1
|
Yang J, Wang D, Yu H, Wang L, Wang Y, Liu X, Huang Y, Ouyang C, Hong Y, Ren S, Wang Y, Jin Y, Hu J, Feng J. Lauric acid-mediated gelatin/hyaluronic acid composite hydrogel with effective antibacterial and immune regulation for accelerating MRSA-infected diabetic wound healing. Int J Biol Macromol 2024; 290:138792. [PMID: 39689796 DOI: 10.1016/j.ijbiomac.2024.138792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/02/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
The infected diabetic wound healing is an increasingly severe healthcare problem worldwide. Bacterial infection and the inflammatory microenvironment hinder diabetic wound healing. Meanwhile, the combination of inhibiting bacterial growth and promoting macrophage polarization in the wound microenvironment is beneficial for treating diabetic wounds. Nowadays, hydrogels, as an emerging wound dressing, have great potential to replace or supplement traditional bandages or gauze. Here, glycyl methacrylate gelatin (Gel-Gym), oxidized hyaluronic acid (HA-CHO) and lauric acid (LA) were used to prepare the composite hydrogel (GH/LA) in addressing the clinical dilemma. The hydrogel could withstand 50 % compression deformation, its swelling rate was as low as 18 %, and its adhesion to pig skin reached 14 kPa. Moreover, a diabetic infected wound model was used to evaluate the feasibility of GH/LA hydrogel in vivo. The hydrogels' antimicrobial, anti-inflammatory and prorestitutive potentials were further investigated, and GH/LA showed a therapeutic effect on diabetic wounds. Interestingly, macrophage polarization into the M2 phenotype was significantly enhanced in the presence of GH/LA via GPR40/NF-κB pathway. This study provided a new avenue for treating methicillin-resistant staphylococcus aureus (MRSA) infected diabetic wounds.
Collapse
Affiliation(s)
- Jian Yang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Dongyu Wang
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China.
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China.
| | - Yun Wang
- Zhejiang TUANYUAN Composite Materials Co., Ltd., Pinghu 314200, PR China
| | - Xiaowei Liu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Yudi Huang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Chenguang Ouyang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Yichuan Hong
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Shuning Ren
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Yu Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Yang Jin
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Jian Hu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310003, PR China
| | - Jingyi Feng
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, PR China
| |
Collapse
|
2
|
Araujo-Abad S, Berna JM, Lloret-Lopez E, López-Cortés A, Saceda M, de Juan Romero C. Exosomes: from basic research to clinical diagnostic and therapeutic applications in cancer. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00990-2. [PMID: 39298081 DOI: 10.1007/s13402-024-00990-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2024] [Indexed: 09/21/2024] Open
Abstract
Cancer continues to pose a global threat despite potent anticancer drugs, often accompanied by undesired side effects. To enhance patient outcomes, sophisticated multifunctional approaches are imperative. Small extracellular vesicles (EVs), a diverse family of naturally occurring vesicles derived from cells, offer advantages over synthetic carriers. Among the EVs, the exosomes are facilitating intercellular communication with minimal toxicity, high biocompatibility, and low immunogenicity. Their tissue-specific targeting ability, mediated by surface molecules, enables precise transport of biomolecules to cancer cells. Here, we explore the potential of exosomes as innovative therapeutic agents, including cancer vaccines, and their clinical relevance as biomarkers for clinical diagnosis. We highlight the cargo possibilities, including nucleic acids and drugs, which make them a good delivery system for targeted cancer treatment and contrast agents for disease monitoring. Other general aspects, sources, and the methodology associated with therapeutic cancer applications are also reviewed. Additionally, the challenges associated with translating exosome-based therapies into clinical practice are discussed, together with the future prospects for this innovative approach.
Collapse
Affiliation(s)
- Salomé Araujo-Abad
- Cancer Research Group, Faculty of Engineering and Applied Sciences, Universidad de Las Américas, Quito, 170124, Ecuador
| | - José Marcos Berna
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Elena Lloret-Lopez
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, 170124, Ecuador
| | - Miguel Saceda
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain
| | - Camino de Juan Romero
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l'Almazara 11, Elche, Alicante, 03203, Spain.
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández, Avda. Universidad s/n, Ed. Torregaitán, Elche, Alicante, 03202, Spain.
| |
Collapse
|
3
|
Yang H, Niu L, Jia J, Liang W, Li Q, Pan Y. Extracellular vesicles: Mediators of microenvironment in hypoxia-associated neurological diseases. Clin Neurol Neurosurg 2024; 240:108250. [PMID: 38552364 DOI: 10.1016/j.clineuro.2024.108250] [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: 11/14/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 04/29/2024]
Abstract
Hypoxia is a prevalent characteristic of numerous neurological disorders including stroke, Alzheimer's disease, and Parkinson's disease. Extracellular vesicles (EVs) are minute particles released by cells that contain diverse biological materials, including proteins, lipids, and nucleic acids. They have been implicated in a range of physiological and pathological processes including intercellular communication, immune responses, and disease progression. EVs are believed to play a pivotal role in modulating the microenvironment of hypoxia-associated neurological diseases. These EVs are capable of transporting hypoxia-inducible factors such as proteins and microRNAs to neighboring or remote cells, thereby influencing their behavior. Furthermore, EVs can traverse the blood-brain barrier, shielding the brain from detrimental substances in the bloodstream. This enables them to deliver their payload directly to the brain cells, potentially intensifying the effects of hypoxia. Nonetheless, the capacity of EVs to breach the blood-brain barrier presents new opportunities for drug delivery. The objective of this study was to elucidate the role of EVs as mediators of information exchange during tissue hypoxia, a pathophysiological process in ischemic stroke and malignant gliomas. We also investigated their involvement in the progression and regression of major diseases of the central nervous system, which are pertinent to the development of therapeutic interventions for neurological disorders.
