1
|
Kim GB, Hu Q, Rommel PC. Special issue: Recent advances in immunotherapy and immunoengineering. Bioact Mater 2025; 48:529-530. [PMID: 40104023 PMCID: PMC11919249 DOI: 10.1016/j.bioactmat.2025.01.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Affiliation(s)
- Gloria B Kim
- Department of Physiology and Biomedical Engineering, Mayo Clinic Arizona, Scottsdale, AZ, USA
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Philipp C Rommel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
2
|
Liu C, Cheng C, Cheng K, Gao AS, Li Q, Atala A, Zhang Y. Precision exosome engineering for enhanced wound healing and scar revision. J Transl Med 2025; 23:578. [PMID: 40410904 PMCID: PMC12103044 DOI: 10.1186/s12967-025-06578-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
The dysfunction of wound-healing processes can result in chronic non-healing wounds and pathological scar formation. Current treatment options often fall short, necessitating innovative approaches. Exosomes, extracellular vesicles secreted by various cells, have emerged as promising therapeutic agents serving as an intercellular communication system. By engineering exosomes, their cargo and surface properties can be tailored to enhance therapeutic efficacy and specificity. Engineered exosomes (eExo) are emerging as a favorable tool for treating non-healing wounds and pathological scars. In this review, we delve into the underlying mechanisms of non-healing wounds and pathological scars, outline the current state of engineering strategies, and explore the clinical potential of eExo based on preclinical and clinical studies. In addition, we address the current challenges and future research directions, including standardization, safety and efficacy assessments, and potential immune responses. In conclusion, eExo hold great promise as a novel therapeutic approach for non-healing wounds and non-healing wounds and pathological scars. Further research and clinical trials are warranted to translate preclinical findings into effective clinical treatments.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Chen Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Kun Cheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, 64108-2718, USA
| | - Allen S Gao
- Department of Urologic Surgery, School of Medicine, University of California, Davis Sacramento, CA, 95817, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Anthony Atala
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27151, USA
| | - Yuanyuan Zhang
- Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27151, USA.
| |
Collapse
|
3
|
Hassan M, Elzallat M, Mohammed DM, Balata M, El-Maadawy WH. Exploiting regulatory T cells (Tregs): Cutting-edge therapy for autoimmune diseases. Int Immunopharmacol 2025; 155:114624. [PMID: 40215774 DOI: 10.1016/j.intimp.2025.114624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/11/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025]
Abstract
Regulatory T cells (Tregs) are a specialized subset of suppressive T cells that are essential for maintaining self-tolerance, regulating effector T cells, managing microbial infections, preventing tumors, allergies, and autoimmune disorders, and facilitating allograft transplantation. Disruptions in Treg function or abundance contribute to an imbalance between pathogenic and protective immune cells in autoimmune diseases. Recently, one promising treatment strategy to restore immune balance involves the selective expansion or manipulation of Tregs using low-dose IL-2 therapy, adoptive Treg cell transfer, and chimeric antigen receptor (CAR)-Treg approaches. Tregs have been shown in an increasing number of research studies to prevent or even treat a variety of disorders, such as tumors, autoimmune and allergic diseases, transplant rejection, and graft-versus-host disease. A thorough comprehension of Treg function is anticipated to provide clear prospects for effective Treg immunotherapy in the treatment of a wide range of diseases. This review provides an overview of Tregs biology, including their functions, suppressive mechanisms, phenotypic markers, as well as their involvement in disease settings. Furthermore, we discuss the therapeutic potential of different Treg subpopulations and their translational applications in the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Marwa Hassan
- Immunology Department, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El Hadar, Imbaba, P.O. 30, Giza 12411, Egypt
| | - Mohamed Elzallat
- Immunology Department, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El Hadar, Imbaba, P.O. 30, Giza 12411, Egypt
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, 12622, Egypt.
| | - Mahmoud Balata
- University hospital bonn. Venusberg-Campus 1, 53127 Bonn, Germany.
| | - Walaa H El-Maadawy
- Pharmacology Department, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El-Hadar, Imbaba, P.O. 30, Giza, 12411, Egypt
| |
Collapse
|
4
|
Khalilzad MA, Mohammadi J, Amirsaadat S, Najafi S, Zare S, Nilforoushzadeh MA, Khalilzad M, Amirkhani MA, Peyrovan A, Khalili SFS, Farahani A, Zare S. Therapeutic potential of apoptotic vesicles in modulating inflammation, immune responses, and tissue regeneration. J Nanobiotechnology 2025; 23:260. [PMID: 40170079 PMCID: PMC11960034 DOI: 10.1186/s12951-025-03278-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
The process of apoptosis plays a crucial role in tissue homeostasis, immune system regulation, and organ formation. Apoptotic vesicles (ApoEVs) are involved in efferocytosis, the process by which phagocytes ingest dead cells. ApoEVs also have potential therapeutic applications in cancer treatment, ischemic diseases, and their anti-inflammatory properties make them incredibly versatile for medical applications. These vesicles can induce apoptosis in cancer cells, provide tumor antigens for cancer vaccines, and even serve as effective drug delivery systems. Moreover, they can target hypoxic cells, inhibit inflammatory cell death pathways, and promote tissue regeneration. Also, their potential in addressing inflammatory disorders such as gastrointestinal ailments, osteoarthritis, and diabetes is promising. Additionally, ApoEVs can polarize anti-inflammatory immune cells and suppress inflammatory immune responses which make them a viable option for addressing the unmet need for novel anti-inflammatory medications. Despite a wealth of reviews examining the applications of ApoEVs, very few have thoroughly investigated the mechanisms underlying their anti-inflammatory effects. This distinctive approach positions the current review as timely and immensely relevant, illuminating the intriguing ways these entities function beyond their established advantages.
Collapse
Affiliation(s)
- Mohammad Amin Khalilzad
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Mohammadi
- Department of Life Sciences Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran.
| | - Soumayeh Amirsaadat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Laserin Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Stem Cell and Regenerative Medicine Institute, Sharif University of Technology, Tehran, Iran.
