curr Stem Cell Res Ther 2021; 17:91-102. [PMID: 34161212 DOI: 10.2174/1574888x16666210622123942] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/16/2020] [Accepted: 03/11/2021] [Indexed: 11/22/2022]
Abstract
A tumor is an abnormal growth of cells within a tissue that can lead to death due to late diagnosis, poor prognosis, drug resistance, and finally enhanced metastasis formation. Exosomes are nanovesicles that have been derived from all the different cell types. These vesicles can transfer various molecules, including the distinct form of nucleic acids (mRNA, miRNA, and circRNA) and proteins. Tumor-derived exosomes (TEXs) have exceptionally important roles through multiple molecular and cellular pathways like progression, tumorigenesis, drug resistance, and as well as metastasis. TEXs are detectable in all body fluids, such as serum and urine, a convenient and non-invasive way to access these nano-sized vesicles. TEXs lead to the symptom expression of genetic aberrations in the tumor cell population, making them an accurate and sensitive biomarker for the diagnosis and prognosis of tumors. On the other hand, TEXs contain major histocompatibility complexes (MHCs) and play important dual roles in regulating tumor immune responses; they can mediate both immune activation and suppression through tumor-associated immunity. Despite numerous scientific studies, there are still many technical barriers to distinguish TEXs from non-tumor-derived exosomes. Removing exosomes lead to a wide difference in outcomes inside a patient's body. Hence, controversial pieces of evidence have demonstrated the vital role of TEXs as hopeful biomarkers for the early detection of cancers, evaluation of therapeutic effects, and monitoring of the patient.
Collapse
Affiliation(s)
- Javad Verdi
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Ketabchi
- Department of Medical Laboratory Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Negar Noorbakhsh
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mahshid Saleh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iman Seyhoun
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maria Kavianpour
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
Review of novel liquid-based biomarkers for prostate cancer: towards personalised and targeted medicine. JOURNAL OF RADIOTHERAPY IN PRACTICE 2021. [DOI: 10.1017/s1460396921000248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Abstract
Background:
Prostate cancer is the most commonly diagnosed cancer in men and it is responsible for about 10% of all cancer mortalities in both American and Canadian men. At present, serum prostate-specific antigen levels remain the most commonly used test to detect prostate cancer, and the standard and definitive diagnosis of the disease is via prostate biopsy. Conventional tissue biopsies are usually invasive, expensive, painful, time-consuming, and unsuitable for screening and need to be consistently evaluated by expert pathologists and have limited repeatability. Consequently, liquid biopsies are emerging as a favourable alternative to conventional tissue biopsies, providing a non-invasive and cost-effective approach for screening, diagnosis, treatment and monitoring of prostate cancer patients.
Materials and methods:
We searched several databases from August to December 2020 for relevant studies published in English between 2000 and 2020 and reporting on liquid-based biomarkers available in detectable quantities in patient bodily fluid samples. In this narrative review paper, we describe seven novel and promising liquid-based biomarkers that potentially account for individual patient variability as well as used in disease risk assessment, screening for early disease detection and diagnosis, identification of patients’ risk for metastatic disease and subsequent relapse, monitoring patient response to specific treatment and providing clinicians the potential to stratify patients likely to benefit from a particular treatment.
Conclusions:
The concept of precision medicine from prevention to treatment techniques that take individual patient variability into account will depend on the development of effective clinical biomarkers that interrogate key aberrant pathways potentially targetable with molecular targets or immunologic therapies. Liquid-based biomarkers with high sensitivity and specificity for prostate cancer are emerging as minimally invasive, lower risk, readily obtainable and easily repeatable technique for screening for early disease detection and diagnosis, patient stratification at diagnosis into different risk categories, identification of patients’ risk for metastatic disease and subsequent relapse, and real-time monitoring of patient response to specific treatment. Thus, effective liquid-based biomarkers will potentially shift the treatment paradigm of prostate cancer towards more personalised and targeted medicine.
Collapse
|
13
|
Lo ST, Parrott D, Jordan MVC, Joseph DB, Strand D, Lo UG, Lin H, Darehshouri A, Sherry AD. The Roles of ZnT1 and ZnT4 in Glucose-Stimulated Zinc Secretion in Prostate Epithelial Cells. Mol Imaging Biol 2021; 23:230-240. [PMID: 33140261 PMCID: PMC7914160 DOI: 10.1007/s11307-020-01557-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE We have previously demonstrated by MRI that high glucose stimulates efflux of zinc ions from the prostate. To our knowledge, this phenomena had not been reported previously and the mechanism remains unknown. Here, we report some initial observations that provide new insights into zinc processing during glucose-stimulated zinc secretion (GSZS) in the immortalized human prostate epithelial cell line, PNT1A. Additionally, we identified the subtypes of zinc-containing cells in human benign prostatic hyperplasia (BPH) tissue to further identify which cell types are likely responsible for zinc release in vivo. PROCEDURE An intracellular fluorescence marker, FluoZin-1-AM, was used to assess the different roles of ZnT1 and ZnT4 in zinc homeostasis in wild type (WT) and mRNA knockdown PNT1A cell lines. Additionally, Bafilomycin A1 (Baf) was used to disrupt lysosomes and assess the role of lysosomal storage during GSZS. ZIMIR, an extracellular zinc-responsive fluorescent marker, was used to assess dynamic zinc efflux of WT and ZnT1 mRNA knockdown cells exposed to high glucose. Electron microscopy was used to assess intracellular zinc storage in response to high glucose and evaluate how Bafilomycin A1 affects zinc trafficking. BPH cells were harvested from transurtheral prostatectomy tissue and stained with fluorescent zinc granule indicator (ZIGIR), an intracellular zinc-responsive fluorescent marker, before being sorted for cell types using flow cytometry. RESULTS Fluorescent studies demonstrate that ZnT1 is the major zinc efflux transporter in prostate epithelial cells and that loss of ZnT1 via mRNA knockdown combined with lysosomal storage disruption results in a nearly 4-fold increase in cytosolic zinc. Knockdown of ZnT1 dramatically reduces zinc efflux during GSZS. Electron microscopy (EM) reveals that glucose stimulation significantly increases lysosomal storage of zinc; disruption of lysosomes via Baf or ZnT4 mRNA knockdown increases multi-vesicular body (MVB) formation and cytosolic zinc levels. In human BPH tissue, only the luminal epithelial cells contained significant amounts of zinc storage granules. CONCLUSIONS Exposure of prostate epithelial cells to high glucose alters zinc homeostasis by inducing efflux of zinc ions via ZnT1 channels and increasing lysosomal storage via ZnT4. Given that prostate cancer cells undergo profound metabolic changes that result in reduced levels of total zinc, understanding the complex interplay between glucose exposure and zinc homeostasis in the prostate may provide new insights into the development of prostate carcinogenesis.
Collapse
Affiliation(s)
- Su-Tang Lo
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, NE 4.210, Dallas, TX, 75390-8568, USA
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, 75390-8896, USA
| | - Daniel Parrott
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, NE 4.210, Dallas, TX, 75390-8568, USA
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, 75390-8896, USA
| | - M Veronica Clavijo Jordan
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, NE 4.210, Dallas, TX, 75390-8568, USA
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, 75390-8896, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Diya Binoy Joseph
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Douglas Strand
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - U-Ging Lo
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, 75390-9110, USA
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung City, 402, Taiwan
| | - Anza Darehshouri
- Electron Microscopy Core Facility, UT Southwestern Medical Center, Dallas, TX, 75390-9039, USA
| | - A Dean Sherry
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, NE 4.210, Dallas, TX, 75390-8568, USA.
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, 75390-8896, USA.
- Department of Chemistry, University of Texas at Dallas, Richardson, TX, 75083, USA.
| |
Collapse
|
14
|
Lim HK, Jeffrey GP, Ramm GA, Soekmadji C. Pathogenesis of Viral Hepatitis-Induced Chronic Liver Disease: Role of Extracellular Vesicles. Front Cell Infect Microbiol 2020; 10:587628. [PMID: 33240824 PMCID: PMC7683521 DOI: 10.3389/fcimb.2020.587628] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles are encapsulated lipid nanoparticles secreted by a variety of cell types in living organisms. They are known to carry proteins, metabolites, nucleic acids, and lipids as their cargoes and are important mediators of intercellular communication. The role of extracellular vesicles in chronic liver disease has been reported. Chronic liver disease such as viral hepatitis accounts for a significant mortality and morbidity burden worldwide. Hepatic fibrosis has been commonly associated with the chronic form of viral hepatitis, which results in end-stage liver disease, including cirrhosis, liver failure, and carcinoma in some patients. In this review, we discuss the potential role of extracellular vesicles in mediating communication between infectious agents (hepatitis B and C viruses) and host cells, and how these complex cell-cell interactions may facilitate the development of chronic liver disease. We will further discuss how understanding their biological mechanism of action might be beneficial for developing therapeutic strategies to treat chronic liver disease.
