1
|
Li D, Qu Y, Wang B, Zhang H, Qin L. Spatio-temporal expression of Sox2 + progenitor cells regulates the regeneration of rat submandibular gland. Arch Oral Biol 2024; 168:106080. [PMID: 39217919 DOI: 10.1016/j.archoralbio.2024.106080] [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: 02/28/2024] [Revised: 04/12/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE Sox2 plays crucial roles in tissues homeostasis and regeneration. However, there are lack of a comprehensive examination of Sox2 expression and its functional role in submandibular gland regeneration. Therefore, we aimed to elucidate the impact of Sox2 on submandibular gland regeneration. MATERIALS AND METHODS A Sprague-Dawley rat submandibular gland duct ligation/de-ligation regeneration model was conducted in this study. Sox2-shRNA vectors were retro-ductally administered into the submandibular gland to establish a stable Sox2 knockdown model. Conventional histopathological and molecular biological methods were used to investigate phenotypic changes. RESULTS The submandibular gland normalized completely 28 days after ligature removal (following 7 days of duct ligation). AQP5 expression gradually increased after ligation removal until returning to normal levels. In submandibular gland regeneration, Sox2 re-expressed and co-expressed with AQP5+ acinar cells, and Sox2 expression peaked on day 14, recovered to normal on day 28, reproducing the developmental pattern. Sox2 knockdown hindered gland regeneration and induced irreversible fibrosis. The AQP5 expression was significantly lower than the contemporaneous solely ligated group, while the blue collagen deposition and the Vimentin expression increased prominently. The expression of CD68, IL-1β, TNF-α and IL-17A increased significantly, and epithelial cells in the Sox2 knockdown group expressed higher levels of IL-17A. CONCLUSIONS These findings highlight Sox2 as a crucial regulator of the acinar cell lineage. Sox2+ progenitor cells are pivotal for acinar cell maintenance, which is indispensable for submandibular gland regeneration. Collectively, our findings may help develop targeted interventions for enhancing tissue repair and preventing irreversible fibrosis in salivary gland disorders.
Collapse
Affiliation(s)
- Dan Li
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, China
| | - Yi Qu
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, China
| | - Bin Wang
- Department of Head and Neck Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Workers' New Village No.3, Taiyuan, 030013, China
| | - Haoyang Zhang
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, China
| | - Lizheng Qin
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Tian Tan Xi Li No.4, Beijing 100050, China.
| |
Collapse
|
2
|
Zhang YX, Zhou Y, Xiong YY, Li YM. Beyond skin deep: Revealing the essence of iPS cell-generated skin organoids in regeneration. Burns 2024; 50:107194. [PMID: 39317530 DOI: 10.1016/j.burns.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/13/2024] [Accepted: 06/23/2024] [Indexed: 09/26/2024]
Abstract
Various methods have been used for in vivo and in vitro skin regeneration, including stem cell therapy, tissue engineering, 3D printing, and platelet-rich plasma (PRP) injection therapy. However, these approaches are rooted in the existing knowledge of skin structures, which overlook the normal physiological processes of skin development and fall short of replicating the skin's regenerative processes outside the body. This comprehensive review primarily focuses on skin organoids derived from human pluripotent stem cells, which have the capacity to regenerate human skin tissue by restoring the embryonic skin structure, thus offering a novel avenue for producing in vitro skin substitutes. Furthermore, they contribute to the repair of damaged skin lesions in patients with systemic sclerosis or severe burns. Particular emphasis will be placed on the origins, generations, and applications of skin organoids, especially in dermatology, and the challenges that must be addressed before clinical implementation.
Collapse
Affiliation(s)
- Yu-Xuan Zhang
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Yuan Zhou
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Yu-Yun Xiong
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.
| | - Yu-Mei Li
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
3
|
Grosu-Bularda A, Lita FF, Hodea FV, Bordeanu-Diaconescu EM, Cretu A, Dumitru CS, Cacior S, Marinescu BM, Lascar I, Hariga CS. Navigating the Complexities of Radiation Injuries: Therapeutic Principles and Reconstructive Strategies. J Pers Med 2024; 14:1100. [PMID: 39590592 PMCID: PMC11595796 DOI: 10.3390/jpm14111100] [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: 09/24/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Radiation injuries, particularly those resulting from therapeutic or accidental exposure, present complex challenges for medical management. These injuries can manifest localized skin damage or extend to deeper tissues, presenting as various clinical entities that require treatment strategies, ranging from conservative management to complex surgical interventions. Radiation treatment constitutes a fundamental component of neoplastic management, with nearly two out of three oncological instances undergoing it as an element of their therapeutic strategy. The therapeutic approach to radiation injury consists of expanding prophylactic measures while maintaining the efficacy of treatment, such as conservative treatment or local debridement followed by reconstruction. The armamentarium of reconstructive methods available for plastic surgeons, from secondary healing to free tissue transfer, can be successfully applied to radiation injuries. However, the unique pathophysiological changes induced by radiation necessitate a careful and specialized approach for their application, considering the altered tissue characteristics and healing dynamics. The therapeutic strategy is guided by both the severity and progression of the injury, with the primary aim of restoring functionality and aesthetic aspects while simultaneously minimizing the risk of complications. This paper explores the various conditions encompassed by the term "radiation injury," reviews both non-surgical and surgical therapeutic strategies for managing these injuries, and highlights the unique challenges associated with treating irradiated tissues within specific oncological contexts.
