1
|
Li S, Cai X, Guo J, Li X, Li W, Liu Y, Qi M. Cell communication and relevant signaling pathways in osteogenesis-angiogenesis coupling. Bone Res 2025; 13:45. [PMID: 40195313 PMCID: PMC11977258 DOI: 10.1038/s41413-025-00417-0] [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: 07/07/2024] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Osteogenesis is the process of bone formation mediated by the osteoblasts, participating in various bone-related physiological processes including bone development, bone homeostasis and fracture healing. It exhibits temporal and spatial interconnectivity with angiogenesis, constructed by multiple forms of cell communication occurring between bone and vascular endothelial cells. Molecular regulation among different cell types is crucial for coordinating osteogenesis and angiogenesis to facilitate bone remodeling, fracture healing, and other bone-related processes. The transmission of signaling molecules and the activation of their corresponding signal pathways are indispensable for various forms of cell communication. This communication acts as a "bridge" in coupling osteogenesis to angiogenesis. This article reviews the modes and processes of cell communication in osteogenesis-angiogenesis coupling over the past decade, mainly focusing on interactions among bone-related cells and vascular endothelial cells to provide insights into the mechanism of cell communication of osteogenesis-angiogenesis coupling in different bone-related contexts. Moreover, clinical relevance and applications are also introduced in this review.
Collapse
Affiliation(s)
- Shuqing Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xinjia Cai
- Central Laboratory, Peking University School and Hospital for Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Jiahe Guo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xiaolu Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Wen Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital for Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Mengchun Qi
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China.
| |
Collapse
|
2
|
Shen YQ, Sun L, Wang SM, Zheng XY, Xu R. Exosomal integrins in tumor progression, treatment and clinical prediction (Review). Int J Oncol 2024; 65:118. [PMID: 39540373 PMCID: PMC11575930 DOI: 10.3892/ijo.2024.5706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Integrins are a large family of cell adhesion molecules involved in tumor cell differentiation, migration, proliferation and neovascularization. Tumor cell‑derived exosomes carry a large number of integrins, which are closely associated with tumor progression. As crucial mediators of intercellular communication, exosomal integrins have gained attention in the field of cancer biology. The present review examined the regulatory mechanisms of exosomal integrins in tumor cell proliferation, migration and invasion, and emphasized their notable roles in tumor initiation and progression. The potential of exosomal integrins as drug delivery systems in cancer treatment was explored. Additionally, the potential of exosomal integrins in clinical tumor prediction was considered, while summarizing their applications in diagnosis, prognosis assessment and treatment response prediction. Thus, the present review aimed to provide guidance and insights for future basic research and the clinical translation of exosomal integrins. The study of exosomal integrins is poised to offer new perspectives and methods for precise cancer treatment and clinical prediction.
Collapse
Affiliation(s)
- Yu-Qing Shen
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Lei Sun
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Shi-Ming Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Xian-Yu Zheng
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, Anhui 230032, P.R. China
| | - Rui Xu
- College & Hospital of Stomatology, Anhui Medical University, Key Laboratory of Oral Diseases Research of Anhui Province, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
3
|
Cerrotti G, Buratta S, Latella R, Calzoni E, Cusumano G, Bertoldi A, Porcellati S, Emiliani C, Urbanelli L. Hitting the target: cell signaling pathways modulation by extracellular vesicles. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:527-552. [PMID: 39697631 PMCID: PMC11648414 DOI: 10.20517/evcna.2024.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 07/10/2024] [Accepted: 09/18/2024] [Indexed: 12/20/2024]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed nanoparticles released outside the cell. EVs have drawn attention not only for their role in cell waste disposal, but also as additional tools for cell-to-cell communication. Their complex contents include not only lipids, but also proteins, nucleic acids (RNA, DNA), and metabolites. A large part of these molecules are involved in mediating or influencing signal transduction in target cells. In multicellular organisms, EVs have been suggested to modulate signals in cells localized either in the neighboring tissue or in distant regions of the body by interacting with the cell surface or by entering the cells via endocytosis or membrane fusion. Most of the EV-modulated cell signaling pathways have drawn considerable attention because they affect morphogenetic signaling pathways, as well as pathways activated by cytokines and growth factors. Therefore, they are implicated in relevant biological processes, such as embryonic development, cancer initiation and spreading, tissue differentiation and repair, and immune response. Furthermore, it has recently emerged that multicellular organisms interact with and receive signals through EVs released by their microbiota as well as by edible plants. This review reports studies investigating EV-mediated signaling in target mammalian cells, with a focus on key pathways for organism development, organ homeostasis, cell differentiation and immune response.
Collapse
Affiliation(s)
- Giada Cerrotti
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Raffaella Latella
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Eleonora Calzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Gaia Cusumano
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Agnese Bertoldi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Serena Porcellati
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Perugia 06123, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia 06123, Italy
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Perugia 06123, Italy
| |
Collapse
|
4
|
Kim J, Ro J, Cho YK. Vascularized platforms for investigating cell communication via extracellular vesicles. BIOMICROFLUIDICS 2024; 18:051504. [PMID: 39323481 PMCID: PMC11421861 DOI: 10.1063/5.0220840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
The vascular network plays an essential role in the maintenance of all organs in the body via the regulated delivery of oxygen and nutrients, as well as tissue communication via the transfer of various biological signaling molecules. It also serves as a route for drug administration and affects pharmacokinetics. Due to this importance, engineers have sought to create physiologically relevant and reproducible vascular systems in tissue, considering cell-cell and extracellular matrix interaction with structural and physical conditions in the microenvironment. Extracellular vesicles (EVs) have recently emerged as important carriers for transferring proteins and genetic material between cells and organs, as well as for drug delivery. Vascularized platforms can be an ideal system for studying interactions between blood vessels and EVs, which are crucial for understanding EV-mediated substance transfer in various biological situations. This review summarizes recent advances in vascularized platforms, standard and microfluidic-based techniques for EV isolation and characterization, and studies of EVs in vascularized platforms. It provides insights into EV-related (patho)physiological regulations and facilitates the development of EV-based therapeutics.
Collapse
|
5
|
Aghakhani A, Pezeshki PS, Rezaei N. The role of extracellular vesicles in immune cell exhaustion and resistance to immunotherapy. Expert Opin Investig Drugs 2024; 33:721-740. [PMID: 38795060 DOI: 10.1080/13543784.2024.2360209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/22/2024] [Indexed: 05/27/2024]
Abstract
INTRODUCTION Extracellular vesicles (EVs) are membrane-bound nanoparticles for intercellular communication. Subtypes of EVs, namely exosomes and microvesicles transfer diverse, bioactive cargo to their target cells and eventually interfere with immune responses. Despite being a promising approach, cancer immunotherapy currently faces several challenges including immune resistance. EVs secreted from various sources in the tumor microenvironment provoke immune cell exhaustion and lower the efficacy of immunological treatments, such as CAR T cells and immune checkpoint inhibitors. AREAS COVERED This article goes through the mechanisms of action of various types of EVs in inhibiting immune response and immunotherapies, and provides a comprehensive review of EV-based treatments. EXPERT OPINION By making use of the distinctive features of EVs, natural or modified EVs are innovatively utilized as novel cancer therapeutics. They are occasionally coupled with currently established treatments to overcome their inadequacies. Investigating the properties and interactions of EVs and EV-based treatments is crucial for determining future steps in cancer therapeutics.
Collapse
Affiliation(s)
- Ava Aghakhani
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Parmida Sadat Pezeshki
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Wen J. Single-cell transcriptomics reveals antigen-presenting capacity and therapeutic resistance potential of immunomodulatory endothelial cells in colorectal cancer. Immun Inflamm Dis 2024; 12:e1311. [PMID: 38874280 PMCID: PMC11177288 DOI: 10.1002/iid3.1311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/10/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND The heterogeneity of tumor endothelial cells (TECs) hinders the efficacy of antiangiogenic therapies (AATs). Only a small percentage of angiogenic TECs are considered effective targets for AATs. Immunomodulatory ECs (IMECs), as a newly focused functional subgroup of endothelial cells (ECs), are being evaluated for their ability to regulate tumor immune balance and influence existing AATs. METHODS Based on single-cell transcriptome data from colorectal cancer in a publicly available database, we conducted a wide array of bioinformatic approaches to study EC subsets that meet the IMECs definition. Our investigation encompassed the gene expression signatures of these subsets, cellular composition differences, cell-cell interactions. RESULTS Two subsets that meet the IMECs definition were found in tumors and para-cancerous tissues. Combined with the results of gene ontological analysis and interaction with CD4+ T cells, we found that IMECs can present MHC-II antigens to mature CD4+ T cells. There were differences in the level of interaction between IMECs and different types of mature CD4+ T cell subsets. In addition, IMEC subsets had different expression levels of angiogenesis related genes. The angiogenesis score of IMECs decreased after patients received immunotherapy. IMEC subsets do not depend on a single proangiogenic receptor and are involved in regulating angiogenesis, which may reduce the efficacy of AATs. The adverse effects of specific IMEC subsets on AATs were validated in the RNA-seq dataset of the bevacizumab treatment group. CONCLUSION Our study suggests the potential MHC-II antigen presentation capacity of IMECs and the enhanced angiogenesis characteristics within tumors. The function of IMECs in the vascular network may have a potentially adverse effect on AATs. Controlling the functional properties of IMECs may be a new angle for tumor therapy.
Collapse
Affiliation(s)
- Jingyi Wen
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouGuangdongChina
| |
Collapse
|
7
|
Kalele K, Nyahatkar S, Mirgh D, Muthuswamy R, Adhikari MD, Anand K. Exosomes: A Cutting-Edge Theranostics Tool for Oral Cancer. ACS APPLIED BIO MATERIALS 2024; 7:1400-1415. [PMID: 38394624 DOI: 10.1021/acsabm.3c01243] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Exosomes are a subpopulation of extracellular vesicles (EVs) secreted by cells. In cancer, they are key cellular messengers during cancer development and progression. Tumor-derived exosomes (TEXs) promote cancer progression. In oral cancer, the major complication is oral squamous cell carcinoma (OSCC). Exosomes show strong participation in several OSCC-related activities such as uncontrolled cell growth, immune suppression, angiogenesis, metastasis, and drug and therapeutic resistance. It is also a potential biomarker source for oral cancer. Some therapeutic exosome sources such as stem cells, plants (it is more effective compared to others), and engineered exosomes reduce oral cancer development. This therapeutic approach is effective because of its specificity, biocompatibility, and cell-free therapy (it reduced side effects in cancer treatment). This article highlights exosome-based theranostics signatures in oral cancer, clinical trials, challenges of exosome-based oral cancer research, and future improvements. In the future, exosomes may become an effective and affordable solution for oral cancer.
