1
|
Liu Y, Pierre CJ, Joshi S, Sun L, Li Y, Guan J, Favor JDL, Holmes C. Cell-Specific Impacts of Surface Coating Composition on Extracellular Vesicle Secretion. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29737-29759. [PMID: 38805212 DOI: 10.1021/acsami.4c03213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Biomaterial properties have recently been shown to modulate extracellular vesicle (EV) secretion and cargo; however, the effects of substrate composition on EV production remain underexplored. This study investigates the impacts of surface coatings composed of collagen I (COLI), fibronectin (FN), and poly l-lysine (PLL) on EV secretion for applications in therapeutic EV production and to further understanding of how changes in the extracellular matrix microenvironment affect EVs. EV secretion from primary bone marrow-derived mesenchymal stromal cells (BMSCs), primary adipose-derived stem cells (ASCs), HEK293 cells, NIH3T3 cells, and RAW264.7 cells was characterized on the different coatings. Expression of EV biogenesis genes and cellular adhesion genes was also analyzed. COLI coatings significantly decreased EV secretion in RAW264.7 cells, with associated decreases in cell viability and changes in EV biogenesis-related and cell adhesion genes at day 4. FN coatings increased EV secretion in NIH3T3 cells, while PLL coatings increased EV secretion in ASCs. Surface coatings had significant effects on the capacity of EVs derived from RAW264.7 and NIH3T3 cells to impact in vitro macrophage proliferation. Overall, surface coatings had different cell-specific effects on EV secretion and in vitro functional capacity, thus highlighting the potential of substrate coatings to further the development of clinical EV production systems.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida A&M University, Florida State University, 2525 Pottsdamer Street, Tallahasee, Florida 32310-6046, United States
| | - Clifford J Pierre
- Department of Health, Nutrition, and Food Science, College of Education, Health and Human Sciences, Florida State University, 1114 West Call Street, Tallahasee, Florida 32306, United States
| | - Sailesti Joshi
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida A&M University, Florida State University, 2525 Pottsdamer Street, Tallahasee, Florida 32310-6046, United States
| | - Li Sun
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida A&M University, Florida State University, 2525 Pottsdamer Street, Tallahasee, Florida 32310-6046, United States
- Department of Biomedical Sciences, College of Medicine, Florida State University, 1115 West Call Street, Tallahasee, Florida 32306-4300, United States
| | - Yan Li
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida A&M University, Florida State University, 2525 Pottsdamer Street, Tallahasee, Florida 32310-6046, United States
| | - Jingjiao Guan
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida A&M University, Florida State University, 2525 Pottsdamer Street, Tallahasee, Florida 32310-6046, United States
| | - Justin D La Favor
- Department of Health, Nutrition, and Food Science, College of Education, Health and Human Sciences, Florida State University, 1114 West Call Street, Tallahasee, Florida 32306, United States
| | - Christina Holmes
- Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida A&M University, Florida State University, 2525 Pottsdamer Street, Tallahasee, Florida 32310-6046, United States
| |
Collapse
|
2
|
Huang MB, Brena D, Wu JY, Shelton M, Bond VC. SMR peptide antagonizes Staphylococcus aureus biofilm formation. Microbiol Spectr 2024; 12:e0258323. [PMID: 38170991 PMCID: PMC10846015 DOI: 10.1128/spectrum.02583-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/07/2023] [Indexed: 01/05/2024] Open
Abstract
The emergence and international dissemination of multi-drug resistant Staphylococcus aureus (S. aureus) strains challenge current antibiotic-based therapies, representing an urgent threat to public health worldwide. In the U.S. alone, S. aureus infections are responsible for 11,000 deaths and 500,000 hospitalizations annually. Biofilm formation is a major contributor to antibiotic tolerance and resistance-induced delays in empirical therapy with increased infection severity, frequency, treatment failure, and mortality. Developing novel treatment strategies to prevent and disrupt biofilm formation is imperative. In this article, we test the Secretion Modification Region (SMR) peptides for inhibitory effects on resistant S. aureus biofilm-forming capacity by targeting the molecular chaperone DnaK. The dose effect of SMR peptides on biofilm formation was assessed using microtiter plate methods and confocal microscopy. Interaction between the antagonist and DnaK was determined by immune precipitation with anti-Flag M2 Affinity and Western blot analysis. Increasing SMR peptide concentrations exhibited increasing blockade of S. aureus biofilm formation with significant inhibition found at 18 µM, 36 µM, and 72 µM. This work supports the potential therapeutic benefit of SMR peptides in reducing biofilm viability and could improve the susceptibility to antimicrobial agents.IMPORTANCEThe development of anti-biofilm agents is critical to restoring bacterial sensitivity, directly combating the evolution of resistance, and overall reducing the clinical burden related to pervasive biofilm-mediated infections. Thus, in this study, the SMR peptide, a novel small molecule derived from the HIV Nef protein, was preliminarily explored for anti-biofilm properties. The SMR peptide was shown to effectively target the molecular chaperone DnaK and inhibit biofilm formation in a dose-dependent manner. These results support further investigation into the mechanism of SMR peptide-mediated biofilm formation and inhibition to benefit rational drug design and the identification of therapeutic targets.
Collapse
Affiliation(s)
- Ming-Bo Huang
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Dara Brena
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Jennifer Y. Wu
- Columbia University School of International and Public Affairs, Columbia University, New York, New York, USA
| | - Martin Shelton
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
- NanoString Technologies, Inc, Seattle, Washington, USA
| | - Vincent C. Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Li J, Wang J, Chen Z, Hu P, Zhang X, Guo X, Zhu X, Huang Y. An Exosome-Related Long Non-coding RNA (lncRNA)-Based Signature for Prognosis and Therapeutic Interventions in Lung Adenocarcinoma. Cureus 2023; 15:e47574. [PMID: 38021786 PMCID: PMC10666655 DOI: 10.7759/cureus.47574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background The poor prognosis of lung adenocarcinoma (LUAD) has been confirmed by a large number of studies, so it is necessary to construct a prognosis model. In addition, exosome is closely related to tumors, but there are few studies on exosome-related long non-coding RNA (lncRNA) (ExolncRNA). Methods In this study, we designed a prognostic model, exosome-related lncRNA-based signature (ExoLncSig), using ExolncRNA expression profiles of LUAD patients from The Cancer Genome Atlas (TCGA). ExolncRNAs were identified through univariate and multivariate and Lasso analyses. Subsequently, based on the ExoLncSig, gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, immune function and immunotherapy analysis, drug screening, and so on were performed. Results AC026355.2, AC108136.1, AL590428.1, and LINC01312 were examined to establish the ExoLncSig. Gene enrichment analysis identified potential prognostic markers and therapeutic targets, including human leukocyte antigen (HLA), parainflammation, chemokine receptor (CCR), antigen-presenting cell (APC) co-inhibition, cancer-associated fibroblast (CAF), and myeloid-derived suppressor cell (MDSC). Moreover, we ascertained that the high-risk subgroup exhibits heightened susceptibility to pharmaceutical agents. Conclusion Our findings indicate that ExoLncSig holds promise as a valuable prognostic marker in LUAD. Furthermore, the immunogenic properties of ExolncRNAs may pave the way for the development of a therapeutic vaccine against LUAD.
Collapse
Affiliation(s)
- Jinghong Li
- Computational Oncology Laboratory, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, CHN
| | - Junhua Wang
- Oncology Department, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, CHN
| | - Zhihong Chen
- Computational Oncology Laboratory, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, CHN
| | - Pan Hu
- Computational Oncology Laboratory, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, CHN
| | - Xiaodan Zhang
- Computational Oncology Laboratory, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, CHN
| | - Xiaojun Guo
- Computational Oncology Laboratory, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, CHN
| | - Xiao Zhu
- Genetics Department, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, CHN
| | - Yongmei Huang
- Computational Oncology Laboratory, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, CHN
| |
Collapse
|
4
|
Loric S, Denis JA, Desbene C, Sabbah M, Conti M. Extracellular Vesicles in Breast Cancer: From Biology and Function to Clinical Diagnosis and Therapeutic Management. Int J Mol Sci 2023; 24:7208. [PMID: 37108371 PMCID: PMC10139222 DOI: 10.3390/ijms24087208] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer (BC) is the first worldwide most frequent cancer in both sexes and the most commonly diagnosed in females. Although BC mortality has been thoroughly declining over the past decades, there are still considerable differences between women diagnosed with early BC and when metastatic BC is diagnosed. BC treatment choice is widely dependent on precise histological and molecular characterization. However, recurrence or distant metastasis still occurs even with the most recent efficient therapies. Thus, a better understanding of the different factors underlying tumor escape is mainly mandatory. Among the leading candidates is the continuous interplay between tumor cells and their microenvironment, where extracellular vesicles play a significant role. Among extracellular vesicles, smaller ones, also called exosomes, can carry biomolecules, such as lipids, proteins, and nucleic acids, and generate signal transmission through an intercellular transfer of their content. This mechanism allows tumor cells to recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. By reciprocity, stromal cells can also use exosomes to profoundly modify tumor cell behavior. This review intends to cover the most recent literature on the role of extracellular vesicle production in normal and cancerous breast tissues. Specific attention is paid to the use of extracellular vesicles for early BC diagnosis, follow-up, and prognosis because exosomes are actually under the spotlight of researchers as a high-potential source of liquid biopsies. Extracellular vesicles in BC treatment as new targets for therapy or efficient nanovectors to drive drug delivery are also summarized.