Collapse
Affiliation(s)
- Andreea Grosu-Bularda
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Flavia-Francesca Lita
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
- Clinical Department Plastic Surgery and Reconstructive Microsurgery, Central Military Emergency University Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Florin-Vlad Hodea
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Eliza-Maria Bordeanu-Diaconescu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Andrei Cretu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Catalina-Stefania Dumitru
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Stefan Cacior
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Bogdan-Mihai Marinescu
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinical Department Plastic Surgery and Reconstructive Microsurgery, Central Military Emergency University Hospital “Dr. Carol Davila”, 010825 Bucharest, Romania
| | - Ioan Lascar
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| | - Cristian-Sorin Hariga
- Department 11, Discipline Plastic and Reconstructive Surgery, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (I.L.); (C.-S.H.)
- Clinic of Plastic Surgery and Reconstructive Microsurgery, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
4
|
Lan J, Ren Y, Liu Y, Chen L, Liu J. A bibliometric analysis of radiation-induced brain injury: a research of the literature from 1998 to 2023. Discov Oncol 2024; 15:364. [PMID: 39172266 PMCID: PMC11341524 DOI: 10.1007/s12672-024-01223-6] [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: 04/02/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury (RIBI) is a debilitating sequela after cranial radiotherapy. Research on the topic of RIBI has gradually entered the public eye, with more innovations and applications of evidence-based research and biological mechanism research in the field of that. This was the first bibliometric analysis on RIBI, assessing brain injury related to radiation articles that were published during 1998-2023, to provide an emerging theoretical basis for the future development of RIBI. METHODS Literature were obtained from the Web of Science Core Collection (WOSCC) from its inception to December 31, 2023. The column of publications, author details, affiliated institutions and countries, publication year, and keywords were also recorded. RESULTS A total of 2543 journal articles were selected. The annual publications on RIBI fluctuated within a certain range. Journal of Neuro-oncology was the most published journal and Radiation Oncology was the most impactful one. LIMOLI CL was the most prolific author with 37 articles and shared the highest h-index with BARNETT GH. The top one country and institutions were the USA and the University of California System, respectively. Clusters analysis of co-keywords demonstrated that the temporal research trends in this field primarily focused on imaging examination and therapy for RIBI. CONCLUSION This study collects, visualizes, and analyzes the literature within the field of RIBI over the last 25 years to map the development process, research frontiers and hotspots, and cutting-edge directions in clinical practice and mechanisms related to RIBI.
Collapse
Affiliation(s)
- Jinxin Lan
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yifan Ren
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yuyang Liu
- Department of Neurosurgery, The 920th Hospital of Joint Logistics Support Force, Kunming, 650032, Yunnan, China
| | - Ling Chen
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China.
- Chinese PLA General Hospital, Chinese PLA Institute of Neurosurgery, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Jialin Liu
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China.
- Chinese PLA General Hospital, Chinese PLA Institute of Neurosurgery, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
5
|
Gupta K, Perkerson RB, Parsons TM, Angom R, Amerna D, Burgess JD, Ren Y, McLean PJ, Mukhopadhyay D, Vibhute P, Wszolek ZK, Zubair AC, Quiñones-Hinojosa A, Kanekiyo T. Secretome from iPSC-derived MSCs exerts proangiogenic and immunosuppressive effects to alleviate radiation-induced vascular endothelial cell damage. Stem Cell Res Ther 2024; 15:230. [PMID: 39075600 PMCID: PMC11287895 DOI: 10.1186/s13287-024-03847-5] [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: 03/06/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Radiation therapy is the standard of care for central nervous system tumours. Despite the success of radiation therapy in reducing tumour mass, irradiation (IR)-induced vasculopathies and neuroinflammation contribute to late-delayed complications, neurodegeneration, and premature ageing in long-term cancer survivors. Mesenchymal stromal cells (MSCs) are adult stem cells that facilitate tissue integrity, homeostasis, and repair. Here, we investigated the potential of the iPSC-derived MSC (iMSC) secretome in immunomodulation and vasculature repair in response to radiation injury utilizing human cell lines. METHODS We generated iPSC-derived iMSC lines and evaluated the potential of their conditioned media (iMSC CM) to treat IR-induced injuries in human monocytes (THP1) and brain vascular endothelial cells (hCMEC/D3). We further assessed factors in the iMSC secretome, their modulation, and the molecular pathways they elicit. RESULTS Increasing doses of IR disturbed endothelial tube and spheroid formation in hCMEC/D3. When IR-injured hCMEC/D3 (IR ≤ 5 Gy) were treated with iMSC CM, endothelial cell viability, adherence, spheroid compactness, and proangiogenic sprout formation were significantly ameliorated, and IR-induced ROS levels were reduced. iMSC CM augmented tube formation in cocultures of hCMEC/D3 and iMSCs. Consistently, iMSC CM facilitated angiogenesis in a zebrafish model in vivo. Furthermore, iMSC CM suppressed IR-induced NFκB activation, TNF-α release, and ROS production in THP1 cells. Additionally, iMSC CM diminished NF-kB activation in THP1 cells cocultured with irradiated hCMEC/D3, iMSCs, or HMC3 microglial lines. The cytokine array revealed that iMSC CM contains the proangiogenic and immunosuppressive factors MCP1/CCL2, IL6, IL8/CXCL8, ANG (Angiogenin), GROα/CXCL1, and RANTES/CCL5. Common promoter regulatory elements were enriched in TF-binding motifs such as androgen receptor (ANDR) and GATA2. hCMEC/D3 phosphokinome profiling revealed increased expression of pro-survival factors, the PI3K/AKT/mTOR modulator PRAS40 and β-catenin in response to CM. The transcriptome analysis revealed increased expression of GATA2 in iMSCs and the enrichment of pathways involved in RNA metabolism, translation, mitochondrial respiration, DNA damage repair, and neurodevelopment. CONCLUSIONS The iMSC secretome is a comodulated composite of proangiogenic and immunosuppressive factors that has the potential to alleviate radiation-induced vascular endothelial cell damage and immune activation.