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran.
| | - Mohammad Ali Nilforoushzadeh
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Skin Repair Research Center, Jordan Dermatology and Hair Transplantation Center, Tehran, Iran.
| | - Mitra Khalilzad
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Aysan Peyrovan
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Atefeh Farahani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Zare
- Laserin Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Mukerjee N, Bhattacharya A, Maitra S, Kaur M, Ganesan S, Mishra S, Ashraf A, Rizwan M, Kesari KK, Tabish TA, Thorat ND. Exosome isolation and characterization for advanced diagnostic and therapeutic applications. Mater Today Bio 2025; 31:101613. [PMID: 40161926 PMCID: PMC11950786 DOI: 10.1016/j.mtbio.2025.101613] [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: 09/22/2024] [Revised: 02/01/2025] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Advancements in exosome isolation technologies are pivotal for transforming personalized medicine and enhancing clinical diagnostics. Exosomes, small extracellular vesicles with diameters ranging between 30 and 150 nm, are secreted into bodily fluids by a variety of cells and play essential roles in intercellular communication. These vesicles facilitate the transfer of nucleic acids, lipids, and proteins, affecting a wide range of biological and pathological processes. Given their importance in disease diagnostics, therapy, and as biomarkers, there has been a surge in developing methods to isolate them from fluids such as urine, saliva, blood, and cerebrospinal fluid. While traditional isolation techniques like ultracentrifugation and polymer-based precipitation have been foundational, recent technological advances have introduced more precise methods like microfluidics and immunoaffinity capture. These newer methods enable high-throughput and specific exosome isolation by targeting surface markers, thus enhancing purity. However, challenges such as balancing purity with yield and the lack of standardized protocols across different laboratories persist, impacting the consistency of findings. By integrating advanced isolation techniques and discussing their implications in diagnostics and therapy, this review aims to catalyze further research and adoption of exosome-based technologies in medicine, marking a significant stride towards tailored healthcare solutions.
Collapse
Affiliation(s)
- Nobendu Mukerjee
- Centre for Infectious Diseases & Microbiology, School of Public Health Sciences and Technology, Malla Reddy Vishwavidyapeeth, Hyderabad 500 055, Telangana, India
| | - Arghya Bhattacharya
- Department of Pharmacology, Bengal School of Technology, West Bengal, Kolkata, 712102, India
| | - Swastika Maitra
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Shivang Mishra
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Ayash Ashraf
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, 140307, Punjab, India
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Department of Ophthalmology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Tanveer A. Tabish
- Radcliffe Department of Medicine, University of Oxford, OX3 7BN, United Kingdom
| | - Nanasaheb D. Thorat
- Department of Physics and Bernal Institute, University of Limerick, Castletroy, Limerick V94T9PX, Ireland
- Limerick Digital Cancer Research Centre (LDCRC) University of Limerick, Castletroy, Limerick, V94T9PX, Ireland
| |
Collapse
|
6
|
Hsu CY, Altalbawy FMA, Oghenemaro EF, Uthirapathy S, Chandra M, Nathiya D, Kaur P, Ravi Kumar M, Kadhim AJ, Kariem M. Exosomal lncRNAs in the Tumor Angiogenesis: As Therapeutic Targets in Cancer Treatment. Arch Pharm (Weinheim) 2025; 358:e202400940. [PMID: 40165644 DOI: 10.1002/ardp.202400940] [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: 12/12/2024] [Revised: 02/12/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
Exosomes, as mediators of intercellular communication, can be released from different types of cells and regulate the function of the target cell by transferring cargo, such as proteins, DNA, and RNA. Recent investigations have revealed a preponderance of long noncoding RNAs (lncRNAs), a subclass of noncoding RNAs, within exosomes, where they exhibit notable stability and are implicated in the development and progression of neoplastic processes, such as tumor angiogenesis. Angiogenesis, as a hallmark of cancer, provides diffusible nutrients and oxygen to the distant cells and guarantees tumorigenesis and metastasis. Exosomal lncRNAs, including MALAT1, OIP5-AS1, PART1, SNHG family, FAM225A, ATB, RAMP2-AS1, UCA1, TRPM2-AS, FGD5-AS1, and LINC0016, could modulate tumor angiogenesis by activating signaling cascades and mediators within the target cells, such as microRNAs (miRNAs). Regulation of tumor angiogenesis through modulation of exosomal lncRNAs could be a reliable strategy for cancer therapy. In this review, we discuss the characteristics and biogenesis of exosomes and lncRNAs and how exosomal lncRNAs are involved in various processes of tumorigenesis. Our primary focus is on exosomal lncRNAs, their impact on tumor angiogenesis, and their potential as novel diagnostic markers and therapeutic targets for various cancers.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, USA
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, Egypt
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
7
|
Alfawaz Altamimi AS, Arockia Babu M, Afzal M, Bishoyi AK, Roopashree R, Saini S, Sharma RSK, Pathak PK, Chauhan AS, Goyal K, Ali H, Khan NH, Balaraman AK. Exosomes derived from natural killer cells: transforming immunotherapy for aggressive breast cancer. Med Oncol 2025; 42:114. [PMID: 40100465 DOI: 10.1007/s12032-025-02647-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
Natural killer cell-derived exosomes (NK-Exos) hold great promise as immune modulators and immunotherapeutics against cancer due to their intrinsically latent anti-tumor effects. They use these nanosized vesicles to deliver cytotoxic molecules, such as perforin, granzymes, and miRNAs, directly to cancer cells to kill them, avoiding immune suppression. NK-Exos has particular efficacy for treating aggressive breast cancer by modulating the TME to activate the immune response and suppress immunosuppressive factors. Bioengineering advances have extended the therapeutic potential of NK-Exos, which permits precise tumor cell targeting and efficient delivery of therapeutic payloads, including small RNAs and chemotherapeutic agents. In engineered NK-Exos, sensitization of cancer cells to apoptosis, reduction of tumor growth, and resistance to drugs have been demonstrated to be highly effective. When combined, NK-Exos synergizes with radiotherapy, chemotherapy, or checkpoint inhibitors, enhancing therapeutic efficacy, and minimizing systemic toxicity. This review emphasizes the critical role of NK-Exos in breast cancer treatment, their integration into combination therapies, and the need for further research to overcome existing limitations and fully realize their clinical potential.