Collapse
Affiliation(s)
- Hu Yang
- Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Liang Niu
- Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China; The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Juan Jia
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Department of Anesthesiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Wentao Liang
- Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Qiang Li
- Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China.
| | - Yawen Pan
- Department of Neurosurgery, The Second Hospital of Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 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
|
5
|
Kumar S, Dhar R, Kumar LBSS, Shivji GG, Jayaraj R, Devi A. Theranostic signature of tumor-derived exosomes in cancer. Med Oncol 2023; 40:321. [PMID: 37798480 DOI: 10.1007/s12032-023-02176-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023]
Abstract
Cancer is the most challenging global health crisis. In the recent times, studies on extracellular vesicles (EVs) are adding a new chapter to cancer research and reports on EVs explores cancer in a new dimension. Exosomes are a group of subpopulations of EVs. It originates from the endosomes and carries biologically active molecules to the neighboring cells which in turn transforms the recipient cell activity. In general, it plays a role in cellular communication. The correlation between exosomes and cancer is fascinating. Tumor-derived exosomes (TEXs) play a dynamic role in cancer progression and are associated with uncontrolled cell growth, angiogenesis, immune suppression, and metastasis. Its molecular cargo is an excellent source of cancer biomarkers. Several advanced molecular profiling approaches assist in exploring the TEXs in depth. This paves the way for a strong foundation for identifying and detecting more specific and efficient biomarkers. TEXs are also gaining importance in scientific society for its role in cancer therapy and several clinical trials based on TEXs is a proof of its significance. In this review, we have highlighted the role of TEXs in mediating immune cell reprogramming, cancer development, metastasis, EMT, organ-specific metastasis, and its clinical significance in cancer theranostics. TEXs profiling is an effective method to understand the complications associated with cancer leading to good health and well-being of the individual and society as a whole.
Collapse
Affiliation(s)
- Samruti Kumar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Lokesh Babu Sirkali Suresh Kumar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Gauresh Gurudas Shivji
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India
| | - Rama Jayaraj
- Jindal Institute of Behavioral Sciences (JIBS), Jindal Global Institution of Eminence Deemed to Be University, 28, Sonipat, 131001, India
- Director of Clinical Sciences, Northern Territory Institute of Research and Training, Darwin, NT, 0909, Australia
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Chengalpattu District, Kattankulathur, Tamil Nadu, 603203, India.
| |
Collapse
|
6
|
Moeinabadi-Bidgoli K, Rezaee M, Hossein-Khannazer N, Babajani A, Aghdaei HA, Arki MK, Afaghi S, Niknejad H, Vosough M. Exosomes for angiogenesis induction in ischemic disorders. J Cell Mol Med 2023; 27:763-787. [PMID: 36786037 PMCID: PMC10003030 DOI: 10.1111/jcmm.17689] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
Ischaemic disorders are leading causes of morbidity and mortality worldwide. While the current therapeutic approaches have improved life expectancy and quality of life, they are unable to "cure" ischemic diseases and instate regeneration of damaged tissues. Exosomes are a class of extracellular vesicles with an average size of 100-150 nm, secreted by many cell types and considered a potent factor of cells for paracrine effects. Since exosomes contain multiple bioactive components such as growth factors, molecular intermediates of different intracellular pathways, microRNAs and nucleic acids, they are considered as cell-free therapeutics. Besides, exosomes do not rise cell therapy concerns such as teratoma formation, alloreactivity and thrombotic events. In addition, exosomes are stored and utilized more convenient. Interestingly, exosomes could be an ideal complementary therapeutic tool for ischemic disorders. In this review, we discussed therapeutic functions of exosomes in ischemic disorders including angiogenesis induction through various mechanisms with specific attention to vascular endothelial growth factor pathway. Furthermore, different delivery routes of exosomes and different modification strategies including cell preconditioning, gene modification and bioconjugation, were highlighted. Finally, pre-clinical and clinical investigations in which exosomes were used were discussed.
Collapse
Affiliation(s)
- Kasra Moeinabadi-Bidgoli
- Basic and Molecular Epidemiology of Gastroenterology Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Malihe Rezaee
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastroenterology Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siamak Afaghi
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
7
|
Jin M, Zhang S, Wang M, Li Q, Ren J, Luo Y, Sun X. Exosomes in pathogenesis, diagnosis, and therapy of ischemic stroke. Front Bioeng Biotechnol 2022; 10:980548. [PMID: 36588958 PMCID: PMC9800834 DOI: 10.3389/fbioe.2022.980548] [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: 06/28/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Ischemic stroke is one of the major contributors to death and disability worldwide. Thus, there is an urgent need to develop early brain tissue perfusion therapies following acute stroke and to enhance functional recovery in stroke survivors. The morbidity, therapy, and recovery processes are highly orchestrated interactions involving the brain with other tissues. Exosomes are natural and ideal mediators of intercellular information transfer and recognized as biomarkers for disease diagnosis and prognosis. Changes in exosome contents express throughout the physiological process. Accumulating evidence demonstrates the use of exosomes in exploring unknown cellular and molecular mechanisms of intercellular communication and organ homeostasis and indicates their potential role in ischemic stroke. Inspired by the unique properties of exosomes, this review focuses on the communication, diagnosis, and therapeutic role of various derived exosomes, and their development and challenges for the treatment of cerebral ischemic stroke.
Collapse
Affiliation(s)
- Meiqi Jin
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Shuxia Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Mengchen Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Qiaoyu Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Jiahui Ren
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China,*Correspondence: Yun Luo, ; Xiaobo Sun,
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China,Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China,NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China,*Correspondence: Yun Luo, ; Xiaobo Sun,
| |
Collapse
|
8
|
Huda MN, Nurunnabi M. Potential Application of Exosomes in Vaccine Development and Delivery. Pharm Res 2022; 39:2635-2671. [PMID: 35028802 PMCID: PMC8757927 DOI: 10.1007/s11095-021-03143-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023]
Abstract
Exosomes are cell-derived components composed of proteins, lipid, genetic information, cytokines, and growth factors. They play a vital role in immune modulation, cell-cell communication, and response to inflammation. Immune modulation has downstream effects on the regeneration of damaged tissue, promoting survival and repair of damaged resident cells, and promoting the tumor microenvironment via growth factors, antigens, and signaling molecules. On top of carrying biological messengers like mRNAs, miRNAs, fragmented DNA, disease antigens, and proteins, exosomes modulate internal cell environments that promote downstream cell signaling pathways to facilitate different disease progression and induce anti-tumoral effects. In this review, we have summarized how vaccines modulate our immune response in the context of cancer and infectious diseases and the potential of exosomes as vaccine delivery vehicles. Both pre-clinical and clinical studies show that exosomes play a decisive role in processes like angiogenesis, prognosis, tumor growth metastasis, stromal cell activation, intercellular communication, maintaining cellular and systematic homeostasis, and antigen-specific T- and B cell responses. This critical review summarizes the advancement of exosome based vaccine development and delivery, and this comprehensive review can be used as a valuable reference for the broader delivery science community.