Collapse
Affiliation(s)
- Hong Kiat Lim
- Hepatic Fibrosis Group, Department of Cellular and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Gary P Jeffrey
- Faculty of Health and Medical Sciences, University of Western Australia, Perth, WA, Australia.,Sir Charles Gairdner Hospital, Nedlands, Hepatology Department and Liver Transplant Service, Perth, WA, Australia
| | - Grant A Ramm
- Hepatic Fibrosis Group, Department of Cellular and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Carolina Soekmadji
- Hepatic Fibrosis Group, Department of Cellular and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
15
|
Dong M, Lih TM, Chen SY, Cho KC, Eguez RV, Höti N, Zhou Y, Yang W, Mangold L, Chan DW, Zhang Z, Sokoll LJ, Partin A, Zhang H. Urinary glycoproteins associated with aggressive prostate cancer. Am J Cancer Res 2020; 10:11892-11907. [PMID: 33204318 PMCID: PMC7667684 DOI: 10.7150/thno.47066] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background: There is an urgent need for the detection of aggressive prostate cancer. Glycoproteins play essential roles in cancer development, while urine is a noninvasive and easily obtainable biological fluid that contains secretory glycoproteins from the urogenital system. Therefore, here we aimed to identify urinary glycoproteins that are capable of differentiating aggressive from non-aggressive prostate cancer. Methods: Quantitative mass spectrometry data of glycopeptides from a discovery cohort comprised of 74 aggressive (Gleason score ≥8) and 68 non-aggressive (Gleason score = 6) prostate cancer urine specimens were acquired via a data independent acquisition approach. The glycopeptides showing distinct expression profiles in aggressive relative to non-aggressive prostate cancer were further evaluated for their performance in distinguishing the two groups either individually or in combination with others using repeated 5-fold cross validation with logistic regression to build predictive models. Predictive models showing good performance from the discovery cohort were further evaluated using a validation cohort. Results: Among the 20 candidate glycoproteins, urinary ACPP outperformed the other candidates. Urinary ACPP can also serve as an adjunct to serum PSA to further improve the discrimination power for aggressive prostate cancer (AUC= 0.82, 95% confidence interval 0.75 to 0.89). A three-signature panel including urinary ACPP, urinary CLU, and serum PSA displayed the ability to distinguish aggressive prostate cancer from non-aggressive prostate cancer with an AUC of 0.86 (95% confidence interval 0.8 to 0.92). Another three-signature panel containing urinary ACPP, urinary LOX, and serum PSA also demonstrated its ability in recognizing aggressive prostate cancer (AUC=0.82, 95% confidence interval 0.75 to 0.9). Moreover, consistent performance was observed from each panel when evaluated using a validation cohort. Conclusion: We have identified glycopeptides of urinary glycoproteins associated with aggressive prostate cancer using a quantitative mass spectrometry-based glycoproteomic approach and demonstrated their potential to serve as noninvasive urinary glycoprotein biomarkers worthy of further validation by a multi-center study.
Collapse
|
16
|
Zhao Y, Wang Y, Zhao E, Tan Y, Geng B, Kang C, Li X. PTRF/CAVIN1, regulated by SHC1 through the EGFR pathway, is found in urine exosomes as a potential biomarker of ccRCC. Carcinogenesis 2020; 41:274-283. [PMID: 31605605 DOI: 10.1093/carcin/bgz147] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 01/10/2023] Open
Abstract
Polymerase I and transcript release factor (PTRF)/Cavin1 regulates RNA polymerase I during transcription and plays a critical role in endocytosis. Abnormal expressions of PTRF were detected in multiple cancers according to increasing research. PTRF has been showed to involve in the formation and secretion of exosomes and can be detected in the exosomes, which suggests that PTRF would be a potential biomarker for diagnosis of clear cell renal cell carcinoma (ccRCC) using urine samples. Approximately 50-90% of ccRCC cases suffered abnormal epidermal growth factor receptor (EGFR), which activates a variety of signaling pathways, including the mitogen-activated protein kinase/extracellular signal-regulated kinase and Phosphoinositide 3-Kinase/Akt pathway. According to bioinformatic analysis of gene expression arrays of kidney clear cell carcinoma from The Cancer Genome Atlas, we found SHC1 was significantly overexpressed in high-grade ccRCC and correlated to poor prognosis, and also SHC1 was annotated in extracellular matrix process, which was regulated by EGFR. Further studies showed that the expression of PTRF was regulated by SHC1 through EGFR-Phosphoinositide 3-Kinase/Akt pathway. PTRF was detected in the exosomes isolated from ccRCC patients' urine and ccRCC cancer cells culture medium. It suggested that the abnormal SHC1-increased PTRF, which is detected in exosomes from urine, would be a potential marker for ccRCC diagnose and treatment.
Collapse
Affiliation(s)
- Yubo Zhao
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunfei Wang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital; Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Enyang Zhao
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanli Tan
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Bo Geng
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital; Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Xuedong Li
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Liao Y, Xu K. Epigenetic regulation of prostate cancer: the theories and the clinical implications. Asian J Androl 2020; 21:279-290. [PMID: 30084432 PMCID: PMC6498736 DOI: 10.4103/aja.aja_53_18] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epigenetics is the main mechanism that controls transcription of specific genes with no changes in the underlying DNA sequences. Epigenetic alterations lead to abnormal gene expression patterns that contribute to carcinogenesis and persist throughout disease progression. Because of the reversible nature, epigenetic modifications emerge as promising anticancer drug targets. Several compounds have been developed to reverse the aberrant activities of enzymes involved in epigenetic regulation, and some of them show encouraging results in both preclinical and clinical studies. In this article, we comprehensively review the up-to-date roles of epigenetics in the development and progression of prostate cancer. We especially focus on three epigenetic mechanisms: DNA methylation, histone modifications, and noncoding RNAs. We elaborate on current models/theories that explain the necessity of these epigenetic programs in driving the malignant phenotypes of prostate cancer cells. In particular, we elucidate how certain epigenetic regulators crosstalk with critical biological pathways, such as androgen receptor (AR) signaling, and how the cooperation dynamically controls cancer-oriented transcriptional profiles. Restoration of a "normal" epigenetic landscape holds promise as a cure for prostate cancer, so we concluded by highlighting particular epigenetic modifications as diagnostic and prognostic biomarkers or new therapeutic targets for treatment of the disease.
Collapse
Affiliation(s)
- Yiji Liao
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Kexin Xu
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, TX 78229, USA
| |
Collapse
|
18
|
Exosomes in Prostate Cancer Diagnosis, Prognosis and Therapy. Int J Mol Sci 2020; 21:ijms21062118. [PMID: 32204455 PMCID: PMC7139716 DOI: 10.3390/ijms21062118] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the second most common cause of cancer-related mortality among men in the developed world. Conventional anti-PCa therapies are not effective for patients with advanced and/or metastatic disease. In most cases, cancer therapies fail due to an incomplete depletion of tumor cells, resulting in tumor relapse. Exosomes are involved in tumor progression, promoting the angiogenesis and migration of tumor cells during metastasis. These structures contribute to the dissemination of pathogenic agents through interaction with recipient cells. Exosomes may deliver molecules that are able to induce the transdifferentiation process, known as “epithelial to mesenchymal transition”. The composition of exosomes and the associated possibilities of interacting with cells make exosomes multifaceted regulators of cancer development. Extracellular vesicles have biophysical properties, such as stability, biocompatibility, permeability, low toxicity and low immunogenicity, which are key for the successful development of an innovative drug delivery system. They have an enhanced circulation stability and bio-barrier permeation ability, and they can therefore be used as effective chemotherapeutic carriers to improve the regulation of target tissues and organs. Exosomes have the capacity to deliver different types of cargo and to target specific cells. Chemotherapeutics, natural products and RNA have been encapsulated for the treatment of prostate cancers.