Collapse
Affiliation(s)
- Kshama Gupta
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
| | - Ralph B Perkerson
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Tammee M Parsons
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Ramacharan Angom
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Danilyn Amerna
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Jeremy D Burgess
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Debabrata Mukhopadhyay
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Prasanna Vibhute
- Department of Radiology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Zbigniew K Wszolek
- Department of Neurology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Abba C Zubair
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Alfredo Quiñones-Hinojosa
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
- Department of Neurosurgery, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
- Center of Regenerative Biotherapeutics, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL, 32224, USA.
| |
Collapse
|
6
|
Sharma P, Maurya DK. Wharton's jelly mesenchymal stem cells: Future regenerative medicine for clinical applications in mitigation of radiation injury. World J Stem Cells 2024; 16:742-759. [PMID: 39086560 PMCID: PMC11287430 DOI: 10.4252/wjsc.v16.i7.742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/28/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Wharton's jelly mesenchymal stem cells (WJ-MSCs) are gaining significant attention in regenerative medicine for their potential to treat degenerative diseases and mitigate radiation injuries. WJ-MSCs are more naïve and have a better safety profile, making them suitable for both autologous and allogeneic transplantations. This review highlights the regenerative potential of WJ-MSCs and their clinical applications in mitigating various types of radiation injuries. In this review, we will also describe why WJ-MSCs will become one of the most probable stem cells for future regenerative medicine along with a balanced view on their strengths and weaknesses. Finally, the most updated literature related to both preclinical and clinical usage of WJ-MSCs for their potential application in the regeneration of tissues and organs will also be compiled.
Collapse
Affiliation(s)
- Prashasti Sharma
- Life Sciences, Homi Bhabha National Institute, Mumbai 400094, Maharashtra, India
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
| | - Dharmendra Kumar Maurya
- Life Sciences, Homi Bhabha National Institute, Mumbai 400094, Maharashtra, India
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India.
| |
Collapse
|
7
|
Huang H, Zheng Y, Chang M, Song J, Xia L, Wu C, Jia W, Ren H, Feng W, Chen Y. Ultrasound-Based Micro-/Nanosystems for Biomedical Applications. Chem Rev 2024; 124:8307-8472. [PMID: 38924776 DOI: 10.1021/acs.chemrev.4c00009] [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: 06/28/2024]
Abstract
Due to the intrinsic non-invasive nature, cost-effectiveness, high safety, and real-time capabilities, besides diagnostic imaging, ultrasound as a typical mechanical wave has been extensively developed as a physical tool for versatile biomedical applications. Especially, the prosperity of nanotechnology and nanomedicine invigorates the landscape of ultrasound-based medicine. The unprecedented surge in research enthusiasm and dedicated efforts have led to a mass of multifunctional micro-/nanosystems being applied in ultrasound biomedicine, facilitating precise diagnosis, effective treatment, and personalized theranostics. The effective deployment of versatile ultrasound-based micro-/nanosystems in biomedical applications is rooted in a profound understanding of the relationship among composition, structure, property, bioactivity, application, and performance. In this comprehensive review, we elaborate on the general principles regarding the design, synthesis, functionalization, and optimization of ultrasound-based micro-/nanosystems for abundant biomedical applications. In particular, recent advancements in ultrasound-based micro-/nanosystems for diagnostic imaging are meticulously summarized. Furthermore, we systematically elucidate state-of-the-art studies concerning recent progress in ultrasound-based micro-/nanosystems for therapeutic applications targeting various pathological abnormalities including cancer, bacterial infection, brain diseases, cardiovascular diseases, and metabolic diseases. Finally, we conclude and provide an outlook on this research field with an in-depth discussion of the challenges faced and future developments for further extensive clinical translation and application.