Collapse
Affiliation(s)
- Sylvain Loric
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | | | - Cédric Desbene
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Michèle Sabbah
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Marc Conti
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
- INTEGRACELL SAS, 91160 Longjumeau, France
| |
Collapse
|
5
|
Huang MB, Brena D, Wu JY, Roth WW, Owusu S, Bond VC. Novel secretion modification region (SMR) peptide exhibits anti-metastatic properties in human breast cancer cells. Sci Rep 2022; 12:13204. [PMID: 35915218 PMCID: PMC9343421 DOI: 10.1038/s41598-022-17534-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/27/2022] [Indexed: 12/01/2022] Open
Abstract
Breast cancer is the second leading cause of cancer-related mortality in women worldwide, with nearly 90% attributed to metastatic progression. Exosomes containing epithelial–mesenchymal transition (EMT) ‘programs’ transmit pro-metastatic phenotypes. Our group discovered and developed a novel anti-cancer SMR peptide that antagonizes breast cancer cell exosome release resulting in cell cycle arrest and tumor growth suppression. This study aims to evaluate the anti-metastatic capabilities of the SMR peptide, focusing on exosomes and EMT. Breast cancer cell lines MDA-MB-231 and MCF-7 were treated with the SMRwt peptide, and the following assays were performed: cell wound-healing, migration, invasion. The SMRwt peptide consists of the following amino acid sequence VGFPVAAVGFPVDYKDDDDK and contains the SMR domain (66VGFPV70) of the HIV-1 Nef protein. Western blot analysis detected epithelial and mesenchymal markers to evaluate EMT progression. Extracellular vesicle type and quantity were assessed through NanoSight analysis. Mortalin and Vimentin knockdown was achieved through antibody targeting and miRNAs. Data gathered demonstrated that the SMR peptide interacts with Mortalin and Vimentin to inhibit pro-EMT exosome release and induce EMT tumor suppressor protein expression. Specifically, SMRwt treatment reduced mesenchymal markers Mortalin and Vimentin expression, while the epithelial marker E-cadherin expression was increased in breast cancer cells and breast cancer-derived exosomes. The SMR peptide specificity was identified as no effect was observed for MCF-10A exosome release or function. Direct Mortalin knockdown paralleled the results of SMR peptide treatment with an effective blockade of breast cancer cell migration. Conversely, the invasion assay differed between breast cancer cell lines with invasion blocked for in MCF-7 but not in MDA-MB-231. These results reinforce the therapeutic value of targeting breast cancer exosome release and reinforce Mortalin and Vimentin as critical regulators and therapeutic targets in breast cancer cell progression, EMT, and metastatic potential. A greater understanding of the SMR peptide mechanism of action will benefit the therapeutic design of anti-metastatic agents.
Collapse
Affiliation(s)
- Ming-Bo Huang
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| | - Dara Brena
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Jennifer Y Wu
- Columbia College, Columbia University, New York, NY, 10027, USA
| | - William W Roth
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Samuel Owusu
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Vincent C Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| |
Collapse
|
6
|
Alberti G, Vergilio G, Paladino L, Barone R, Cappello F, Conway de Macario E, Macario AJL, Bucchieri F, Rappa F. The Chaperone System in Breast Cancer: Roles and Therapeutic Prospects of the Molecular Chaperones Hsp27, Hsp60, Hsp70, and Hsp90. Int J Mol Sci 2022; 23:ijms23147792. [PMID: 35887137 PMCID: PMC9324353 DOI: 10.3390/ijms23147792] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/30/2022] [Accepted: 07/10/2022] [Indexed: 12/26/2022] Open
Abstract
Breast cancer (BC) is a major public health problem, with key pieces of information needed for developing preventive and curative measures still missing. For example, the participation of the chaperone system (CS) in carcinogenesis and anti-cancer responses is poorly understood, although it can be predicted to be a crucial factor in these mechanisms. The chief components of the CS are the molecular chaperones, and here we discuss four of them, Hsp27, Hsp60, Hsp70, and Hsp90, focusing on their pro-carcinogenic roles in BC and potential for developing anti-BC therapies. These chaperones can be targets of negative chaperonotherapy, namely the elimination/blocking/inhibition of the chaperone(s) functioning in favor of BC, using, for instance, Hsp inhibitors. The chaperones can also be employed in immunotherapy against BC as adjuvants, together with BC antigens. Extracellular vesicles (EVs) in BC diagnosis and management are also briefly discussed, considering their potential as easily accessible carriers of biomarkers and as shippers of anti-cancer agents amenable to manipulation and controlled delivery. The data surveyed from many laboratories reveal that, to enhance the understanding of the role of the CS in BS pathogenesis, one must consider the CS as a physiological system, encompassing diverse members throughout the body and interacting with the ubiquitin–proteasome system, the chaperone-mediated autophagy machinery, and the immune system (IS). An integrated view of the CS, including its functional partners and considering its highly dynamic nature with EVs transporting CS components to reach all the cell compartments in which they are needed, opens as yet unexplored pathways leading to carcinogenesis that are amenable to interference by anti-cancer treatments centered on CS components, such as the molecular chaperones.
Collapse
Affiliation(s)
- Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
| | - Giuseppe Vergilio
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
| | - Letizia Paladino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
- Correspondence:
| | - Rosario Barone
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
| | - Francesco Cappello
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
| | - Everly Conway de Macario
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA;
| | - Alberto J. L. Macario
- Euro-Mediterranean Institute of Science and Technology (IEMEST), 90139 Palermo, Italy;
- Department of Microbiology and Immunology, School of Medicine, University of Maryland at Baltimore-Institute of Marine and Environmental Technology (IMET), Baltimore, MD 21202, USA;
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy; (G.A.); (G.V.); (R.B.); (F.C.); (F.B.); (F.R.)
| |
Collapse
|
7
|
Yoon AR, Wadhwa R, Kaul SC, Yun CO. Why is Mortalin a Potential Therapeutic Target for Cancer? Front Cell Dev Biol 2022; 10:914540. [PMID: 35859897 PMCID: PMC9290191 DOI: 10.3389/fcell.2022.914540] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide, accounting for nearly 10 million deaths in 2020. Therefore, cancer therapy is a priority research field to explore the biology of the disease and identify novel targets for the development of better treatment strategies. Mortalin is a member of the heat shock 70 kDa protein family. It is enriched in several types of cancer and contributes to carcinogenesis in various ways, including inactivation of the tumor suppressor p53, deregulation of apoptosis, induction of epithelial–mesenchymal transition, and enhancement of cancer stemness. It has been studied extensively as a therapeutic target for cancer treatment, and several types of anti-mortalin molecules have been discovered that effectively suppress the tumor cell growth. In this review, we 1) provide a comprehensive sketch of the role of mortalin in tumor biology; 2) discuss various anti-mortalin molecules, including natural compounds, synthetic small molecules, peptides, antibodies, and nucleic acids, that have shown potential for cancer treatment in laboratory studies; and 3) provide future perspectives in cancer treatment.
Collapse
Affiliation(s)
- A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul, South Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
| | - Renu Wadhwa
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Sunil C Kaul
- AIST-INDIA DAILAB, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul, South Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
- GeneMedicine CO, Ltd, Seoul, South Korea
- *Correspondence: Chae-Ok Yun,
| |
Collapse
|
8
|
Huang MB, Gao Z, Xia M, Zhao X, Fan X, Lin S, Zhang L, Huang L, Wei A, Zhou H, Wu JY, Roth WW, Bond VC, Leng J. Improved Aitongxiao prescription (I-ATXP) induces apoptosis, cell cycle arrest and blocks exosomes release in hepatocellular carcinoma (HCC) cells. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2022; 14:90-113. [PMID: 35619665 PMCID: PMC9123477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/13/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the second most common malignancy globally, after lung cancer, accounting for 85-90% of primary liver cancer. Hepatitis B virus (HBV) infection is considered the leading risk factor for HCC development in China. HCC is a highly malignant cancer whose metastasis is primarily influenced by the tumor microenvironment. The role of exosomes in cancer development has become the focus of much research due to the many newly described contents of exosomes, which may contribute to tumorigenesis. However, the possible role exosomes play in the interactions between HCC cells and their surrounding hepatic milieu is mainly unknown. We discovered an Improved Aitongxiao Prescription (I-ATXP): an 80% alcohol extract from a mix of 15 specific plant and animal compounds, which had been shown to have an anticancer effect through inducing apoptosis and cell cycle arrest and blocking exosomes release in HCC cells. However, the anticancer mechanism of I-ATXP on human liver carcinoma is still unclear. OBJECTIVE Due to its inhibitory effects on chemical carcinogenesis and inflammation, I-ATXP has been proposed as an effective agent for preventing or treating human liver carcinoma. In this study, we aimed to explore the effect of I-ATXP on proliferation, apoptosis, and cell cycles of different HCC cell lines. We investigated the impact of I-ATXP on exosomes' secretion derived from these HCC cells. METHODS The inhibitory effect of I-ATXP on proliferation and cytotoxicity of HepG2, SMMC7721, HKCL-C3 HCC cell lines, and MIHA immortalized hepatocyte cell line was assessed by CCK-8 assay. The cell cycle distribution and cell apoptosis were determined by flow cytometry using Annexin V-FITC/PI staining. The expression of Alix and CD63 of exosome marker proteins was detected by western blotting. The exosome protein concentration was measured by a fluorescent plate reader. The exosome-specific enzyme activity was measured by acetylcholinesterase (AchE) assay, and exosome morphological characteristics were identified by transmission electron microscopy (TEM). RESULTS I-ATXP inhibited the growth of HCC cells in a dose and time-dependent manner. Flow cytometry analysis showed that I-ATXP induced G0/G1 phase arrest and cell apoptosis. The I-ATX reduced HepG2, SMMC7721, and HKCI-C HCC cell lines exosomes release and low-dose I-ATXP significantly enhanced the growth inhibition induced by 5-Fu. Western blot analysis shows that after HCC cell lines were treated with various concentrations of I-ATXP (0.125-1 mg/ml) for 24 h, exosomes derived from three different HCC cells expressed exosome-specific proteins Alix and CD63. Compared with the untreated group, with the increment of the concentration of I-ATXP, the expression of exosome-specific proteins Alix and CD63 were reduced. These results suggest that I-ATXP can inhibit the release of exosomes with Alix and CD63 protein from HCC cells. CONCLUSIONS I-ATXP is a traditional Chinese medicine that acts as an effective agent for preventing or treating human liver carcinoma. (i) I-ATXP can effectively inhibit cell proliferation of different HCC cells in a time and dose-dependent manner. Compared with 5-Fu, I-ATXP exhibited more selective proliferation inhibition in HCC cells, displaying traditional Chinese medicine advantages on tumor therapy and providing the experimental basis for I-ATXP clinical application. (ii) I-ATXP can induce apoptosis and cell cycle arrest in HCC cells. The CCK-8 assay results indicated that I-ATXP could inhibit HCC cell proliferation mediated by apoptosis and cell cycle arrest. (iii) I-ATXP can inhibit both the exosome releases and expression of CD63, and Alix derived from HCC cells, but the exosomes derived from liver cancer cells affect liver cancer cells' biological properties such as proliferation, invasion, and migration. These suggest that I-ATXP may affect HCC cells via regulation of exosomes of HCC cells, further indicating the potential clinical values of I-ATXP for the prevention or treatment of human liver carcinoma.