Collapse
Affiliation(s)
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ashok Kumar Bishoyi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - R S K Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Division of Research and Innovation, Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Haider Ali
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, 63000, Cyberjaya, Selangor, Malaysia.
| |
Collapse
|
8
|
Zhou T, Fang G, Wang Z, Qiao Z, Nie N, Fu B, Tseng PH, Sun X, Chen YC. Digital Lasing Biochip for Tumor-Derived Exosome Analysis. Anal Chem 2025; 97:5605-5611. [PMID: 40042136 PMCID: PMC11923948 DOI: 10.1021/acs.analchem.4c06172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/03/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025]
Abstract
Digital microfluidics represents an emerging versatile platform that offers numerous advantages in biomolecule detection. However, conventional probes often lack high-intensity and high-sensitivity signals, making it challenging for precise and automatic analysis. Recently, optical microresonators stand as a prominent high-sensitivity detection in the biological field. Here we introduce whispering gallery mode (WGM) microlasers into the microwell array, forming a digital lasing detection system. The lasing signal makes it highly sensitive, which amplifies the subtle changes via the strong interactions of light and matter. The microfluidic droplet technique further allowed microlasers with uniform laser thresholds and high-throughput fabrication. We utilized this tool for the analysis of exosomes derived from tumor spheroids. We believe that this digital optofluidic system could serve as a promising tool in diverse biomolecule assays and various biomedical applications.
Collapse
Affiliation(s)
- Tian Zhou
- School
of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Guocheng Fang
- School
of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Ziyihui Wang
- School
of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
| | - Zhen Qiao
- School
of Optical-Electrical and Computer Engineering, University of Shanghai for Science and Technology, No. 516 Jun Gong Road, Shanghai 200093, China
| | - Ningyuan Nie
- School
of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Bowen Fu
- School
of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Po-Hao Tseng
- School
of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Xiyu Sun
- School
of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Yu-Cheng Chen
- School
of Electrical and Electronics Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
9
|
Zhu L, Ahn BC. Natural Killer Cell-Derived Exosome Mimetics as Natural Nanocarriers for In Vitro Delivery of Chemotherapeutics to Thyroid Cancer Cells. Exp Oncol 2025; 46:358-367. [PMID: 39985349 DOI: 10.15407/exp-oncology.2024.04.358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Exosomes have become a potential field of nanotechnology for the treatment and identification of many disorders. However, the generation of exosomes is a difficult, time-consuming, and low-yielding procedure. At the same time, exosome mimetics (EM) resemble exosomes in their characteristics but have higher production yields. The aim of this study was to produce natural killer (NK) cell-derived EM (NKEM) loaded with sorafenib and test their killing ability against thyroid cancer cell lines. MATERIALS AND METHODS Sorafenib was loaded into NKEM by mixing sorafenib with NK cells during NKEM production (NKEM-S). Then, these two types of nanoparticles were characterized with nanoparticle tracking analysis (NTA) to measure their sizes. In addition, the cellular uptake and in vitro killing effect of NKEM-S on thyroid cancer cell lines were investigated using confocal laser microscopy and bioluminescence imaging (BLI) techniques. RESULTS The uptake of NKEM and NKEM-S by the thyroid cancer cells was observed. Moreover, BLI confirmed the killing and anti-proliferation effect of NKEM-S on two thyroid cancer cell lines. Especially important, the NKEM-S demonstrated a desirable killing effect even for anaplastic thyroid cancer (ATC) cells. CONCLUSION Sorafenib-loaded NKEM showed the ability to kill thyroid cancer cells in vitro, even against ATC. This provides a new opportunity for drug delivery systems and thyroid cancer treatment.
Collapse
Affiliation(s)
- L Zhu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, South Korea
| | - B-C Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
10
|
Tufail M, Jiang CH, Li N. Tumor dormancy and relapse: understanding the molecular mechanisms of cancer recurrence. Mil Med Res 2025; 12:7. [PMID: 39934876 PMCID: PMC11812268 DOI: 10.1186/s40779-025-00595-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/26/2025] [Indexed: 02/13/2025] Open
Abstract
Cancer recurrence, driven by the phenomenon of tumor dormancy, presents a formidable challenge in oncology. Dormant cancer cells have the ability to evade detection and treatment, leading to relapse. This review emphasizes the urgent need to comprehend tumor dormancy and its implications for cancer recurrence. Despite notable advancements, significant gaps remain in our understanding of the mechanisms underlying dormancy and the lack of reliable biomarkers for predicting relapse. This review provides a comprehensive analysis of the cellular, angiogenic, and immunological aspects of dormancy. It highlights the current therapeutic strategies targeting dormant cells, particularly combination therapies and immunotherapies, which hold promise in preventing relapse. By elucidating these mechanisms and proposing innovative research methodologies, this review aims to deepen our understanding of tumor dormancy, ultimately facilitating the development of more effective strategies for preventing cancer recurrence and improving patient outcomes.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, 410008, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Oral Precancerous Lesions, Central South University, Changsha, 410008, China.
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
11
|
Guo X, Piao H, Sui R. Exosomes in the Chemoresistance of Glioma: Key Point in Chemoresistance. J Cell Mol Med 2025; 29:e70401. [PMID: 39950738 PMCID: PMC11826829 DOI: 10.1111/jcmm.70401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/17/2025] Open
Abstract
Gliomas are the most ordinary primary virulent brain tumours and commonly used clinical treatments include tumour resection, radiation therapy and chemotherapy. Although significant progress has been made in recent years in progression-free survival (PFS) and overall survival (OS) for patients with high-grade gliomas, the prognosis for patients remains poor. Chemoresistance refers to the phenomenon of decreased sensitivity of tumour cells to drugs, resulting in reduced or ineffective drug efficacy, and is an important cause of failure of tumour chemotherapy. Exosomes, a type of extracellular vesicle, are secreted by cancer cells and various stromal cells in the tumour microenvironment (TME) and transfer their inclusions to cancer cells, increasing chemoresistance. Furthermore, depletion of exosomes reverses certain detrimental effects on tumour metabolism and restores sensitivity to chemotherapeutic agents. Here, we summarised the correlation between exosomes and resistance to chemotherapeutic agents in glioma patients, the mechanisms of action of exosomes involved in resistance and their clinical value. We aimed to afford new thoughts for research, clinical diagnosis and intervention in the mechanisms of chemoresistance in glioma patients.