Collapse
Affiliation(s)
- Hui Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yi Zheng
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P. R. China
| | - Jun Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wencong Jia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Hongze Ren
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Wei Feng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| | - Yu Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, P. R. China
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
8
|
Christy BA, Herzig MC, Wu X, Mohammadipoor A, McDaniel JS, Bynum JA. Cell Therapies for Acute Radiation Syndrome. Int J Mol Sci 2024; 25:6973. [PMID: 39000080 PMCID: PMC11241804 DOI: 10.3390/ijms25136973] [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: 05/10/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
The risks of severe ionizing radiation exposure are increasing due to the involvement of nuclear powers in combat operations, the increasing use of nuclear power, and the existence of terrorist threats. Exposure to a whole-body radiation dose above about 0.7 Gy results in H-ARS (hematopoietic acute radiation syndrome), which is characterized by damage to the hematopoietic system; higher doses result in further damage to the gastrointestinal and nervous systems. Only a few medical countermeasures for ARS are currently available and approved for use, although others are in development. Cell therapies (cells or products produced by cells) are complex therapeutics that show promise for the treatment of radiation injury and have been shown to reduce mortality and morbidity in animal models. Since clinical trials for ARS cannot be ethically conducted, animal testing is extremely important. Here, we describe cell therapies that have been tested in animal models. Both cells and cell products appear to promote survival and lessen tissue damage after whole-body irradiation, although the mechanisms are not clear. Because radiation exposure often occurs in conjunction with other traumatic injuries, animal models of combined injury involving radiation and future countermeasure testing for these complex medical problems are also discussed.
Collapse
Affiliation(s)
- Barbara A Christy
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Maryanne C Herzig
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Xiaowu Wu
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Arezoo Mohammadipoor
- Hemorrhage and Vascular Dysfunction, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Jennifer S McDaniel
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - James A Bynum
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
- Department of Surgery, UT Health San Antonio, San Antonio, TX 78229, USA
- Trauma Research and Combat Casualty Care Collaborative, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
9
|
Xiao W, Zhao L, Sun Y, Yang X, Fu Q. Stimuli-Responsive Nanoradiosensitizers for Enhanced Cancer Radiotherapy. SMALL METHODS 2024; 8:e2301131. [PMID: 37906050 DOI: 10.1002/smtd.202301131] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/10/2023] [Indexed: 11/02/2023]
Abstract
Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.
Collapse
Affiliation(s)
- Wenjing Xiao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Lin Zhao
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yang Sun
- Department of Radiotherapy, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266021, China
| | - Xiao Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Qinrui Fu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| |
Collapse
|
10
|
Wang Z, Wang D, Ren X, Liu Z, Liu A, Li X, Guan L, Shen Y, Jin S, Zvyagin AV, Yang B, Wang T, Lin Q. One Stone, Three Birds: Multifunctional Nanodots as "Pilot Light" for Guiding Surgery, Enhanced Radiotherapy, and Brachytherapy of Tumors. ACS CENTRAL SCIENCE 2023; 9:1976-1988. [PMID: 37901175 PMCID: PMC10604975 DOI: 10.1021/acscentsci.3c00994] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Indexed: 10/31/2023]
Abstract
Surgery, radiotherapy (RT), and brachytherapy are crucial treatments for localized deep tumors. However, imprecise tumor location often leads to issues such as positive surgical margins, extended radiotherapy target volumes, and radiation damage to healthy tissues. Reducing side effects in healthy tissue and enhancing RT efficacy are critical challenges. To address these issues, we developed a multifunctional theranostic platform using Au/Ag nanodots (Au/AgNDs) that act as a "pilot light" for real-time guided surgery, high-efficiency RT, and brachytherapy, achieving a strategy of killing three birds with one stone. First, dual-mode imaging of Au/AgNDs enabled precision RT, minimizing damage to adjacent normal tissue during X-ray irradiation. Au/AgNDs enhanced ionizing radiation energy deposition, increased intracellular reactive oxygen species (ROS) generation, regulated the cell cycle, promoted DNA damage formation, and inhibited DNA repair in tumor cells, significantly improving RT efficacy. Second, in brachytherapy, precise guidance provided by dual-mode imaging addressed challenges related to non-visualization of existing interstitial brachytherapy and multiple adjustments of insertion needle positions. Meanwhile, the effect of brachytherapy was improved. Third, the excellent fluorescence imaging of Au/AgNDs accurately distinguished tumors from normal tissue, facilitating their use as a powerful tool for assisting surgeons during tumor resection. Taken together, our multifunctional theranostic platform offers real-time guidance for surgery and high-efficiency RT, and improves brachytherapy precision, providing a novel strategy and vision for the clinical diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Ze Wang
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Dongzhou Wang
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Xiaojun Ren
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Zhongshan Liu
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Annan Liu
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Xingchen Li
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Lin Guan