Collapse
Affiliation(s)
- Ming-Bo Huang
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of MedicineAtlanta, Georgia 30310, USA
| | - Zhao Gao
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| | - Meng Xia
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| | - Xiaoqing Zhao
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| | - Xiaoyuan Fan
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| | - Shijie Lin
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| | - Lifeng Zhang
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| | - Li Huang
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| | - Ailing Wei
- The First Affiliated Hospital of Guangxi University of Chinese MedicineNanning 530023, Guangxi, China
| | - Hu Zhou
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| | - Jennifer Y Wu
- Columbia College, Columbia UniversityNew York, NY 10027, USA
| | - William W Roth
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of MedicineAtlanta, Georgia 30310, USA
| | - Vincent C Bond
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of MedicineAtlanta, Georgia 30310, USA
| | - Jing Leng
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese MedicineNanning 530200, Guangxi, China
| |
Collapse
|
9
|
Exosomal non-coding RNAs: Emerging roles in bilateral communication between cancer cells and macrophages. Mol Ther 2022; 30:1036-1053. [PMID: 34864204 PMCID: PMC8899606 DOI: 10.1016/j.ymthe.2021.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/28/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is a dynamic network of cellular organization that comprises diverse cell types and significantly contributes to cancer development. As pivotal immune stromal cells in the TME, macrophages are extensively heterogeneous and exert both antitumor and protumor functions. Exosomes are nanosized extracellular membranous vesicles with diameters between 30 and 150 nm. By transferring multiple bioactive substances such as proteins, lipids, and nucleic acids, exosomes play an important role in the communication between cells. Recently, growing evidence has demonstrated that non-coding RNAs (ncRNAs) are enriched in exosomes and that exosomal ncRNAs are involved in the crosstalk between cancer cells and macrophages. Furthermore, circulating exosomal ncRNAs can be detected in biofluids, serving as promising noninvasive biomarkers for the early diagnosis and prognostic prediction of cancer. Exosome-based therapies are emerging as potent strategies that can be utilized to alleviate tumor progression. Herein, the present knowledge of exosomal ncRNAs and their vital roles in regulating the interplay between cancer cells and macrophages, as well as their clinical applications are summarized.
Collapse
|
10
|
Extracellular Vesicles as Mediators of Therapy Resistance in the Breast Cancer Microenvironment. Biomolecules 2022; 12:biom12010132. [PMID: 35053279 PMCID: PMC8773878 DOI: 10.3390/biom12010132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/17/2022] Open
Abstract
Resistance to various therapies, including novel immunotherapies, poses a major challenge in the management of breast cancer and is the leading cause of treatment failure. Bidirectional communication between breast cancer cells and the tumour microenvironment is now known to be an important contributor to therapy resistance. Several studies have demonstrated that crosstalk with the tumour microenvironment through extracellular vesicles is an important mechanism employed by cancer cells that leads to drug resistance via changes in protein, lipid and nucleic acid cargoes. Moreover, the cargo content enables extracellular vesicles to be used as effective biomarkers for predicting response to treatments and as potential therapeutic targets. This review summarises the literature to date regarding the role of extracellular vesicles in promoting therapy resistance in breast cancer through communication with the tumour microenvironment.
Collapse
|
11
|
Kabakov AE, Gabai VL. HSP70s in Breast Cancer: Promoters of Tumorigenesis and Potential Targets/Tools for Therapy. Cells 2021; 10:cells10123446. [PMID: 34943954 PMCID: PMC8700403 DOI: 10.3390/cells10123446] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/25/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022] Open
Abstract
The high frequency of breast cancer worldwide and the high mortality among women with this malignancy are a serious challenge for modern medicine. A deeper understanding of the mechanisms of carcinogenesis and emergence of metastatic, therapy-resistant breast cancers would help development of novel approaches to better treatment of this disease. The review is dedicated to the role of members of the heat shock protein 70 subfamily (HSP70s or HSPA), mainly inducible HSP70, glucose-regulated protein 78 (GRP78 or HSPA5) and GRP75 (HSPA9 or mortalin), in the development and pathogenesis of breast cancer. Various HSP70-mediated cellular mechanisms and pathways which contribute to the oncogenic transformation of mammary gland epithelium are reviewed, as well as their role in the development of human breast carcinomas with invasive, metastatic traits along with the resistance to host immunity and conventional therapeutics. Additionally, intracellular and cell surface HSP70s are considered as potential targets for therapy or sensitization of breast cancer. We also discuss a clinical implication of Hsp70s and approaches to targeting breast cancer with gene vectors or nanoparticles downregulating HSP70s, natural or synthetic (small molecule) inhibitors of HSP70s, HSP70-binding antibodies, HSP70-derived peptides, and HSP70-based vaccines.
Collapse
Affiliation(s)
- Alexander E. Kabakov
- Department of Radiation Biochemistry, A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Koroleva 4, 249036 Obninsk, Russia;
| | - Vladimir L. Gabai
- CureLab Oncology Inc., Dedham, MA 02026, USA
- Correspondence: ; Tel.: +1-617-319-7314
| |
Collapse
|
12
|
Brena D, Huang MB, Bond V. Extracellular vesicle-mediated transport: Reprogramming a tumor microenvironment conducive with breast cancer progression and metastasis. Transl Oncol 2021; 15:101286. [PMID: 34839106 PMCID: PMC8636863 DOI: 10.1016/j.tranon.2021.101286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles’ (EVs) role in breast tumor microenvironment and pre-metastatic niche development. Breast cancer EV-mediated transmission of pro-metastatic and drug-resistant phenotypes. Precision medicine with EVs as biomarkers and delivery vehicles for drug and anticancer genetic material.
Breast cancer metastatic progression to critical secondary sites is the second leading cause of cancer-related mortality in women. While existing therapies are highly effective in combating primary tumors, metastatic disease is generally deemed incurable with a median survival of only 2, 3 years. Extensive efforts have focused on identifying metastatic contributory targets for therapeutic antagonism and prevention to improve patient survivability. Excessive breast cancer release of extracellular vesicles (EVs), whose contents stimulate a metastatic phenotype, represents a promising target. Complex breast cancer intercellular communication networks are based on EV transport and transference of molecular information is in bulk resulting in complete reprogramming events within recipient cells. Other breast cancer cells can acquire aggressive phenotypes, endothelial cells can be induced to undergo tubule formation, and immune cells can be neutralized. Recent advancements continue to implicate the critical role EVs play in cultivating a tumor microenvironment tailored to cancer proliferation, metastasis, immune evasion, and conference of drug resistance. This literature review serves to frame the role of EV transport in breast cancer progression and metastasis. The following five sections will be addressed: (1) Intercellular communication in developing a tumor microenvironment & pre-metastatic niche. (2) Induction of the epithelial-to-mesenchymal transition (EMT). (3). Immune suppression & evasion. (4) Transmission of drug resistance mechanisms. (5) Precision medicine: clinical applications of EVs.
Collapse
Affiliation(s)
- Dara Brena
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| | - Ming-Bo Huang
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| | - Vincent Bond
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States
| |
Collapse
|
13
|
Mammes A, Pasquier J, Mammes O, Conti M, Douard R, Loric S. Extracellular vesicles: General features and usefulness in diagnosis and therapeutic management of colorectal cancer. World J Gastrointest Oncol 2021; 13:1561-1598. [PMID: 34853637 PMCID: PMC8603448 DOI: 10.4251/wjgo.v13.i11.1561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/29/2021] [Accepted: 09/08/2021] [Indexed: 02/06/2023] Open
Abstract
In the world, among all type of cancers, colorectal cancer (CRC) is the third most commonly diagnosed in males and the second in females. In most of cases, (RP1) patients’ prognosis limitation with malignant tumors can be attributed to delayed diagnosis of the disease. Identification of patients with early-stage disease leads to more effective therapeutic interventions. Therefore, new screening methods and further innovative treatment approaches are mandatory as they may lead to an increase in progression-free and overall survival rates. For the last decade, the interest in extracellular vesicles (EVs) research has exponentially increased as EVs generation appears to be a universal feature of every cell that is strongly involved in many mechanisms of cell-cell communication either in physiological or pathological situations. EVs can cargo biomolecules, such as lipids, proteins, nucleic acids and generate transmission signal through the intercellular transfer of their content. By this mechanism, tumor cells can recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. This review intends to cover the most recent literature on the role of EVs production in colorectal normal and cancer tissues. Specific attention is paid to the use of EVs for early CRC diagnosis, follow-up, and prognosis as EVs have come into the spotlight of research as a high potential source of ‘liquid biopsies’. The use of EVs as new targets or nanovectors as drug delivery systems for CRC therapy is also summarized.