Collapse
Affiliation(s)
- Ketki Kalele
- Neuron Institute of Applied Research, Rajapeth-Irwin Square Flyover, Amravati, Maharashtra 444601, India
| | - Sidhanti Nyahatkar
- VYWS Dental College & Hospital, WQMV+7X6, Tapovan-Wadali Road, Camp Rd, SRPF Colony, Amravati, Maharashtra 444602, India
| | - Divya Mirgh
- Department of Infectious Diseases, Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Raman Muthuswamy
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Manab Deb Adhikari
- Department of Biotechnology, University of North Bengal, Darjeeling, West Bengal 734013, India
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
8
|
Miyamura Y, Kamei S, Matsuo M, Yamazaki M, Usuki S, Yasunaga K, Uemura A, Satou Y, Ohguchi H, Minami T. FOXO1 stimulates tip cell-enriched gene expression in endothelial cells. iScience 2024; 27:109161. [PMID: 38444610 PMCID: PMC10914484 DOI: 10.1016/j.isci.2024.109161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 11/29/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
Forkhead box O (FOXO) family proteins are expressed in various cells, and play crucial roles in cellular metabolism, apoptosis, and aging. FOXO1-null mice exhibit embryonic lethality due to impaired endothelial cell (EC) maturation and vascular remodeling. However, FOXO1-mediated genome-wide regulation in ECs remains unclear. Here, we demonstrate that VEGF dynamically regulates FOXO1 cytosol-nucleus translocation. FOXO1 re-localizes to the nucleus via PP2A phosphatase. RNA-seq combined with FOXO1 overexpression/knockdown in ECs demonstrated that FOXO1 governs the VEGF-responsive tip cell-enriched genes, and further inhibits DLL4-NOTCH signaling. Endogenous FOXO1 ChIP-seq revealed that FOXO1 binds to the EC-unique tip-enriched genes with co-enrichment of EC master regulators, and the condensed chromatin region as a pioneer factor. We identified new promoter/enhancer regions of the VEGF-responsive tip cell genes regulated by FOXO1: ESM1 and ANGPT2. This is the first study to identify cell type-specific FOXO1 functions, including VEGF-mediated tip cell definition in primary cultured ECs.
Collapse
Affiliation(s)
- Yuri Miyamura
- Divison of Molecular and Vascular Biology, IRDA, Kumamoto University, Kumamoto 860-0811, Japan
| | - Shunsuke Kamei
- Divison of Molecular and Vascular Biology, IRDA, Kumamoto University, Kumamoto 860-0811, Japan
| | - Misaki Matsuo
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-8556, Japan
| | - Masaya Yamazaki
- Division of Medical Biochemistry, Graduate School of Medical Science, Kumamoto University, Kumamoto 860-8556, Japan
| | - Shingo Usuki
- Liaison Laboratory Research Promotion Center, IMEG, Kumamoto University, Kumamoto 860-8556, Japan
| | - Keiichiro Yasunaga
- Liaison Laboratory Research Promotion Center, IMEG, Kumamoto University, Kumamoto 860-8556, Japan
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Yorifumi Satou
- Division of Genomics and Transcriptomics, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hiroto Ohguchi
- Division of Disease Epigenetics, IRDA, Kumamoto University, Kumamoto 860-0811, Japan
| | - Takashi Minami
- Divison of Molecular and Vascular Biology, IRDA, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
9
|
Zhang C, Qin C, Dewanjee S, Bhattacharya H, Chakraborty P, Jha NK, Gangopadhyay M, Jha SK, Liu Q. Tumor-derived small extracellular vesicles in cancer invasion and metastasis: molecular mechanisms, and clinical significance. Mol Cancer 2024; 23:18. [PMID: 38243280 PMCID: PMC10797874 DOI: 10.1186/s12943-024-01932-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 01/02/2024] [Indexed: 01/21/2024] Open
Abstract
The production and release of tumor-derived small extracellular vesicles (TDSEVs) from cancerous cells play a pivotal role in the propagation of cancer, through genetic and biological communication with healthy cells. TDSEVs are known to orchestrate the invasion-metastasis cascade via diverse pathways. Regulation of early metastasis processes, pre-metastatic niche formation, immune system regulation, angiogenesis initiation, extracellular matrix (ECM) remodeling, immune modulation, and epithelial-mesenchymal transition (EMT) are among the pathways regulated by TDSEVs. MicroRNAs (miRs) carried within TDSEVs play a pivotal role as a double-edged sword and can either promote metastasis or inhibit cancer progression. TDSEVs can serve as excellent markers for early detection of tumors, and tumor metastases. From a therapeutic point of view, the risk of cancer metastasis may be reduced by limiting the production of TDSEVs from tumor cells. On the other hand, TDSEVs represent a promising approach for in vivo delivery of therapeutic cargo to tumor cells. The present review article discusses the recent developments and the current views of TDSEVs in the field of cancer research and clinical applications.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neuro-Oncology at Hunan Province, Changsha, 410008, China
| | - Chaoying Qin
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neuro-Oncology at Hunan Province, Changsha, 410008, China
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India.
| | - Hiranmoy Bhattacharya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Niraj Kumar Jha
- Centre of Research Impact and Outreach, Chitkara University Institute of Engineering and Technology, Chitkara University, Punjab, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India
| | - Moumita Gangopadhyay
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat, Kolkata, 700126, West Bengal, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, New Delhi, Delhi, 110008, India.
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- The Institute of Skull Base Surgery and Neuro-Oncology at Hunan Province, Changsha, 410008, China.
| |
Collapse
|
10
|
Wei YN, Yan CY, Zhao ML, Zhao XH. The role and application of vesicles in triple-negative breast cancer: Opportunities and challenges. Mol Ther Oncolytics 2023; 31:100752. [PMID: 38130701 PMCID: PMC10733704 DOI: 10.1016/j.omto.2023.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Extracellular vesicles (EVs) carry DNA, RNA, protein, and other substances involved in intercellular crosstalk and can be used for the targeted delivery of drugs. Triple-negative breast cancer (TNBC) is rich in recurrent and metastatic disease and lacks therapeutic targets. Studies have proved the role of EVs in the different stages of the genesis and development of TNBC. Cancer cells actively secrete various biomolecules, which play a significant part establishing the tumor microenvironment via EVs. In this article, we describe the roles of EVs in the tumor immune microenvironment, metabolic microenvironment, and vascular remodeling, and summarize the application of EVs for objective delivery of chemotherapeutic drugs, immune antigens, and cancer vaccine adjuvants. EVs-based therapy may represent the next-generation tool for targeted drug delivery for the cure of a variety of diseases lacking effective drug treatment.
Collapse
Affiliation(s)
- Ya-Nan Wei
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Chun-Yan Yan
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Meng-Lu Zhao
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| | - Xi-He Zhao
- Department of Clinical Oncology, Sheng jing Hospital of China Medical University, Shenyang 110022, People’s Republic of China
| |
Collapse
|
11
|
Cai F, Chen W, Zhao R, Liu Y. The capacity of exosomes derived from adipose-derived stem cells to enhance wound healing in diabetes. Front Pharmacol 2023; 14:1063458. [PMID: 37808198 PMCID: PMC10551633 DOI: 10.3389/fphar.2023.1063458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
The slow healing and nonhealing of diabetic wounds have long posed challenges for clinical practitioners. In the presence of elevated glucose levels, the body's regulatory mechanisms undergo alterations that impede normal wound healing processes, including cell proliferation, cytokine release, and growth factor activity. Consequently, the advancement of stem cell technology has sparked growing interest in utilizing stem cells and their derivatives as potential therapeutic agents to enhance diabetic wound healing. This paper aims to provide an academic review of the therapeutic effects of adipose-derived stem cell-EXOs (ADSC-EXOs) in diabetic wound healing. As a cell-free therapy, exosomes (EXOs) possess a multitude of proteins and growth factors that have been shown to be advantageous in promoting wound healing and mitigating the potential risks associated with stem cell therapy. By examining the current knowledge on ADSC-EXOs, this review seeks to offer insights and guidance for the potential application of EXOs in the treatment of diabetic wounds.
Collapse
Affiliation(s)
| | | | | | - Yi Liu
- Department of Burns and Plastic Surgery, and Wound Repair Surgery, The Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
12
|
Liu JL, Zhang L, Huang Y, Li XH, Liu YF, Zhang SM, Zhao YE, Chen XJ, Liu Y, He LY, Dong Z, Liu FY, Sun L, Xiao L. Epsin1-mediated exosomal sorting of Dll4 modulates the tubular-macrophage crosstalk in diabetic nephropathy. Mol Ther 2023; 31:1451-1467. [PMID: 37016580 PMCID: PMC10188907 DOI: 10.1016/j.ymthe.2023.03.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/18/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
Tubular epithelial cells (TECs) play critical roles in the development of diabetic nephropathy (DN), and can activate macrophages through the secretion of exosomes. However, the mechanism(s) of TEC-exosomes in macrophage activation under DN remains unknown. By mass spectrometry, 1,644 differentially expressed proteins, especially Dll4, were detected in the urine exosomes of DN patients compared with controls, which was confirmed by western blot assay. Elevated Epsin1 and Dll4/N1ICD expression was observed in kidney tissues in both DN patients and db/db mice and was positively associated with tubulointerstitial damage. Exosomes from high glucose (HG)-treated tubular cells (HK-2) with Epsin1 knockdown (KD) ameliorated macrophage activation, TNF-α, and IL-6 expression, and tubulointerstitial damage in C57BL/6 mice in vivo. In an in vitro study, enriched Dll4 was confirmed in HK-2 cells stimulated with HG, which was captured by THP-1 cells and promoted M1 macrophage activation. In addition, Epsin1 modulated the content of Dll4 in TEC-exosomes stimulated with HG. TEC-exosomes with Epsin1-KD significantly inhibited N1ICD activation and iNOS expression in THP-1 cells compared with incubation with HG alone. These findings suggested that Epsin1 could modulate tubular-macrophage crosstalk in DN by mediating exosomal sorting of Dll4 and Notch1 activation.
Collapse
Affiliation(s)
- Jia-Lu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Zhang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Huang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao-Hui Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi-Fei Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shu-Min Zhang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue-E Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao-Jun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li-Yu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Fu-You Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
13
|
Extracellular Vesicles: New Classification and Tumor Immunosuppression. BIOLOGY 2023; 12:biology12010110. [PMID: 36671802 PMCID: PMC9856004 DOI: 10.3390/biology12010110] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
Extracellular vesicles (EVs) are cell-derived membrane-surrounded vesicles carrying various types of molecules. These EV cargoes are often used as pathophysiological biomarkers and delivered to recipient cells whose fates are often altered in local and distant tissues. Classical EVs are exosomes, microvesicles, and apoptotic bodies, while recent studies discovered autophagic EVs, stressed EVs, and matrix vesicles. Here, we classify classical and new EVs and non-EV nanoparticles. We also review EVs-mediated intercellular communication between cancer cells and various types of tumor-associated cells, such as cancer-associated fibroblasts, adipocytes, blood vessels, lymphatic vessels, and immune cells. Of note, cancer EVs play crucial roles in immunosuppression, immune evasion, and immunotherapy resistance. Thus, cancer EVs change hot tumors into cold ones. Moreover, cancer EVs affect nonimmune cells to promote cellular transformation, including epithelial-to-mesenchymal transition (EMT), chemoresistance, tumor matrix production, destruction of biological barriers, angiogenesis, lymphangiogenesis, and metastatic niche formation.