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yannan Shen
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Shunzi Jin
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Andrei V. Zvyagin
- Australian
Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW 2109, Australia
- Institute
of Biology and Biomedicine, Lobachevsky
Nizhny Novgorod State University, 603105 Nizhny Novgorod, Russia
| | - Bai Yang
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Tiejun Wang
- Department
of Radiation Oncology, The Second Affiliated
Hospital of Jilin University, Changchun 130041, P. R. China
- NHC
Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, P. R. China
| | - Quan Lin
- State
Key Laboratory of Supramolecular Structure and Materials, College
of Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
11
|
Satyamitra MM, Andres DK, Bergmann JN, Hoffman CM, Hogdahl T, Homer MJ, Hu TC, Rios CI, Yeung DT, DiCarlo AL. Overlapping Science in Radiation and Sulfur Mustard Exposures of Skin and Lung: Consideration of Models, Mechanisms, Organ Systems, and Medical Countermeasures: Overlapping science in radiation and sulfur mustard injuries to lung and skin. Disaster Med Public Health Prep 2023; 17:e552. [PMID: 37852927 PMCID: PMC10843005 DOI: 10.1017/dmp.2023.176] [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] [Indexed: 10/20/2023]
Abstract
PURPOSE To summarize presentations and discussions from the 2022 trans-agency workshop titled "Overlapping science in radiation and sulfur mustard (SM) exposures of skin and lung: Consideration of models, mechanisms, organ systems, and medical countermeasures." METHODS Summary on topics includes: (1) an overview of the radiation and chemical countermeasure development programs and missions; (2) regulatory and industry perspectives for drugs and devices; 3) pathophysiology of skin and lung following radiation or SM exposure; 4) mechanisms of action/targets, biomarkers of injury; and 5) animal models that simulate anticipated clinical responses. RESULTS There are striking similarities between injuries caused by radiation and SM exposures. Primary outcomes from both types of exposure include acute injuries, while late complications comprise chronic inflammation, oxidative stress, and vascular dysfunction, which can culminate in fibrosis in both skin and lung organ systems. This workshop brought together academic and industrial researchers, medical practitioners, US Government program officials, and regulators to discuss lung-, and skin- specific animal models and biomarkers, novel pathways of injury and recovery, and paths to licensure for products to address radiation or SM injuries. CONCLUSIONS Regular communications between the radiological and chemical injury research communities can enhance the state-of-the-science, provide a unique perspective on novel therapeutic strategies, and improve overall US Government emergency preparedness.
Collapse
Affiliation(s)
- Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH)
| | | | - Julie N. Bergmann
- Radiological/Nuclear Medical Countermeasures Program, Biomedical Advanced Research and Development Authority (BARDA)
| | - Corey M. Hoffman
- Radiological/Nuclear Medical Countermeasures Program, Biomedical Advanced Research and Development Authority (BARDA)
| | | | - Mary J. Homer
- Radiological/Nuclear Medical Countermeasures Program, Biomedical Advanced Research and Development Authority (BARDA)
| | - Tom C. Hu
- Chemical Medical Countermeasures Program, BARDA
| | - Carmen I. Rios
- Radiation and Nuclear Countermeasures Program (RNCP), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH)
| | - David T. Yeung
- Chemical Countermeasures Research Program (CCRP), NIAID, NIH
| | - Andrea L. DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH)
| |
Collapse
|
12
|
Singh PV, Singh PV, Anjankar A. Harnessing the Therapeutic Potential of Stem Cells in the Management of Chronic Obstructive Pulmonary Disease: A Comprehensive Review. Cureus 2023; 15:e44498. [PMID: 37711945 PMCID: PMC10497883 DOI: 10.7759/cureus.44498] [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: 07/31/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent and debilitating respiratory condition with limited treatment options. Stem cell therapy has emerged as a promising approach for COPD management due to its regenerative and immunomodulatory properties. This review article aims to comprehensively explore the therapeutic potential of stem cells in COPD management. The introduction provides background on COPD, highlighting its impact on health and the need for novel therapies. The different types of stem cells relevant to COPD, including embryonic stem cells, adult stem cells, and induced pluripotent stem cells, are described along with their properties and characteristics. The pathogenesis of COPD is discussed, emphasizing the key mechanisms involved in disease development and progression. Subsequently, the role of stem cells in tissue repair, regeneration, and immunomodulation is examined, highlighting their ability to address specific pathological processes in COPD. Mechanisms of action, such as paracrine signaling, immunomodulation, anti-inflammatory effects, and tissue regeneration, are explored. The interaction between stem cells and the host environment, which promotes lung repair, is also discussed. Challenges in stem cell therapy for COPD, including optimal cell sources, delivery methods, safety, and efficacy, are identified. Regulatory considerations and the importance of standardization are emphasized. Potential strategies for optimizing the therapeutic potential of stem cells in COPD management, such as combination therapies and preconditioning techniques, are outlined. Emerging trends and future directions are highlighted, including advanced cell engineering and patient-specific induced pluripotent stem cells. In conclusion, stem cell therapy holds significant promise for COPD management, addressing the limitations of current treatments. Continued research and development are necessary to overcome challenges, optimize therapies, and realize stem cells' full potential in improving the lives of patients with COPD.