Collapse
Affiliation(s)
- Aurelien Mammes
- INSERM UMR-938, Cancer Biology and Therapeutics Unit, Saint-Antoine Research Center, Saint Antoine University Hospital, Paris 75012, France
| | - Jennifer Pasquier
- INSERM UMR-938, Cancer Biology and Therapeutics Unit, Saint-Antoine Research Center, Saint Antoine University Hospital, Paris 75012, France
| | | | - Marc Conti
- INSERM UMR-938, Cancer Biology and Therapeutics Unit, Saint-Antoine Research Center, Saint Antoine University Hospital, Paris 75012, France
- Metabolism Research Unit, Integracell SAS, Longjumeau 91160, France
| | - Richard Douard
- UCBM, Necker University Hospital, Paris 75015, France
- Gastrointestinal Surgery Department, Clinique Bizet, Paris 75016, France
| | - Sylvain Loric
- INSERM UMR-938, Cancer Biology and Therapeutics Unit, Saint-Antoine Research Center, Saint Antoine University Hospital, Paris 75012, France
| |
Collapse
|
14
|
Rai R, Kennedy AL, Isingizwe ZR, Javadian P, Benbrook DM. Similarities and Differences of Hsp70, hsc70, Grp78 and Mortalin as Cancer Biomarkers and Drug Targets. Cells 2021; 10:cells10112996. [PMID: 34831218 PMCID: PMC8616428 DOI: 10.3390/cells10112996] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Upregulation of Heath Shock Protein 70 (HSP70) chaperones supports cancer cell survival. Their high homology causes a challenge to differentiate them in experimental or prevention and treatment strategies. The objective of this investigation was to determine similarities and differences of Hsp70, hsc70, Grp78 and Mortalin members of the HSP70 family encoded by HSPA1, HSPA8, HSPA5 and HSPA9 genes, respectively. Methods: Literature reviews were conducted using HSPA1, HSPA5, HSPA8 and HSPA9 gene or protein names or synonyms combined with biological or cancer-relevant terms. Ingenuity Pathway Analysis was used to identify and compare profiles of proteins that directly bind individual chaperones and their associated pathways. TCGA data was probed to identify associations of hsc70 with cancer patient survival. ClinicalTrials.gov was used to identify HSP70 family studies. Results: The chaperones have similar protein folding functions. Their different cellular effects are determined by co-chaperones and client proteins combined with their intra- and extra-cellular localizations. Their upregulation is associated with worse patient prognosis in multiple cancers and can stimulate tumor immune responses or drug resistance. Their inhibition selectively kills cancer over healthy cells. Conclusions: Differences in Hsp70, hsc70, Grp78 and mortalin provide opportunities to calibrate HSP70 inhibitors for individual cancers and combination therapies.
Collapse
Affiliation(s)
- Rajani Rai
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (P.J.)
| | - Amy L. Kennedy
- Pathology Department, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Zitha Redempta Isingizwe
- Pharmaceutical Sciences Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Pouya Javadian
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (P.J.)
| | - Doris Mangiaracina Benbrook
- Gynecologic Oncology Section, Obstetrics and Gynecology Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.R.); (P.J.)
- Pathology Department, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Pharmaceutical Sciences Department, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Correspondence: ; Tel.: +1-405-271-5523
| |
Collapse
|
15
|
Hosseini R, Asef-Kabiri L, Yousefi H, Sarvnaz H, Salehi M, Akbari ME, Eskandari N. The roles of tumor-derived exosomes in altered differentiation, maturation and function of dendritic cells. Mol Cancer 2021; 20:83. [PMID: 34078376 PMCID: PMC8170799 DOI: 10.1186/s12943-021-01376-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor-derived exosomes (TDEs) have been shown to impede anti-tumor immune responses via their immunosuppressive cargo. Since dendritic cells (DCs) are the key mediators of priming and maintenance of T cell-mediated responses; thus it is logical that the exosomes released by tumor cells can exert a dominant influence on DCs biology. This paper intends to provide a mechanistic insight into the TDEs-mediated DCs abnormalities in the tumor context. More importantly, we discuss extensively how tumor exosomes induce subversion of DCs differentiation, maturation and function in separate sections. We also briefly describe the importance of TDEs at therapeutic level to help guide future treatment options, in particular DC-based vaccination strategy, and review advances in the design and discovery of exosome inhibitors. Understanding the exosomal content and the pathways by which TDEs are responsible for immune evasion may help to revise treatment rationales and devise novel therapeutic approaches to overcome the hurdles in cancer treatment.
Collapse
Affiliation(s)
- Reza Hosseini
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Leila Asef-Kabiri
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, USA
| | - Hamzeh Sarvnaz
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
16
|
Esposito CL, Quintavalle C, Ingenito F, Rotoli D, Roscigno G, Nuzzo S, Thomas R, Catuogno S, de Franciscis V, Condorelli G. Identification of a novel RNA aptamer that selectively targets breast cancer exosomes. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:982-994. [PMID: 33614245 PMCID: PMC7868932 DOI: 10.1016/j.omtn.2021.01.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/14/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is a leading cause of cancer mortality in women. Despite advances in its management, the identification of new options for early-stage diagnosis and therapy of this tumor still represents a crucial challenge. Increasing evidence indicates that extracellular vesicles called exosomes may have great potential as early diagnostic biomarkers and regulators of many cancers, including breast cancer. Therefore, exploiting molecules able to selectively recognize them is of great interest. Here, we developed a novel differential SELEX strategy, called Exo-SELEX, to isolate nucleic acid aptamers against intact exosomes derived from primary breast cancer cells. Among the obtained sequences, we optimized a high-affinity aptamer (ex-50.T) able to specifically recognize exosomes from breast cancer cells or patient serum samples. Furthermore, we demonstrated that the ex.50.T is a functional inhibitor of exosome cellular uptake and antagonizes cancer exosome-induced cell migration in vitro. This molecule provides an innovative tool for the specific exosome detection and the development of new therapeutic approaches for breast cancer.
Collapse
Affiliation(s)
- Carla Lucia Esposito
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Cristina Quintavalle
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Francesco Ingenito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II,” 80100 Naples, Italy
| | - Deborah Rotoli
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Giuseppina Roscigno
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II,” 80100 Naples, Italy
| | | | | | - Silvia Catuogno
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Vittorio de Franciscis
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
| | - Gerolama Condorelli
- Institute for Experimental Endocrinology and Oncology, “G.Salvatore” IEOS, Consiglio Nazionale delle Ricerche (CNR), 80131 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II,” 80100 Naples, Italy
| |
Collapse
|
17
|
Sinha D, Roy S, Saha P, Chatterjee N, Bishayee A. Trends in Research on Exosomes in Cancer Progression and Anticancer Therapy. Cancers (Basel) 2021; 13:cancers13020326. [PMID: 33477340 PMCID: PMC7829710 DOI: 10.3390/cancers13020326] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Intensive research in the field of cancer biology has discovered a unique mode of interplay between cells via extracellular bioactive vesicles called exosomes. Exosomes serve as intermediators among cells via their cargoes that, in turn, contribute in the progression of cancer. They are ubiquitously present in all body fluids as they are secreted from both normal and tumor cells. These minuscules exhibit multiple unique properties that facilitate their migration to distant locations and modulate the microenvironment for progression of cancer. This review summarizes the multifarious role of exosomes in various aspects of cancer research with its pros and cons. It discusses biogenesis of exosomes, their functional role in cancer metastasis, both protumorigenic and antitumorigenic, and also their applications in anticancer therapy. Abstract Exosomes, the endosome-derived bilayered extracellular nanovesicles with their contribution in many aspects of cancer biology, have become one of the prime foci of research. Exosomes derived from various cells carry cargoes similar to their originator cells and their mode of generation is different compared to other extracellular vesicles. This review has tried to cover all aspects of exosome biogenesis, including cargo, Rab-dependent and Rab-independent secretion of endosomes and exosomal internalization. The bioactive molecules of the tumor-derived exosomes, by virtue of their ubiquitous presence and small size, can migrate to distal parts and propagate oncogenic signaling and epigenetic regulation, modulate tumor microenvironment and facilitate immune escape, tumor progression and drug resistance responsible for cancer progression. Strategies improvised against tumor-derived exosomes include suppression of exosome uptake, modulation of exosomal cargo and removal of exosomes. Apart from the protumorigenic role, exosomal cargoes have been selectively manipulated for diagnosis, immune therapy, vaccine development, RNA therapy, stem cell therapy, drug delivery and reversal of chemoresistance against cancer. However, several challenges, including in-depth knowledge of exosome biogenesis and protein sorting, perfect and pure isolation of exosomes, large-scale production, better loading efficiency, and targeted delivery of exosomes, have to be confronted before the successful implementation of exosomes becomes possible for the diagnosis and therapy of cancer.
Collapse
Affiliation(s)
- Dona Sinha
- Department of Receptor Biology and Tumour Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India; (S.R.); (P.S.); (N.C.)
- Correspondence: or (D.S.); or (A.B.)
| | - Sraddhya Roy
- Department of Receptor Biology and Tumour Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India; (S.R.); (P.S.); (N.C.)
| | - Priyanka Saha
- Department of Receptor Biology and Tumour Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India; (S.R.); (P.S.); (N.C.)
| | - Nabanita Chatterjee
- Department of Receptor Biology and Tumour Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India; (S.R.); (P.S.); (N.C.)
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
- Correspondence: or (D.S.); or (A.B.)
| |
Collapse
|
18
|
Praharaj PP, Patra S, Panigrahi DP, Patra SK, Bhutia SK. Clusterin as modulator of carcinogenesis: A potential avenue for targeted cancer therapy. Biochim Biophys Acta Rev Cancer 2020; 1875:188500. [PMID: 33385484 DOI: 10.1016/j.bbcan.2020.188500] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 12/30/2022]
Abstract
Clusterin (CLU) is an evolutionary conserved molecular chaperone present in different human tissues and fluids and established to be a significant cancer regulator. It controls several cancer-associated cellular events, including cancer cell proliferation, stemness, survival, metastasis, epithelial-mesenchymal transition, therapy resistance, and inhibition of programmed cell death to support cancer growth and recurrence. This multifunctional role of CLU makes it an ideal target for cancer control. More importantly, genetic and antisense-mediated (OGX-011) inhibition of CLU enhances the anticancer potential of different FDA-approved chemotherapeutic drugs at the clinical level, improving patient's survival. In this review, we have discussed the detailed mechanism of CLU-mediated modulation of different cancer-associated signaling pathways. We have also provided updated information on the current preclinical and clinical findings that drive trials in various cancer types for potential targeted cancer therapy.