Collapse
|
14
|
Brain Microvascular Endothelial Cell-Derived Exosomes Protect Neurons from Ischemia–Reperfusion Injury in Mice. Pharmaceuticals (Basel) 2022; 15:ph15101287. [PMID: 36297399 PMCID: PMC9608440 DOI: 10.3390/ph15101287] [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: 09/26/2022] [Revised: 10/14/2022] [Accepted: 10/16/2022] [Indexed: 11/17/2022] Open
Abstract
Stroke often results in neurological and neuropsychiatric sequela. Exosomes derived from brain endothelial cells (EC-Exo) protect neurons from hypoxic injury. However, the biological role of exosomes in apoptosis and synaptic plasticity remains unclear. This research aimed to assess whether cerebral microvascular endothelial cells inhibit apoptosis and promote synaptic remodeling through exosome-mediated cell–cell interaction after the ischemic attack. The effects of EC-Exo on primary neuronal apoptosis and synapses in oxyglucose deprivation reoxygenation (OGD/R) injury were first assessed in vitro. Animal experiments were performed using C57BL/6J mice, divided into three groups: a sham group, a model (middle cerebral artery occlusion/reperfusion, MCAO/R) group, and an EC-Exo group (tail vein injection of EC-Exo, once/2 days for 14 days) to evaluate the neuromotor and exploratory abilities of mice after MCAO/R. Apoptosis and synaptic protein expression levels were detected. The results demonstrated that EC-Exo inhibited neuronal apoptosis and increased synaptic length after OGD/R. In vivo, EC-Exo not only improved neural motor behavior and increased regional cerebral blood flow (rCBF) in MCAO/R-injured mice but also promoted the expression of synaptic regulatory proteins and inhibited apoptosis in the brain. These results suggest that EC-Exo may provide neuroprotection against stroke by promoting synaptic remodeling and inhibiting apoptosis from protecting neurons.
Collapse
|
15
|
Non-classical Notch signaling by MDA-MB-231 breast cancer cell-derived small extracellular vesicles promotes malignancy in poorly invasive MCF-7 cells. Cancer Gene Ther 2022; 29:1056-1069. [PMID: 35022518 DOI: 10.1038/s41417-021-00411-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/23/2021] [Accepted: 11/18/2021] [Indexed: 11/08/2022]
Abstract
Aberrant Notch signaling is implicated in breast cancer progression, and recent studies have demonstrated links between the Notch pathway components Notch1 and Notch1 intracellular domain (N1ICD) with poor clinical outcomes. Growing evidence suggests that Notch signaling can be regulated by small extracellular vesicles (SEVs). Here, we used breast cancer cell models to examine whether SEVs are involved in functional Notch signaling. We found that Notch components are packaged into MDA-MB-231- and MCF-7-derived SEVs, although higher levels of N1ICD were detected in SEVs from the more aggressive MDA-MB-231 cell line than from poorly invasive MCF-7 cells. SEV-Notch components were functional, as SEVs cargo from MDA-MB-231 cells induced the expression of Notch target genes in MCF-7 cells and triggered a more invasive and proliferative phenotype concomitant with the acquisition of mesenchymal features. Neutralization of the N1ICD cargo in MDA-MB-231-derived SEVs significantly reduced their potential to enhance the aggressiveness of MCF-7 cells in vitro and in a xenograft model. Overall, our results indicate that a SEV-mediated non-classical pathway of Notch signal transduction in breast cancer models bypasses the need for classical ligand-receptor interactions, which may have important implications in cancer.
Collapse
|
16
|
Gao J, Zhang X, Jiang L, Li Y, Zheng Q. Tumor endothelial cell-derived extracellular vesicles contribute to tumor microenvironment remodeling. Cell Commun Signal 2022; 20:97. [PMID: 35752798 PMCID: PMC9233793 DOI: 10.1186/s12964-022-00904-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/22/2022] [Indexed: 11/12/2022] Open
Abstract
Cancer progression involves several biological steps where angiogenesis is a key tumorigenic phenomenon. Extracellular vesicles (EVs) derived from tumor cells and other cells in the tumor microenvironment (TME) help modulate and maintain favorable microenvironments for tumors. Endothelial cells (ECs) activated by cancer-derived EVs have important roles in tumor angiogenesis. Interestingly, EVs from ECs activate tumor cells, i.e. extracellular matrix (ECM) remodeling and provide more supplements for tumor cells. Thus, EV communications between cancer cells and ECs may be effective therapeutic targets for controlling cancer progression. In this review, we describe the current knowledge on EVs derived from ECs and we examine how these EVs affect TME remodeling. Video abstract
Collapse
Affiliation(s)
- Jian Gao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, 110122, China.,Science Experiment Center of China Medical University, Shenyang, 110122, China
| | - Xiaodong Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100000, China.,National Clinical Research Center for Digestive Diseases, Beijing, 100000, China
| | - Lei Jiang
- Department of General Surgery, Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China
| | - Yan Li
- Department of Radiotherapy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, China.
| | - Qianqian Zheng
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
17
|
Autocrine Activity of Extracellular Vesicles Induced by Icariin and Its Effectiveness in Glucocorticoid-Induced Injury of Bone Microvascular Endothelial Cells. Cells 2022; 11:cells11121921. [PMID: 35741052 PMCID: PMC9221798 DOI: 10.3390/cells11121921] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/31/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023] Open
Abstract
Glucocorticoids could induce injury and apoptosis of bone microvascular endothelial cells (BMECs) in the femoral head, which is associated with the development of osteonecrosis and osteoporosis. Icariin is a prenylated flavonol glycoside isolated from Epimedium brevicornum, serving as the main active pharmaceutical constituent to treat bone loss. Currently, the impact of the autocrine activity of extracellular vesicles (EVs) induced by icariin on the glucocorticoid-induced injury of BMECs is still to be confirmed. In this study, EVs were isolated from BMECs treated with and without icariin by super-speed centrifugation. Although icariin treatment would not significantly change the size and total protein content of BMECs-derived EVs, expression of EVs-carried vascular endothelial growth factor (VEGF) and transforming growth factor β1 (TGF-β1) was enhanced and numerous miRNAs involved in cell proliferation and apoptosis were upregulated (e.g., hsa-miR-1469 and hsa-miR-133a-5p) or downregulated (e.g., hsa-miR-10b-5p) (p < 0.05). A total of 29 differentially expressed inflammatory factors were detected between the EVs secreted by BMECs from the Icariin-treated group and the Model group. The EVs secreted by BMECs could improve cell viability, decrease cell apoptosis, and promote cell migration and angiogenesis under the intervention of glucocorticoids. Meanwhile, icariin intervention could reinforce these protective effects of BMECs-derived EVs. To sum up, the present study indicates that icariin acts as a promising candidate for treating glucocorticoid-induced injury of BMECs and bone diseases, partially through the autocrine activity of EVs. In vivo or animal studies are still required to better understand the function of BMECs-derived EVs.
Collapse
|
18
|
Tong KL, Tan KE, Lim YY, Tien XY, Wong PF. CircRNA-miRNA interactions in atherogenesis. Mol Cell Biochem 2022; 477:2703-2733. [PMID: 35604519 DOI: 10.1007/s11010-022-04455-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 04/27/2022] [Indexed: 11/30/2022]
Abstract
Atherosclerosis is the major cause of coronary artery disease (CAD) which includes unstable angina, myocardial infarction, and heart failure. The onset of atherogenesis, a process of atherosclerotic lesion formation in the intima of arteries, is driven by lipid accumulation, a vicious cycle of reactive oxygen species (ROS)-induced oxidative stress and inflammatory reactions leading to endothelial cell (EC) dysfunction, vascular smooth muscle cell (VSMC) activation, and foam cell formation which further fuel plaque formation and destabilization. In recent years, there is a surge in the number of publications reporting the involvement of circular RNAs (circRNAs) in the pathogenesis of cardiovascular diseases, cancers, and metabolic syndromes. These studies have advanced our understanding on the biological functions of circRNAs. One of the most common mechanism of action of circRNAs reported is the sponging of microRNAs (miRNAs) by binding to the miRNAs response element (MRE), thereby indirectly increases the transcription of their target messenger RNAs (mRNAs). Individual networks of circRNA-miRNA-mRNA associated with atherogenesis have been extensively reported, however, there is a need to connect these findings for a complete overview. This review aims to provide an update on atherogenesis-related circRNAs and analyze the circRNA-miRNA-mRNA interactions in atherogenesis. The atherogenic mechanisms and clinical relevance of each atherogenesis-related circRNA were systematically discussed for better understanding of the knowledge gap in this area.
Collapse
Affiliation(s)
- Kind-Leng Tong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ke-En Tan
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yat-Yuen Lim
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Xin-Yi Tien
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
19
|
Jiang L, Chen W, Ye J, Wang Y. Potential Role of Exosomes in Ischemic Stroke Treatment. Biomolecules 2022; 12:115. [PMID: 35053263 PMCID: PMC8773818 DOI: 10.3390/biom12010115] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/01/2022] [Accepted: 01/07/2022] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is a life-threatening cerebral vascular disease and accounts for high disability and mortality worldwide. Currently, no efficient therapeutic strategies are available for promoting neurological recovery in clinical practice, except rehabilitation. The majority of neuroprotective drugs showed positive impact in pre-clinical studies but failed in clinical trials. Therefore, there is an urgent demand for new promising therapeutic approaches for ischemic stroke treatment. Emerging evidence suggests that exosomes mediate communication between cells in both physiological and pathological conditions. Exosomes have received extensive attention for therapy following a stroke, because of their unique characteristics, such as the ability to cross the blood brain-barrier, low immunogenicity, and low toxicity. An increasing number of studies have demonstrated positively neurorestorative effects of exosome-based therapy, which are largely mediated by the microRNA cargo. Herein, we review the current knowledge of exosomes, the relationships between exosomes and stroke, and the therapeutic effects of exosome-based treatments in neurovascular remodeling processes after stroke. Exosomes provide a viable and prospective treatment strategy for ischemic stroke patients.
Collapse
Affiliation(s)
- Lingling Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (L.J.); (W.C.); (J.Y.)
- Chinese Institute for Brain Research, Beijing 102206, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| | - Weiqi Chen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (L.J.); (W.C.); (J.Y.)
- Chinese Institute for Brain Research, Beijing 102206, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| | - Jinyi Ye
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (L.J.); (W.C.); (J.Y.)
- Chinese Institute for Brain Research, Beijing 102206, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; (L.J.); (W.C.); (J.Y.)
- Chinese Institute for Brain Research, Beijing 102206, China
- China National Clinical Research Center for Neurological Diseases, Beijing 100070, China
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100070, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing 100070, China
| |
Collapse
|
20
|
Hadjivasiliou Z, Hunter G. Talking to your neighbors across scales: Long-distance Notch signaling during patterning. Curr Top Dev Biol 2022; 150:299-334. [DOI: 10.1016/bs.ctdb.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
21
|
Abstract
Exosomes are a new horizon in modern therapy, presenting exciting new opportunities for advanced drug delivery and targeted release. Exosomes are small extracellular vesicles with a size range of 30-100 nm, secreted by all cell types in the human body and carrying a unique collection of DNA fragments, RNA species, lipids, protein biomarkers, transcription factors and metabolites. miRNAs are one of the most common RNA species in exosomes, and they play a role in a variety of biological processes including exocytosis, hematopoiesis and angiogenesis, as well as cellular communication via exosomes. Exosomes can act as cargo to transport this information from donor cells to near and long-distance target cells, participating in the reprogramming of recipient cells.