Collapse
Affiliation(s)
- Parth V Singh
- Internal Medicine, Indira Gandhi Government Medical College, Nagpur, IND
| | - Prateesh V Singh
- Medicine and Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ashish Anjankar
- Biochemistry, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
13
|
Kim WH, Yoo JH, Yoo IK, Kwon CI, Hong SP. Effects of Mesenchymal Stem Cells Treatment on Radiation-Induced Proctitis in Rats. Yonsei Med J 2023; 64:167-174. [PMID: 36825342 PMCID: PMC9971437 DOI: 10.3349/ymj.2022.0342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 02/25/2023] Open
Abstract
PURPOSE There are no effective treatment methods with which to control complications of radiation proctitis with fistula or recurrent bleeding following radiation treatment for prostate, cervical, or rectal cancer. Mesenchymal stem cells (MSCs) can induce immune modification, resulting in tissue repair and regeneration. Therefore, we used a rat model of radiation-induced proctitis and observed the effects of using human placenta-derived (PD) and adipose tissue-derived (AD) MSCs. MATERIALS AND METHODS Female Sprague Dawley rats were irradiated at the pelvic area with 25 Gy. We injected 1×106 cells of human PD-MSCs, human AD-MSCs, human foreskin fibroblasts, and control media into the rectal submucosa following irradiation. We sacrificed rats for pathologic evaluation. RESULTS Fibrosis on the rectum was reduced in both MSC groups, compared to the control group. Mucosal Ki-67 indices of both MSC injected groups were higher than those in the control group. Although caspase-3 positive cells in the mucosa gradually increased and decreased in the control group, those in both MSC injected groups increased rapidly and decreased thereafter. CONCLUSION We demonstrated the effects of regional MSC injection treatment for radiation-induced proctitis in rats. MSC injection reduced fibrosis and increased proliferation in rat mucosa. Human AD-MSCs and PD-MSCs had similar effectiveness.
Collapse
Affiliation(s)
- Won Hee Kim
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Jun Hwan Yoo
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - In Kyung Yoo
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Chang Il Kwon
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Sung Pyo Hong
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, Korea.
| |
Collapse
|
14
|
Kano F, Hashimoto N, Liu Y, Xia L, Nishihara T, Oki W, Kawarabayashi K, Mizusawa N, Aota K, Sakai T, Azuma M, Hibi H, Iwasaki T, Iwamoto T, Horimai N, Yamamoto A. Therapeutic benefits of factors derived from stem cells from human exfoliated deciduous teeth for radiation-induced mouse xerostomia. Sci Rep 2023; 13:2706. [PMID: 36792628 PMCID: PMC9932159 DOI: 10.1038/s41598-023-29176-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Radiation therapy for head and neck cancers is frequently associated with adverse effects on the surrounding normal tissue. Irreversible damage to radiation-sensitive acinar cells in the salivary gland (SG) causes severe radiation-induced xerostomia (RIX). Currently, there are no effective drugs for treating RIX. We investigated the efficacy of treatment with conditioned medium derived from stem cells from human exfoliated deciduous teeth (SHED-CM) in a mouse RIX model. Intravenous administration of SHED-CM, but not fibroblast-CM (Fibro-CM), prevented radiation-induced cutaneous ulcer formation (p < 0.0001) and maintained SG function (p < 0.0001). SHED-CM treatment enhanced the expression of multiple antioxidant genes in mouse RIX and human acinar cells and strongly suppressed radiation-induced oxidative stress. The therapeutic effects of SHED-CM were abolished by the superoxide dismutase inhibitor diethyldithiocarbamate (p < 0.0001). Notably, quantitative liquid chromatography-tandem mass spectrometry shotgun proteomics of SHED-CM and Fibro-CM identified eight proteins activating the endogenous antioxidant system, which were more abundant in SHED-CM than in Fibro-CM (p < 0.0001). Neutralizing antibodies against those activators reduced antioxidant activity of SHED-CM (anti-PDGF-D; p = 0.0001, anti-HGF; p = 0.003). Our results suggest that SHED-CM may provide substantial therapeutic benefits for RIX primarily through the activation of multiple antioxidant enzyme genes in the target tissue.
Collapse
Affiliation(s)
- Fumiya Kano
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Noboru Hashimoto
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Yao Liu
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Linze Xia
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Takaaki Nishihara
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Wakana Oki
- grid.267335.60000 0001 1092 3579Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504 Japan
| | - Keita Kawarabayashi
- grid.267335.60000 0001 1092 3579Department of Pediatric Dentistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Noriko Mizusawa
- grid.267335.60000 0001 1092 3579Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Keiko Aota
- grid.267335.60000 0001 1092 3579Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takayoshi Sakai
- grid.136593.b0000 0004 0373 3971Department of Oral-Facial Disorders, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masayuki Azuma
- grid.267335.60000 0001 1092 3579Department of Oral Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hideharu Hibi
- grid.27476.300000 0001 0943 978XDepartment of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonori Iwasaki
- grid.267335.60000 0001 1092 3579Department of Pediatric Dentistry, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tsutomu Iwamoto
- grid.265073.50000 0001 1014 9130Department of Pediatric Dentistry/Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Akihito Yamamoto
- Department of Tissue Regeneration, Tokushima University Graduate School of Biomedical Sciences, Tokushima, 770-8504, Japan.