Collapse
Affiliation(s)
- Xu Guo
- Department of NeurosurgeryCancer Hospital of Daflian University of Technology, Liaoning Cancer Hospital & InstituteShenyangLiaoningChina
| | - Haozhe Piao
- Department of NeurosurgeryCancer Hospital of Daflian University of Technology, Liaoning Cancer Hospital & InstituteShenyangLiaoningChina
| | - Rui Sui
- Department of NeurosurgeryCancer Hospital of Daflian University of Technology, Liaoning Cancer Hospital & InstituteShenyangLiaoningChina
| |
Collapse
|
12
|
Sethi M, Mohanty TK, Shah N, Bhakat M, Kumar N, Baithalu RK. Understanding the Crucial Role of Seminal Plasma Exosomes in Bull Fertility: A Review. Reprod Domest Anim 2024; 59:e70000. [PMID: 39714190 DOI: 10.1111/rda.70000] [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: 09/17/2024] [Revised: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024]
Abstract
Bull fertility is a multi-factorial trait and is affected by many factors, such as nutrition, genetics, and epigenetics. Superior quality male germplasm with high genetic merit helps to improve the livestock production trait. To achieve the target of livestock production, the availability of superior male germplasm is a great concern. In developing countries, there is a gap between the highly fertile frozen semen doses produced and the highly fertile frozen semen doses required. Improving the quality of existing low-fertile semen from high genetic merit bulls seems to play a promising role in filling this gap. Seminal exosomes are extracellular vesicles secreted by the epithelial cells of the testis, epididymis, and accessory sex glands such as the prostate gland. They contain a cargo of bioactive molecules such as proteins, nucleic acids and various metabolites. These molecules are transferred to the spermatozoa during its maturation and help in sperm capacitation, maturation, acrosome reaction, and fertilisation. Studies reveal that seminal exosomes help to improve the sperm functionality of low-quality sperm. Identification of the molecular profile of exosomes of bulls with proven fertility and their addition in an extender containing low-fertile semen may help to ameliorate the sperm quality of low-fertile semen, which may eventually aid in generating quantities of highly fertile ejaculates.
Collapse
Affiliation(s)
- Manisha Sethi
- Animal Reproduction, Gynaecology and Obstetrics, Artificial Breeding Research Centre (ABRC), ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Tushar Kumar Mohanty
- Animal Reproduction, Gynaecology and Obstetrics, Artificial Breeding Research Centre (ABRC), ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Nadeem Shah
- Sardar Vallabhbhai Patel University of Agriculture & Technology, Meerut, Uttar Pradesh, India
| | - Mukesh Bhakat
- Central Institute for Research on Goats, Makhdum, Uttar Pradesh, India
| | - Nishant Kumar
- Animal Reproduction, Gynaecology and Obstetrics, Artificial Breeding Research Centre (ABRC), ICAR-National Dairy Research Institute, Karnal, Haryana, India
| | - Rubina Kumari Baithalu
- Animal Reproduction, Gynaecology and Obstetrics, Artificial Breeding Research Centre (ABRC), ICAR-National Dairy Research Institute, Karnal, Haryana, India
| |
Collapse
|
13
|
Zabeti Touchaei A, Norollahi SE, Najafizadeh A, Babaei K, Bakhshalipour E, Vahidi S, Samadani AA. Therapeutic combinations of exosomes alongside cancer stem cells (CSCs) and of CSC-derived exosomes (CSCEXs) in cancer therapy. Cancer Cell Int 2024; 24:334. [PMID: 39369258 PMCID: PMC11453077 DOI: 10.1186/s12935-024-03514-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/22/2024] [Indexed: 10/07/2024] Open
Abstract
Exosomes which are membrane vesicles released by cells have gained significant interest in the field of cancer therapy as a novel means of intercellular communication. Their role in immune activation and their pathophysiological functions in cancer therapy have been recognized. Exosomes carry diverse bioactive components including proteins, mRNA, microRNAs, and bioactive lipids. These molecules have therapeutic potential in promoting tissue regeneration, supporting stem cell activity, preventing cell death, modulating immune responses, and promoting the growth of new blood vessels. However, the precise roles of exosomes derived from mesenchymal stem cells (MSCs) in the treatment of various cancers are still not fully understood. Consequently, cancer stem cells (CSCs) can self-renew and differentiate into various cell types. Understanding the mechanisms that sustain their persistence is crucial for developing effective therapies. Exosomes have recently gained interest as vehicles for intercellular communication between CSCs and non-CSCs, influencing cancer progression and the microenvironment. Research is ongoing on the utilization of exosomes derived from cancer stem cells (CSC-Exosome) for cancer treatment. The composition of extracellular vesicles is influenced by the specific type and condition of the cells from which they are secreted. Circulating exosomes contain stable RNA molecules such as mRNAs, microRNAs, and long non-coding RNAs (lncRNAs). In this review, we will explore the significance of exosomes and their diverse cellular combinations in the context of cancer therapy.