Collapse
Affiliation(s)
- Nihat Dilsiz
- Molecular Biology & Genetics, Faculty of Engineering & Natural Sciences, Istanbul Medeniyet University, Istanbul, 34700, Turkey
| |
Collapse
|
22
|
Li J, Gao X, Tian S, Tang M, Liu W. Exploring exosome data to identify prognostic gene signatures for lung adenocarcinoma. Future Oncol 2021; 17:4745-4756. [PMID: 34658257 DOI: 10.2217/fon-2021-0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Background: Exosomes are involved in tumorigenesis, growth and metastasis. However, the prognostic value of exosome-related genes in lung adenocarcinoma (LUAD) remains unclear. Methods: Clinical and transcriptome data from The Cancer Genome Atlas LUAD cohort were used to construct a model based on exosome-related genes, which was validated with LUAD data from the Gene Expression Omnibus (GEO). Gene ontology and Kyoto Encyclopedia of Genes and Genomes analysis were used to explore underlying mechanisms; the single-sample gene set enrichment analysis score was used to determine immune functions. Results: A 19-exosome-related gene signature for overall survival in LUAD was predictive in both The Cancer Genome Atlas and GEO LUAD cohorts. Immune-related and extracellular matrix-related pathways were enriched in differentially expressed genes. Immune states differed between high- and low-risk groups. Conclusion: The novel signature can be used to predict outcomes in LUAD.
Collapse
Affiliation(s)
- Jialin Li
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Xinliang Gao
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Suyan Tian
- Department of Division of Clinical Research, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Wei Liu
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| |
Collapse
|
23
|
Hong S, You JY, Paek K, Park J, Kang SJ, Han EH, Choi N, Chung S, Rhee WJ, Kim JA. Inhibition of tumor progression and M2 microglial polarization by extracellular vesicle-mediated microRNA-124 in a 3D microfluidic glioblastoma microenvironment. Am J Cancer Res 2021; 11:9687-9704. [PMID: 34646393 PMCID: PMC8490520 DOI: 10.7150/thno.60851] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/14/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Glioblastoma (GBM) is one of the most aggressive types of brain cancer. GBM progression is closely associated with microglia activation; therefore, understanding the regulation of the crosstalk between human GBM and microglia may help develop effective therapeutic strategies. Elucidation of efficient delivery of microRNA (miRNA) via extracellular vesicles (EVs) and their intracellular communications is required for therapeutic applications in GBM treatment. Methods: We used human GBM cells (U373MG) and human microglia. MiRNA-124 was loaded into HEK293T-derived EVs (miR-124 EVs). Various anti-tumor effects (proliferation, metastasis, chemosensitivity, M1/M2 microglial polarization, and cytokine profile) were investigated in U373MG and microglia. Anti-tumor effect of miR-124 EVs was also investigated in five different patient-derived GBM cell lines (SNU-201, SNU-466, SNU-489, SNU-626, and SNU-1105). A three-dimensional (3D) microfluidic device was used to investigate the interactive microenvironment of the tumor and microglia. Results: MiR-124 EVs showed highly efficient anti-tumor effects both in GBM cells and microglia. The mRNA expression levels of tumor progression and M2 microglial polarization markers were decreased in response to miR-124 EVs. The events were closely related to signal transducer and activator of transcription (STAT) 3 signaling in both GBM and microglia. In 3D microfluidic experiments, both U373MG and microglia migrated to a lesser extent and showed less-elongated morphology in the presence of miR-124 EVs compared to the control. Analyses of changes in cytokine levels in the microfluidic GBM-microglia environment showed that the treatment with miR-124 EVs led to tumor suppression and anti-cancer immunity, thereby recruiting natural killer (NK) cells into the tumor. Conclusions: In this study, we demonstrated that EV-mediated miR-124 delivery exerted synergistic anti-tumor effects by suppressing the growth of human GBM cells and inhibiting M2 microglial polarization. These findings provide new insights toward a better understanding of the GBM microenvironment and provide substantial evidence for the development of potential therapeutic strategies using miRNA-loaded EVs.
Collapse
|
24
|
Gomez AH, Joshi S, Yang Y, Tune JD, Zhao MT, Yang H. Bioengineering Systems for Modulating Notch Signaling in Cardiovascular Development, Disease, and Regeneration. J Cardiovasc Dev Dis 2021; 8:125. [PMID: 34677194 PMCID: PMC8541010 DOI: 10.3390/jcdd8100125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
The Notch intercellular signaling pathways play significant roles in cardiovascular development, disease, and regeneration through modulating cardiovascular cell specification, proliferation, differentiation, and morphogenesis. The dysregulation of Notch signaling leads to malfunction and maldevelopment of the cardiovascular system. Currently, most findings on Notch signaling rely on animal models and a few clinical studies, which significantly bottleneck the understanding of Notch signaling-associated human cardiovascular development and disease. Recent advances in the bioengineering systems and human pluripotent stem cell-derived cardiovascular cells pave the way to decipher the role of Notch signaling in cardiovascular-related cells (endothelial cells, cardiomyocytes, smooth muscle cells, fibroblasts, and immune cells), and intercellular crosstalk in the physiological, pathological, and regenerative context of the complex human cardiovascular system. In this review, we first summarize the significant roles of Notch signaling in individual cardiac cell types. We then cover the bioengineering systems of microfluidics, hydrogel, spheroid, and 3D bioprinting, which are currently being used for modeling and studying Notch signaling in the cardiovascular system. At last, we provide insights into ancillary supports of bioengineering systems, varied types of cardiovascular cells, and advanced characterization approaches in further refining Notch signaling in cardiovascular development, disease, and regeneration.
Collapse
Affiliation(s)
- Angello Huerta Gomez
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (A.H.G.); (S.J.); (Y.Y.)
| | - Sanika Joshi
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (A.H.G.); (S.J.); (Y.Y.)
- Texas Academy of Mathematics and Science, University of North Texas, Denton, TX 76201, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (A.H.G.); (S.J.); (Y.Y.)
| | - Johnathan D. Tune
- Department of Physiology & Anatomy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
| | - Ming-Tao Zhao
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43215, USA;
- The Heart Center, Nationwide Children’s Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (A.H.G.); (S.J.); (Y.Y.)
| |
Collapse
|
25
|
Romano E, Netti PA, Torino E. A High Throughput Approach Based on Dynamic High Pressure for the Encapsulation of Active Compounds in Exosomes for Precision Medicine. Int J Mol Sci 2021; 22:9896. [PMID: 34576059 PMCID: PMC8470411 DOI: 10.3390/ijms22189896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 12/18/2022] Open
Abstract
In recent decades, endogenous nanocarrier-exosomes have received considerable scientific interest as drug delivery systems. The unique proteo-lipid architecture allows the crossing of various natural barriers and protects exosomes cargo from degradation in the bloodstream. However, the presence of this bilayer membrane as well as their endogenous content make loading of exogenous molecules challenging. In the present work, we will investigate how to promote the manipulation of vesicles curvature by a high-pressure microfluidic system as a ground-breaking method for exosomes encapsulation. Exosomes isolated from Uppsala 87 Malignant Glioma (U87-MG) cell culture media were characterized before and after the treatment with high-pressure homogenization. Once their structural and biological stability were validated, we applied this novel method for the encapsulation in the lipidic exosomal bilayer of the chemotherapeutic Irinotecan HCl Trihydrate-CPT 11. Finally, we performed in vitro preliminary test to validate the nanobiointeraction of exosomes, uptake mechanisms, and cytotoxic effect in cell culture model.
Collapse
Affiliation(s)
- Eugenia Romano
- Department of Chemical, Materials Engineering & Industrial Production, University of Naples Federico II, Piazzale Tecchio 80, 80125 Naples, Italy; (E.R.); (P.A.N.)
- Interdisciplinary Research Center on Biomaterials, CRIB, Piazzale Tecchio 80, 80125 Naples, Italy
- Center for Advanced Biomaterials for Health Care, CABHC, Istituto Italiano di Tecnologia, IIT@CRIB, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials Engineering & Industrial Production, University of Naples Federico II, Piazzale Tecchio 80, 80125 Naples, Italy; (E.R.); (P.A.N.)
- Interdisciplinary Research Center on Biomaterials, CRIB, Piazzale Tecchio 80, 80125 Naples, Italy
- Center for Advanced Biomaterials for Health Care, CABHC, Istituto Italiano di Tecnologia, IIT@CRIB, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| | - Enza Torino
- Department of Chemical, Materials Engineering & Industrial Production, University of Naples Federico II, Piazzale Tecchio 80, 80125 Naples, Italy; (E.R.); (P.A.N.)
- Interdisciplinary Research Center on Biomaterials, CRIB, Piazzale Tecchio 80, 80125 Naples, Italy
- Center for Advanced Biomaterials for Health Care, CABHC, Istituto Italiano di Tecnologia, IIT@CRIB, Largo Barsanti e Matteucci 53, 80125 Naples, Italy
| |
Collapse
|
26
|
Zhou W, Dou M, Timilsina SS, Xu F, Li X. Recent innovations in cost-effective polymer and paper hybrid microfluidic devices. LAB ON A CHIP 2021; 21:2658-2683. [PMID: 34180494 PMCID: PMC8360634 DOI: 10.1039/d1lc00414j] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Hybrid microfluidic systems that are composed of multiple different types of substrates have been recognized as a versatile and superior platform, which can draw benefits from different substrates while avoiding their limitations. This review article introduces the recent innovations of different types of low-cost hybrid microfluidic devices, particularly focusing on cost-effective polymer- and paper-based hybrid microfluidic devices. In this article, the fabrication of these hybrid microfluidic devices is briefly described and summarized. We then highlight various hybrid microfluidic systems, including polydimethylsiloxane (PDMS)-based, thermoplastic-based, paper/polymer hybrid systems, as well as other emerging hybrid systems (such as thread-based). The special benefits of using these hybrid systems have been summarized accordingly. A broad range of biological and biomedical applications using these hybrid microfluidic devices are discussed in detail, including nucleic acid analysis, protein analysis, cellular analysis, 3D cell culture, organ-on-a-chip, and tissue engineering. The perspective trends of hybrid microfluidic systems involving the improvement of fabrication techniques and broader applications are also discussed at the end of the review.
Collapse
Affiliation(s)
- Wan Zhou
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA.
| | - Maowei Dou
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA.
| | - Sanjay S Timilsina
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA.