| |
Collapse
|
15
|
Wang Z, Ren X, Wang D, Guan L, Li X, Zhao Y, Liu A, He L, Wang T, Zvyagin AV, Yang B, Lin Q. Novel strategies for tumor radiosensitization mediated by multifunctional gold-based nanomaterials. Biomater Sci 2023; 11:1116-1136. [PMID: 36601661 DOI: 10.1039/d2bm01496c] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Radiotherapy (RT) is one of the most effective and commonly used cancer treatments for malignant tumors. However, the existing radiosensitizers have a lot of side effects and poor efficacy, which limits the curative effect and further application of radiotherapy. In recent years, emerging nanomaterials have shown unique advantages in enhancing radiosensitization. In particular, gold-based nanomaterials, with high X-ray attenuation capacity, good biocompatibility, and promising chemical, electronic and optical properties, have become a new type of radiotherapy sensitizer. In addition, gold-based nanomaterials can be used as a carrier to load a variety of drugs and immunosuppressants; in particular, its photothermal therapy, photodynamic therapy and multi-mode imaging functions aid in providing excellent therapeutic effect in coordination with RT. Recently, many novel strategies of radiosensitization mediated by multifunctional gold-based nanomaterials have been reported, which provides a new idea for improving the efficacy and reducing the side effects of RT. In this review, we systematically summarize the recent progress of various new gold-based nanomaterials that mediate radiosensitization and describe the mechanism. We further discuss the challenges and prospects in the field. It is hoped that this review will help researchers understand the latest progress of gold-based nanomaterials for radiosensitization, and encourage people to optimize the existing methods or explore novel approaches for radiotherapy.
Collapse
Affiliation(s)
- Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Xiaojun Ren
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Dongzhou Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Lin Guan
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Xingchen Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Yue Zhao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Annan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Liang He
- Department of Urology, the First Hospital of Jilin University, Changchun 130021, Jilin, China.
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Andrei V Zvyagin
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW 2109, Australia
- Institute of Biology and Biomedicine, Lobachevsky Nizhny Novgorod State University, 603105, Nizhny Novgorod, Russia
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, P. R. China.
| |
Collapse
|
16
|
Circulating microvesicles correlate with radiation proctitis complication after radiotherapy. Sci Rep 2023; 13:2033. [PMID: 36739457 PMCID: PMC9899237 DOI: 10.1038/s41598-022-21726-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/30/2022] [Indexed: 02/05/2023] Open
Abstract
In a large retrospective study, we assessed the putative use of circulating microvesicles (MVs), as innovative biomarkers of radiation toxicity in a cohort of 208 patients with prostate adenocarcinoma overexposed to radiation. The level of platelet (P)-, monocyte (M)- and endothelial (E)-derived MVs were assessed by flow cytometry. Rectal bleeding toxicity scores were collected at the time of blood sampling and during the routine follow-up and were tested for association with MVs using a multivariate logistic regression. MVs dosimetric correlation was investigated using dose volume histograms information available for a subset of 36 patients. The number of PMVs was significantly increased in patients with highest toxicity grades compared to lower grades. Risk prediction analysis revealed that increased numbers of PMVs, and an increased amount of MMVs relative to EMVs, were associated with worst rectal bleeding grade compared to the time of blood sampling. Moreover, a significant correlation was found between PMV and MMV numbers, with the range of doses up to the median exposure (40 Gy) of bladder/rectum and anterior rectal wall, respectively. MVs could be considered as new biomarkers to improve the identification of patients with high toxicity grade and may be instrumental for the prognosis of radiation therapy complications.
Collapse
|
17
|
Ji C, Zhao M, Wang C, Liu R, Zhu S, Dong X, Su C, Gu Z. Biocompatible Tantalum Nanoparticles as Radiosensitizers for Enhancing Therapy Efficacy in Primary Tumor and Metastatic Sentinel Lymph Nodes. ACS NANO 2022; 16:9428-9441. [PMID: 35666259 DOI: 10.1021/acsnano.2c02314] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Metastasis of breast carcinoma is commonly realized through lymphatic circulation, which seriously threatens the lives of breast cancer patients. Therefore, efficient therapy for both primary tumor and metastatic sentinel lymph nodes (SLNs) is highly desired to inhibit cancer growth and metastasis. During breast cancer treatment, radiotherapy (RT) is a common clinical method. However, the efficacy of RT is decreased by the radioresistance to a hypoxic microenvironment and inevitable side effects for healthy issues at high radiation doses. Considering the above-mentioned, we provide high biocompatible poly(vinylpyrrolidone) coated Ta nanoparticles (Ta@PVP NPs) for photothermal therapy (PTT) assisted RT for primary tumor and metastatic SLNs. On the one hand, for primary tumor treatment, Ta@PVP NPs with a high X-ray mass attenuation coefficient (4.30 cm2/kg at 100 keV) can deposit high radiation doses within tumors. On the other hand, for metastatic SLNs treatment, the effective delivery of Ta@PVP NPs from the primary tumor into SLNs is monitored by computed tomography and photoacoustic imaging, which greatly benefit the prognosis and treatment for metastatic SLNs. Moreover, Ta@PVP NPs-mediated PTT could enhance the RT effect, and immunogenic cell death caused by RT/PTT could induce an immune response to improve the therapeutic effect of metastatic SLNs. This study not only explores the potential of Ta@PVP NPs as effective radiosensitizers and photothermal agents for combined RT and PTT but also offers an efficient strategy to cure both primary tumor and metastatic SLNs in breast carcinoma.