Collapse
Affiliation(s)
| | - Seyedeh Elham Norollahi
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Najafizadeh
- School of Paramedicine Sciences, Guilan University of Medical Sciences, Langarud, Iran
| | - Kosar Babaei
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Elahe Bakhshalipour
- School of Paramedicine Sciences, Guilan University of Medical Sciences, Langarud, Iran
| | - Sogand Vahidi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Ali Akbar Samadani
- Neuroscience Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
14
|
Liu Y, Zhang J, Peng X, Yan S. Deciphering the Evolution of Inertial Migration in Serpentine Channels. Anal Chem 2024; 96:14306-14314. [PMID: 39165174 PMCID: PMC11375625 DOI: 10.1021/acs.analchem.4c03474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Serpentine channels coupling inertial and secondary flows enable effective particle focusing and separation, showing great potential in clinical diagnostics and drug screening. However, the nonsteady secondary flows in the serpentine channel make the evolution of inertial migration unclear, hindering the development and application of the serpentine channel. Herein, to refine the inertial migration mechanism, we established a model with varying curvature ratios to study the effect of secondary flow on particle migration in the serpentine channel. This method used direct numerical simulation (DNS) to calculate inertial lift, mapped the inertial lift to cross sections of the serpentine channel, and deciphered the inertial migration by using the Lagrangian particle tracking (LPT) method. The inertial migration of microparticles is experimentally investigated to validate the established numerical model. The results indicate that particle migration in serpentine channels follows a two-stage migration. An increase in secondary flow accelerates the second stage of the migration process while slowing the first stage process. Subsequently, we investigated the effects of different parameters, including Reynolds number, aspect ratio, and blockage ratio, on the equilibrium positions of particles, providing guidelines for the high-resolution separation of particles. Taking flow resistance into account, the dimensionless study makes the separation of arbitrary-sized particles possible. This work reveals the migration mechanism in serpentine channels, paving the way for the inertial separation of the particles.
Collapse
Affiliation(s)
- Yong Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jun Zhang
- Queensland Micro- and Nanotechnology Centre, Nathan, Queensland 4111, Australia
- School of Engineering and Built Environment, Griffith University, Nathan, Queensland 4111, Australia
| | - Xiaobo Peng
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen 518060, China
| | - Sheng Yan
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
15
|
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
|
16
|
Miura A, Manabe Y, Suzuki KGN, Shomura H, Okamura S, Shirakawa A, Yano K, Miyake S, Mayusumi K, Lin CC, Morimoto K, Ishitobi J, Nakase I, Arai K, Kobayashi S, Ishikawa U, Kanoh H, Miyoshi E, Yamaji T, Kabayama K, Fukase K. De Novo Glycan Display on Cell Surfaces Using HaloTag: Visualizing the Effect of the Galectin Lattice on the Lateral Diffusion and Extracellular Vesicle Loading of Glycosylated Membrane Proteins. J Am Chem Soc 2024; 146:22193-22207. [PMID: 38963258 DOI: 10.1021/jacs.4c02040] [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: 07/05/2024]
Abstract
Glycans cover the cell surface to form the glycocalyx, which governs a myriad of biological phenomena. However, understanding and regulating glycan functions is extremely challenging due to the large number of heterogeneous glycans that engage in intricate interaction networks with diverse biomolecules. Glycocalyx-editing techniques offer potent tools to probe their functions. In this study, we devised a HaloTag-based technique for glycan manipulation, which enables the introduction of chemically synthesized glycans onto a specific protein (protein of interest, POI) and concurrently incorporates fluorescent units to attach homogeneous, well-defined glycans to the fluorescence-labeled POIs. Leveraging this HaloTag-based glycan-display system, we investigated the influence of the interactions between Gal-3 and various N-glycans on protein dynamics. Our analyses revealed that glycosylation modulates the lateral diffusion of the membrane proteins in a structure-dependent manner through interaction with Gal-3, particularly in the context of the Gal-3-induced formation of the glycan network (galectin lattice). Furthermore, N-glycan attachment was also revealed to have a significant impact on the extracellular vesicle-loading of membrane proteins. Notably, our POI-specific glycan introduction does not disrupt intact glycan structures, thereby enabling a functional analysis of glycans in the presence of native glycan networks. This approach complements conventional glycan-editing methods and provides a means for uncovering the molecular underpinnings of glycan functions on the cell surface.
Collapse
Affiliation(s)
- Ayane Miura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Kenichi G N Suzuki
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu 501-1193, Japan
- National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | - Hiroki Shomura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Soichiro Okamura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Asuka Shirakawa
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Kumpei Yano
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Shuto Miyake
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Koki Mayusumi
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing-Hua University, Hsinchu 30013, Taiwan
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Kenta Morimoto
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Jojiro Ishitobi
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Ikuhiko Nakase
- Department of Biological Chemistry, Graduate School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Biological Chemistry, School of Science, Osaka Metropolitan University, 1-1, Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Kenta Arai
- Advanced ICT Research Institute, National Institute of Information and Communications Technology, 588-2 Iwaoka, Iwaoka-cho, Nishi-ku, Kobe, Hyogo 651-2492, Japan
| | - Shouhei Kobayashi
- Advanced ICT Research Institute, National Institute of Information and Communications Technology, 588-2 Iwaoka, Iwaoka-cho, Nishi-ku, Kobe, Hyogo 651-2492, Japan
| | - Ushio Ishikawa
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsushima, Aoba Ward, Sendai, Miyagi 981-8558, Japan
| | - Hirotaka Kanoh
- Division of Glycopathology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsushima, Aoba Ward, Sendai, Miyagi 981-8558, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Interdisciplinary Research Center for Radiation Sciences, Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Center for Advanced Modalities and DDS, Osaka University, 1-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
17
|
Liang L, Peng W, Qin A, Zhang J, Chen R, Zhou D, Zhang X, Zhou N, Yu XY, Zhang L. Intracellularly Synthesized Artificial Exosome Treats Acute Lung Injury. ACS NANO 2024; 18:21009-21023. [PMID: 39087239 DOI: 10.1021/acsnano.4c01900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), induce high morbidity and mortality rates, which challenge the present approaches for the treatment of ALI/ARDS. The clinically used photosensitizer verteporfin (VER) exhibits great potential in the treatment of acute lung injury and acute respiratory distress syndrome (ALI/ARDS) by regulating macrophage polarization and reducing inflammation. Nevertheless, its hydrophobic characteristics, nonspecificity, and constrained bioavailability hinder its therapeutic efficacy. In this work, we developed a type of VER-cored artificial exosome (EVM), which was produced by using mesoporous silica nanoparticles (MSNs) to load VER, followed by the exocytosis of internalized VER-MSNs from mouse bone marrow-derived mesenchymal stem cells (mBMSCs) without further modification. Both in vitro and in vivo assessments confirmed the powerful anti-inflammation induced by EVM. EVM also showed significant higher accumulation to inflammatory lungs compared with healthy ones, which was beneficial to the treatment of ALI/ARDS. EVM improved pulmonary function, attenuated lung injury, and reduced mortality in ALI mice with high levels of biocompatibility, exhibiting a 5-fold higher survival rate than the control. This type of artificial exosome emitted near-infrared light in the presence of laser activation, which endowed EVM with trackable ability both in vitro and in vivo. Our work developed a type of clinically used photosensitizer-loaded artificial exosome with membrane integrity and traceability. To the best of our knowledge, this kind of intracellularly synthesized artificial exosome was developed and showed great potential in ALI/ARDS therapy.