Collapse
Affiliation(s)
- Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Debasna Pritimanjari Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology, Rourkela, 769008, Odisha, India.
| |
Collapse
|
19
|
Ibáñez F, Ureña-Peralta JR, Costa-Alba P, Torres JL, Laso FJ, Marcos M, Guerri C, Pascual M. Circulating MicroRNAs in Extracellular Vesicles as Potential Biomarkers of Alcohol-Induced Neuroinflammation in Adolescence: Gender Differences. Int J Mol Sci 2020; 21:ijms21186730. [PMID: 32937997 PMCID: PMC7555060 DOI: 10.3390/ijms21186730] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/27/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023] Open
Abstract
Current studies evidence the role of miRNAs in extracellular vesicles (EVs) as key regulators of pathological processes, including neuroinflammation and neurodegeneration. As EVs can cross the blood–brain barrier, and EV miRNAs are very stable in peripheral circulation, we evaluated the potential gender differences in inflammatory-regulated miRNAs levels in human and murine plasma EVs derived from alcohol-intoxicated female and male adolescents, and whether these miRNAs could be used as biomarkers of neuroinflammation. We demonstrated that while alcohol intoxication lowers anti-inflammatory miRNA (mir-146a-5p, mir-21-5p, mir-182-5p) levels in plasma EVs from human and mice female adolescents, these EV miRNAs increased in males. In mice brain cortices, ethanol treatment lowers mir-146a-5p and mir-21-5p levels, while triggering a higher expression of inflammatory target genes (Traf6, Stat3, and Camk2a) in adolescent female mice. These results indicate, for the first time, that female and male adolescents differ as regards the ethanol effects associated with the inflammatory-related plasma miRNAs EVs profile, and suggest that female adolescents are more vulnerable than males to the inflammatory effects of binge alcohol drinking. These findings also support the view that circulating miRNAs in EVs could be useful biomarkers for screening ethanol-induced neuroinflammation and brain damage in adolescence.
Collapse
Affiliation(s)
- Francesc Ibáñez
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012 Valencia, Spain; (F.I.); (J.R.U.-P.); (C.G.)
| | - Juan R. Ureña-Peralta
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012 Valencia, Spain; (F.I.); (J.R.U.-P.); (C.G.)
| | - Pilar Costa-Alba
- Emergency Department, University Hospital of Salamanca-IBSAL, 37007 Salamanca, Spain;
| | - Jorge-Luis Torres
- Department of Internal Medicine, University Hospital of Salamanca, University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (J.-L.T.); (F.-J.L.); (M.M.)
| | - Francisco-Javier Laso
- Department of Internal Medicine, University Hospital of Salamanca, University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (J.-L.T.); (F.-J.L.); (M.M.)
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca, University of Salamanca, Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (J.-L.T.); (F.-J.L.); (M.M.)
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012 Valencia, Spain; (F.I.); (J.R.U.-P.); (C.G.)
| | - María Pascual
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, 46012 Valencia, Spain; (F.I.); (J.R.U.-P.); (C.G.)
- Department of Physiology, School of Medicine and Dentistry, University of Valencia, 46010 Valencia, Spain
- Correspondence: ; Tel.: +34-961-625-635; Fax: +34-963-864-642
| |
Collapse
|
20
|
Shi ZY, Yang XX, Malichewe C, Li YS, Guo XL. Exosomal microRNAs-mediated intercellular communication and exosome-based cancer treatment. Int J Biol Macromol 2020; 158:530-541. [PMID: 32360962 DOI: 10.1016/j.ijbiomac.2020.04.228] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/26/2020] [Accepted: 04/25/2020] [Indexed: 02/06/2023]
Abstract
Exosomes are extracellular vesicles with a diameter of about 30 to 100 nm, which play a crucial role in intercellular communication. Compared with normal cells, the release rate of tumor-derived exosomes (TDEs) significantly increased, and exosomal contents, especially microRNAs (miRNAs), greatly changed. TDEs contribute to the proliferation, metastasis and resistance of tumor cells, regulate immune response and tumor autophagy, and mediate tumor-stroma communication. In addition, exosomes may be involved in tumor complications. In view of the role of exosomes in intercellular communication, exosomes have been developed as tumor biomarkers, therapeutic targets, and drug delivery systems for tumor diagnosis, prognosis and treatment. Despite the many advantages of exosomes, there are many challenges in exosomal development and application, such as incomprehensive understanding of biological functions, safety and specificity for therapeutic use. This article reviews the biogenesis of TDEs and focuses on the role of exosomal miRNAs in intercellular communication and exosome-based treatment for cancer.
Collapse
Affiliation(s)
- Zhao-Yu Shi
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Xiao-Xia Yang
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - ChristinaYallen Malichewe
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Ying-Shuang Li
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Xiu-Li Guo
- Department of Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China..
| |
Collapse
|
21
|
Liu X, Wang L, Ma C, Wang G, Zhang Y, Sun S. Exosomes derived from platelet-rich plasma present a novel potential in alleviating knee osteoarthritis by promoting proliferation and inhibiting apoptosis of chondrocyte via Wnt/β-catenin signaling pathway. J Orthop Surg Res 2019; 14:470. [PMID: 31888697 PMCID: PMC6936129 DOI: 10.1186/s13018-019-1529-7] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/19/2019] [Indexed: 03/20/2023] Open
Abstract
Background Platelet-rich plasma (PRP) provides a nonsurgical approach for treating osteoarthritis (OA). Exosomes that play vital roles in intercellular communication have been studied extensively. Here, we investigated the therapeutic potential and molecular mechanism of exosomes derived from PRP (PRP-Exos) in alleviating OA. Methods Exosomes derived from PRP(PRP-Exos) were isolated and purified using the exoEasy Maxi Kit and then identified and analyzed. Primary rabbit chondrocytes were isolated and treated with interleukin 1 beta (IL-1β) to establish the OA model in vitro. Proliferation, migration, and apoptosis assays were measured and compared between PRP-Exos and activated PRP (PRP-As) to evaluate the therapeutic effects on OA. The mechanism involving the Wnt/β-catenin signaling pathway was investigated by Western blot analysis. In vivo, we established animal knee OA model by surgery to compare the therapeutic effect of PRP-Exos and PRP-As. Results We successfully isolated and purified exosomes from PRP using the exoEasy Maxi Kit. We also isolated and identified chondrocytes from the New Zealand white rabbit and established the IL-1β-induced OA model; meanwhile, PRP-Exos and PRP-As both inhibited the release of tumor necrosis factor-α(TNF-α) and there was no statistically significant difference between the two. In proliferation, migration, scratch assay, the promoting effect of PRP-Exos was significantly more better than PRP-As. Furthermore, PRP-Exos could significantly decreased apoptotic rate of OA chondrocyte compared with PRP-As. In Western blot analysis, the expression of β-catenin, and RUNX2, Wnt5a were increased in IL-1β-treated chondrocytes, but PRP-Exos and PRP-As could both reverse these changes, and the reversal effect of the former was better than the latter. In vivo, we found that both PRP-Exos and PRP-As displayed the progression of OA, and the effect of PRP-Exos was obviously better than PRP-As by chondrocyte count and Osteoarthritis Research Society International (OARSI) scoring system. Conclusion The therapeutic effects of PRP-Exos on OA were similar or better compared with those of PRP-As in vitro or in vivo. PRP-Exos acting as carriers containing growth factors derived from PRP present a novel therapy for OA by activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xuchang Liu
- Department of Emergency Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.,School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Taian, 271016, Shandong, China
| | - Lubo Wang
- Department of Trauma Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Chengshan Ma
- Department of Emergency Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Guozong Wang
- School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China
| | - Yuanji Zhang
- School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
22
|
SMR peptide antagonizes mortalin promoted release of extracellular vesicles and affects mortalin protection from complement-dependent cytotoxicity in breast cancer cells and leukemia cells. Oncotarget 2019; 10:5419-5438. [PMID: 31534628 PMCID: PMC6739210 DOI: 10.18632/oncotarget.27138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Mortalin/GRP-75/mt-hsp70 is a mitochondrial chaperone protein, found in the cytoplasm, endoplasmic reticulum and cytoplasmic vesicles. It functions in many cellular processes such as mitochondrial biogenesis, intracellular trafficking, cell proliferation, signaling, immortalization and tumorigenesis. Thus, inhibition of mortalin is a promising avenue for cancer therapy. Previous studies in our lab have suggested that mortalin contributes to breast cancer development and progression. We showed that tumor extracellular vesicle secretion was decreased by knockdown of mortalin expression using HIV-1 Nef SMR peptides. Specifically, these peptides can block extracellular vesicle secretion and mediate cell cycle arrest in MDA-MB-231 and MCF-7 breast cancer cells.
Aims: This study aims to investigate further the function and mechanism of interaction of PEG-SMR-CLU and SMR-CPP peptides with the chaperone protein mortalin and to explore the effect of SMR-derived peptides and mortalin expression on extracellular vesicle release and complement dependent cell toxicity in human breast cancer and leukemia cell lines.
Results: Our results demonstrated additional effects reversing the tumorigenicity of these cells. First, the modified SMRwt peptides reduced the expression of the mesenchymal marker vimentin (VIM). Second, exposure to the SMRwt peptide inhibited mortalin and complement C9 expression in MDA-MB-231, MCF-7 breast cancer cells and K562 leukemia cells as measured by the Western blot analysis. Third, the SMRwt peptides blocked the cancer cells’ ability to release extracellular vesicles, which we observed blocked extracellular vesicle-mediated release of complement, re-establishing complements mediated cell death in those peptide-treated cells.