Collapse
Affiliation(s)
- Md Nurul Huda
- Department of Pharmaceutical Sciences, University of Texas at El Paso School of Pharmacy, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Enviromental Science and Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, University of Texas at El Paso School of Pharmacy, 1101 N. Campbell St, El Paso, TX, 79902, USA.
- Enviromental Science and Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA.
- Biomedical Engineering, University of Texas at El Paso, El Paso, TX, 79968, USA.
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
9
|
Karami Fath M, Azami J, Masoudi A, Mosaddeghi Heris R, Rahmani E, Alavi F, Alagheband Bahrami A, Payandeh Z, Khalesi B, Dadkhah M, Pourzardosht N, Tarhriz V. Exosome-based strategies for diagnosis and therapy of glioma cancer. Cancer Cell Int 2022; 22:262. [PMID: 35989351 PMCID: PMC9394011 DOI: 10.1186/s12935-022-02642-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/26/2022] [Indexed: 11/10/2022] Open
Abstract
Glioblastoma belongs to the most aggressive type of cancer with a low survival rate that is characterized by the ability in forming a highly immunosuppressive tumor microenvironment. Intercellular communication are created via exosomes in the tumor microenvironment through the transport of various biomolecules. They are primarily involved in tumor growth, differentiation, metastasis, and chemotherapy or radiation resistance. Recently several studies have highlighted the critical role of tumor-derived exosomes against immune cells. According to the structural and functional properties, exosomes could be essential instruments to gain a better molecular mechanism for tumor understanding. Additionally, they are qualified as diagnostic/prognostic markers and therapeutic tools for specific targeting of invasive tumor cells such as glioblastomas. Due to the strong dependency of exosome features on the original cells and their developmental status, it is essential to review their critical modulating molecules, clinical relevance to glioma, and associated signaling pathways. This review is a non-clinical study, as the possible role of exosomes and exosomal microRNAs in glioma cancer are reported. In addition, their content to overcome cancer resistance and their potential as diagnostic biomarkers are analyzed.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Jalil Azami
- Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Alireza Masoudi
- Department of Laboratory Sciences, Faculty of Alied Medical Sciences, Qom University of Medical Sciences, Qom, Iran
| | | | - Elnaz Rahmani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Alavi
- Department of Pathobiology, Faculty of Specialized Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Armina Alagheband Bahrami
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Payandeh
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research, Tabriz, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Navid Pourzardosht
- Biochemistry Department, Guilan University of Medical Sciences, Rasht, Iran
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Zhao G, Yu H, Ding L, Wang W, Wang H, Hu Y, Qin L, Deng G, Xie B, Li G, Qi L. microRNA-27a-3p delivered by extracellular vesicles from glioblastoma cells induces M2 macrophage polarization via the EZH1/KDM3A/CTGF axis. Cell Death Dis 2022; 8:260. [PMID: 35568721 PMCID: PMC9107457 DOI: 10.1038/s41420-022-01035-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 11/09/2022]
Abstract
Glioblastoma (GBM) cell-derived extracellular vesicles (EVs) have been demonstrated to modulate tumor microenvironment. In the present study, we attempted to discuss the role of hsa-microRNA-27a-3p (miR-27a-3p) delivered by GBM-EVs in M2 macrophage polarization. The isolated GBM-EVs were co-cultured with macrophages. After co-culture under normoxia/hypoxia, the effect of EV-derived hsa-miR-27a-3p on GBM cell biological processes was analyzed. Additionally, the target genes of hsa-miR-27a-3p were predicted. Moreover, the binding of enhancer of zeste homologue 1 (EZH1) to lysine-specific demethylase 3A (KDM3A) promoter region and the interaction between KDM3A and connective tissue growth factor (CTGF) were analyzed. GBM mouse models were established to verify the functions of EV-derived hsa-miR-27a-3p in vivo. We found increased hsa-miR-27a-3p in GBM tissues as well as GBM-EVs, which induced M2 polarization, thus promoting proliferative, migrative and invasive potentials of GBM cells. hsa-miR-27a-3p targeted EZH1 and promoted KDM3A expression to elevate the CTGF expression. GBM-EV-delivered hsa-miR-27a-3p promoted the KDM3A-upregulated CTGF by downregulating EZH1, thereby promoting M2 macrophage polarization and development of GBM in vivo. We demonstrated that EV-derived hsa-miR-27a-3p may promote M2 macrophage polarization to induce GBM.
Collapse
Affiliation(s)
- Guifang Zhao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.,Jilin Medical University, Jilin, 132013, China
| | - Hongquan Yu
- Department of Oncological Neurosurgery, the First Hospital of Jilin University, Changchun, 130021, China
| | - Lijuan Ding
- Department of Oncological Neurosurgery, the First Hospital of Jilin University, Changchun, 130021, China
| | - Weiyao Wang
- Jilin Medical University, Jilin, 132013, China
| | - Huan Wang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Yao Hu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Lingsha Qin
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Guangce Deng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Buqing Xie
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Guofeng Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China
| | - Ling Qi
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, China.
| |
Collapse
|
11
|
Molecular Profile Study of Extracellular Vesicles for the Identification of Useful Small “Hit” in Cancer Diagnosis. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app112210787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor-secreted extracellular vesicles (EVs) are the main mediators of cell-cell communication, permitting cells to exchange proteins, lipids, and metabolites in varying physiological and pathological conditions. They contain signature tumor-derived molecules that reflect the intracellular status of their cell of origin. Recent studies have shown that tumor cell-derived EVs can aid in cancer metastasis through the modulation of the tumor microenvironment, suppression of the immune system, pre-metastatic niche formation, and subsequent metastasis. EVs can easily be isolated from a variety of biological fluids, and their content makes them useful biomarkers for the diagnosis, prognosis, monitorization of cancer progression, and response to treatment. This review aims to explore the biomarkers of cancer cell-derived EVs obtained from liquid biopsies, in order to understand cancer progression and metastatic evolution for early diagnosis and precision therapy.