Collapse
|
19
|
Amirrad F, Pytak PA, Sadeghiani-Pelar N, Nguyen JPT, Cauble EL, Jones AC, Bisoffi M. Prostate field cancerization and exosomes: Association between CD9, early growth response 1 and fatty acid synthase. Int J Oncol 2020; 56:957-968. [PMID: 32319557 DOI: 10.3892/ijo.2020.4980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 01/23/2020] [Indexed: 11/06/2022] Open
Abstract
Intracapsular and well‑defined adenocarcinomas of the prostate are often surrounded by tissue areas that harbor molecular aberrations, including those of genetic, epigenetic and biochemical nature. This is known as field cancerization, or a field effect and denotes a state of pre‑malignancy. Such alterations in histologically normal tumor‑adjacent prostatic tissues have been recognized as clinically important and are potentially exploitable as biomarkers of disease and/or targets for preventative/therapeutic intervention. The authors have previously identified and validated two protein markers of field cancerization: The expressional upregulation of the transcription factor early growth response 1 (EGR‑1) and the lipogenic enzyme fatty acid synthase (FASN). However, the molecular etiology of prostate field cancerization, including EGR‑1 and FASN upregulation, remains largely unknown. It was thus hypothesized that extracellular vesicles, notably exosomes, released by tumor lesions may induce molecular alterations in the surrounding tissues, resulting in field cancerization, priming the tissue, and ultimately promoting multifocal tumorigenesis, which is often observed in prostate cancer. Towards testing this hypothesis, the current study, to the best of our knowledge, for the first time, presents correlative protein expression data, generated in disease‑free, tumor‑adjacent and cancerous human prostate tissues by quantitative immunofluorescence, between the exosomal marker CD9, and EGR‑1 and FASN. Despite the pilot character of the present study, and the static nature and heterogeneity of human tissues, the data suggest that CD9 expression itself is part of a field effect. In support of this hypothesis, the results suggest a possible contribution of exosomes to the induction of field cancerization in the prostate, particularly for EGR‑1. These findings were corroborated in established cell models of cancerous (LNCaP) and non‑cancerous (RWPE‑1) human prostate epithelial cells. The findings of this study warrant further investigation into the functional interface between exosomes and field cancerization, as a detailed understanding of this characterization may lead to the development of clinical applications related to diagnosis and/or prognosis and targeted intervention to prevent progression from pre‑malignancy to cancer.
Collapse
Affiliation(s)
- Farideh Amirrad
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Philip A Pytak
- Division of Chemistry and Biochemistry, Chapman University Schmid College of Science and Technology, Keck Center for Science and Engineering, Orange, CA 92866, USA
| | - Neda Sadeghiani-Pelar
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Julie P T Nguyen
- Division of Chemistry and Biochemistry, Chapman University Schmid College of Science and Technology, Keck Center for Science and Engineering, Orange, CA 92866, USA
| | - Emily L Cauble
- Division of Biological Sciences, Chapman University Schmid College of Science and Technology, Keck Center for Science and Engineering, Orange, CA 92866, USA
| | - Anna C Jones
- University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87102, USA
| | - Marco Bisoffi
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| |
Collapse
|
20
|
Archana SS, Selvaraju S, Binsila BK, Arangasamy A, Krawetz SA. Immune regulatory molecules as modifiers of semen and fertility: A review. Mol Reprod Dev 2019; 86:1485-1504. [DOI: 10.1002/mrd.23263] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 08/22/2019] [Indexed: 12/16/2022]
Affiliation(s)
- S. Siddalingappa Archana
- Reproductive Physiology Laboratory, Animal Physiology DivisionICAR‐National Institute of Animal Nutrition and Physiology Bengaluru India
- Department of BiochemistryJain University Bengaluru India
| | - Sellappan Selvaraju
- Reproductive Physiology Laboratory, Animal Physiology DivisionICAR‐National Institute of Animal Nutrition and Physiology Bengaluru India
| | - B. Krishnan Binsila
- Reproductive Physiology Laboratory, Animal Physiology DivisionICAR‐National Institute of Animal Nutrition and Physiology Bengaluru India
| | - Arunachalam Arangasamy
- Reproductive Physiology Laboratory, Animal Physiology DivisionICAR‐National Institute of Animal Nutrition and Physiology Bengaluru India
| | - Stephen A. Krawetz
- Department of Obstetrics and GynecologyWayne State University School of Medicine Detroit Michigan
- Center for Molecular Medicine and GeneticsC.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine Detroit Michigan
| |
Collapse
|
21
|
Balachandran B, Yuana Y. Extracellular vesicles-based drug delivery system for cancer treatment. COGENT MEDICINE 2019. [DOI: 10.1080/2331205x.2019.1635806] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Banuja Balachandran
- Division of Imaging, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Yuana Yuana
- Division of Imaging, University Medical Centre Utrecht, Utrecht, The Netherlands
- Faculty of Biomedical Engineering, Technical University Eindhoven, Eindhoven, The Netherlands
| |
Collapse
|
22
|
Jablonska J, Pietrowska M, Ludwig S, Lang S, Thakur BK. Challenges in the Isolation and Proteomic Analysis of Cancer Exosomes-Implications for Translational Research. Proteomes 2019; 7:proteomes7020022. [PMID: 31096692 PMCID: PMC6631388 DOI: 10.3390/proteomes7020022] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/21/2022] Open
Abstract
Exosomes belong to the group of extracellular vesicles (EVs) that derive from various cell populations and mediate intercellular communication in health and disease. Like hormones or cytokines, exosomes released by cells can play a potent role in the communication between the cell of origin and distant cells in the body to maintain homeostatic or pathological processes, including tumorigenesis. The nucleic acids, and lipid and protein cargo present in the exosomes are involved in a myriad of carcinogenic processes, including cell proliferation, tumor angiogenesis, immunomodulation, and metastasis formation. The ability of exosomal proteins to mediate direct functions by interaction with other cells qualifies them as tumor-specific biomarkers and targeted therapeutic approaches. However, the heterogeneity of plasma-derived exosomes consistent of (a) exosomes derived from all kinds of body cells, including cancer cells and (b) contamination of exosome preparation with other extracellular vesicles, such as apoptotic bodies, makes it challenging to obtain solid proteomics data for downstream clinical application. In this manuscript, we review these challenges beginning with the choice of different isolation methods, through the evaluation of obtained exosomes and limitations in the process of proteome analysis of cancer-derived exosomes to identify novel protein targets with functional impact in the context of translational oncology.
Collapse
Affiliation(s)
- Jadwiga Jablonska
- Translational Oncology, Department of Otorhinolaryngology, University Hospital Essen, 45147 Essen, Germany.
| | - Monika Pietrowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie Institute⁻Oncology Center, Gliwice Branch, 44-100 Gliwice, Poland.
| | - Sonja Ludwig
- Translational Oncology, Department of Otorhinolaryngology, University Hospital Essen, 45147 Essen, Germany.
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Essen, 45147 Essen, Germany.
| | - Stephan Lang
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Essen, 45147 Essen, Germany.
| | - Basant Kumar Thakur
- Cancer Exosome Research Lab, Department of Pediatric Hematology and Oncology, University Hospital Essen, 45147 Essen, Germany.
| |
Collapse
|
23
|
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med 2019; 17:3. [PMID: 30602375 PMCID: PMC6317263 DOI: 10.1186/s12967-018-1760-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
Milk consumption is a hallmark of western diet. According to common believes, milk consumption has beneficial effects for human health. Pasteurization of cow's milk protects thermolabile vitamins and other organic compounds including bioactive and bioavailable exosomes and extracellular vesicles in the range of 40-120 nm, which are pivotal mediators of cell communication via systemic transfer of specific micro-ribonucleic acids, mRNAs and regulatory proteins such as transforming growth factor-β. There is compelling evidence that human and bovine milk exosomes play a crucial role for adequate metabolic and immunological programming of the newborn infant at the beginning of extrauterine life. Milk exosomes assist in executing an anabolic, growth-promoting and immunological program confined to the postnatal period in all mammals. However, epidemiological and translational evidence presented in this review indicates that continuous exposure of humans to exosomes of pasteurized milk may confer a substantial risk for the development of chronic diseases of civilization including obesity, type 2 diabetes mellitus, osteoporosis, common cancers (prostate, breast, liver, B-cells) as well as Parkinson's disease. Exosomes of pasteurized milk may represent new pathogens that should not reach the human food chain.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7A, 49076 Osnabrück, Germany
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, University of Regensburg, Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
24
|
Alharbi M, Zuñiga F, Elfeky O, Guanzon D, Lai A, Rice GE, Perrin L, Hooper J, Salomon C. The potential role of miRNAs and exosomes in chemotherapy in ovarian cancer. Endocr Relat Cancer 2018; 25:R663-R685. [PMID: 30400025 DOI: 10.1530/erc-18-0019] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 07/26/2018] [Indexed: 12/20/2022]
Abstract
Chemoresistance is one of the major obstacles in the treatment of cancer patients. It poses a fundamental challenge to the effectiveness of chemotherapy and is often linked to relapse in patients. Chemoresistant cells can be identified in different types of cancers; however, ovarian cancer has one of the highest rates of chemoresistance-related relapse (50% of patients within 5 years). Resistance in cells can either develop through prolonged cycles of treatment or through intrinsic pathways. Mechanistically, the problem of drug resistance is complex mainly because numerous factors are involved, such as overexpression of drug efflux pumps, drug inactivation, DNA repair mechanisms and alterations to and/or mutations in the drug target. Additionally, there is strong evidence that circulating miRNAs participate in the development of chemoresistance. Recently, miRNAs have been identified in exosomes, where they are encapsulated and hence protected from degradation. These miRNAs within exosomes (exo-miRNAs) can regulate the gene expression of target cells both locally and systemically. Exo-miRNAs play an important role in disease progression and can potentially facilitate chemoresistance in cancer cells. In addition, and from a diagnostic perspective, exo-miRNAs profiles may contribute to the development of predictive models to identify responder and non-responder chemotherapy. Such model may also be used for monitoring treatment response and disease progression. Exo-miRNAs may ultimately serve as both a predictive biomarker for cancer response to therapy and as a prognostic marker for the development of chemotherapy resistance. Therefore, this review examines the potential role of exo-miRNAs in chemotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Mona Alharbi