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - XiuJun Li
- Department of Chemistry and Biochemistry, University of Texas at El Paso, 500 W University Ave., El Paso, TX 79968, USA. and Border Biomedical Research Center, Biomedical Engineering, University of Texas at El Paso, 500 West University Ave., El Paso, TX 79968, USA and Environmental Science and Engineering, University of Texas at El Paso, 500 West University Ave., El Paso, TX 79968, USA
| |
Collapse
|
27
|
Bermick JR, Lincoln PM, Allen RM, Kunkel SL, Schaller MA. Elevated Notch ligands in serum are associated with HIV/TB coinfection. J Clin Tuberc Other Mycobact Dis 2021; 24:100258. [PMID: 34307905 PMCID: PMC8258674 DOI: 10.1016/j.jctube.2021.100258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Objective There is a clear need for improved biomarkers to diagnose HIV/TB coinfection. Although numerous tests can identify the existence of both of these microbes within the host, a parallel assessment of the host response to HIV/TB coinfection may prove as useful confirmation in cases where microbiological tests are inconclusive. To this end we assessed the levels of Notch ligands found in serum samples of patients with TB, HIV or HIV/TB coinfection. The Notch system is involved in almost every stage of development, including the maturation of the immune response. Upon exposure to a pathogen, the innate immune system will increase expression of Notch ligands Delta-like 1 and Delta-like 4. Previous research has demonstrated that Notch ligand expression is increased on monocytes from patients diagnosed with tuberculosis. We hypothesized that if Notch ligands were present in the peripheral blood of individuals diagnosed with TB, they may serve as a novel marker for infection. Design: Serum samples from patients with HIV, TB or HIV/TB coinfection were compared to serum from uninfected individuals to determine levels of DLL1 and DLL4 in a case controlled study. Methods DLL1 and DLL4 were measured by ELISA. Linear regression with post tests were used to determine if levels of DLL1 and DLL4 were increased in individuals with HIV/TB coinfection as compared to individuals infected with either HIV or TB or healthy controls. Results Delta-like 1 and Delta-like 4 were significantly increased in the serum of patients with HIV and HIV/ M. tuberculosis coinfection compared to other groups. Conclusions Assessment of Notch ligands in peripheral blood may enhance the diagnosis of individuals with active TB that are co-infected with HIV. The study will also need to be validated in in a larger cohort.
Collapse
Affiliation(s)
- Jennifer R Bermick
- Department of Pediatrics, Division of Neonatology, University of Iowa, Iowa City, IA, USA.,Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Pamela M Lincoln
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Ronald M Allen
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Steven L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Matthew A Schaller
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
28
|
Bhat A, Yadav J, Thakur K, Aggarwal N, Tripathi T, Chhokar A, Singh T, Jadli M, Bharti AC. Exosomes from cervical cancer cells facilitate pro-angiogenic endothelial reconditioning through transfer of Hedgehog-GLI signaling components. Cancer Cell Int 2021; 21:319. [PMID: 34167524 PMCID: PMC8223267 DOI: 10.1186/s12935-021-02026-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 06/15/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Angiogenic switch is a hallmark feature of transition from low-grade to high-grade cervical intraepithelial neoplasia (CIN) in cervical cancer progression. Therefore, early events leading to locally-advanced cervical metastatic lesions demand a greater understanding of the underlying mechanisms. Recent leads indicate the role of tumor-derived exosomes in altering the functions of endothelial cells in cervical cancer, which needs further investigation. METHODS Exosomes isolated from cervical cancer cell lines were assessed for their angiogenic effect on the human umbilical vein endothelial cells (HUVEC) using tube formation and wound healing assay. The exosomal uptake by HUVEC cells was monitored using PKH-67 labelling followed by fluorescence microscopy. Alterations in Hh-GLI signaling components, PTCH1 and GLI1, in HUVEC were measured by immunoblotting. Changes in angiogenesis-related transcripts of vascular endothelial growth factor VEGF-A, VEGF-B, VEGFR2 and angiopoietin-1, angiopoietin-2, osteopontin were measured in exosome-treated HUVEC and in the exosomal RNA by RT-PCR. RESULTS Enhanced tube formation, with an increased number of nodes and branching was observed in HUVEC's treated with exosomes derived from different cervical cancer cell lines. HPV-positive (SiHa and HeLa) cells' exosomes were more angiogenic. Exosome-treated HUVEC showed increased migration rate. PKH-67 labelled exosomes were found internalized in HUVEC. A high level of PTCH1 protein was detected in the exosome-treated endothelial cells. Subsequent RT-PCR analysis showed increased transcripts of Hh-GLI downstream target genes VEGF-A, VEGFR2, angiopoietin-2, and decreased expression of VEGF-B, and angiopoietin-1, suggestive of active Hh-GLI signaling. These effects were more pronounced in HUVEC's treated with exosomes of HPV-positive cells. However, these effects were independent of tumor-derived VEGF-A as exosomal cargo lacked VEGF-A transcripts or proteins. CONCLUSION Overall, the data showed cervical cancer exosomes promote pro-angiogenic response in endothelial cells via upregulation of Hh-GLI signaling and modulate downstream angiogenesis-related target genes. The study provides a novel exosome-mediated mechanism potentially favoring cervical angiogenesis and thus identifies the exosomes as potential pharmacological targets against locally-advanced metastatic cervical lesions.
Collapse
Affiliation(s)
- Anjali Bhat
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Joni Yadav
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Kulbhushan Thakur
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Nikita Aggarwal
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Tanya Tripathi
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Arun Chhokar
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Tejveer Singh
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Mohit Jadli
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India
| | - Alok Chandra Bharti
- Molecular Oncology Laboratory, Department of Zoology, University of Delhi (North Campus), Delhi, 110007, India.
| |
Collapse
|
29
|
Song H, Liu B, Dong B, Xu J, Zhou H, Na S, Liu Y, Pan Y, Chen F, Li L, Wang J. Exosome-Based Delivery of Natural Products in Cancer Therapy. Front Cell Dev Biol 2021; 9:650426. [PMID: 33738290 PMCID: PMC7960777 DOI: 10.3389/fcell.2021.650426] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
A rapidly growing research evidence has begun to shed light on the potential application of exosome, which modulates intercellular communications. As donor cell released vesicles, exosomes could play roles as a regulator of cellular behaviors in up-taken cells, as well as a delivery carrier of drugs for targeted cells. Natural product is an invaluable drug resources and it is used widely as therapeutic agents in cancers. This review summarizes the most recent advances in exosomes as natural product delivery carriers in cancer therapy from the following aspects: composition of exosomes, biogenesis of exosomes, and its functions in cancers. The main focus is the advantages and applications of exosomes for drug delivery in cancer therapy. This review also summarizes the isolation and application of exosomes as delivery carriers of natural products in cancer therapy. The recent progress and challenges of using exosomes as drug delivery vehicles for five representative anti-cancer natural products including paclitaxel, curcumin, doxorubicin, celastrol, and β-Elemene. Based on the discussion on the current knowledge about exosomes as delivery vehicles for drugs and natural compounds to the targeted site, this review delineates the landscape of the recent research, challenges, trends and prospects in exosomes as delivery vehicles for drugs and natural compounds for cancer treatment.
Collapse
Affiliation(s)
- Hang Song
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Bin Liu
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Bin Dong
- Neurology Department, The Hefei First People's Hospital, Hefei, China
| | - Jing Xu
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hui Zhou
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Sha Na
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Yanyan Liu
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Yunxia Pan
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Fengyuan Chen
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Lu Li
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Jinghui Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
30
|
Buchroithner B, Mayr S, Hauser F, Priglinger E, Stangl H, Santa-Maria AR, Deli MA, Der A, Klar TA, Axmann M, Sivun D, Mairhofer M, Jacak J. Dual Channel Microfluidics for Mimicking the Blood-Brain Barrier. ACS NANO 2021; 15:2984-2993. [PMID: 33480670 PMCID: PMC7905877 DOI: 10.1021/acsnano.0c09263] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/19/2021] [Indexed: 05/25/2023]
Abstract
High-resolution imaging is essential for analysis of the steps and way stations of cargo transport in in vitro models of the endothelium. In this study, we demonstrate a microfluidic system consisting of two channels horizontally separated by a cell-growth-promoting membrane. Its design allows for high-resolution (down to single-molecule level) imaging using a high numerical aperture objective with a short working distance. To reduce optical aberrations and enable single-molecule-sensitive imaging, an observation window was constructed in the membrane via laser cutting with subsequent structuring using 3D multiphoton lithography for improved cell growth. The upper channel was loaded with endothelial cells under flow conditions, which showed polarization and junction formation. A coculture of human vascular endothelial cells with pericytes was developed that mimics the blood-brain barrier. Finally, this dual channel microfluidics system enabled 3D localization microscopy of the cytoskeleton and 3D single-molecule-sensitive tracing of lipoprotein particles.
Collapse
Affiliation(s)
- Boris Buchroithner
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Sandra Mayr
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Fabian Hauser
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Eleni Priglinger
- Ludwig
Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria
| | - Herbert Stangl
- Institute
of Medical Chemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währingerstrasse 10, 1090 Vienna, Austria
| | - Ana Raquel Santa-Maria
- Institute
of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Maria A. Deli
- Institute
of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Andras Der
- Institute
of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Thomas A. Klar
- Institute
of Applied Physics, Johannes Kepler University
Linz, Altenberger Straße 69, 4040 Linz, Austria
| | - Markus Axmann
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Dmitry Sivun
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Mario Mairhofer
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| | - Jaroslaw Jacak
- Department
of Medical Engineering, University of Applied
Sciences Upper Austria, Garnisonstraße 21, 4020 Linz, Austria
| |
Collapse
|
31
|
Tuttolomondo A, Puleo MG, Velardo MC, Corpora F, Daidone M, Pinto A. Molecular Biology of Atherosclerotic Ischemic Strokes. Int J Mol Sci 2020; 21:9372. [PMID: 33317034 PMCID: PMC7763838 DOI: 10.3390/ijms21249372] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Among the causes of global death and disability, ischemic stroke (also known as cerebral ischemia) plays a pivotal role, by determining the highest number of worldwide mortality, behind cardiomyopathies, affecting 30 million people. The etiopathogenetic burden of a cerebrovascular accident could be brain ischemia (~80%) or intracranial hemorrhage (~20%). The most common site when ischemia occurs is the one is perfused by middle cerebral arteries. Worse prognosis and disablement consequent to brain damage occur in elderly patients or affected by neurological impairment, hypertension, dyslipidemia, and diabetes. Since, in the coming years, estimates predict an exponential increase of people who have diabetes, the disease mentioned above constitutes together with stroke a severe social and economic burden. In diabetic patients after an ischemic stroke, an exorbitant activation of inflammatory molecular pathways and ongoing inflammation is responsible for more severe brain injury and impairment, promoting the advancement of ischemic stroke and diabetes. Considering that the ominous prognosis of ischemic brain damage could by partially clarified by way of already known risk factors the auspice would be modifying poor outcome in the post-stroke phase detecting novel biomolecules associated with poor prognosis and targeting them for revolutionary therapeutic strategies.