Collapse
|
12
|
The Renin-Angiotensin System in the Tumor Microenvironment of Glioblastoma. Cancers (Basel) 2021; 13:cancers13164004. [PMID: 34439159 PMCID: PMC8392691 DOI: 10.3390/cancers13164004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Glioblastoma (GB) is the most aggressive brain cancer in humans. Patient survival outcomes have remained dismal despite intensive research over the past 50 years, with a median overall survival of only 14.6 months. We highlight the critical role of the renin–angiotensin system (RAS) on GB cancer stem cells and the tumor microenvironment which, in turn, influences cancer stem cells in driving tumorigenesis and treatment resistance. We present recent developments and underscore the need for further research into the GB tumor microenvironment. We discuss the novel therapeutic targeting of the RAS using existing commonly available medications and utilizing model systems to further this critical investigation. Abstract Glioblastoma (GB) is an aggressive primary brain tumor. Despite intensive research over the past 50 years, little advance has been made to improve the poor outcome, with an overall median survival of 14.6 months following standard treatment. Local recurrence is inevitable due to the quiescent cancer stem cells (CSCs) in GB that co-express stemness-associated markers and components of the renin–angiotensin system (RAS). The dynamic and heterogeneous tumor microenvironment (TME) plays a fundamental role in tumor development, progression, invasiveness, and therapy resistance. There is increasing evidence showing the critical role of the RAS in the TME influencing CSCs via its upstream and downstream pathways. Drugs that alter the hallmarks of cancer by modulating the RAS present a potential new therapeutic alternative or adjunct to conventional treatment of GB. Cerebral and GB organoids may offer a cost-effective method for evaluating the efficacy of RAS-modulating drugs on GB. We review the nexus between the GB TME, CSC niche, and the RAS, and propose re-purposed RAS-modulating drugs as a potential therapeutic alternative or adjunct to current standard therapy for GB.
Collapse
|
13
|
Han J, Shi Y, Willis G, Imani J, Kwon MY, Li G, Ayaub E, Ghanta S, Ng J, Hwang N, Tsoyi K, El-Chemaly S, Kourembanas S, Mitsialis SA, Rosas IO, Liu X, Perrella MA. Mesenchymal stromal cell-derived syndecan-2 regulates the immune response during sepsis to foster bacterial clearance and resolution of inflammation. FEBS J 2021; 289:417-435. [PMID: 34355516 PMCID: PMC8766882 DOI: 10.1111/febs.16154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/28/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Sepsis is a life-threatening process related to a dysregulated host response to an underlying infection, which results in organ dysfunction and poor outcomes. Therapeutic strategies using mesenchymal stromal cells (MSCs) are under investigation for sepsis, with efforts to improve cellular utility. Syndecan (SDC) proteins are transmembrane proteoglycans involved with cellular signaling events including tissue repair and modulating inflammation. Bone marrow-derived human MSCs express syndecan-2 (SDC2) at a level higher than other SDC family members; thus, we explored SDC2 in MSC function. Administration of human MSCs silenced for SDC2 in experimental sepsis resulted in decreased bacterial clearance, and increased tissue injury and mortality compared with wild-type MSCs. These findings were associated with a loss of resolution of inflammation in the peritoneal cavity, and higher levels of proinflammatory mediators in organs. MSCs silenced for SDC2 had a decreased ability to promote phagocytosis of apoptotic neutrophils by macrophages in the peritoneum, and also a diminished capability to convert macrophages from a proinflammatory to a proresolution phenotype via cellular or paracrine actions. Extracellular vesicles are a paracrine effector of MSCs that may contribute to resolution of inflammation, and their production was dramatically reduced in SDC2-silenced human MSCs. Collectively, these data demonstrate the importance of SDC2 for cellular and paracrine function of human MSCs during sepsis.
Collapse
Affiliation(s)
- Junwen Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,School of Life Sciences, Beijing University of Chinese Medicine, China
| | - Yuanyuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, China
| | - Gareth Willis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, MA, USA
| | - Jewel Imani
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Min-Young Kwon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gu Li
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ehab Ayaub
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Sailaja Ghanta
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie Ng
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Narae Hwang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Konstantin Tsoyi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, MA, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, MA, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Seifali E, Hassanzadeh G, Mahdavipour M, Mortezaee K, Moini A, Satarian L, Shekari F, Nazari A, Movassaghi S, Akbari M. Extracellular Vesicles Derived from Human Umbilical Cord Perivascular Cells Improve Functional Recovery in Brain Ischemic Rat via the Inhibition of Apoptosis. IRANIAN BIOMEDICAL JOURNAL 2021; 24:347-60. [PMID: 32872749 PMCID: PMC7601540 DOI: 10.29252/ibj.24.6.342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Ischemic stroke, as a health problem caused by the reduced blood supply to the brain, can lead to the neuronal death. The number of reliable therapies for stroke is limited. MSCs exhibit therapeutic achievement. A major limitation of MSC application in cell therapy is the short survival span. MSCs affect target tissues through the secretion of many paracrine agents including EVs. This study aimed to investigate the effect of HUCPVCs-derived EVs on apoptosis, functional recovery, and neuroprotection. Methods: Ischemia was induced by MCAO in male Wistar rats. Animals were classified into sham, MCAO, MCAO + HUCPVC, and MCAO + EV groups. Treatments began at two hours after ischemia. Expressions of apoptotic-related proteins (BAX/BCl-2 and caspase-3 and -9), the amount of TUNEL-positive cells, neuronal density (MAP2), and dead neurons (Nissl staining) were assessed on day seven post MCAO. Results: Administration of EVs improved the sensorimotor function (p < 0.001) and reduced the apoptotic rate of Bax/Bcl-2 ratio (p < 0.001), as well as caspases and TUNEL-positive cells (p < 0.001) in comparison to the MCAO group. EV treatment also reduced the number of dead neurons and increased the number of MAP2+ cells in the IBZ (p < 0.001), as compared to the MCAO group. Conclusion: Our findings showed that HUCPVCs-derived EVs are more effective than their mother’s cells in improving neural function, possibly via the regulation of apoptosis in the ischemic rats. The strategy of cell-free extracts is, thus, helpful in removing the predicaments surrounding cell therapy in targeting brain diseases.