Collapse
Affiliation(s)
- Lu Liang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA, and the State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Weijie Peng
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA, and the State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Aiping Qin
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA, and the State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Jiandong Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA, and the State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Rongqi Chen
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA, and the State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Dazhi Zhou
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA, and the State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Xin Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Na Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Xi-Yong Yu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA, and the State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Lingmin Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA, and the State Key Laboratory of Respiratory Disease, The School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P.R. China
| |
Collapse
|
18
|
Chen Y, Tang S, Cai F, Wan Y. Strategies for Small Extracellular Vesicle-Based Cancer Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0421. [PMID: 39040921 PMCID: PMC11260559 DOI: 10.34133/research.0421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed vesicles released by cells. EVs encapsulate proteins and nucleic acids of their parental cell and efficiently deliver the cargo to recipient cells. These vesicles act as mediators of intercellular communication and thus play a crucial role in various physiological and pathological processes. Moreover, EVs hold promise for clinical use. They have been explored as drug delivery vehicles, therapeutic agents, and targets for disease diagnosis. In the landscape of cancer research, while strides have been made in EV-focused cancer physiopathology, liquid biopsy, and drug delivery, the exploration of EVs as immunotherapeutic agents may not have seen substantial progress to date. Despite promising findings reported in cell and animal studies, the clinical translation of EV-based cancer immunotherapeutics encounters challenges. Here, we review the existing strategies used in EV-based cancer immunotherapy, aiming to propel the development of this emerging yet crucial field.
Collapse
Affiliation(s)
- Yundi Chen
- Department of Breast Surgery, Tongji Hospital, School of Medicine,
Tongji University, Shanghai, China
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering,
Binghamton University, Binghamton, NY, USA
| | - Shasha Tang
- Department of Breast Surgery, Tongji Hospital, School of Medicine,
Tongji University, Shanghai, China
| | - Fengfeng Cai
- Department of Breast Surgery, Tongji Hospital, School of Medicine,
Tongji University, Shanghai, China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering,
Binghamton University, Binghamton, NY, USA
| |
Collapse
|
19
|
Wu H, Chen Q, Wang S, Yang C, Xu L, Xiao H, Xie T, Pan Q. Serum exosomes lncRNAs: TCONS_I2_00013502 and ENST00000363624 are new diagnostic markers for rheumatoid arthritis. Front Immunol 2024; 15:1419683. [PMID: 39044812 PMCID: PMC11263027 DOI: 10.3389/fimmu.2024.1419683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
The lack of diagnostic markers limits the window of effectiveness for rheumatoid arthritis (RA) therapies. Here, we isolated exosomes of serum samples from four distinct groups RA patients, according to disease activity and with/without medication. Then, total RNA of exosomes was extracted for whole-transcriptome sequencing. Focusing on lncRNA sequencing, gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analyses were performed. We found that the number of upregulated lncRNAs were significantly higher than that of downregulated lncRNAs in each four RA groups. And most importantly, we identified two specific lncRNAs from differentially expressed lncRNAs, TCONS_I2_00013502 (up-regulated) and ENST00000363624 (down-regulated) in RA. Receiver Operating Characteristic (ROC) curve analysis showed that the two lncRNAs were promising biomarkers for RA diagnosis. These findings highlight lncRNAs of the serum exosome are important biomarkers and provide application potential for diagnosis of RA.
Collapse
Affiliation(s)
- Han Wu
- Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Clinical Laboratory, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qiuhua Chen
- Department of Immunology and Rheumatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Sijie Wang
- Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Chunlong Yang
- Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Li Xu
- Clinical Laboratory, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Haiyan Xiao
- Department of Cellular Biology and Anatomy, James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Tong Xie
- Department of Immunology and Rheumatology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qingjun Pan
- Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
20
|
Javdani-Mallak A, Salahshoori I. Environmental pollutants and exosomes: A new paradigm in environmental health and disease. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 925:171774. [PMID: 38508246 DOI: 10.1016/j.scitotenv.2024.171774] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/16/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
This study investigates the intricate interplay between environmental pollutants and exosomes, shedding light on a novel paradigm in environmental health and disease. Cellular stress, induced by environmental toxicants or disease, significantly impacts the production and composition of exosomes, crucial mediators of intercellular communication. The heat shock response (HSR) and unfolded protein response (UPR) pathways, activated during cellular stress, profoundly influence exosome generation, cargo sorting, and function, shaping intercellular communication and stress responses. Environmental pollutants, particularly lipophilic ones, directly interact with exosome lipid bilayers, potentially affecting membrane stability, release, and cellular uptake. The study reveals that exposure to environmental contaminants induces significant changes in exosomal proteins, miRNAs, and lipids, impacting cellular function and health. Understanding the impact of environmental pollutants on exosomal cargo holds promise for biomarkers of exposure, enabling non-invasive sample collection and real-time insights into ongoing cellular responses. This research explores the potential of exosomal biomarkers for early detection of health effects, assessing treatment efficacy, and population-wide screening. Overcoming challenges requires advanced isolation techniques, standardized protocols, and machine learning for data analysis. Integration with omics technologies enhances comprehensive molecular analysis, offering a holistic understanding of the complex regulatory network influenced by environmental pollutants. The study underscores the capability of exosomes in circulation as promising biomarkers for assessing environmental exposure and systemic health effects, contributing to advancements in environmental health research and disease prevention.