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane Queensland, Australia
| | - Felipe Zuñiga
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepción, Chile
| | - Omar Elfeky
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane Queensland, Australia
| | - Dominic Guanzon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane Queensland, Australia
| | - Andrew Lai
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane Queensland, Australia
| | - Gregory E Rice
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane Queensland, Australia
- Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
- Perinatology Research Branch, NICHD/NIH, Wayne State University, Detroit, Michigan, USA
| | - Lewis Perrin
- Mater Research Institute, University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland, Australia
| | - John Hooper
- Mater Research Institute, University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland, Australia
| | - Carlos Salomon
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland, Brisbane Queensland, Australia
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción, Concepción, Chile
- Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Ochsner Clinic Foundation, New Orleans, Louisiana, USA
| |
Collapse
|
25
|
Prostate cancer sheds the αvβ3 integrin in vivo through exosomes. Matrix Biol 2018; 77:41-57. [PMID: 30098419 DOI: 10.1016/j.matbio.2018.08.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/03/2018] [Accepted: 08/05/2018] [Indexed: 12/14/2022]
Abstract
The αvβ3 integrin has been shown to promote aggressive phenotypes in many types of cancers, including prostate cancer. We show that GFP-labeled αvβ3 derived from cancer cells circulates in the blood and is detected in distant lesions in NOD scid gamma (NSG) mice. We, therefore, hypothesized that αvβ3 travels through exosomes and tested its levels in pools of vesicles, which we designate extracellular vesicles highly enriched in exosomes (ExVs), and in exosomes isolated from the plasma of prostate cancer patients. Here, we show that the αvβ3 integrin is found in patient blood exosomes purified by sucrose or iodixanol density gradients. In addition, we provide evidence that the αvβ3 integrin is transferred through ExVs isolated from prostate cancer patient plasma to β3-negative recipient cells. We also demonstrate the intracellular localization of β3-GFP transferred via cancer cell-derived ExVs. We show that the ExVs present in plasma from prostate cancer patients contain higher levels of αvβ3 and CD9 as compared to plasma ExVs from age-matched subjects who are not affected by cancer. Furthermore, using PSMA antibody-bead mediated immunocapture, we show that the αvβ3 integrin is expressed in a subset of exosomes characterized by PSMA, CD9, CD63, and an epithelial-specific marker, Trop-2. Finally, we present evidence that the levels of αvβ3, CD63, and CD9 remain unaltered in ExVs isolated from the blood of prostate cancer patients treated with enzalutamide. Our results suggest that detecting exosomal αvβ3 integrin in prostate cancer patients could be a clinically useful and non-invasive biomarker to follow prostate cancer progression. Moreover, the ability of αvβ3 integrin to be transferred from ExVs to recipient cells provides a strong rationale for further investigating the role of αvβ3 integrin in the pathogenesis of prostate cancer and as a potential therapeutic target.
Collapse
|
26
|
Lee J, Kwon MH, Kim JA, Rhee WJ. Detection of exosome miRNAs using molecular beacons for diagnosing prostate cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S52-S63. [PMID: 30033809 DOI: 10.1080/21691401.2018.1489263] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Prostate cancer is the fifth leading cause of cancer-related deaths among males worldwide. However, the biomarker for diagnosing prostate cancer that is used currently has limitations that must be overcome. Recently, several studies have demonstrated that the cancer liquid biopsy can be implemented by using exosome miRNAs. However, the current methods for the detection of exosome miRNAs are time-consuming, expensive, and laborious. Thus, we investigated a novel method for diagnosing prostate cancer that involves the use of molecular beacons for the in situ detection of miRNAs in exosomes from prostate cancer cells. We chose miRNA-375 and miRNA-574-3p as the target miRNAs for prostate cancer, and these markers in exosomes produced by prostate cancer cells including DU145 and PC-3 were successfully detected using molecular beacons. High fluorescent signals were obtained from MB and miRNA hybridization in exosomes in a concentration-dependent manner. In addition, exosome miRNAs can be detected even in the presence of human urine, so this method can be applied directly using human urine to perform liquid biopsies for prostate cancer. Overall, the in situ detection of exosome miRNAs using molecular beacons can be developed as a simple, cost effective, and non-invasive liquid biopsy for diagnosing prostate cancer.
Collapse
Affiliation(s)
- Jinhee Lee
- a Division of Bioengineering , Incheon National University , Incheon , Republic of Korea
| | - Min Hee Kwon
- a Division of Bioengineering , Incheon National University , Incheon , Republic of Korea
| | - Jeong Ah Kim
- b Biomedical Omics Group , Korea Basic Science Institute , Cheongju , Republic of Korea
| | - Won Jong Rhee
- a Division of Bioengineering , Incheon National University , Incheon , Republic of Korea
| |
Collapse
|
27
|
Soekmadji C, Rockstroh A, Ramm GA, Nelson CC, Russell PJ. Extracellular Vesicles in the Adaptive Process of Prostate Cancer during Inhibition of Androgen Receptor Signaling by Enzalutamide. Proteomics 2018; 17. [PMID: 29105980 DOI: 10.1002/pmic.201600427] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 09/11/2017] [Indexed: 01/05/2023]
Abstract
Current treatments for advanced prostate cancer focus on inhibition of the androgen receptor (AR) by androgen deprivation therapy (ADT). However, complex interactions mediated by tumor suppressors, oncogenes, aberrations of AR expression, or de novo androgen production have been shown to induce the adaptive response of prostate cancer, leading to the development of castration resistant prostate cancer. In this study, we report the effects of AR antagonist, enzalutamide on the protein contents of extracellular vesicles (EVs). EVs mediate cell-to-cell communication and increasing evidence shows the role of EVs in promoting cancer survival and metastasis. We found that treatment with enzalutamide alters the secretion of EVs, one of which is a plasma membrane calcium pump, ATP2B1/PMCA ATPase, as an AR-regulated EV protein. We highlight the networks of interactions between AR, Ca2+ , and ATP2B1, where the extracellular proteins thrombospondin-1, gelsolin, and integrinß1 were previously reported as regulators for cancer progression and metastasis, indicating the potential role of EV-derived proteins in mediating calcium homoeostasis under AR inhibition by enzalutamide. Our data further highlight the cross-talk between AR signaling and EV pathways in mediating resistance toward ADT.
Collapse
Affiliation(s)
- Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anja Rockstroh
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT),, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| | - Grant A Ramm
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT),, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| | - Pamela J Russell
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT),, Princess Alexandra Hospital, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
28
|
Jantaratrirat S, Boonarkart C, Ruangrung K, Suptawiwat O, Auewarakul P. Microparticle Release from Cell Lines and Its Anti-Influenza Activity. Viral Immunol 2018; 31:447-456. [DOI: 10.1089/vim.2017.0201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Saharat Jantaratrirat
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chompunuch Boonarkart
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanyarat Ruangrung
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ornpreya Suptawiwat
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Prasert Auewarakul
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
29
|
Safdar A, Tarnopolsky MA. Exosomes as Mediators of the Systemic Adaptations to Endurance Exercise. Cold Spring Harb Perspect Med 2018; 8:a029827. [PMID: 28490541 PMCID: PMC5830902 DOI: 10.1101/cshperspect.a029827] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Habitual endurance exercise training is associated with multisystemic metabolic adaptations that lower the risk of inactivity-associated disorders such as obesity and type 2 diabetes mellitus (T2DM). Identification of complex systemic signaling networks responsible for these benefits are of great interest because of their therapeutic potential in metabolic diseases; however, specific signals that modulate the multisystemic benefits of exercise in multiple tissues and organs are only recently being discovered. Accumulated evidence suggests that muscle and other tissues have an endocrine function and release peptides and nucleic acids into the circulation in response to acute endurance exercise to mediate the multisystemic adaptations. Factors released from skeletal muscle have been termed myokines and we propose that the total of all factors released in response to endurance exercise (including peptides, nucleic acids, and metabolites) be termed, "exerkines." We propose that many of the exerkines are released within extracellular vesicles called exosomes, which regulate peripheral organ cross talk. Exosomes (30-140 nm) and larger microvesicles [MVs] (100-1000 nm) are subcategories of extracellular vesicles that are released into the circulation. Exosomes contain peptides and several nucleic acids (microRNA [miRNA], messenger RNA [mRNA], mitochondrial DNA [mtDNA]) and are involved in intercellular/tissue exchange of their contents. An acute bout of endurance exercise increases circulating exosomes that are hypothesized to mediate organ cross talk to promote systemic adaptation to endurance exercise. Further support for the role of exosomes (and possibly MVs) in mediating the systemic benefits of exercise comes from the fact that the majority of the previously reported myokines/exerkines are found in extracellular vesicles databases (Vesiclepedia and ExoCarta). We propose that exosomes isolated from athletes following exercise or exosomes bioengineered to incorporate one or many of known exerkines will be therapeutically useful in the treatment of obesity, T2DM, and other aging-associated metabolic disorders.