Collapse
Affiliation(s)
- Antonino Tuttolomondo
- Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialties, “G. D’Alessandro”, University of Palermo, Piazza delle Cliniche n.2, 90127 Palermo, Italy; (M.G.P.); (M.C.V.); (F.C.); (M.D.); (A.P.)
| | | | | | | | | | | |
Collapse
|
32
|
Notch Signaling Function in the Angiocrine Regulation of Tumor Development. Cells 2020; 9:cells9112467. [PMID: 33198378 PMCID: PMC7697556 DOI: 10.3390/cells9112467] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022] Open
Abstract
The concept of tumor growth being angiogenesis dependent had its origin in the observations of Judah Folkman in 1969 of a retinoblastoma in a child. Tumor angiogenesis is initiated when endothelial cells (ECs) respond to local stimuli and migrate towards the growing mass, which results in the formation of tubular structures surrounded by perivascular support cells that transport blood to the inner tumor. In turn, the neo-vasculature supports tumor development and eventual metastasis. This process is highly regulated by several signaling pathways. Central to this process is the Notch signaling pathway. Beyond the role of Notch signaling in tumor angiogenesis, a major hallmark of cancer development, it has also been implicated in the regulation of tumor cell proliferation and survival, in epithelial-to-mesenchymal transition, invasion and metastasis and in the regulation of cancer stem cells, in a variety of hematologic and solid malignancies. There is increasing evidence for the tumor vasculature being important in roles other than those linked to blood perfusion. Namely, endothelial cells act on and influence neighboring tumor cells by use of angiocrine factors to generate a unique cellular microenvironment, thereby regulating tumor stem-like cells’ homeostasis, modulating tumor progression, invasiveness, trafficking and metastasis. This review will focus on Notch signaling components that play a part in angiocrine signaling in a tumor setting.
Collapse
|
33
|
Roefs MT, Sluijter JPG, Vader P. Extracellular Vesicle-Associated Proteins in Tissue Repair. Trends Cell Biol 2020; 30:990-1013. [PMID: 33069512 DOI: 10.1016/j.tcb.2020.09.009] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
The administration of (stem) cell-derived extracellular vesicles (EVs) promotes tissue repair through management of different inflammatory, proliferative and remodeling processes in the body. Despite the widely observed biological and therapeutic roles of EVs in wound healing and tissue repair, knowledge on how EVs activate recipient cells and which EV cargo is responsible for the subsequent functional effects is limited. Recent studies hint toward an important role for proteins as functional EV cargo. Here, we provide an overview of how EV-associated proteins promote tissue repair processes and discuss current challenges in evaluating their contribution to EV function and future directions for translating fundamental insights into clinically relevant EV therapies.
Collapse
Affiliation(s)
- Marieke T Roefs
- Department of Cardiology, Experimental Cardiology Laboratory, University Utrecht, University Medical Center Utrecht, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, University Utrecht, University Medical Center Utrecht, The Netherlands.
| | - Pieter Vader
- Department of Cardiology, Experimental Cardiology Laboratory, University Utrecht, University Medical Center Utrecht, The Netherlands; CDL Research, University Medical Center Utrecht, The Netherlands.
| |
Collapse
|
34
|
Robust and Scalable Angiogenesis Assay of Perfused 3D Human iPSC-Derived Endothelium for Anti-Angiogenic Drug Screening. Int J Mol Sci 2020; 21:ijms21134804. [PMID: 32645937 PMCID: PMC7370283 DOI: 10.3390/ijms21134804] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/28/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022] Open
Abstract
To advance pre-clinical vascular drug research, in vitro assays are needed that closely mimic the process of angiogenesis in vivo. Such assays should combine physiological relevant culture conditions with robustness and scalability to enable drug screening. We developed a perfused 3D angiogenesis assay that includes endothelial cells (ECs) from induced pluripotent stem cells (iPSC) and assessed its performance and suitability for anti-angiogenic drug screening. Angiogenic sprouting was compared with primary ECs and showed that the microvessels from iPSC-EC exhibit similar sprouting behavior, including tip cell formation, directional sprouting and lumen formation. Inhibition with sunitinib, a clinically used vascular endothelial growth factor (VEGF) receptor type 2 inhibitor, and 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3PO), a transient glycolysis inhibitor, both significantly reduced the sprouting of both iPSC-ECs and primary ECs, supporting that both cell types show VEGF gradient-driven angiogenic sprouting. The assay performance was quantified for sunitinib, yielding a minimal signal window of 11 and Z-factor of at least 0.75, both meeting the criteria to be used as screening assay. In conclusion, we have developed a robust and scalable assay that includes physiological relevant culture conditions and is amenable to screening of anti-angiogenic compounds.
Collapse
|
35
|
Rana U, Callan E, Entringer B, Michalkiewicz T, Joshi A, Parchur AK, Teng RJ, Konduri GG. AMP-Kinase Dysfunction Alters Notch Ligands to Impair Angiogenesis in Neonatal Pulmonary Hypertension. Am J Respir Cell Mol Biol 2020; 62:719-731. [PMID: 32048878 PMCID: PMC7258820 DOI: 10.1165/rcmb.2019-0275oc] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/26/2019] [Indexed: 12/18/2022] Open
Abstract
Decreased angiogenesis contributes to persistent pulmonary hypertension of the newborn (PPHN); mechanisms remain unclear. AMPK (5'AMP activated protein kinase) is a key regulator of cell metabolism. We investigated the hypothesis that a decrease in AMPK function leads to mitochondrial dysfunction and altered balance of notch ligands delta-like 4 (DLL4) and Jagged 1 (Jag1) to impair angiogenesis in PPHN. Studies were done in fetal lambs with PPHN induced by prenatal ductus arteriosus constriction and gestation-matched control lambs. PPHN lambs were treated with saline or AMPK agonist metformin. Angiogenesis was assessed in lungs with micro-computed tomography angiography and histology. AMPK function; expression of mitochondrial electron transport chain (ETC) complex proteins I-V, Dll4, and Jag1; mitochondrial number; and in vitro angiogenesis function were assessed in pulmonary artery endothelial cells (PAEC) from control and PPHN lambs. AMPK function was decreased in PPHN PAEC and lung sections. Expression of mitochondrial transcription factor, PGC-1α, ETC complex proteins I-V, and mitochondrial number were decreased in PPHN. In vitro angiogenesis of PAEC and capillary number and vessel volume fraction in the lung were decreased in PPHN. Expression of DLL4 was increased and Jag1 was decreased in PAEC from PPHN lambs. AMPK agonists A769662 and metformin increased the mitochondrial complex proteins and number, in vitro angiogenesis, and Jag1 levels and decreased DLL4 levels in PPHN PAEC. Infusion of metformin in vivo increased the vessel density in PPHN lungs. Decreased AMPK function contributes to impaired angiogenesis in PPHN by altered balance of notch ligands in PPHN.
Collapse
Affiliation(s)
- Ujala Rana
- Department of Pediatrics and Children’s Research Institute, and
| | - Emily Callan
- Department of Pediatrics and Children’s Research Institute, and
| | | | | | - Amit Joshi
- Department of Radiology and Center for Imaging, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Abdul K. Parchur
- Department of Radiology and Center for Imaging, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ru-Jeng Teng
- Department of Pediatrics and Children’s Research Institute, and
| | | |
Collapse
|
36
|
Jurj A, Zanoaga O, Braicu C, Lazar V, Tomuleasa C, Irimie A, Berindan-Neagoe I. A Comprehensive Picture of Extracellular Vesicles and Their Contents. Molecular Transfer to Cancer Cells. Cancers (Basel) 2020; 12:cancers12020298. [PMID: 32012717 PMCID: PMC7072213 DOI: 10.3390/cancers12020298] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
Critical processes such as growth, invasion, and metastasis of cancer cells are sustained via bidirectional cell-to-cell communication in tissue complex environments. Such communication involves the secretion of soluble factors by stromal cells and/or cancer cells within the tumor microenvironment (TME). Both stromal and cancer cells have been shown to export bilayer nanoparticles: encapsulated regulatory molecules that contribute to cell-to-cell communication. These nanoparticles are known as extracellular vesicles (EVs) being classified into exosomes, microvesicles, and apoptotic bodies. EVs carry a vast repertoire of molecules such as oncoproteins and oncopeptides, DNA fragments from parental to target cells, RNA species (mRNAs, microRNAs, and long non-coding RNA), and lipids, initiating phenotypic changes in TME. According to their specific cargo, EVs have crucial roles in several early and late processes associated with tumor development and metastasis. Emerging evidence suggests that EVs are being investigated for their implication in early cancer detection, monitoring cancer progression and chemotherapeutic response, and more relevant, the development of novel targeted therapeutics. In this study, we provide a comprehensive understanding of the biophysical properties and physiological functions of EVs, their implications in TME, and highlight the applicability of EVs for the development of cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Oana Zanoaga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
| | - Vladimir Lazar
- Worldwide Innovative Network for Personalized Cancer Therapy, 94800 Villejuif, France;
| | - Ciprian Tomuleasa
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
- Department of Hematology, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Alexandru Irimie
- 11th Department of Surgical Oncology and Gynaecological Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania
- Department of Surgery, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
- Correspondence: (A.I.); (I.B.-N.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.J.); (O.Z.); (C.B.); (C.T.)
- MEDFUTURE—Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute Prof. Dr. Ion Chiricuta, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
- Correspondence: (A.I.); (I.B.-N.)
| |
Collapse
|
37
|
Tjakra M, Wang Y, Vania V, Hou Z, Durkan C, Wang N, Wang G. Overview of Crosstalk Between Multiple Factor of Transcytosis in Blood Brain Barrier. Front Neurosci 2020; 13:1436. [PMID: 32038141 PMCID: PMC6990130 DOI: 10.3389/fnins.2019.01436] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022] Open
Abstract
Blood brain barrier (BBB) conserves unique regulatory system to maintain barrier tightness while allowing adequate transport between neurovascular units. This mechanism possess a challenge for drug delivery, while abnormality may result in pathogenesis. Communication between vascular and neural system is mediated through paracellular and transcellular (transcytosis) pathway. Transcytosis itself showed dependency with various components, focusing on caveolae-mediated. Among several factors, intense communication between endothelial cells, pericytes, and astrocytes is the key for a normal development. Regulatory signaling pathway such as VEGF, Notch, S1P, PDGFβ, Ang/Tie, and TGF-β showed interaction with the transcytosis steps. Recent discoveries showed exploration of various factors which has been proven to interact with one of the process of transcytosis, either endocytosis, endosomal rearrangement, or exocytosis. As well as providing a hypothetical regulatory pathway between each factors, specifically miRNA, mechanical stress, various cytokines, physicochemical, basement membrane and junctions remodeling, and crosstalk between developmental regulatory pathways. Finally, various hypotheses and probable crosstalk between each factors will be expressed, to point out relevant research application (Drug therapy design and BBB-on-a-chip) and unexplored terrain.