Collapse
Affiliation(s)
- Afsaneh Javdani-Mallak
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Iman Salahshoori
- Department of Polymer Processing, Iran Polymer and Petrochemical Institute, Tehran, Iran; Department of Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
21
|
Moghaddam MM, Behzadi E, Sedighian H, Goleij Z, Kachuei R, Heiat M, Fooladi AAI. Regulation of immune responses to infection through interaction between stem cell-derived exosomes and toll-like receptors mediated by microRNA cargoes. Front Cell Infect Microbiol 2024; 14:1384420. [PMID: 38756232 PMCID: PMC11096519 DOI: 10.3389/fcimb.2024.1384420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
Infectious diseases are among the factors that account for a significant proportion of disease-related deaths worldwide. The primary treatment approach to combat microbial infections is the use of antibiotics. However, the widespread use of these drugs over the past two decades has led to the emergence of resistant microbial species, making the control of microbial infections a serious challenge. One of the most important solutions in the field of combating infectious diseases is the regulation of the host's defense system. Toll-like receptors (TLRs) play a crucial role in the first primary defense against pathogens by identifying harmful endogenous molecules released from dying cells and damaged tissues as well as invading microbial agents. Therefore, they play an important role in communicating and regulating innate and adaptive immunity. Of course, excessive activation of TLRs can lead to disruption of immune homeostasis and increase the risk of inflammatory reactions. Targeting TLR signaling pathways has emerged as a new therapeutic approach for infectious diseases based on host-directed therapy (HDT). In recent years, stem cell-derived exosomes have received significant attention as factors regulating the immune system. The regulation effects of exosomes on the immune system are based on the HDT strategy, which is due to their cargoes. In general, the mechanism of action of stem cell-derived exosomes in HDT is by regulating and modulating immunity, promoting tissue regeneration, and reducing host toxicity. One of their most important cargoes is microRNAs, which have been shown to play a significant role in regulating immunity through TLRs. This review investigates the therapeutic properties of stem cell-derived exosomes in combating infections through the interaction between exosomal microRNAs and Toll-like receptors.
Collapse
Affiliation(s)
- Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- The Academy of Medical Sciences of I.R. Iran, Tehran, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zoleikha Goleij
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Albarrán V, San Román M, Pozas J, Chamorro J, Rosero DI, Guerrero P, Calvo JC, González C, García de Quevedo C, Pérez de Aguado P, Moreno J, Cortés A, Soria A. Adoptive T cell therapy for solid tumors: current landscape and future challenges. Front Immunol 2024; 15:1352805. [PMID: 38550594 PMCID: PMC10972864 DOI: 10.3389/fimmu.2024.1352805] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
Adoptive cell therapy (ACT) comprises different strategies to enhance the activity of T lymphocytes and other effector cells that orchestrate the antitumor immune response, including chimeric antigen receptor (CAR) T-cell therapy, T-cell receptor (TCR) gene-modified T cells, and therapy with tumor-infiltrating lymphocytes (TILs). The outstanding results of CAR-T cells in some hematologic malignancies have launched the investigation of ACT in patients with refractory solid malignancies. However, certain characteristics of solid tumors, such as their antigenic heterogeneity and immunosuppressive microenvironment, hamper the efficacy of antigen-targeted treatments. Other ACT modalities, such as TIL therapy, have emerged as promising new strategies. TIL therapy has shown safety and promising activity in certain immunogenic cancers, mainly advanced melanoma, with an exciting rationale for its combination with immune checkpoint inhibitors. However, the implementation of TIL therapy in clinical practice is hindered by several biological, logistic, and economic challenges. In this review, we aim to summarize the current knowledge, available clinical results, and potential areas of future research regarding the use of T cell therapy in patients with solid tumors.
Collapse
Affiliation(s)
- Víctor Albarrán
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - María San Román
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - Javier Pozas
- Department of Medical Oncology, The Royal Marsden Hospital, London, United Kingdom
| | - Jesús Chamorro
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - Diana Isabel Rosero
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - Patricia Guerrero
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - Juan Carlos Calvo
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - Carlos González
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | | | | | - Jaime Moreno
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - Alfonso Cortés
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| | - Ainara Soria
- Department of Medical Oncology, Ramon y Cajal University Hospital, Madrid, Spain
| |
Collapse
|
23
|
Zhang C, Cao Q, Li Y, Lu J, Xiong S, Yue Y. Exosome co-delivery of a STING agonist augments immunogenicity elicited by CVB3 VP1 vaccine via promoting antigen cross-presentation of CD8 + DCs. Int J Biol Macromol 2024; 261:129518. [PMID: 38244740 DOI: 10.1016/j.ijbiomac.2024.129518] [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: 11/01/2023] [Revised: 01/09/2024] [Accepted: 01/13/2024] [Indexed: 01/22/2024]
Abstract
The induction of a robust CD8+ T cell response is critical for the success of an antiviral vaccine. In this study, we incorporated a STING agonist (SA) 2'3'-cGAMP into a previously developed exosome-based CVB3 viral myocarditis vaccine (Exo-VP1) to enhance its ability to induce CD8+ T cell responses and immunoprotection. Our results showed that compared to free SA adjuvant, exosome-mediated co-delivery (ExoSA-VP1) significantly enhanced SA uptake by dendritic cells (DCs) and more potently stimulated DC maturation. Immunization of mice showed that the ExoSA-VP1 vaccine-induced higher levels of CVB3-specific T cell proliferation and cytotoxicity, significantly increased the percentage of IFN-γ+CD8+ rather than CD4+ T cells, effectively reduced cardiac viral loads, attenuated myocarditis and improved survival in mice compared to the previous Exo-VP1 vaccine. Further investigation showed that ExoSA-VP1 significantly increased both the percentage and antigen cross-presentation capacity of splenic CD8+ DCs. Depletion of these CD8+ DCs by cytochrome C administration nearly abolished the advantage of ExoSA-VP1 in dominantly inducing IFN-γ+CD8+ cytotoxic T lymphocyte (CTL) production in immunized mice. Taken together, our results demonstrated the potential of ExoSA-VP1 as a promising candidate for anti-CVB3 vaccines and provide insights into immune-enhancing strategies aiming at augmenting antigen cross-presentation by DCs and enhancing potent CTL responses.