Collapse
Affiliation(s)
- Adeel Safdar
- Department of Pediatrics, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
- Department of Pediatrics & Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada
| |
Collapse
|
30
|
Christenson JL, Trepel JB, Ali HY, Lee S, Eisner JR, Baskin-Bey ES, Elias AD, Richer JK. Harnessing a Different Dependency: How to Identify and Target Androgen Receptor-Positive Versus Quadruple-Negative Breast Cancer. Discov Oncol 2018; 9:82-94. [PMID: 29340907 DOI: 10.1007/s12672-017-0314-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/07/2017] [Indexed: 12/12/2022] Open
Abstract
The androgen receptor (AR) is a promising therapeutic target for a subset of triple-negative breast cancers (TNBCs) in which AR is expressed. However, the mechanistic action of AR and the degree to which primary and metastatic tumors depend on AR, both before and after conventional treatment, remain to be defined. We discuss preclinical and clinical data for AR+ TNBC, the difficulties in monitoring AR protein levels, new methods for determining AR status, the influence of AR on "stemness" in the context of TNBC, the role of combined inhibition of sex steroid production and AR, and the role of AR in regulation of the immune system. Although the exact role of AR in subsets of TNBC is still being characterized, new therapies that target AR and the production of androgens may provide additional options for patients with TNBC for whom chemotherapy is currently the sole treatment option.
Collapse
Affiliation(s)
- Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jane B Trepel
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Sunmin Lee
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | | | - Anthony D Elias
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
31
|
Soekmadji C, Corcoran NM, Oleinikova I, Jovanovic L, Ramm GA, Nelson CC, Jenster G, Russell PJ. Extracellular vesicles for personalized therapy decision support in advanced metastatic cancers and its potential impact for prostate cancer. Prostate 2017; 77:1416-1423. [PMID: 28856701 DOI: 10.1002/pros.23403] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/03/2017] [Indexed: 12/31/2022]
Abstract
The use of circulating tumor cells (CTCs) and circulating extracellular vesicles (EVs), such as exosomes, as liquid biopsy-derived biomarkers for cancers have been investigated. CTC enumeration using the CellSearch based platform provides an accurate insight on overall survival where higher CTC counts indicate poor prognosis for patients with advanced metastatic cancer. EVs provide information based on their lipid, protein, and nucleic acid content and can be isolated from biofluids and analyzed from a relatively small volume, providing a routine and non-invasive modality to monitor disease progression. Our pilot experiment by assessing the level of two subpopulations of small EVs, the CD9 positive and CD63 positive EVs, showed that the CD9 positive EV level is higher in plasma from patients with advanced metastatic prostate cancer with detectable CTCs. These data show the potential utility of a particular EV subpopulation to serve as biomarkers for advanced metastatic prostate cancer. EVs can potentially be utilized as biomarkers to provide accurate genotypic and phenotypic information for advanced prostate cancer, where new strategies to design a more personalized therapy is currently the focus of considerable investigation.
Collapse
Affiliation(s)
- Carolina Soekmadji
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Niall M Corcoran
- Australian Prostate Cancer Research Centre Epworth, and Department of Surgery, University of Melbourne, Australia
| | - Irina Oleinikova
- Department of Urology, Queensland Health, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Lidija Jovanovic
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| | - Grant A Ramm
- Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| | - Guido Jenster
- Department of Urology, Erasmus Medical Centre, R,otterdam, The Netherlands
| | - Pamela J Russell
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
32
|
Pan J, Ding M, Xu K, Yang C, Mao LJ. Exosomes in diagnosis and therapy of prostate cancer. Oncotarget 2017; 8:97693-97700. [PMID: 29228644 PMCID: PMC5722596 DOI: 10.18632/oncotarget.18532] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 06/02/2017] [Indexed: 01/16/2023] Open
Abstract
Exosomes are small vesicular bodies released by a variety of cells. Exosomes contain miRNAs, mRNAs and proteins with the potential to regulate signaling pathways in recipient cells. Exosomes deliver nucleic acids and proteins to mediate the communication between cancer cells and stroma cells. In this review, we summarize recent progress in our understanding of the role of exosomes in prostate cancer. The tumorigenesis, metastasis and drug resistance of prostate cancer are associated with the cargos of exosomes such as miRNAs, lncRNAs and proteins. In addition, prostate cancer cells modulate surrounding stromal cells via the exosomes. Affected stromal cells employ the exosomes to modulate microenvironment and promote tumor growth and metastasis. Exosomes derived from prostate cancer cells contribute to cancer chemoresistance. The lipid bilayer membrane of the exosomes makes them promising carriers of drugs and other therapeutic molecules targeting prostate cancer. Furthermore, exosomes can be detected and isolated from various body fluids for the diagnosis of prostate cancer.
Collapse
Affiliation(s)
- Jun Pan
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Meng Ding
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Kai Xu
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Chunhua Yang
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.,Radiotherapy Department, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Li-Jun Mao
- Department of Urinary Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| |
Collapse
|
33
|
|
34
|
Yang JS, Lee JC, Byeon SK, Rha KH, Moon MH. Size Dependent Lipidomic Analysis of Urinary Exosomes from Patients with Prostate Cancer by Flow Field-Flow Fractionation and Nanoflow Liquid Chromatography-Tandem Mass Spectrometry. Anal Chem 2017; 89:2488-2496. [PMID: 28192938 DOI: 10.1021/acs.analchem.6b04634] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Exosomes are membrane-bound extracellular vesicles involved in intercellular communication and tumor cell metastasis. In this study, flow field-flow fractionation (FlFFF) was utilized to separate urinary exosomes by size, demonstrating a significant difference in exosome sizes between healthy controls and patients with prostate cancer (PCa). Exosome fractions of different sizes were collected for microscopic analysis during an FlFFF run and evaluated with exosome marker proteins using Western blot analysis. The results indicated that exosomes of different sizes originated from different types of cells. Collected exosome fractions were further examined using nanoflow ultrahigh performance liquid chromatography-electrospray ionization-tandem mass spectrometry (nUPLC-ESI-MS/MS) for lipidomic analysis. A total of 162 lipids (from 286 identified) were quantified using a selected reaction monitoring (SRM) method. The overall amount of lipids increased by 1.5- to 2-fold in patients with PCa and degree of increase was more significant in the smaller fractions (diameter <150 nm) than in the larger ones (diameter >150 nm) some classes of lipids. In addition, neutral lipids like diacylglycerol (DAG) and triacylglycerol (TAG) decreased in all exosomes without size dependency. Moreover, a dramatic increase in 22:6/22:6-phosphatidylglycerol (PG) was observed and significant decrease in (16:0,16:0)- and (16:1, 18:1)-DAG species (nearly 5-fold) and high abundant TAG species (>2.5-fold) was observed in patients with PCa. The results of this study indicate that FlFFF can be employed for the high-speed screening of urinary exosome sizes in patients with PCa and lipidomic analysis of the fractionated exosomes has potential for developing and distinguishing biomarkers of PCa.
Collapse
Affiliation(s)
- Joon Seon Yang
- Department of Chemistry, Yonsei University , 50 Yonsei-Ro, Seoul, 03722 South Korea
| | - Jong Cheol Lee
- Department of Chemistry, Yonsei University , 50 Yonsei-Ro, Seoul, 03722 South Korea
| | - Seul Kee Byeon
- Department of Chemistry, Yonsei University , 50 Yonsei-Ro, Seoul, 03722 South Korea
| | - Koon Ho Rha
- Department of Urology, Yonsei University College of Medicine , 50-1 Yonsei-Ro, Seoul, 03722 South Korea
| | - Myeong Hee Moon
- Department of Chemistry, Yonsei University , 50 Yonsei-Ro, Seoul, 03722 South Korea
| |
Collapse
|
35
|
Tumor Microenvironment Modulation via Gold Nanoparticles Targeting Malicious Exosomes: Implications for Cancer Diagnostics and Therapy. Int J Mol Sci 2017; 18:ijms18010162. [PMID: 28098821 PMCID: PMC5297795 DOI: 10.3390/ijms18010162] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 12/18/2022] Open
Abstract
Exosomes are nanovesicles formed in the endosomal pathway with an important role in paracrine and autocrine cell communication. Exosomes secreted by cancer cells, malicious exosomes, have important roles in tumor microenvironment maturation and cancer progression. The knowledge of the role of exosomes in tumorigenesis prompted a new era in cancer diagnostics and therapy, taking advantage of the use of circulating exosomes as tumor biomarkers due to their stability in body fluids and targeting malignant exosomes’ release and/or uptake to inhibit or delay tumor development. In recent years, nanotechnology has paved the way for the development of a plethora of new diagnostic and therapeutic platforms, fostering theranostics. The unique physical and chemical properties of gold nanoparticles (AuNPs) make them suitable vehicles to pursuit this goal. AuNPs’ properties such as ease of synthesis with the desired shape and size, high surface:volume ratio, and the possibility of engineering their surface as desired, potentiate AuNPs’ role in nanotheranostics, allowing the use of the same formulation for exosome detection and restraining the effect of malicious exosomes in cancer progression.