Collapse
Affiliation(s)
- Marco Tjakra
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Vicki Vania
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Zhengjun Hou
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Colm Durkan
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
38
|
Baruah J, Wary KK. Exosomes in the Regulation of Vascular Endothelial Cell Regeneration. Front Cell Dev Biol 2020; 7:353. [PMID: 31998716 PMCID: PMC6962177 DOI: 10.3389/fcell.2019.00353] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022] Open
Abstract
Exosomes have been described as nanoscale membranous extracellular vesicles that emerge from a variety of cells and tissues and are enriched with biologically active genomic and non-genomic biomolecules capable of transducing cell to cell communication. Exosome release, and exosome mediated signaling and cross-talks have been reported in several pathophysiological states. Therefore, exosomes have the potential to become suitable for the diagnosis, prognosis and treatment of specific diseases, including endothelial cell (EC) dysfunction and regeneration. The role of EC-derived exosomes in the mechanisms of cardiovascular tissue regenerative processes represents currently an area of intense research activity. Recent studies have described the potential of exosomes to influence the pathophysiology of immune signaling, tumor metastasis, and angiogenesis. In this review, we briefly discuss progress made in our understanding of the composition and the roles of exosomes in relation to EC regeneration as well as revascularization of ischemic tissues.
Collapse
Affiliation(s)
- Jugajyoti Baruah
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,Angiogenesis and Brain Development Laboratory, Division of Basic Neuroscience, McLean Hospital, Belmont, MA, United States
| | - Kishore K Wary
- Department of Pharmacology, The University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
39
|
Whitehead CA, Kaye AH, Drummond KJ, Widodo SS, Mantamadiotis T, Vella LJ, Stylli SS. Extracellular vesicles and their role in glioblastoma. Crit Rev Clin Lab Sci 2019:1-26. [PMID: 31865806 DOI: 10.1080/10408363.2019.1700208] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Research on the role of extracellular vesicles (EVs) in disease pathogenesis has been rapidly growing over the last two decades. As EVs can mediate intercellular communication, they can ultimately facilitate both normal and pathological processes through the delivery of their bioactive cargo, which may include nucleic acids, proteins and lipids. EVs have emerged as important regulators of brain tumors, capable of transferring oncogenic proteins, receptors, and small RNAs that may support brain tumor progression, including in the most common type of brain cancer, glioma. Investigating the role of EVs in glioma is crucial, as the most malignant glioma, glioblastoma (GBM), is incurable with a dismal median survival of 12-15 months. EV research in GBM has primarily focused on circulating brain tumor-derived vesicles in biofluids, such as blood and cerebrospinal fluid (CSF), investigating their potential as diagnostic and prognostic biomarkers. Gaining a greater understanding of the role of EVs and their cargo in brain tumor progression may contribute to the discovery of novel diagnostics and therapeutics. In this review, we summarize the known and emerging functions of EVs in glioma biology and pathogenesis, as well as their emerging biomarker potential.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia
| | - Andrew H Kaye
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Katharine J Drummond
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Australia
| | - Samuel S Widodo
- Department of Microbiology & Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, Australia
| | - Theo Mantamadiotis
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Department of Microbiology & Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, Australia
| | - Laura J Vella
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Stanley S Stylli
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia.,Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| |
Collapse
|
40
|
Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer 2019; 18:75. [PMID: 30940145 PMCID: PMC6444571 DOI: 10.1186/s12943-019-0991-5] [Citation(s) in RCA: 995] [Impact Index Per Article: 165.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/27/2019] [Indexed: 12/21/2022] Open
Abstract
Tumor-derived exosomes (TDEs) participate in formation and progression of different cancer processes, including tumor microenvironment (TME) remodeling, angiogenesis, invasion, metastasis and drug-resistance. Exosomes initiate or suppress various signaling pathways in the recipient cells via transmitting heterogeneous cargoes. In this review we discuss exosome biogenesis, exosome mediated metastasis and chemoresistance. Furthermore, tumor derived exosomes role in tumor microenvironment remodeling, and angiogenesis is reviewed. Also, exosome induction of epithelial mesenchymal transition (EMT) is highlighted. More importantly, we discuss extensively how exosomes regulate drug resistance in several cancers. Thus, understanding exosome biogenesis, their contents and the molecular mechanisms and signaling pathways that are responsible for metastasis and drug-resistance mediated by TDEs may help to devise novel therapeutic approaches for cancer progression particularly to overcome therapy-resistance and preventing metastasis as major factors of cancer mortality.
Collapse
Affiliation(s)
- Ladan Mashouri
- Department of Genetics, Faculty of Science, Shahrekord University, Shahrekord, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, USA
| | - Amir Reza Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Ali Mohammad Ahadi
- Department of Genetics, Faculty of Science, Shahrekord University, Shahrekord, Iran
| | - Fatemeh Molaei
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, USA.
| |
Collapse
|
41
|
Li X, Corbett AL, Taatizadeh E, Tasnim N, Little JP, Garnis C, Daugaard M, Guns E, Hoorfar M, Li ITS. Challenges and opportunities in exosome research-Perspectives from biology, engineering, and cancer therapy. APL Bioeng 2019; 3:011503. [PMID: 31069333 PMCID: PMC6481742 DOI: 10.1063/1.5087122] [Citation(s) in RCA: 371] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/08/2019] [Indexed: 12/11/2022] Open
Abstract
Exosomes are small (∼30-140 nm) lipid bilayer-enclosed particles of endosomal origin. They are a subset of extracellular vesicles (EVs) that are secreted by most cell types. There has been growing interest in exosome research in the last decade due to their emerging role as intercellular messengers and their potential in disease diagnosis. Indeed, exosomes contain proteins, lipids, and RNAs that are specific to their cell origin and could deliver cargo to both nearby and distant cells. As a result, investigation of exosome cargo contents could offer opportunities for disease detection and treatment. Moreover, exosomes have been explored as natural drug delivery vehicles since they can travel safely in extracellular fluids and deliver cargo to destined cells with high specificity and efficiency. Despite significant efforts made in this relatively new field of research, progress has been held back by challenges such as inefficient separation methods, difficulties in characterization, and lack of specific biomarkers. In this review, we summarize the current knowledge in exosome biogenesis, their roles in disease progression, and therapeutic applications and opportunities in bioengineering. Furthermore, we highlight the established and emerging technological developments in exosome isolation and characterization. We aim to consider critical challenges in exosome research and provide directions for future studies.
Collapse
Affiliation(s)
- Xia Li
- Department of Chemistry, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Alexander L. Corbett
- Department of Chemistry, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | | | - Nishat Tasnim
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Jonathan P. Little
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Cathie Garnis
- Department of Integrative Oncology, BC Cancer Agency, Vancouver, British Columbia, V5Z 1L3, Canada, and Department of Surgery, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Mads Daugaard
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada, and Department of Urologic Sciences, University of British Columbia, Vancouver, Vancouver, BC V5Z 1M9, Canada
| | - Emma Guns
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada, and Department of Urologic Sciences, University of British Columbia, Vancouver, Vancouver, BC V5Z 1M9, Canada
| | - Mina Hoorfar
- School of Engineering, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| | - Isaac T. S. Li
- Department of Chemistry, University of British Columbia, Kelowna, British Columbia V1V 1V7, Canada
| |
Collapse
|
42
|
Evidence of Extracellular Vesicles Biogenesis and Release in Mouse Embryonic Stem Cells. Stem Cell Rev Rep 2018; 14:262-276. [PMID: 29032399 DOI: 10.1007/s12015-017-9776-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Extracellular vesicles (EVs) released by mouse embryonic stem cells (mESCs) are considered a source of bioactive molecules that modulate their microenvironment by acting on intercellular communication. Either intracellular endosomal machinery or their derived EVs have been considered a relevant system of signal circuits processing. Herein, we show that these features are found in mESCs. Ultrastructural analysis revealed structures and organelles of the endosomal system such as coated pits and endocytosis-related vesicles, prominent rough endoplasmic reticulum and Golgi apparatus, and multivesicular bodies (MVBs) containing either few or many intraluminal vesicles (ILVs) that could be released as exosomes to extracellular milieu. Besides, budding vesicles shed from the plasma membrane to the extracellular space is suggestive of microvesicle biogenesis in mESCs. mESCs and mouse blastocyst express specific markers of the Endosomal Sorting Complex Required for Transport (ESCRT) system. Ultrastructural analysis and Nanoparticle Tracking Analysis (NTA) of isolated EVs revealed a heterogeneous population of exosomes and microvesicles released by mESCs. These vesicles contain Wnt10b and the Notch ligand Delta-like 4 (DLL4) and also the co-chaperone stress inducible protein 1 (STI1) and its partner Hsp90. Wnt10b and Dll4 colocalize with EVs biogenesis markers in mESCs. Overall, the present study supports the function of the mESCs endocytic network and their EVs as players in stem cell biology.
Collapse
|
43
|
Role of tumor-derived exosomes in cancer metastasis. Biochim Biophys Acta Rev Cancer 2018; 1871:12-19. [PMID: 30419312 DOI: 10.1016/j.bbcan.2018.10.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022]
Abstract
The highlights of cancer research include the discovery of exosomes, which are small (30-100 nm) sized vesicular nanoparticles released virtually by all cells. Tumor-derived exosomes (TDEs) are notoriously known for orchestrating the invasion-metastasis cascade via systemic pathways that we have previously proposed (1), resulting in a paradigm shift of our understanding about the pathobiology of metastases. In principle, exosomes serve as transport medium for proteins, mRNAs and miRNAs to transmit targeted cues from the primary cell to distant sites via horizontal transfer or cell-receptor interaction. In this chapter, we seek to explore in-depth the mechanisms engendering TDE in the metastatic cascade, along with experimental models to augment our understanding. The aforementioned has also paved way for parallel advancements in the therapeutic armamentarium, as evident from pronounced efforts to exploit the metastatic process for therapeutic targeting. In this light, we aim to examine potential anti-metastatic therapeutic opportunities derived from exosomal research. Lastly, exosomes may play a crucial role in the contemporary era of "liquid biopsies", given the array of molecular information with diagnostic and predictive indications. We thus intend to end this chapter off by exploring future applications of exosomes that could illuminate shortcomings and propel advancements in biomarker research.
Collapse
|
44
|
Luhtala N, Hunter T. Failure to detect functional transfer of active K-Ras protein from extracellular vesicles into recipient cells in culture. PLoS One 2018; 13:e0203290. [PMID: 30192821 PMCID: PMC6128481 DOI: 10.1371/journal.pone.0203290] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 08/17/2018] [Indexed: 12/14/2022] Open
Abstract
Exosomes, extracellular nanovesicles that carry nucleic acids, lipids, and proteins, have been the subject of several studies to assess their ability to transfer functional cargoes to cells. We recently characterized extracellular nanovesicles released from glioblastoma cells that carry active Ras in complex with proteins regulating exosome biogenesis. Here, we investigated whether a functional transfer of Ras from exosomes to other cells can initiate intercellular signaling. We observed that treatment of serum-starved, cultured glioblastoma cells with exogenous glioblastoma exosomes caused a significant increase in cellular viability over time. Moreover, we detected fluorescent signal transfer from lipophilic dye-labeled exogenous glioblastoma exosomes into cultured glioblastoma cells. To probe possible signaling from cell-to-cell, we utilized bimolecular luciferase complementation to examine the ability of K-Ras in exosomes to interact with the Raf-Ras Binding domain (Raf-RBD) expressed in a recipient cell line. Although the K-Ras/Raf-RBD interaction was readily detectable upon co-expression in a single cell line, or following lysis of co-cultured cell lines separately expressing K-Ras and RBD, bearing in mind the limitations of our assay, we were unable to detect the interaction in the intact, co-cultured cell lines or upon treatment of the Raf-RBD-expressing cells with exosomes containing K-Ras. Furthermore, HA-Tag-BFP fused to the K-Ras hypervariable region and CAAX sequence failed to be transferred at significant levels from extracellular vesicles into recipient cells, but remained detectable in the cell supernatants even after 96 hours of culture of naïve cells with extracellular vesicles. We conclude that if transfer of functional K-Ras from extracellular vesicles into the cytoplasm of recipient cells occurs, it must do so at an extremely low efficiency and therefore is unlikely to initiate Ras-ERK MAP kinase pathway signaling. These results suggest that studies claiming functional transfer of protein cargoes from exosomes should be interpreted with caution.