Collapse
Affiliation(s)
- Changwei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Qinghui Cao
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Yuanyu Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Juan Lu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China.
| | - Yan Yue
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
24
|
Wang Y, Dong H, Qu H, Cheng W, Chen H, Gu Y, Jiang H, Xue X, Hu R. Biomimetic Lung-Targeting Nanoparticles with Antioxidative and Nrf2 Activating Properties for Treating Ischemia/Reperfusion-Induced Acute Lung Injury. NANO LETTERS 2024; 24:2131-2141. [PMID: 38227823 DOI: 10.1021/acs.nanolett.3c03671] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Ischemia/reperfusion (IR)-induced acute lung injury (ALI) has a high mortality rate. Reactive oxygen species (ROS) play a crucial role in causing cellular damage and death in IR-induced ALI. In this work, we developed a biomimetic lung-targeting nanoparticle (PC@MB) as an antioxidative lung protector for treating IR-induced ALI. PC@MBs showed excellent ROS scavenging and Nrf2 activation properties, along with a lung-targeting function through autologous cell membrane coating. The PC@MBs exhibited an impressive antioxidative and pulmonary protective role via redox homeostasis recovery through Nrf2 and heme oxygenase-1 activation. PC@MBs could maintain cell viability by effectively scavenging the intracellular ROS and restoring the redox equilibrium in the lesion. In the IR mouse model, the PC@MBs preferentially accumulated in the lung and distinctly repaired the pneumonic damage. Our strategy has the potential to offer a promising therapeutic paradigm for treating IR-induced ALI through the incorporation of different therapeutic mechanisms.
Collapse
Affiliation(s)
- Yanjun Wang
- Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- School of Pharmaceutical Sciences, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hui Dong
- Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Haijing Qu
- School of Pharmaceutical Sciences, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Cheng
- School of Pharmaceutical Sciences, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Chen
- School of Pharmaceutical Sciences, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yunfan Gu
- Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Hong Jiang
- Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiangdong Xue
- School of Pharmaceutical Sciences, Shanghai Frontiers Science Center for Drug Target Identification and Drug Delivery, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Rong Hu
- Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
25
|
Liu Y, Wu H, Sang Y, Chong W, Shang L, Li L. Research progress of exosomes in the angiogenesis of digestive system tumour. Discov Oncol 2024; 15:33. [PMID: 38341827 PMCID: PMC10859358 DOI: 10.1007/s12672-024-00879-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/30/2024] [Indexed: 02/13/2024] Open
Abstract
Malignant tumours of the digestive system cover a wide range of diseases that affect the health of people to a large extent. Angiogenesis is indispensable in the development, and metastasis of tumours, mainly in two ways: occupation or formation. Vessels can provide nutrients, oxygen, and growth factors for tumours to encourage growth and metastasis, so cancer progression depends on simultaneous angiogenesis. Recently, exosomes have been proven to participate in the angiogenesis of tumours. They influence angiogenesis by binding to tyrosine kinase receptors (VEGFR)-1, VEGFR-2, and VEGFR-3 with different affinities, regulating Yap-VEGF pathway, Akt pathway or other signaling pathway. Additionally, exosomes are potential therapeutic vectors that can deliver many types of cargoes to different cells. In this review, we summarize the roles of exosomes in the angiogenesis of digestive system tumours and highlight the clinical application prospects, directly used as targers or delivery vehicles, in antiangiogenic therapy.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- Department of Gastrointestinal Surgery, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital, Jinan, 250021, China
- Department of Gastrointestinal Surgery, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Hao Wu
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yaodong Sang
- Department of Gastrointestinal Surgery, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital, Jinan, 250021, China
- Department of Gastrointestinal Surgery, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital, Jinan, 250021, China.
- Department of Gastrointestinal Surgery, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China.
| | - Liang Shang
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital, Jinan, 250021, China.
- Department of Gastrointestinal Surgery, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China.
| | - Leping Li
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Key Laboratory of Engineering of Shandong Province, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital, Jinan, 250021, China.
- Department of Gastrointestinal Surgery, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, China.
| |
Collapse
|
26
|
Yue Y, Liu Y, Lin Y, Guo F, Cai K, Chen S, Zhang W, Tang S. A carboxymethyl chitosan/oxidized hyaluronic acid composite hydrogel dressing loading with stem cell exosome for chronic inflammation wounds healing. Int J Biol Macromol 2024; 257:128534. [PMID: 38048924 DOI: 10.1016/j.ijbiomac.2023.128534] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/14/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
Stem cell exosomes (Exo) play an important role in the transformation of macrophages, but the rapid clearance of Exo in vivo limits their therapeutic effects for chronic inflammation wounds healing. Here, stem cell Exo was isolated and introduced to a composite hydrogel including carboxymethyl chitosan (CMCS) and oxidized hyaluronic acid (OHA) through chemical cross-linking, which formed an Exo-loaded (CMCS/OHA/Exo) hydrogel. The CMCS/OHA/Exo hydrogel exhibited a function of Exo sustained release and an Exo protection within 6 days. This CMCS/OHA/Exo hydrogel was much better than CMCS/OHA hydrogel or Exo solution in macrophage cell phagocytosis, proliferation and migration in vitro, especially, played an obviously positive role in the transformation of macrophages compared with the reference groups. For the treatment of the chronic inflammation wounds in vivo, the CMCS/OHA/Exo hydrogel had the best results at wound heal rate and inhibiting the secretion of inflammatory factors, and it was far superior to reference groups in wound re-epithelization and collagen production. CMCS/OHA/Exo hydrogels can promote Exo release based on hydrogel degradation to regulate macrophages transformation and accelerate chronic wound healing. The study offers a method for preparing Exo-loaded hydrogels that effectively promote the transformation of macrophages and accelerate chronic inflammatory wound healing.
Collapse
Affiliation(s)
- Yan Yue
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Yang Liu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China.
| | - Yukai Lin
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Fengbiao Guo
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Kun Cai
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Shengqin Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Department of Biology, College of Science, Shantou University, Shantou, Guangdong 515063, PR China
| | - Wancong Zhang
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou University, Shantou, Guangdong 515063, PR China
| | - Shijie Tang
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou University, Shantou, Guangdong 515063, PR China
| |
Collapse
|