Collapse
|
36
|
Mallik S, Bhadra T, Maulik U. Identifying Epigenetic Biomarkers using Maximal Relevance and Minimal Redundancy Based Feature Selection for Multi-Omics Data. IEEE Trans Nanobioscience 2017; 16:3-10. [PMID: 28092570 DOI: 10.1109/tnb.2017.2650217] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Epigenetic Biomarker discovery is an important task in bioinformatics. In this article, we develop a new framework of identifying statistically significant epigenetic biomarkers using maximal-relevance and minimal-redundancy criterion based feature (gene) selection for multi-omics dataset. Firstly, we determine the genes that have both expression as well as methylation values, and follow normal distribution. Similarly, we identify the genes which consist of both expression and methylation values, but do not follow normal distribution. For each case, we utilize a gene-selection method that provides maximal-relevant, but variable-weighted minimum-redundant genes as top ranked genes. For statistical validation, we apply t-test on both the expression and methylation data consisting of only the normally distributed top ranked genes to determine how many of them are both differentially expressed andmethylated. Similarly, we utilize Limma package for performing non-parametric Empirical Bayes test on both expression and methylation data comprising only the non-normally distributed top ranked genes to identify how many of them are both differentially expressed and methylated. We finally report the top-ranking significant gene-markerswith biological validation. Moreover, our framework improves positive predictive rate and reduces false positive rate in marker identification. In addition, we provide a comparative analysis of our gene-selection method as well as othermethods based on classificationperformances obtained using several well-known classifiers.
Collapse
|
37
|
Sunkara V, Woo HK, Cho YK. Emerging techniques in the isolation and characterization of extracellular vesicles and their roles in cancer diagnostics and prognostics. Analyst 2017; 141:371-81. [PMID: 26535415 DOI: 10.1039/c5an01775k] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) are cell-derived nanovesicles, present in almost all types of body fluids, which play an important role in intercellular communication and are involved in the transport of biological signals for regulating diverse cellular functions. Due to the increasing clinical interest in the role of EVs in tumor promotion, various techniques for their isolation, detection, and characterization are being developed. In this review, we present an overview of the current EV isolation and characterization methods in addition to their applications and limitations. Furthermore, EVs as the potential emerging biomarkers in cancer management and their clinical implementation are briefly discussed.
Collapse
Affiliation(s)
- Vijaya Sunkara
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan 689-798, Republic of Korea.
| | - Hyun-Kyung Woo
- Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan 689-798, Republic of Korea.
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), UNIST-gil 50, Ulsan 689-798, Republic of Korea. and Department of Biomedical Engineering, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), UNIST-gil 50, Ulsan 689-798, Republic of Korea.
| |
Collapse
|
38
|
Foster BP, Balassa T, Benen TD, Dominovic M, Elmadjian GK, Florova V, Fransolet MD, Kestlerova A, Kmiecik G, Kostadinova IA, Kyvelidou C, Meggyes M, Mincheva MN, Moro L, Pastuschek J, Spoldi V, Wandernoth P, Weber M, Toth B, Markert UR. Extracellular vesicles in blood, milk and body fluids of the female and male urogenital tract and with special regard to reproduction. Crit Rev Clin Lab Sci 2016; 53:379-95. [PMID: 27191915 DOI: 10.1080/10408363.2016.1190682] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Extracellular vesicles (EVs) are released from almost all cells and tissues. They are able to transport substances (e.g. proteins, RNA or DNA) at higher concentrations than in their environment and may adhere in a receptor-controlled manner to specific cells or tissues in order to release their content into the respective target structure. Blood contains high concentrations of EVs mainly derived from platelets, and, at a smaller amount, from erythrocytes. The female and male reproductive tracts produce EVs which may be associated with fertility or infertility and are released into body fluids and mucosas of the urogenital organs. In this review, the currently relevant detection methods are presented and critically compared. During pregnancy, placenta-derived EVs are dynamically detectable in peripheral blood with changing profiles depending upon progress of pregnancy and different pregnancy-associated pathologies, such as preeclampsia. EVs offer novel non-invasive diagnostic tools which may reflect the situation of the placenta and the foetus. EVs in urine have the potential of reflecting urogenital diseases including cancers of the neighbouring organs. Several methods for detection, quantification and phenotyping of EVs have been established, which include electron microscopy, flow cytometry, ELISA-like methods, Western blotting and analyses based on Brownian motion. This review article summarises the current knowledge about EVs in blood and cord blood, in the different compartments of the male and female reproductive tracts, in trophoblast cells from normal and pre-eclamptic pregnancies, in placenta ex vivo perfusate, in the amniotic fluid, and in breast milk, as well as their potential effects on natural killer cells as possible targets.
Collapse
Affiliation(s)
- B P Foster
- a Maternal and Fetal Health Research Centre, School of Biomedicine, University of Manchester, and Manchester Academic Health Sciences Centre, University Research , Manchester , UK
| | - T Balassa
- b Department of Medical Microbiology and Immunology , Medical School, University of Pécs , Pécs , Hungary
| | - T D Benen
- c Microtrac GmbH , Krefeld , Germany
| | - M Dominovic
- d Department of Physiology and Immunology , Medical Faculty, University of Rijeka , Rijeka , Croatia
| | - G K Elmadjian
- e Repro Inova Immunology Laboratory , Sofia , Bulgaria
| | - V Florova
- f Department of Obstetrics , Gynecology and Perinatology, First Moscow State Medical University , Moscow , Russia
| | - M D Fransolet
- g Laboratory of Tumor and Development Biology , GIGA-R, University of Liège , Liège , Belgium
| | - A Kestlerova
- h Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and First Faculty of Medicine , Charles University Prague , Czech Republic
- i Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University , Prague , Czech Republic
| | - G Kmiecik
- j Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero , Brescia , Italy
| | - I A Kostadinova
- k Department of Immunoneuroendocrinology , Institute of Biology and Immunology of Reproduction , Sofia , Bulgaria
| | - C Kyvelidou
- l Department of Biology , University of Crete , Crete , Greece
| | - M Meggyes
- b Department of Medical Microbiology and Immunology , Medical School, University of Pécs , Pécs , Hungary
| | - M N Mincheva
- m Repro Inova Immunology Laboratory , Sofia , Bulgaria
| | - L Moro
- n ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic- Universitat de Barcelona , Barcelona , Spain
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - J Pastuschek
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - V Spoldi
- j Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero , Brescia , Italy
| | - P Wandernoth
- p Institute of Anatomy, University Hospital, University Duisburg-Essen , Essen , Germany
| | - M Weber
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| | - B Toth
- q Department of Gynecological Endocrinology and Fertility Disorders , Ruprecht-Karls University of Heidelberg , Heidelberg , Germany
| | - U R Markert
- o Department of Obstetrics , Placenta-Lab, University Hospital Jena , Jena , Germany
| |
Collapse
|
39
|
Xie Y, Chen Y, Zhang L, Ge W, Tang P. The roles of bone-derived exosomes and exosomal microRNAs in regulating bone remodelling. J Cell Mol Med 2016; 21:1033-1041. [PMID: 27878944 PMCID: PMC5387131 DOI: 10.1111/jcmm.13039] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/19/2016] [Indexed: 12/17/2022] Open
Abstract
Pathological destructive bone diseases are primarily caused by the failure of a lifelong self-renewal process of the skeletal system called bone remodelling. The mechanisms underlying this process include enhanced osteoclast activity and decreased generation of the osteoblast lineage. Intercellular interaction and crosstalk among these cell types are crucial for the maintenance of bone remodelling, either through the secretion of growth factors or direct cell-cell physical engagement. Recent studies have revealed that exosomes derived from bone cells, including osteoclasts, osteoblasts and their precursors, play pivotal roles on bone remodelling by transferring biologically active molecules to target cells, especially in the processes of osteoclast and osteoblast differentiation. Here, we review the contents of bone-derived exosomes and their functions in the regulatory processes of differentiation and communication of osteoclasts and osteoblasts. In addition, we highlight the characteristics of microRNAs of bone-derived exosomes involved in the regulation of bone remodelling, as well as the potential clinical applications of bone-derived exosomes in bone remodelling disorders.