Collapse
Affiliation(s)
- Natalie Luhtala
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States of America
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, United States of America
| |
Collapse
|
45
|
Li Y, Liang J, Hu J, Ren X, Sheng Y. Down-regulation of exosomal miR-106b-5p derived from cholesteatoma perimatrix fibroblasts promotes angiogenesis in endothelial cells by overexpression of Angiopoietin 2. Cell Biol Int 2018; 42:1300-1310. [PMID: 29905392 DOI: 10.1002/cbin.11002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/02/2018] [Indexed: 12/17/2022]
Abstract
Human cholesteatoma perimatrix fibroblasts (hCPFs) can stimulate the endothelial cells of nearby microvessels to proliferate and migrate in a paracrine manner. Exosomes, secreted from various cell types, are one of the most important paracrine factors and play critical roles in intercellular communication. However, whether exosomes derived from human cholesteatoma perimatrix fibroblasts (hCPFs-Exo) can promote angiogenesis has not been reported. In this study, we isolated exosomes secreted by hCPFs and observed that hCPFs-Exo was able to promote migration and tube formation in human umbilical vein endothelial cells (HUVECs). Advanced studies revealed hCPFs-Exo with low expression of miR-106b-5p was transferred into HUVECs, and decreased expression of miR-106b-5p could promote angiogenesis by targeting Angiopoietin 2 (Angpt2) via binding to its 3'-UTR. Furthermore, low levels of miR-106b-5p triggered overexpression of Angpt2, and significantly increased HUVEC migration and tube formation. Taken together, our results suggest that hCPFs-Exo transports low expressed exosomal miR-106b-5p to endothelial cells and promotes angiogenesis by overexpression of Angpt2.
Collapse
Affiliation(s)
- Yang Li
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xi Wu Lu, Xi'an 710004, Shaanxi Province, China
| | - JianMin Liang
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xi Wu Lu, Xi'an 710004, Shaanxi Province, China
| | - Juan Hu
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xi Wu Lu, Xi'an 710004, Shaanxi Province, China
| | - Xiaoyong Ren
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xi Wu Lu, Xi'an 710004, Shaanxi Province, China
| | - Ying Sheng
- Department of Otolaryngology-Head and Neck Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, No.157, Xi Wu Lu, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
46
|
Rackov G, Garcia-Romero N, Esteban-Rubio S, Carrión-Navarro J, Belda-Iniesta C, Ayuso-Sacido A. Vesicle-Mediated Control of Cell Function: The Role of Extracellular Matrix and Microenvironment. Front Physiol 2018; 9:651. [PMID: 29922170 PMCID: PMC5996101 DOI: 10.3389/fphys.2018.00651] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/14/2018] [Indexed: 12/20/2022] Open
Abstract
Extracellular vesicles (EVs) — including exosomes, microvesicles and apoptotic bodies — have received much scientific attention last decade as mediators of a newly discovered cell-to-cell communication system, acting at short and long distances. EVs carry biologically active molecules, thus providing signals that influence a spectrum of functions in recipient cells during various physiological and pathological processes. Recent findings point to EVs as very attractive immunomodulatory therapeutic agents, vehicles for drug delivery and diagnostic and prognostic biomarkers in liquid biopsies. In addition, EVs interact with and regulate the synthesis of extracellular matrix (ECM) components, which is crucial for organ development and wound healing, as well as bone and cardiovascular calcification. EVs carrying matrix metalloproteinases (MMPs) are involved in ECM remodeling, thus modifying tumor microenvironment and contributing to premetastatic niche formation and angiogenesis. Here we review the role of EVs in control of cell function, with emphasis on their interaction with ECM and microenvironment in health and disease.
Collapse
Affiliation(s)
| | | | - Susana Esteban-Rubio
- Fundación de Investigación HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad CEU San Pablo, Madrid, Spain
| | | | | | - Angel Ayuso-Sacido
- IMDEA Nanoscience Institute, Madrid, Spain.,Fundación de Investigación HM Hospitales, Madrid, Spain.,Facultad de Medicina (IMMA), Universidad CEU San Pablo, Madrid, Spain
| |
Collapse
|
47
|
Abstract
NOTCH signalling is an evolutionarily conserved juxtacrine signalling pathway that is essential in development. Jagged1 (JAG1) and Delta-like ligand 4 (DLL4) are transmembrane NOTCH ligands that regulate angiogenesis by controlling endothelial cell (EC) differentiation, vascular development and maturation. In addition, DLL4 could bypass its canonical cell-cell contact-dependent signalling to influence NOTCH signalling and angiogenesis at a distance when it is packaged into extracellular vesicles (EVs). However, it is not clear whether JAG1 could also be packaged into EVs to influence NOTCH signalling and angiogenesis. In this work, we demonstrate that JAG1 is also packaged into EVs. We present evidence that JAG1-EVs inhibit NOTCH signalling and regulate EC behaviour and function. JAG1-EVs inhibited VEGF-induced HUVEC proliferation and migration in 2D culture condition and suppressed sprouting in a 3D microfluidic microenvironment. JAG1-EV treatment of HUVECs leads to a reduction of Notch1 intracellular domain (N1-ICD), and the proteasome and the intracellular domain of JAG1 (JAG1-ICD) are both required for this reduction to occur. These findings reveal a novel mechanism of JAG1 function in NOTCH signalling and ECs through EVs.
Collapse
|
48
|
Su SA, Xie Y, Fu Z, Wang Y, Wang JA, Xiang M. Emerging role of exosome-mediated intercellular communication in vascular remodeling. Oncotarget 2018; 8:25700-25712. [PMID: 28147325 PMCID: PMC5421963 DOI: 10.18632/oncotarget.14878] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 01/18/2017] [Indexed: 12/17/2022] Open
Abstract
Vascular remodeling refers to the alternations of function and structure in vasculature. A complex autocrine/paracrine set of cellular interaction is involved in vascular remodeling. Exosome, a newly identified natural nanocarrier and intercellular messenger, plays a pivotal role in regulating cell-to-cell communication. Exosome emerges as an important mediator in the process of vascular remodeling, showing the most prognostic and therapeutic potent in vascular diseases. Benefiting from exosomal trafficking, the vasculature can not only maintain its function and structure in physiological condition, but also adapt itself in pathological status. In this review, we will represent the roles of exosomes in angiogenesis, endothelial function and cardiac regeneration. In addition, greatly depending on the pathophysiological status of donor cells and peripheral micro-circumstance, the exosomal content could alter, which makes exosomes exhibit pleiotropic effects in vascular diseases. Hence, the diverse effects of exosomes in vascular diseases including atherosclerosis, neointima formation and vascular repair, primary hypertension, pulmonary artery hypertension, and aortic aneurysm will be discussed. Finally, the translational appliances targeting exosomes will be concluded by providing updated applications of engineered exosomes in clinic.
Collapse
Affiliation(s)
- Sheng-An Su
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| | - Yao Xie
- Cardiovascular Division, King's College London BHF Center, London, United Kingdom
| | - Zurong Fu
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| | - Yaping Wang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| | - Jian-An Wang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| | - Meixiang Xiang
- Department of Cardiology, Cardiovascular Key Lab of Zhejiang Province, Second Affiliated Hospital, Zhejiang University College of Medicine, Hang Zhou, Zhejiang, P.R. China
| |
Collapse
|
49
|
Control of Blood Vessel Formation by Notch Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1066:319-338. [PMID: 30030834 DOI: 10.1007/978-3-319-89512-3_16] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Blood vessels span throughout the body to nourish tissue cells and to provide gateways for immune surveillance. Endothelial cells that line capillaries have the remarkable capacity to be quiescent for years but to switch rapidly into the activated state once new blood vessels need to be formed. In addition, endothelial cells generate niches for progenitor and tumor cells and provide organ-specific paracrine (angiocrine) factors that control organ development and regeneration, maintenance of homeostasis and tumor progression. Recent data indicate a pivotal role for blood vessels in responding to metabolic changes and that endothelial cell metabolism is a novel regulator of angiogenesis. The Notch pathway is the central signaling mode that cooperates with VEGF, WNT, BMP, TGF-β, angiopoietin signaling and cell metabolism to orchestrate angiogenesis, tip/stalk cell selection and arteriovenous specification. Here, we summarize the current knowledge and implications regarding the complex roles of Notch signaling during physiological and tumor angiogenesis, the dynamic nature of tip/stalk cell selection in the nascent vessel sprout and arteriovenous differentiation. Furthermore, we shed light on recent work on endothelial cell metabolism, perfusion-independent angiocrine functions of endothelial cells in organ-specific vascular beds and how manipulation of Notch signaling may be used to target the tumor vasculature.
Collapse
|
50
|
Forghany Z, Robertson F, Lundby A, Olsen JV, Baker DA. Control of endothelial cell tube formation by Notch ligand intracellular domain interactions with activator protein 1 (AP-1). J Biol Chem 2017; 293:1229-1242. [PMID: 29196606 DOI: 10.1074/jbc.m117.819045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/30/2017] [Indexed: 01/08/2023] Open
Abstract
Notch signaling is a ubiquitous signal transduction pathway found in most if not all metazoan cell types characterized to date. It is indispensable for cell differentiation as well as tissue growth, tissue remodeling, and apoptosis. Although the canonical Notch signaling pathway is well characterized, accumulating evidence points to the existence of multiple, less well-defined layers of regulation. In this study, we investigated the function of the intracellular domain (ICD) of the Notch ligand Delta-like 4 (DLL4). We provide evidence that the DLL4 ICD is required for normal DLL4 subcellular localization. We further show that it is cleaved and interacts with the JUN proto-oncogene, which forms part of the activator protein 1 (AP-1) transcription factor complex. Mechanistically, the DLL4 ICD inhibited JUN binding to DNA and thereby controlled the expression of JUN target genes, including DLL4 Our work further demonstrated that JUN strongly stimulates endothelial cell tube formation and that DLL4 constrains this process. These results raise the possibility that Notch/DLL4 signaling is bidirectional and suggest that the DLL4 ICD could represent a point of cross-talk between Notch and receptor tyrosine kinase (RTK) signaling.
Collapse
Affiliation(s)
- Zary Forghany
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| | - Francesca Robertson
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| | - Alicia Lundby
- Novo Nordisk Foundation Center for Protein Research and.,the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | | | - David A Baker
- From the Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands and
| |
Collapse
|