Collapse
Affiliation(s)
- Elham Seifali
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Mahdavipour
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ashraf Moini
- Department of Gynecology and Obstetrics, School of Medicine, Tehran University of Medical Science, Tehran, Iran.,Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,Breast Disease Research Center (BDRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Satarian
- Department of Brain and Cognitive Science, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Abdoreza Nazari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shabnam Movassaghi
- Department of Anatomy and cognitive neuroscience, School of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Akbari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Marsico G, Martin‐Saldaña S, Pandit A. Therapeutic Biomaterial Approaches to Alleviate Chronic Limb Threatening Ischemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003119. [PMID: 33854887 PMCID: PMC8025020 DOI: 10.1002/advs.202003119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/24/2020] [Indexed: 05/14/2023]
Abstract
Chronic limb threatening ischemia (CLTI) is a severe condition defined by the blockage of arteries in the lower extremities that leads to the degeneration of blood vessels and is characterized by the formation of non-healing ulcers and necrosis. The gold standard therapies such as bypass and endovascular surgery aim at the removal of the blockage. These therapies are not suitable for the so-called "no option patients" which present multiple artery occlusions with a likelihood of significant limb amputation. Therefore, CLTI represents a significant clinical challenge, and the efforts of developing new treatments have been focused on stimulating angiogenesis in the ischemic muscle. The delivery of pro-angiogenic nucleic acid, protein, and stem cell-based interventions have limited efficacy due to their short survival. Engineered biomaterials have emerged as a promising method to improve the effectiveness of these latter strategies. Several synthetic and natural biomaterials are tested in different formulations aiming to incorporate nucleic acid, proteins, stem cells, macrophages, or endothelial cells in supportive matrices. In this review, an overview of the biomaterials used alone and in combination with growth factors, nucleic acid, and cells in preclinical models is provided and their potential to induce revascularization and regeneration for CLTI applications is discussed.
Collapse
Affiliation(s)
- Grazia Marsico
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| | - Sergio Martin‐Saldaña
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| |
Collapse
|
16
|
Li C, Kitzerow O, Nie F, Dai J, Liu X, Carlson MA, Casale GP, Pipinos II, Li X. Bioengineering strategies for the treatment of peripheral arterial disease. Bioact Mater 2021; 6:684-696. [PMID: 33005831 PMCID: PMC7511653 DOI: 10.1016/j.bioactmat.2020.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/12/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
Peripheral arterial disease (PAD) is a progressive atherosclerotic disorder characterized by narrowing and occlusion of arteries supplying the lower extremities. Approximately 200 million people worldwide are affected by PAD. The current standard of operative care is open or endovascular revascularization in which blood flow restoration is the goal. However, many patients are not appropriate candidates for these treatments and are subject to continuous ischemia of their lower limbs. Current research in the therapy of PAD involves developing modalities that induce angiogenesis, but the results of simple cell transplantation or growth factor delivery have been found to be relatively poor mainly due to difficulties in stem cell retention and survival and rapid diffusion and enzymolysis of growth factors following injection of these agents in the affected tissues. Biomaterials, including hydrogels, have the capability to protect stem cells during injection and to support cell survival. Hydrogels can also provide a sustained release of growth factors at the injection site. This review will focus on biomaterial systems currently being investigated as carriers for cell and growth factor delivery, and will also discuss biomaterials as a potential stand-alone method for the treatment of PAD. Finally, the challenges of development and use of biomaterials systems for PAD treatment will be reviewed.
Collapse
Affiliation(s)
- Cui Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Oliver Kitzerow
- Department of Genetics Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Fujiao Nie
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Jingxuan Dai
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaoyan Liu
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Omaha VA Medical Center, Omaha, NE, 68105, United States
| | - George P. Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Iraklis I. Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaowei Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| |
Collapse
|
17
|
Zhang S, Yang J, Shen L. Extracellular vesicle-mediated regulation of tumor angiogenesis- implications for anti-angiogenesis therapy. J Cell Mol Med 2021; 25:2776-2785. [PMID: 33586248 PMCID: PMC7957215 DOI: 10.1111/jcmm.16359] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis plays an important role in tumour progression. However, anti‐angiogenesis therapy of inhibiting pro‐angiogenic factors failed to meet expectations in certain types of tumour in clinical trials. Recent studies reveal that tumour‐derived extracellular vesicles (EVs) are essential in tumour angiogenesis and anti‐angiogenesis drug resistance. This function has most commonly been attributed to EV contents including proteins and non‐coding RNAs. Here, we summarize the recent findings of tumour‐derived EV contents associated with regulating angiogenesis and illustrate the underlying mechanisms. In addition, the roles of EVs in tumour microenvironmental cells are also illustrated with a focus on how EVs participate in cell‐cell communication, contributing to tumour‐mediated angiogenesis. It will help offer new perspectives on developing targets of anti‐angiogenesis drugs and improve the efficacy of anti‐angiogenesis therapies based on tumour‐derived EVs.