Methods: We developed a series of peptides derived from the Secretion Modification Region (SMR) of HIV-1 Nef protein, modified by the addition of either a cell-penetrating peptide (CPP), a positively charged arginine-rich peptide derived from HIV-1 regulatory protein Tat, or a Clusterin-binding peptide (CLU), a molecular chaperone involved in protein secretion. Both CPP and CLU peptide sequences were added at the C-terminus of the Nef SMR peptide. The CLU-containing peptides were also modified with polyethylene glycol (PEG) to enhance solubility. After treatment of cells with the peptides, we used the MTT cell viability and complement-mediated cytotoxicity assays to confirm the inhibitory role of modified SMRwt peptides on the proliferation of MDA-MB-231 and MCF-7 breast cancer cells and K562 leukemia cells. Flow cytometry was used to determine complement mediated cell apoptosis and death. Western blot analysis was used to track SMR peptides impact on expression of mortalin, vimentin and complement C9 and to measure the expression of extracellular vesicle proteins. NanoSight analysis and acetylcholinesterase (AChE) assay were used for measuring extracellular vesicles particle size and concentration and acetylcholinesterase.
Conclusions: Mortalin promotes cell proliferation, metastasis, angiogenesis, downregulate apoptotic signaling. Thus, mortalin is a potential therapeutic target for cancer immunotherapy. The novel SMRwt peptides antagonize the functions of mortalin, blocking tumor extracellular vesicle release and extracellular vesicle-mediated release of complement. This leads to decreases in breast cancer cell metastasis and allows standard treatment of these late stage tumor cells, thus having important clinical implications for late stage breast cancer chemotherapy. These findings support further investigation into the therapeutic value of the SMR peptide in cancer metastasis.
Collapse
|
23
|
Patras L, Banciu M. Intercellular Crosstalk Via Extracellular Vesicles in Tumor Milieu as Emerging Therapies for Cancer Progression. Curr Pharm Des 2019; 25:1980-2006. [DOI: 10.2174/1381612825666190701143845] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
:Increasing evidence has suggested that extracellular vesicles (EV) mediated bidirectional transfer of functional molecules (such as proteins, different types of RNA, and lipids) between cancer cells and tumor stromal cells (immune cells, endothelial cells, fibroblasts, stem cells) and strongly contributed to the reinforcement of cancer progression. Thus, intercellular EV-mediated signaling in tumor microenvironment (TME) is essential in the modulation of all processes that support and promote tumor development like immune suppression, angiogenesis, invasion and metastasis, and resistance of tumor cells to anticancer treatments.:Besides EV potential to revolutionize our understanding of the cancer cell-stromal cells crosstalk in TME, their ability to selectively transfer different cargos to recipient cells has created excitement in the field of tumortargeted delivery of specific molecules for anticancer treatments. Therefore, in tight connection with previous findings, this review brought insight into the dual role of EV in modulation of TME. Thus, on one side EV create a favorable phenotype of tumor stromal cells for tumor progression; however, as a future new class of anticancer drug delivery systems EV could re-educate the TME to overcome main supportive processes for malignancy progression.
Collapse
Affiliation(s)
- Laura Patras
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Manuela Banciu
- Department of Molecular Biology and Biotechnology, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
24
|
Cobbs A, Chen X, Zhang Y, George J, Huang MB, Bond V, Thompson W, Zhao X. Saturated fatty acid stimulates production of extracellular vesicles by renal tubular epithelial cells. Mol Cell Biochem 2019; 458:113-124. [PMID: 30993495 PMCID: PMC7027953 DOI: 10.1007/s11010-019-03535-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/10/2019] [Indexed: 12/30/2022]
Abstract
Lipotoxicity, an accumulation of intracellular lipid metabolites, has been proposed as an important pathogenic mechanism contributing to kidney dysfunction in the context of metabolic disease. Palmitic acid, a predominant lipid derivative, can cause lipoapoptosis and the release of inflammatory extracellular vesicles (EVs) in hepatocytes, but the effect of lipids on EV production in chronic kidney disease remains vaguely explored. This study was aimed to investigate whether palmitic acid would stimulate EV release from renal proximal tubular epithelial cells. Human and rat proximal tubular epithelial cells, HK-2 and NRK-52E, were incubated with 1% bovine serum albumin (BSA), BSA-conjugated palmitic acid (PA), and BSA-conjugated oleic acid (OA) for 24-48 h. The EVs released into conditioned media were isolated by ultracentrifugation and quantified by nanoparticle-tracking analysis (NTA). According to NTA, the size distribution of EVs was 30-150 nm with similar mode sizes in all experimental groups. Moreover, BSA-induced EV release was significantly enhanced in the presence of PA, whereas EV release was not altered by the addition of OA. In NRK-52E cells, PA-enhanced EV release was associated with an induction of cell apoptosis reflected by an increase in cleaved caspase-3 protein by Western blot and Annexin V positive cells analyzed by flow cytometry. Additionally, confocal microscopy confirmed the uptake of lipid-induced EVs by recipient renal proximal tubular cells. Collectively, our results indicate that PA stimulates EV release from cultured proximal tubular epithelial cells. Thus, extended characterization of lipid-induced EVs may constitute new signaling paradigms contributing to chronic kidney disease pathology.
Collapse
Affiliation(s)
- Alyssa Cobbs
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Xiaoming Chen
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Yuanyuan Zhang
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Jasmine George
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Ming-Bo Huang
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Vincent Bond
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Winston Thompson
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA
| | - Xueying Zhao
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA, 30310, USA.
| |
Collapse
|
25
|
Khawar MB, Abbasi MH, Siddique Z, Arif A, Sheikh N. An Update on Novel Therapeutic Warfronts of Extracellular Vesicles (EVs) in Cancer Treatment: Where We Are Standing Right Now and Where to Go in the Future. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9702562. [PMID: 31428232 PMCID: PMC6683766 DOI: 10.1155/2019/9702562] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/03/2019] [Accepted: 07/04/2019] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of membrane-bounded vesicles that are believed to be produced and secreted by presumably all cell types under physiological and pathological conditions, including tumors. EVs are very important vehicles in intercellular communications for both shorter and longer distances and are able to deliver a wide range of cargos including proteins, lipids, and various species of nucleic acids effectively. EVs have been emerging as a novel biotherapeutic platform to efficiently deliver therapeutic cargos to treat a broad range of diseases including cancer. This vast potential of drug delivery lies in their abilities to carry a variety of cargos and their ease in crossing the biological membranes. Similarly, their presence in a variety of body fluids makes them a potential biomarker for early diagnosis, prognostication, and surveillance of cancer. Here, we discuss the relatively least and understudied aspects of EV biology and tried to highlight the obstacles and limitations in their clinical applications and also described most of the new warfronts to beat cancer at multiple stages. However, much more challenges still remain to evaluate EV-based therapeutics, and we are very much hopeful that the current work prompts further discovery.
Collapse
Affiliation(s)
- Muhammad Babar Khawar
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Cell & Molecular Biology Lab, Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Muddasir Hassan Abbasi
- Cell & Molecular Biology Lab, Department of Zoology, University of the Punjab, Lahore, Pakistan
- Department of Zoology, University of Okara, Okara, Pakistan
| | - Zerwa Siddique
- Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Amin Arif
- Cell & Molecular Biology Lab, Department of Zoology, University of the Punjab, Lahore, Pakistan
| | - Nadeem Sheikh
- Cell & Molecular Biology Lab, Department of Zoology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
26
|
Milman N, Ginini L, Gil Z. Exosomes and their role in tumorigenesis and anticancer drug resistance. Drug Resist Updat 2019; 45:1-12. [PMID: 31369918 DOI: 10.1016/j.drup.2019.07.003] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/19/2019] [Accepted: 07/21/2019] [Indexed: 12/25/2022]
Abstract
Exosomes are a class of extracellular vesicles ranging in size from 40 to 100 nm, which are secreted by both cancer cells and multiple stromal cells in the tumor microenvironment. Following their secretion, exosomes partake in endocrine, paracrine and autocrine signaling. Internalization of exosomes by tumor cells influences several cellular pathways which alter cancer cell physiology. Tumor-derived exosomes secreted by cancer or stromal cells can also confer anticancer drug-resistant traits upon cancer cells. These exosomes promote chemoresistance by transferring their cargo which includes nucleic acids, proteins, and metabolites to cancer cells or act as a decoy for immunotherapeutic targets. Depletion of exosomes can reverse some of the detrimental effects on tumor metabolism and restore drug sensitivity to chemotherapeutic treatment. Herein we discuss various approaches that have been developed to deplete exosomes for therapeutic purposes. The natural composition, low immunogenicity and cytotoxicity of exosomes, along with their ability to specifically target tumor cells, render them an appealing platform for drug delivery. The ability of exosomes to mediate autocrine and paracrine signaling in target cells, along with their natural structure and low immunogenicity render them an attractive vehicle for the delivery of anticancer drugs to tumors.