Collapse
Affiliation(s)
- Yong Xie
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Yanyu Chen
- National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| | - Wei Ge
- National Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
40
|
Gomzikova MO, Rizvanov AA. Current Trends in Regenerative Medicine: From Cell to Cell-Free Therapy. BIONANOSCIENCE 2016. [DOI: 10.1007/s12668-016-0348-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
41
|
Safdar A, Saleem A, Tarnopolsky MA. The potential of endurance exercise-derived exosomes to treat metabolic diseases. Nat Rev Endocrinol 2016; 12:504-17. [PMID: 27230949 DOI: 10.1038/nrendo.2016.76] [Citation(s) in RCA: 283] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endurance exercise-mediated multisystemic adaptations are known to mitigate metabolism-related disorders such as obesity and type 2 diabetes mellitus (T2DM). However, the underlying molecular mechanisms that promote crosstalk between organs and orchestrate the pro-metabolic effects of endurance exercise remain unclear. Exercise-induced release of peptides and nucleic acids from skeletal muscle and other organs (collectively termed 'exerkines') has been implicated in mediating these systemic adaptations. Given that the extracellular milieu is probably not a hospitable environment for labile exerkines, a lipid vehicle-based mode of delivery has originated over the course of evolution. Two types of extracellular vesicles, exosomes and microvesicles, have been shown to contain proteins and nucleic acids that participate in a variety of physiological and pathological processes. Exosomes, in particular, have been shown to facilitate the exchange of peptides, microRNA, mRNA and mitochondrial DNA between cells and tissues. Intriguingly, circulatory extracellular vesicle content increases in an intensity-dependant manner in response to endurance exercise. We propose that the systemic benefits of exercise are modulated by exosomes and/or microvesicles functioning in an autocrine, paracrine and/or endocrine manner. Furthermore, we posit that native or modified exosomes, and/or microvesicles enriched with exerkines will have therapeutic utility in the treatment of obesity and T2DM.
Collapse
Affiliation(s)
- Adeel Safdar
- Department of Pediatrics, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Ayesha Saleem
- Department of Pediatrics, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University, 1200 Main Street West, Hamilton, Ontario L8N 3Z5, Canada
- Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada
| |
Collapse
|
42
|
Soekmadji C, Riches JD, Russell PJ, Ruelcke JE, McPherson S, Wang C, Hovens CM, Corcoran NM, Hill MM, Nelson CC. Modulation of paracrine signaling by CD9 positive small extracellular vesicles mediates cellular growth of androgen deprived prostate cancer. Oncotarget 2016; 8:52237-52255. [PMID: 28881726 PMCID: PMC5581025 DOI: 10.18632/oncotarget.11111] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 07/16/2016] [Indexed: 02/06/2023] Open
Abstract
Proliferation and maintenance of both normal and prostate cancer (PCa) cells is highly regulated by steroid hormones, particularly androgens, and the extracellular environment. Herein, we identify the secretion of CD9 positive extracellular vesicles (EV) by LNCaP and DUCaP PCa cells in response to dihydrotestosterone (DHT) and use nano-LC–MS/MS to identify the proteins present in these EV. Subsequent bioinformatic and pathway analyses of the mass spectrometry data identified pathologically relevant pathways that may be altered by EV contents. Western blot and CD9 EV TR-FIA assay confirmed a specific increase in the amount of CD9 positive EV in DHT-treated LNCaP and DUCaP cells and treatment of cells with EV enriched with CD9 after DHT exposure can induce proliferation in androgen-deprived conditions. siRNA knockdown of endogenous CD9 in LNCaPs reduced cellular proliferation and expression of AR and prostate specific antigen (PSA) however knockdown of AR did not alter CD9 expression, also implicating CD9 as an upstream regulator of AR. Moreover CD9 positive EV were also found to be significantly higher in plasma from prostate cancer patients in comparison with benign prostatic hyperplasia patients. We conclude that CD9 positive EV are involved in mediating paracrine signalling and contributing toward prostate cancer progression.
Collapse
Affiliation(s)
- Carolina Soekmadji
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Queensland, Australia
| | - James D Riches
- Central Analytical Research Facility, Institute for Future Environments, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Pamela J Russell
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Queensland, Australia
| | - Jayde E Ruelcke
- Translational Research Institute, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Australia
| | - Stephen McPherson
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Queensland, Australia
| | - Chenwei Wang
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Queensland, Australia
| | - Chris M Hovens
- Australian Prostate Cancer Research Centre Epworth, and Department of Surgery, University of Melbourne, Australia
| | - Niall M Corcoran
- Australian Prostate Cancer Research Centre Epworth, and Department of Surgery, University of Melbourne, Australia
| | | | - Michelle M Hill
- Translational Research Institute, Brisbane, Queensland, Australia.,The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Australia
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre-Queensland, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia.,Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
43
|
DeRita RM, Zerlanko B, Singh A, Lu H, Iozzo RV, Benovic JL, Languino LR. c-Src, Insulin-Like Growth Factor I Receptor, G-Protein-Coupled Receptor Kinases and Focal Adhesion Kinase are Enriched Into Prostate Cancer Cell Exosomes. J Cell Biochem 2016; 118:66-73. [PMID: 27232975 DOI: 10.1002/jcb.25611] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 05/25/2016] [Indexed: 12/21/2022]
Abstract
It is well known that Src tyrosine kinase, insulin-like growth factor 1 receptor (IGF-IR), and focal adhesion kinase (FAK) play important roles in prostate cancer (PrCa) development and progression. Src, which signals through FAK in response to integrin activation, has been implicated in many aspects of tumor biology, such as cell proliferation, metastasis, and angiogenesis. Furthermore, Src signaling is known to crosstalk with IGF-IR, which also promotes angiogenesis. In this study, we demonstrate that c-Src, IGF-IR, and FAK are packaged into exosomes (Exo), c-Src in particular being highly enriched in Exo from the androgen receptor (AR)-positive cell line C4-2B and AR-negative cell lines PC3 and DU145. Furthermore, we show that the active phosphorylated form of Src (SrcpY416 ) is co-expressed in Exo with phosphorylated FAK (FAKpY861 ), a known target site of Src, which enhances proliferation and migration. We further demonstrate for the first time exosomal enrichment of G-protein-coupled receptor kinase (GRK) 5 and GRK6, both of which regulate Src and IGF-IR signaling and have been implicated in cancer. Finally, SrcpY416 and c-Src are both expressed in Exo isolated from the plasma of prostate tumor-bearing TRAMP mice, and those same mice have higher levels of exosomal c-Src than their wild-type counterparts. In summary, we provide new evidence that active signaling molecules relevant to PrCa are enriched in Exo, and this suggests that the Src signaling network may provide useful biomarkers detectable by liquid biopsy, and may contribute to PrCa progression via Exo. J. Cell. Biochem. 118: 66-73, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rachel M DeRita
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Brad Zerlanko
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Amrita Singh
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Huimin Lu
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jeffrey L Benovic
- Departments of Biochemistry and Molecular Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Insights into the molecular roles of heparan sulfate proteoglycans (HSPGs—syndecans) in autocrine and paracrine growth factor signaling in the pathogenesis of Hodgkin’s lymphoma. Tumour Biol 2016; 37:11573-11588. [DOI: 10.1007/s13277-016-5118-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 06/09/2016] [Indexed: 12/25/2022] Open
|
45
|
Fonseca P, Vardaki I, Occhionero A, Panaretakis T. Metabolic and Signaling Functions of Cancer Cell-Derived Extracellular Vesicles. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 326:175-99. [PMID: 27572129 DOI: 10.1016/bs.ircmb.2016.04.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles have gained tremendous attention in the recent years as a novel mechanism of cell to cell communication. There are several types of extracellular vesicles, including exosomes, microvesicles, exosome, like vesicles, apoptotic bodies that differ mainly in the mechanism of biogenesis and secretion. The most well studied type of extracellular vesicles are the exosomes which are endosome-derived vesicles with a diameter of 50-150nm and enriched in ESCRT proteins including Alix, TSG101, Hsp70, and tetraspanins. It is now well established that exosomes promote tumor growth, alter the tumor microenvironment, facilitate the dissemination of cancer cells in an organotropic manner, modulate immune responses, and mediate resistance to therapy. Exosomes have also been recently implicated in an emerging hallmark of cancer, the cancer cell metabolism. The metabolic state of the cell defines, to a certain extent, both the rate of secretion and the molecular content of tumor-derived exosomes. Furthermore, exosomes have been shown to possess intrinsic metabolic activity since they can synthesize ATP by glycolysis. It follows that exosomes carry a number of metabolic enzymes and metabolites, including lactate, PGE, LDH isoforms, pyruvate, and monocarboxylate transporters. Last but not the least, exosomes are implicated in fatty acid synthesis and cholesterol metabolism and are thought to be crucial for the transcellular metabolism procedure. Uptake of exosomes is thought to alter the intracellular metabolic state of the cell. In summary, we describe the state of the art on the role of metabolism in the secretion, uptake, and the biological effects of exosomes in the metabolism of recipient cells.