Collapse
Affiliation(s)
- Shuqiong Zhang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junyao Yang
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lisong Shen
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Shi J, Zhang Y, Yao B, Sun P, Hao Y, Piao H, Zhao X. Role of Exosomes in the Progression, Diagnosis, and Treatment of Gliomas. Med Sci Monit 2020; 26:e924023. [PMID: 33245712 PMCID: PMC7706139 DOI: 10.12659/msm.924023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Gliomas are the most common primary malignant brain tumors associated with a low survival rate. Even after surgery, radiotherapy, and chemotherapy, gliomas still have a poor prognosis. Extracellular vesicles are a heterogeneous group of cell-derived membranous structures. Exosomes are a type of extracellular vesicles, their size ranges from 30 nm to 100 nm. Recent studies have proved that glioma cells could release numerous exosomes; therefore, exosomes have gained increasing attention in glioma-related research. Recent studies have confirmed the importance of extracellular vesicles, particularly exosomes, in the development of brain tumors, including gliomas. Exosomes mediate intercellular communication in the tumor microenvironment by transporting biomolecules (proteins, lipids, deoxyribonucleic acid, and ribonucleic acid); thereby playing a prominent role in tumor proliferation, differentiation, metastasis, and resistance to chemotherapy or radiation. Given their nanoscale size, exosomes can traverse the blood-brain barrier and promote tumor progression by modifying the tumor microenvironment. Based on their structural and functional characteristics, exosomes are demonstrating their value not only as diagnostic and prognostic markers, but also as tools in therapies specifically targeting glioma cells. Therefore, exosomes are a promising therapeutic target for the diagnosis, prognosis, and treatment of malignant gliomas. More research will be needed before exosomes can be used in clinical applications. Here, we describe the exosomes, their morphology, and their roles in the diagnosis and progression of gliomas. In addition, we discuss the potential of exosomes as a therapeutic target/drug delivery system for patients with gliomas.
Collapse
Affiliation(s)
- Ji Shi
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Ye Zhang
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Bing Yao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Peixin Sun
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Yuanyuan Hao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Haozhe Piao
- Department of Neurosurgery, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Xi Zhao
- Department of Anesthesia, Cancer Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|
19
|
Majid QA, Fricker ATR, Gregory DA, Davidenko N, Hernandez Cruz O, Jabbour RJ, Owen TJ, Basnett P, Lukasiewicz B, Stevens M, Best S, Cameron R, Sinha S, Harding SE, Roy I. Natural Biomaterials for Cardiac Tissue Engineering: A Highly Biocompatible Solution. Front Cardiovasc Med 2020; 7:554597. [PMID: 33195451 PMCID: PMC7644890 DOI: 10.3389/fcvm.2020.554597] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) constitute a major fraction of the current major global diseases and lead to about 30% of the deaths, i.e., 17.9 million deaths per year. CVD include coronary artery disease (CAD), myocardial infarction (MI), arrhythmias, heart failure, heart valve diseases, congenital heart disease, and cardiomyopathy. Cardiac Tissue Engineering (CTE) aims to address these conditions, the overall goal being the efficient regeneration of diseased cardiac tissue using an ideal combination of biomaterials and cells. Various cells have thus far been utilized in pre-clinical studies for CTE. These include adult stem cell populations (mesenchymal stem cells) and pluripotent stem cells (including autologous human induced pluripotent stem cells or allogenic human embryonic stem cells) with the latter undergoing differentiation to form functional cardiac cells. The ideal biomaterial for cardiac tissue engineering needs to have suitable material properties with the ability to support efficient attachment, growth, and differentiation of the cardiac cells, leading to the formation of functional cardiac tissue. In this review, we have focused on the use of biomaterials of natural origin for CTE. Natural biomaterials are generally known to be highly biocompatible and in addition are sustainable in nature. We have focused on those that have been widely explored in CTE and describe the original work and the current state of art. These include fibrinogen (in the context of Engineered Heart Tissue, EHT), collagen, alginate, silk, and Polyhydroxyalkanoates (PHAs). Amongst these, fibrinogen, collagen, alginate, and silk are isolated from natural sources whereas PHAs are produced via bacterial fermentation. Overall, these biomaterials have proven to be highly promising, displaying robust biocompatibility and, when combined with cells, an ability to enhance post-MI cardiac function in pre-clinical models. As such, CTE has great potential for future clinical solutions and hence can lead to a considerable reduction in mortality rates due to CVD.
Collapse
Affiliation(s)
- Qasim A. Majid
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Annabelle T. R. Fricker
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - David A. Gregory
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Natalia Davidenko
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Olivia Hernandez Cruz
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Richard J. Jabbour
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas J. Owen
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pooja Basnett
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Barbara Lukasiewicz
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Molly Stevens
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Sian E. Harding
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ipsita Roy
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
20
|
mTOR Modulates Intercellular Signals for Enlargement and Infiltration in Glioblastoma Multiforme. Cancers (Basel) 2020; 12:cancers12092486. [PMID: 32887296 PMCID: PMC7564864 DOI: 10.3390/cancers12092486] [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: 08/13/2020] [Revised: 08/26/2020] [Accepted: 08/31/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Glioblastoma multiforme (GBM) is the most aggressive and lethal primary brain tumor. Emerging evidence indicate the multi-faceted role of extracellular vesicles (EVs) in GBM growth and proliferation. In fact, GBM-derived EVs can alter the phenotype of GBM-associated parenchymal cells; thus, promoting tumor growth, angiogenesis, and immune evasion. Remarkably, among several pathways that are frequently deregulated in GBM, mammalian Target of Rapamycin (mTOR) up-regulation, and subsequent autophagy (ATG) depression are considered hallmarks of GBM. In fact, mTOR-dependent ATG inhibition strongly correlates with the presence of EVs, which in turn promotes glioblastoma cancer stem cells (GSCs) self-renewal, proliferation, and infiltration. ATG and exosome release are reciprocally regulated. In detail, a failure in ATG enhances exosomal release. Therefore, strategies aimed at targeting on mTOR-dependent extracellular vesicles could be a promising approach for GBM prevention and treatment. Abstract Recently, exosomal release has been related to the acquisition of a malignant phenotype in glioblastoma cancer stem cells (GSCs). Remarkably, intriguing reports demonstrate that GSC-derived extracellular vesicles (EVs) contribute to glioblastoma multiforme (GBM) tumorigenesis via multiple pathways by regulating tumor growth, infiltration, and immune invasion. In fact, GSCs release tumor-promoting macrovesicles that can disseminate as paracrine factors to induce phenotypic alterations in glioma-associated parenchymal cells. In this way, GBM can actively recruit different stromal cells, which, in turn, may participate in tumor microenvironment (TME) remodeling and, thus, alter tumor progression. Vice versa, parenchymal cells can transfer their protein and genetic contents to GSCs by EVs; thus, promoting GSCs tumorigenicity. Moreover, GBM was shown to hijack EV-mediated cell-to-cell communication for self-maintenance. The present review examines the role of the mammalian Target of Rapamycin (mTOR) pathway in altering EVs/exosome-based cell-to-cell communication, thus modulating GBM infiltration and volume growth. In fact, exosomes have been implicated in GSC niche maintenance trough the modulation of GSCs stem cell-like properties, thus, affecting GBM infiltration and relapse. The present manuscript will focus on how EVs, and mostly exosomes, may act on GSCs and neighbor non tumorigenic stromal cells to modify their expression and translational profile, while making the TME surrounding the GSC niche more favorable for GBM growth and infiltration. Novel insights into the mTOR-dependent mechanisms regulating EV-mediated intercellular communication within GBM TME hold promising directions for future therapeutic applications.