Collapse
Affiliation(s)
- Neta Milman
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, The Clinical Research Institute at Rambam Healthcare Campus, Technion Integrated Cancer Center, Rappaport Institute of Medicine and Research, Technion, Israel Institute of Technology, Haifa, Israel
| | - Lana Ginini
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, The Clinical Research Institute at Rambam Healthcare Campus, Technion Integrated Cancer Center, Rappaport Institute of Medicine and Research, Technion, Israel Institute of Technology, Haifa, Israel
| | - Ziv Gil
- The Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, The Clinical Research Institute at Rambam Healthcare Campus, Technion Integrated Cancer Center, Rappaport Institute of Medicine and Research, Technion, Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
27
|
Huang MB, Xia M, Gao Z, Zhou H, Liu M, Huang S, Zhen R, Wu JY, Roth WW, Bond VC, Xiao J, Leng J. Characterization of Exosomes in Plasma of Patients with Breast, Ovarian, Prostate, Hepatic, Gastric, Colon, and Pancreatic Cancers. JOURNAL OF CANCER THERAPY 2019; 10:382-399. [PMID: 33833900 PMCID: PMC8025783 DOI: 10.4236/jct.2019.105032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Detection of circulating tumor-specific DNA, RNA or proteins can be difficult due to relative scarcity. Exosomes are extracellular vesicles, 30 - 150 nm in diameter derived from fusion of multivesicular bodies with the plasma membrane. They are composed of a lipid bilayer membrane and contain proteins, mRNA and miRNA. Exosomes are secreted by multiple cell types, including cancer cells. However, there is a relative lack of information concerning the contents of exosomes secreted by various tumor cell types. To examine exosomes in cancer, we collected blood plasma samples from patients with breast, ovarian, prostate, hepatic, gastric, colon, and pancreatic cancers. Exosomes were isolated from plasma and confirmed by AchE assay, transmission electron microscopy and expression of the CD63 exosomal marker. Expression of AFP, CA724, CA153, CEA, CA125, CA199 and PSA antigens were determined using an automated electro-chemiluminescence assay. Expression of the tumor-related chaperone protein, mortalin, was determined by Western blot analysis. Levels of exosome secretion were variable among the different tumor types. Both exosome levels and mortalin expression within tumor cell exosomes were higher than in healthy donors, except in pancreatic carcinoma, where exosomes were elevated but mortalin expression was not significantly different from healthy donors. Exosomes provide unique opportunities for the enrichment of tumor-specific materials and may be useful as biomarkers and possibly as tools of cancer therapies. Mortalin, which has been linked to cell proliferation and induction of epithelial-mesenchymal transition of cancer cells, may be useful as a prognostic bio-marker and as a possible therapeutic target.
Collapse
Affiliation(s)
- Ming-Bo Huang
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Meng Xia
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zhao Gao
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Hu Zhou
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Min Liu
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Shan Huang
- Tumor hospital Affiliated to Guangxi Medical University, Nanning, Guangxi, China
| | - Rong Zhen
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jennifer Y. Wu
- Columbia College, Columbia University, New York, NY, USA
| | - William W. Roth
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Vincent C. Bond
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Jian Xiao
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jing Leng
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
28
|
Peng M, Deng J, Zhou S, Tao T, Su Q, Yang X, Yang X. The role of Clusterin in cancer metastasis. Cancer Manag Res 2019; 11:2405-2414. [PMID: 31114318 PMCID: PMC6497892 DOI: 10.2147/cmar.s196273] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
Clusterin is a conserved glycoprotein that has been characterized from almost all human tissues and fluids and plays a key role in cellular stress response and survival. Recently, research efforts have been contributed to explore the function of Clusterin in cancer metastasis, which is particularly important to design the strategies for treating metastatic patients. Evidence collected has demonstrated that Clusterin is overexpressed in tumor metastatic patients and experimental metastasis models. Specifically, Clusterin has been shown to have the role in anti-apoptotic capacities, development of therapy resistance and induction of epithelial–mesenchymal transition, all associated with cancer metastasis. Inhibition of Clusterin is known to increase the cytotoxic effects of chemotherapeutic agents and improves advanced cancer patients survival in clinical trials. Our unpublished data have demonstrated that Clusterin is overexpressed in bladder cancer and metformin, a well-known metabolism modulator specifically targets Clusterin by inhibiting migration of bladder cancer cells. In this review, we provide a general view of how Clusterin modulates cancer metastasis and update current understanding of detailed molecular mechanisms underlying of Clusterin for developing cancer management in future.
Collapse
Affiliation(s)
- Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China.,Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, People's Republic of China
| | - Jun Deng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, People's Republic of China
| | - Sichun Zhou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, People's Republic of China
| | - Ting Tao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, People's Republic of China
| | - Qiongli Su
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, People's Republic of China
| | - Xue Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, Hunan 410013, People's Republic of China
| |
Collapse
|
29
|
Jalalian SH, Ramezani M, Jalalian SA, Abnous K, Taghdisi SM. Exosomes, new biomarkers in early cancer detection. Anal Biochem 2019; 571:1-13. [PMID: 30776327 DOI: 10.1016/j.ab.2019.02.013] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 01/26/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
Abstract
Exosomes are endosomal-derived vesicles, playing a major role in cell-to-cell communication. Multiple cells secret these vesicles to induce and inhibit different cellular and molecular pathways. Cancer-derived exosomes have been shown to affect development of cancer in different stages and contribute to the recruitment and reprogramming of both proximal and distal tissues. The growing interest in defining the clinical relevance of these nano-sized particles in cancers, has led to the identification of either tissue- or disease-specific exosomal contents, such as nucleic acids, proteins and lipids as a source of new biomarkers which propose the diagnostic potentials of exosomes in early detection of cancers. In this review, we have discussed some aspects of exosomes including their contents, applications and isolation techniques in the field of early cancer detection. Although, exosomes are considered as ideal biomarkers in cancer diagnosis, due to their unique characteristics, there is still a long way in the development of exosome-based assays.
Collapse
Affiliation(s)
- Seyed Hamid Jalalian
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Students Research Committee, Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Academic Center for Education, Culture and Research (ACECR)-Mashhad Branch, Mashhad, Iran
| | - Mohammad Ramezani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Ali Jalalian
- Students Research Committee, Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Zong S, Pan F, Zhang R, Chen C, Wang Z, Cui Y. Super blinking and biocompatible nanoprobes based on dye doped BSA nanoparticles for super resolution imaging. NANOTECHNOLOGY 2019; 30:065701. [PMID: 30523996 DOI: 10.1088/1361-6528/aaf03b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
As one of the super-resolved optical imaging techniques, single molecule localization microscopy (SMLM) received considerable attention due to its impressive spatial resolution. Compared with other fluorescence imaging techniques, SMLM has one particular request for the fluorophores, that is, continuous 'on' and 'off' behaviors of their signals (referred to as 'blinking'). Hence, we present here a kind of super blinking and biocompatible nanoprobes (denoted as SBNs) for SMLM. The SBNs have two main advantages, first, they possess an outstanding fluorescence blinking. Second, they are biocompatible since they are based on bovine serum albumin (BSA). The SBNs are fabricated by doping organic dyes into BSA nanoparticles. We fabricated two kinds of SBNs, one was doped with Alexa Fluor 647 (A647) and the other was doped with Alexa Fluor 594 (A594). Especially for A594 doped SBNs, the improved blinking of A594 doped SBNs induced a better localization precision as compared with A594 alone. Moreover, SMLM imaging of breast cancer cells and exosomes using the SBNs was successfully realized with high spatial resolutions. The work demonstrated here provides a new strategy to prepare novel kinds of super blinking fluorescent agents for SMLM, which broadens the selection of suitable fluorophores for SMLM.
Collapse
Affiliation(s)
- Shenfei Zong
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, People's Republic of China
| | | | | | | | | | | |
Collapse
|
31
|
Patters BJ, Kumar S. The role of exosomal transport of viral agents in persistent HIV pathogenesis. Retrovirology 2018; 15:79. [PMID: 30577804 PMCID: PMC6303896 DOI: 10.1186/s12977-018-0462-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection, despite great advances in antiretroviral therapy, remains a lifelong affliction. Though current treatment regimens can effectively suppress viral load to undetectable levels and preserve healthy immune function, they cannot fully alleviate all symptoms caused by the presence of the virus, such as HIV-associated neurocognitive disorders. Exosomes are small vesicles that transport cellular proteins, RNA, and small molecules between cells as a mechanism of intercellular communication. Recent research has shown that HIV proteins and RNA can be packaged into exosomes and transported between cells, to pathogenic effect. This review summarizes the current knowledge on the diverse mechanisms involved in the sorting of viral elements into exosomes and the damage those exosomal agents can inflict. In addition, potential therapeutic options to counteract exosome-mediated HIV pathogenesis are reviewed and considered.
Collapse
Affiliation(s)
- Benjamin J Patters
- Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Santosh Kumar
- Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
32
|
Navarro-Tableros V, Gomez Y, Camussi G, Brizzi MF. Extracellular Vesicles: New Players in Lymphomas. Int J Mol Sci 2018; 20:E41. [PMID: 30583481 PMCID: PMC6337615 DOI: 10.3390/ijms20010041] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022] Open
Abstract
Lymphomas are heterogeneous diseases, and the term includes a number of histological subtypes that are characterized by different clinical behavior and molecular phenotypes. Valuable information on the presence of lymphoma cell-derived extracellular vesicles (LCEVs) in the bloodstream of patients suffering from this hematological cancer has recently been provided. In particular, it has been reported that the number and phenotype of LCEVs can both change as the disease progresses, as well as after treatment. Moreover, the role that LCEVs play in driving tumor immune escape has been reported. This makes LCEVs potential novel clinical tools for diagnosis, disease progression, and chemoresistance. LCEVs express surface markers and convey specific molecules in accordance with their cell of origin, which can be used as targets and thus lead to the development of specific therapeutics. This may be particularly relevant since circulating LCEVs are known to save lymphoma cells from anti-cluster of differentiation (CD)20-induced complement-dependent cytotoxicity. Therefore, effort should be directed toward investigating the feasibility of using LCEVs as predictive biomarkers of disease progression and/or response to treatment that can be translated to clinical use. The use of liquid biopsies in combination with serum EV quantification and cargo analysis have been also considered as potential approaches that can be pursued in the future. Upcoming research will also focus on the identification of specific molecular targets in order to generate vaccines and/or antibodies against LCEVs. Finally, the removal of circulating LCEVs has been proposed as a simple and non-invasive treatment approach. We herein provide an overview of the role of LCEVs in lymphoma diagnosis, immune tolerance, and drug resistance. In addition, alternative protocols that utilize LCEVs as therapeutic targets are discussed.
Collapse
Affiliation(s)
- Victor Navarro-Tableros
- 2i3T Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico Scarl, University of Turin, Turin 10126, Italy.
| | - Yonathan Gomez
- Department of Medical Sciences, University of Turin, Turin 10126, Italy.