Collapse
Affiliation(s)
- P Fonseca
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - I Vardaki
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - A Occhionero
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - T Panaretakis
- Department of Oncology-Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| |
Collapse
|
46
|
Kachalaki S, Ebrahimi M, Mohamed Khosroshahi L, Mohammadinejad S, Baradaran B. Cancer chemoresistance; biochemical and molecular aspects: a brief overview. Eur J Pharm Sci 2016; 89:20-30. [PMID: 27094906 DOI: 10.1016/j.ejps.2016.03.025] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 02/02/2016] [Accepted: 03/31/2016] [Indexed: 01/27/2023]
Abstract
The effectiveness of chemotherapy is one of the main challenges in cancer treatment and resistance to classic drugs and traditional treatment processes is an obstacle to this goal. Drug resistance that may be inherent or adventitious can cause poor treatment outcome and tumor relapse. In most cases, resistance to a drug can lead to resistance to many other drugs structure and function of which is not necessarily similar to the first drug. This phenomenon is the main mechanism behind failure of many of metastatic cancers. There are various molecular mechanisms involved in multidrug resistance, including change in the activity of membrane transporters (such as ABC transporters), increase of drug metabolism, change of the target enzyme (such as mutations that change thymidylate synthase and topoisomerases), promotion of DNA damage repair, and escape from drug induced apoptosis. Clinical and laboratory investigations on biomarkers involved in the response to chemotherapy have characterized the key factors behind the failure of treatments. Knowing the molecular factors involved in drug resistance may help us to develop new strategies for more promising chemotherapy and reduce the rate of relapse. In this brief review, molecular mechanisms and tumor microenvironment leading to decreased drug sensitivity, and strategies of reversing drug resistance are described.
Collapse
Affiliation(s)
- Saeed Kachalaki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mina Ebrahimi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Sina Mohammadinejad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
47
|
Weber M, Göhner C, San Martin S, Vattai A, Hutter S, Parraga M, Jeschke U, Schleussner E, Markert UR, Fitzgerald JS. Unique trophoblast stem cell- and pluripotency marker staining patterns depending on gestational age and placenta-associated pregnancy complications. Cell Adh Migr 2016; 10:56-65. [PMID: 26914354 DOI: 10.1080/19336918.2016.1142035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Preeclampsia (PE) and intrauterine growth retardation (IUGR) are rare but severe pregnancy complications that are associated with placental insufficiency often resulting in premature birth. The clinical pathologies are related to gross placental pathologies and trophoblastic deficiencies that might derive from inflammatory processes and oxidative stress injury. The mesenchymal core of placental villi has been identified as a possible niche for trophoblast progenitor cells that are called upon to replenish the injured syncytiotrophoblast layer. These progenitor cells are known to express trophoblast stem cell (CDX2) and pluripotency (SOX2, NANOG and OCT4A) markers, however only little data is available characterizing the expression of these transcription factors beyond the blastocyst stage. We aimed to describe the expression of these factors in healthy 1st and 3rd trimester placentae as well as PE, IUGR and combined PE+IUGR placentae. We analyzed 8 respective samples derived from 1st trimester (elective abortions), and 3rd trimester (healthy controls, PE, IUGR and combined PE+IUGR). We accomplished immunoperoxidase staining to detect the stem cell markers: CDX2 (trophectoderm), SOX2, NANOG and OCT4A (embryonal). Immunoreative scoring was used for objective analyses of staining patterns. All markers display clearly elevated signals in 1st trimester villous samples as compared to healthy 3rd trimester counterparts. Especially CDX2 and NANOG were specific to the cytotrophoblast layer and the mesenchymal core. Specific and differential expression patterns were visible in the villous/extravillous compartment of each placenta-associated pregnancy complication (PE: pan elevated expression; IUGR elevated SOX2 in basal plate; combined PE+IUGR pan loss of expression). Reduction of stem cell transcription factor expression in term placentae indicates temporal regulation, and probably a specific function which is yet to be elucidated. The differential expression patterns within placentae complicated with placenta-associated pregnancy complications indicate that PE, IUGR and combined PE+IUGR are separate entities. It is unclear whether the alterations are the cause or the effect of the clinical pathology.
Collapse
Affiliation(s)
- Maja Weber
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany
| | - Claudia Göhner
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany.,b Department of Obstetrics and Gynecology , University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - Sebastian San Martin
- c Biomedical Research Center, School of Medicine, Universidad de Valparaiso , Chile
| | - Aurelia Vattai
- d Department of Obstetrics and Gynecology , Ludwig Maximilians University of Munich , Munich , Germany
| | - Stefan Hutter
- d Department of Obstetrics and Gynecology , Ludwig Maximilians University of Munich , Munich , Germany
| | - Mario Parraga
- c Biomedical Research Center, School of Medicine, Universidad de Valparaiso , Chile
| | - Udo Jeschke
- d Department of Obstetrics and Gynecology , Ludwig Maximilians University of Munich , Munich , Germany
| | - Ekkehard Schleussner
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany
| | - Udo R Markert
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany
| | - Justine S Fitzgerald
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany.,e Praxisklinik am Anger, Kinderwunschzentrum Erfurt , Erfurt , Germany
| |
Collapse
|
48
|
Philley JV, Kannan A, Qin W, Sauter ER, Ikebe M, Hertweck KL, Troyer DA, Semmes OJ, Dasgupta S. Complex-I Alteration and Enhanced Mitochondrial Fusion Are Associated With Prostate Cancer Progression. J Cell Physiol 2015; 231:1364-74. [PMID: 26530043 DOI: 10.1002/jcp.25240] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/03/2015] [Indexed: 12/21/2022]
Abstract
Mitochondria (mt) encoded respiratory complex-I (RCI) mutations and their pathogenicity remain largely unknown in prostate cancer (PCa). Little is known about the role of mtDNA loss on mt integrity in PCa. We determined mtDNA mutation in human and mice PCa and assessed the impact of mtDNA depletion on mt integrity. We also examined whether the circulating exosomes from PCa patients are transported to mt and carry mtDNA or mt proteins. We have employed next generation sequencing of the whole mt genome in human and Hi-myc PCa. The impact of mtDNA depletion on mt integrity, presence of mtDNA, and protein in sera exosomes was determined. A co-culture of human PCa cells and the circulating exosomes followed by confocal imaging determined co-localization of exosomes and mt. We observed frequent RCI mutations in human and Hi-myc PCa which disrupted corresponding complex protein expression. Depletion of mtDNA in PCa cells influenced mt integrity, increased expression of MFN1, MFN2, PINK1, and decreased expression of MT-TFA. Increased mt fusion and expression of PINK1 and DNM1L were also evident in the Hi-myc tumors. RCI-mtDNA, MFN2, and IMMT proteins were detected in the circulating exosomes of men with benign prostate hyperplasia (BPH) and progressive PCa. Circulating exosomes and mt co-localized in PCa cells. Our study identified new pathogenic RCI mutations in PCa and defined the impact of mtDNA loss on mt integrity. Presence of mtDNA and mt proteins in the circulating exosomes implicated their usefulness for biomarker development.
Collapse
Affiliation(s)
- Julie V Philley
- Department of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Anbarasu Kannan
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Wenyi Qin
- Department of Surgery, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Edward R Sauter
- Department of Surgery, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Kate L Hertweck
- Department of Biology, The University of Texas at Tyler, Tyler, Texas
| | - Dean A Troyer
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Oliver J Semmes
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Santanu Dasgupta
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas
| |
Collapse
|
49
|
The Emerging Role of Extracellular Vesicle-Mediated Drug Resistance in Cancers: Implications in Advanced Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:454837. [PMID: 26587537 PMCID: PMC4637461 DOI: 10.1155/2015/454837] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Accepted: 07/05/2015] [Indexed: 01/07/2023]
Abstract
Emerging evidence has shown that the extracellular vesicles (EVs) regulate various biological processes and can control cell proliferation and survival, as well as being involved in normal cell development and diseases such as cancers. In cancer treatment, development of acquired drug resistance phenotype is a serious issue. Recently it has been shown that the presence of multidrug resistance proteins such as Pgp-1 and enrichment of the lipid ceramide in EVs could have a role in mediating drug resistance. EVs could also mediate multidrug resistance through uptake of drugs in vesicles and thus limit the bioavailability of drugs to treat cancer cells. In this review, we discussed the emerging evidence of the role EVs play in mediating drug resistance in cancers and in particular the role of EVs mediating drug resistance in advanced prostate cancer. The role of EV-associated multidrug resistance proteins, miRNA, mRNA, and lipid as well as the potential interaction(s) among these factors was probed. Lastly, we provide an overview of the current available treatments for advanced prostate cancer, considering where EVs may mediate the development of resistance against these drugs.
Collapse
|
50
|
Poncelet P, Robert S, Bailly N, Garnache-Ottou F, Bouriche T, Devalet B, Segatchian JH, Saas P, Mullier F. Tips and tricks for flow cytometry-based analysis and counting of microparticles. Transfus Apher Sci 2015; 53:110-26. [DOI: 10.1016/j.transci.2015.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|