Collapse
|
21
|
Ghaemmaghami AB, Mahjoubin-Tehran M, Movahedpour A, Morshedi K, Sheida A, Taghavi SP, Mirzaei H, Hamblin MR. Role of exosomes in malignant glioma: microRNAs and proteins in pathogenesis and diagnosis. Cell Commun Signal 2020; 18:120. [PMID: 32746854 PMCID: PMC7397575 DOI: 10.1186/s12964-020-00623-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
Malignant gliomas are the most common and deadly type of central nervous system tumors. Despite some advances in treatment, the mean survival time remains only about 1.25 years. Even after surgery, radiotherapy and chemotherapy, gliomas still have a poor prognosis. Exosomes are the most common type of extracellular vesicles with a size range of 30 to 100 nm, and can act as carriers of proteins, RNAs, and other bioactive molecules. Exosomes play a key role in tumorigenesis and resistance to chemotherapy or radiation. Recent evidence has shown that exosomal microRNAs (miRNAs) can be detected in the extracellular microenvironment, and can also be transferred from cell to cell via exosome secretion and uptake. Therefore, many recent studies have focused on exosomal miRNAs as important cellular regulators in various physiological and pathological conditions. A variety of exosomal miRNAs have been implicated in the initiation and progression of gliomas, by activating and/or inhibiting different signaling pathways. Exosomal miRNAs could be used as therapeutic agents to modulate different biological processes in gliomas. Exosomal miRNAs derived from mesenchymal stem cells could also be used for glioma treatment. The present review summarizes the exosomal miRNAs that have been implicated in the pathogenesis, diagnosis and treatment of gliomas. Moreover, exosomal proteins could also be involved in glioma pathogenesis. Exosomal miRNAs and proteins could also serve as non-invasive biomarkers for prognosis and disease monitoring. Video Abstract.
Collapse
Affiliation(s)
- Amir B. Ghaemmaghami
- grid.17063.330000 0001 2157 2938Department of Psychology, Behaviour, Genetics and Neurobiology Program, University of Toronto, Toronto, Canada
| | - Maryam Mahjoubin-Tehran
- grid.411583.a0000 0001 2198 6209Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran ,grid.411583.a0000 0001 2198 6209Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Movahedpour
- grid.412571.40000 0000 8819 4698Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Korosh Morshedi
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Sheida
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- grid.444768.d0000 0004 0612 1049Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- grid.38142.3c000000041936754XWellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA ,grid.412988.e0000 0001 0109 131XLaser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, Johannesburg, 2028 South Africa
| |
Collapse
|
22
|
Zuo Z, Hu H, Xu Q, Luo X, Peng D, Zhu K, Zhao Q, Xie Y, Ren J. BBCancer: an expression atlas of blood-based biomarkers in the early diagnosis of cancers. Nucleic Acids Res 2020; 48:D789-D796. [PMID: 31665503 PMCID: PMC7145713 DOI: 10.1093/nar/gkz942] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/28/2019] [Accepted: 10/09/2019] [Indexed: 12/26/2022] Open
Abstract
The early detection of cancer holds the key to combat and control the increasing global burden of cancer morbidity and mortality. Blood-based screenings using circulating DNAs (ctDNAs), circulating RNA (ctRNAs), circulating tumor cells (CTCs) and extracellular vesicles (EVs) have shown promising prospects in the early detection of cancer. Recent high-throughput gene expression profiling of blood samples from cancer patients has provided a valuable resource for developing new biomarkers for the early detection of cancer. However, a well-organized online repository for these blood-based high-throughput gene expression data is still not available. Here, we present BBCancer (http://bbcancer.renlab.org/), a web-accessible and comprehensive open resource for providing the expression landscape of six types of RNAs, including messenger RNAs (mRNAs), long noncoding RNAs (lncRNAs), microRNAs (miRNAs), circular RNAs (circRNAs), tRNA-derived fragments (tRFRNAs) and Piwi-interacting RNAs (piRNAs) in blood samples, including plasma, CTCs and EVs, from cancer patients with various cancer types. Currently, BBCancer contains expression data of the six RNA types from 5040 normal and tumor blood samples across 15 cancer types. We believe this database will serve as a powerful platform for developing blood biomarkers.