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin 10126, Italy.
| | | |
Collapse
|
33
|
Valcz G, Buzás EI, Szállási Z, Kalmár A, Krenács T, Tulassay Z, Igaz P, Molnár B. Perspective: bidirectional exosomal transport between cancer stem cells and their fibroblast-rich microenvironment during metastasis formation. NPJ Breast Cancer 2018; 4:18. [PMID: 30038960 PMCID: PMC6048124 DOI: 10.1038/s41523-018-0071-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/18/2022] Open
Abstract
Carcinomas are complex structures composed of hierarchically organized distinct cell populations such as cancer stem cells and non-stem (bulk) cancer cells. Their genetic/epigenetic makeup and the dynamic interplay between the malignant cell populations and their stromal fibroblasts are important determinants of metastatic tumor invasion. Important mediators of these interactions are the small, membrane-enclosed extracellular vesicles, in particular exosomes. Both cancer cell and fibroblast-derived exosomes carry a set of regulatory molecules, including proteins and different species of RNA, which cooperatively support metastatic tumor spread. Here, we briefly overview potential links between cancer stem cells and the exosome-mediated fibroblast-enriched metastatic niche formation to discuss their role in the promotion of tumor growth and metastatic expansion in breast carcinoma models.
Collapse
Affiliation(s)
- Gábor Valcz
- 1Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary.,22nd Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Edit Irén Buzás
- 3MTA-SE Immuno-Proteogenomics Extracellular Vesicle Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,4Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Zoltán Szállási
- 5Computational Health Informatics Program (CHIP), Boston Children's Hospital, Harvard Medical School, Boston, USA
| | - Alexandra Kalmár
- 1Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary.,22nd Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Tibor Krenács
- 61st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Zsolt Tulassay
- 1Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary.,22nd Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Igaz
- 1Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary.,22nd Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Béla Molnár
- 1Molecular Medicine Research Unit, Hungarian Academy of Sciences, Budapest, Hungary.,22nd Department of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
34
|
Li B, Lyu P, Xi X, Ge L, Mahadevappa R, Shaw C, Kwok HF. Triggering of cancer cell cycle arrest by a novel scorpion venom-derived peptide-Gonearrestide. J Cell Mol Med 2018; 22:4460-4473. [PMID: 29993185 PMCID: PMC6111814 DOI: 10.1111/jcmm.13745] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
In this study, a novel scorpion venom-derived peptide named Gonearrestide was identified in an in-house constructed scorpion venom library through a combination of high-throughput NGS transcriptome and MS/MS proteome platform. In total, 238 novel peptides were discovered from two scorpion species; and 22 peptides were selected for further study after a battery of functional prediction analysis. Following a series of bioinformatics analysis alongside with in vitro biological functional screenings, Gonearrestide was found to be a highly potent anticancer peptide which acts on a broad spectrum of human cancer cells while causing few if any observed cytotoxic effects on epithelial cells and erythrocytes. We further investigated the precise anticancer mechanism of Gonearrestide by focusing on its effects on the colorectal cancer cell line, HCT116. NGS RNA sequencing was employed to obtain full gene expression profiles in HCT116 cells, cultured in the presence and absence of Gonearrestide, to dissect signalling pathway differences. Taken together the in vitro, in vivo and ex vivo validation studies, it was proven that Gonearrestide could inhibit the growth of primary colon cancer cells and solid tumours by triggering cell cycle arrest in G1 phase through inhibition of cyclin-dependent kinases 4 (CDK4) and up-regulate the expression of cell cycle regulators/inhibitors-cyclin D3, p27, and p21. Furthermore, prediction of signalling pathways and potential binding sites used by Gonearrestide are also presented in this study.
Collapse
Affiliation(s)
- Bin Li
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao
| | - Peng Lyu
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao
| | - Xinping Xi
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao.,School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Lilin Ge
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao.,School of Pharmacy, Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Qixia District, Nanjing, China
| | | | - Chris Shaw
- School of Pharmacy, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Hang Fai Kwok
- Faculty of Health Sciences, University of Macau, Taipa, Macau, Macao
| |
Collapse
|
35
|
Moloudizargari M, Asghari MH, Abdollahi M. Modifying exosome release in cancer therapy: How can it help? Pharmacol Res 2018; 134:246-256. [PMID: 29990623 DOI: 10.1016/j.phrs.2018.07.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/08/2018] [Accepted: 07/02/2018] [Indexed: 12/19/2022]
Abstract
The reciprocal interactions of cancer cells with their microenvironment constitute an inevitable aspect of tumor development, progression and response to treatment in all cancers. Such bilateral transactions also serve as the key scenario underlying the development of drug resistance in many cases finally determining the fate of the disease and survival. In this view, a class of extracellular vesicles (EV) known as exosomes (EX) have been shown in the past few years to be important mediators of local and remote cell-to-cell contact changing the activity of their target cells by introducing their content of proteins, non-coding RNAs, and membrane-associated small molecules. In addition to the direct targeting of cancer cells, which has been routinely undertaken by different means to date, parallel attempts to change the signaling network governed by tumor-derived exosomes (TDE) may offer a promising potential to be utilized in cancer therapy. TDE drive diverse functions in the body, most of which have been shown to act to the advantage of tumor progression; however, there are also several studies that report the good aspects of TDE the interruption of which may result in undesirable outcomes. In the present paper, we made an effort to address this important issue by reviewing the very recent literature on different aspects of EX biogenesis and regulation and the various bodily effects of TDE which have been uncovered to date. Moreover, we have reviewed the possible interventions that can be made in TDE release as an important stage of EX biogenesis. Finally, keeping a criticizing view, the advantages and disadvantages of such interventions have been discussed and the future prospect in the field has been outlined.
Collapse
Affiliation(s)
- Milad Moloudizargari
- Department of Immunology, School of Medicine, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Asghari
- Department of Pharmacology, School of Medicine, Babol University of Medical Sciences, Babol, Iran; Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran; Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Abak A, Abhari A, Rahimzadeh S. Exosomes in cancer: small vesicular transporters for cancer progression and metastasis, biomarkers in cancer therapeutics. PeerJ 2018; 6:e4763. [PMID: 29868251 PMCID: PMC5983002 DOI: 10.7717/peerj.4763] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/23/2018] [Indexed: 12/19/2022] Open
Abstract
Cancer progression is a polygenic procedure in which the exosomes can function as substantial roles. Exosomes are tiny, phospholipid bilayer membrane nanovesicles of endocytic derivation with a diameter of 40-100 nm. These nanovesicles can transport bioactive molecules containing mRNAs, proteins, DNA fragments, and non-coding RNAs from a donor cell to recipient cells, and cause the alteration in genetic and epigenetic factors and reprogramming of the target cells. Many diverse cell types such as mesenchymal cells, immune cells, and cancer cells can induce the release of exosomes. Increasing evidence illustrated that the exosomes derived from tumor cells might trigger the tumor initiation, tumor cell growth and progression, metastasis, and drug resistance. The secreted nanovesicles of exosomes can play significant roles in cells communicate via shuttling the nucleic acid molecules and proteins to target cells and tissues. In this review, we discussed multiple mechanisms related to biogenesis, load, and shuttle of the exosomes. Also, we illustrated the diverse roles of exosomes in several types of human cancer development, tumor immunology, angiogenesis, and metastasis. The exosomes may act as the promising biomarkers for the prognosis of various types of cancers which suggested a new pathway for anti-tumor therapeutic of these nanovesicles and promoted exosome-based cancer for clinical diagnostic and remedial procedures.
Collapse
Affiliation(s)
- Atefe Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Abhari
- Department of Biochemistry and Clinical Laboratory, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Rahimzadeh
- Department of Biochemistry and Clinical Laboratory, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
37
|
Effect of exosome biomarkers for diagnosis and prognosis of breast cancer patients. Clin Transl Oncol 2017; 20:906-911. [PMID: 29143228 DOI: 10.1007/s12094-017-1805-0] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 11/09/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE Exosomes are gradually detected as an indicator for diagnosis and prognosis of breast cancer in clinic and a systematic review was conducted. METHODS A search for clinical studies published before July 1, 2017 was performed. Methods of exosome purification and identification from all studies were extracted. For diagnosis evaluation, the comparison of exosome biomarkers expression between breast cancer patients and healthy women was obtained; for prognosis prediction, the correlation between exosome biomarkers expression and chemotherapy resistance, overall survival (OS), disease-free survival (DFS), recurrence and metastasis of breast cancer was also extracted. RESULTS A total of 11 studies with 921 breast cancer patients were included. Ultracentrifugation is the most frequent method to purify exosomes and transmission electron microscopy is commonly used to identify exosomes. Exosome biomarkers (such as HER2, CD47, Del-1, miR-1246 and miR-21) in breast cancer patients are significantly higher than those in healthy controls, exosomal GSTP1 and TRPC5 are related to chemotherapy resistance, exosome-carrying TRPC5, NANOG, NEUROD1, HTR7, KISS1R and HOXC are correlated to PFS, DFS or OS, and some exosomal proteins (HER2, KDR, CD49d, CXCR4 and CD44) as well as miRNAs (miR-340-5p, miR-17-5p, miR-130a-3p, miR-93-5p) are associated with tumor recurrence or distant organ metastasis. CONCLUSIONS Exosome biomarkers can be used for early diagnosis and prognosis of breast cancer patients in clinic.
Collapse
|
38
|
Exosomes in cancer: Use them or target them? Semin Cell Dev Biol 2017; 78:13-21. [PMID: 28803894 DOI: 10.1016/j.semcdb.2017.08.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 12/21/2022]
Abstract
Exosomes are small extracellular vesicles with a significant role in most processes associated with cancer. On one hand, exosomes role in the different hallmarks of cancer has been widely described, highlighting the urge to understand the potential to target communication mediated by exosomes as a novel therapeutic approach in cancer. On the other hand, exosomes stability in circulation and tumor-targeting capacity shows their applicability in the delivery of anti-cancer molecules. This review will discuss the dual applicability of exosomes in cancer focusing on their usage for therapy improvement, or their targeting to block their supportive role in tumor progression and response to therapy. We highlight the current developments and the strategies used to enhance the potential of exosomes to become clinical partners in the treatment of cancer.
Collapse
|