Collapse
Affiliation(s)
- Zhixiang Zuo
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| | - Huanjing Hu
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| | - Qingxian Xu
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| | - Xiaotong Luo
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| | - Di Peng
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| | - Kaiyu Zhu
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| | - Qi Zhao
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| | - Yubin Xie
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| | - Jian Ren
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, School of Life Sciences, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
23
|
Baj-Krzyworzeka M, Mytar B, Weglarczyk K, Szatanek R, Kijowski J, Siedlar M. Protumorogenic Potential of Pancreatic Adenocarcinoma-Derived Extracellular Vesicles. Folia Biol (Praha) 2020; 66:104-110. [PMID: 33069189 DOI: 10.14712/fb2020066030104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Cancer development is a highly complicated process in which tumour growth depends on the development of its vascularization system. To support their own growth, tumour cells significantly modify their microenvironment. One of such modifications inflicted by tumours is stimulation of endothelial cell migration and proliferation. There is accumulating evidence that extracellular vesicles (EVs) secreted by tumour cells (tumour-derived EVs, TEVs) may be regarded as "messengers" with the potential for affecting the biological activities of target cells. Interaction of TEVs with different cell types occurs in an auto- and paracrine manner and may lead to changes in the function of the latter, e.g., promoting motility, proliferation, etc. This study analysed the proangiogenic activity of EVs derived from human pancreatic adenocarcinoma cell line (HPC-4, TEVHPC) in vitro and their effect in vivo on Matrigel matrix vascularization in severe combined immunodeficient (SCID) mice. TEVHPC enhanced proliferation of HPC-4 cells and induced their motility. Moreover, TEVHPC stimulated human umbilical vein endothelial cell (HUVEC) proliferation and migration in vitro. Additionally, TEVHPC influenced secretion of proangiogenic factors (IL-8, VEGF) by HUVEC cells and supported Matrigel matrix haemoglobinization in vivo. These data show that TEVs may support tumour propagation in an autocrine manner and may support vascularization of the tumour. The presented data are in line with the theory that tumour cells themselves are able to modulate the microenvironment via TEVs to maximize their growth potential.
Collapse
Affiliation(s)
- M Baj-Krzyworzeka
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Cracow, Poland
| | - B Mytar
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Cracow, Poland
| | - K Weglarczyk
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Cracow, Poland
| | - R Szatanek
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Cracow, Poland
| | - J Kijowski
- Department of Transplantology, Institute of Paediatrics, Jagiellonian University Medical College, Cracow, Poland
| | - M Siedlar
- Department of Clinical Immunology, Institute of Paediatrics, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
24
|
Miles J, Andre M, Caobi A, Ruiz M, Nair M, Raymond A. Bioengineered Exosomal Extracellular Vesicles in Cancer Therapeutics. Crit Rev Biomed Eng 2020; 48:177-187. [PMID: 33389895 PMCID: PMC11102805 DOI: 10.1615/critrevbiomedeng.2020034847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Liquid or blood-based biopsy is a less invasive and more efficient method in which to clinicians can identify diagnostic, prognostic, and therapeutic responsive biomarkers in cancer patients. Circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), RNAs, proteins, metabolites, and extracellular vesicles (EVs) are all potential biomarkers found in liquid biopsies. All nucleated cells including healthy, virally infected, and cancer cells release EVs. Since the early 1980s, evidence has mounted to support the pathophysiological role of EVs in cancer. Here we focus on the smallest of the EV, the exosome, and their clinical relevance as nanotherapeutics for cancers. Exosomes obtained from tumors have been reported to promote and/or facilitate malignancy of cancers especially in terms of metastatic potential. Exosomal EVs have also contributed to the development of therapeutic resistance. Recent studies demonstrate that intrinsic and bioengineered exosomes can serve as effective therapeutic agents that disrupt cancer progression. Here we review the current literature regarding the utilization of bioengineered exosomes for therapeutics to treat prevalent cancers such as melanoma, glioma, breast, pancreatic, hepatic, cervical, prostate, and colon cancers. Overall, studies reviewed show that bioengineered exosomes are effective and promising for targeted cancer therapy.
Collapse
Affiliation(s)
- J. Miles
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University 11200 SW 8th Street Miami, FL 33199, USA
| | - M. Andre
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University 11200 SW 8th Street Miami, FL 33199, USA
| | - A. Caobi
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University 11200 SW 8th Street Miami, FL 33199, USA
| | - M. Ruiz
- Department of Human and Health Services, Herbert Wertheim College of Medicine, Florida International University 11200 SW 8th Street Miami, FL 33199, USA
- Miami Cancer Institute, Bone Marrow Transplant and CART T-Cell Therapy Department, Baptist Health South Florida, 8900 N Kendall Dr., Miami, FL 33176, USA
| | - M. Nair
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University 11200 SW 8th Street Miami, FL 33199, USA
| | - A.D. Raymond
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University 11200 SW 8th Street Miami, FL 33199, USA
| |
Collapse
|
25
|
Veith AP, Henderson K, Spencer A, Sligar AD, Baker AB. Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev 2019; 146:97-125. [PMID: 30267742 DOI: 10.1016/j.addr.2018.09.010] [Citation(s) in RCA: 492] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 09/15/2018] [Accepted: 09/24/2018] [Indexed: 12/19/2022]
Abstract
The enhancement of wound healing has been a goal of medical practitioners for thousands of years. The development of chronic, non-healing wounds is a persistent medical problem that drives patient morbidity and increases healthcare costs. A key aspect of many non-healing wounds is the reduced presence of vessel growth through the process of angiogenesis. This review surveys the creation of new treatments for healing cutaneous wounds through therapeutic angiogenesis. In particular, we discuss the challenges and advancement that have been made in delivering biologic, pharmaceutical and cell-based therapies as enhancers of wound vascularity and healing.
Collapse
|
26
|
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: 924] [Impact Index Per Article: 154.0] [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
|
27
|
WANG WENTING, LI ZIJIAN, FENG JUAN. The potential role of exosomes in the diagnosis and therapy of ischemic diseases. Cytotherapy 2018; 20:1204-1219. [DOI: 10.1016/j.jcyt.2018.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/14/2018] [Accepted: 06/22/2018] [Indexed: 12/13/2022]
|