Collapse
Affiliation(s)
- Chao Ji
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
- College of Mechanical and Electronic Engineering, Shandong University of Science and Technology, Qingdao 266590, China
| | - Maoru Zhao
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chengyan Wang
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruixue Liu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Zhu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinghua Dong
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
| | - Chunjian Su
- College of Mechanical and Electronic Engineering, Shandong University of Science and Technology, Qingdao 266590, China
| | - Zhanjun Gu
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Institute of High Energy Physics and National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing 100049, China
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Benderitter M, Herrera-Reyes E, Tamarat R. Mesenchymal stromal cells in the regeneration of radiation-induced organ sequelae: will they make the difference? JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2022; 42:024001. [PMID: 35532367 DOI: 10.1088/1361-6498/ac6dd8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/04/2022] [Indexed: 06/14/2023]
Abstract
Mesenchymal stromal cells (MSCs) are a stem cell product with good safety that demonstrate significant clinical efficacy in the treatment of different pathologies, including radiation diseases (e.g. radiological burns, pelvic radiation disease). While the first results for some first human applications for the treatment of radiation disease suggest benefit, larger trials with clinically important endpoints are needed before definitive conclusions can be drawn. However, the supply and cost of MSCs remain the two main limitations for this innovative therapeutic product. Exosomes (EXOs), a stem cell product associated with MSC therapy, have shown promising efficacy and safety in humans. MSC-EXO therapeutics represent a promising next-generation approach for treating radiation diseases involving a primary (major) inflammatory component. Provided that conditions for MSC-EXO production and bio-banking are agreed in the near future, the transition to industrial production of MSC-EXOs will be possible, and this is required to initiate well-controlled clinical trials for approval by the European Medicines Agency (EMA) and US Food and Drug Administration (FDA).
Collapse
Affiliation(s)
- M Benderitter
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Pôle Santé, 31 Avenue de la Division Leclerc, BP17, Fontenay-aux-Roses 92262, France
| | - E Herrera-Reyes
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Pôle Santé, 31 Avenue de la Division Leclerc, BP17, Fontenay-aux-Roses 92262, France
| | - R Tamarat
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Pôle Santé, 31 Avenue de la Division Leclerc, BP17, Fontenay-aux-Roses 92262, France
| |
Collapse
|
19
|
Obrador E, Salvador-Palmer R, Villaescusa JI, Gallego E, Pellicer B, Estrela JM, Montoro A. Nuclear and Radiological Emergencies: Biological Effects, Countermeasures and Biodosimetry. Antioxidants (Basel) 2022; 11:1098. [PMID: 35739995 PMCID: PMC9219873 DOI: 10.3390/antiox11061098] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Atomic and radiological crises can be caused by accidents, military activities, terrorist assaults involving atomic installations, the explosion of nuclear devices, or the utilization of concealed radiation exposure devices. Direct damage is caused when radiation interacts directly with cellular components. Indirect effects are mainly caused by the generation of reactive oxygen species due to radiolysis of water molecules. Acute and persistent oxidative stress associates to radiation-induced biological damages. Biological impacts of atomic radiation exposure can be deterministic (in a period range a posteriori of the event and because of destructive tissue/organ harm) or stochastic (irregular, for example cell mutation related pathologies and heritable infections). Potential countermeasures according to a specific scenario require considering basic issues, e.g., the type of radiation, people directly affected and first responders, range of doses received and whether the exposure or contamination has affected the total body or is partial. This review focuses on available medical countermeasures (radioprotectors, radiomitigators, radionuclide scavengers), biodosimetry (biological and biophysical techniques that can be quantitatively correlated with the magnitude of the radiation dose received), and strategies to implement the response to an accidental radiation exposure. In the case of large-scale atomic or radiological events, the most ideal choice for triage, dose assessment and victim classification, is the utilization of global biodosimetry networks, in combination with the automation of strategies based on modular platforms.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Rosario Salvador-Palmer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - Eduardo Gallego
- Energy Engineering Department, School of Industrial Engineering, Polytechnic University of Madrid, 28040 Madrid, Spain;
| | - Blanca Pellicer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
20
|
Ramos IPR, Dias ML, Nunes De Moraes AC, Meireles Ferreira FG, Souza SAL, Gutfilen B, Barboza T, Ferreira Pimentel C, Paz Batista CM, Kasai-Brunswick TH, Fortes FDSDA, De Andrade CBV, Goldenberg RCDS. Granulocyte Colony-Stimulating Factor Treatment Before Radiotherapy Protects Against Radiation-Induced Liver Disease in Mice. Front Pharmacol 2021; 12:725084. [PMID: 34867327 PMCID: PMC8634713 DOI: 10.3389/fphar.2021.725084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
Radiation-induced liver disease (RILD) remains a major problem resulting from radiotherapy. In this scenario, immunotherapy with granulocyte colony-stimulating factor (G-CSF) arises as an attractive approach that might improve the injured liver. Here, we investigated G-CSF administration’s impact before and after liver irradiation exposure using an association of alcohol consumption and local irradiation to induce liver disease model in C57BL/6 mice. Male and female mice were submitted to a previous alcohol-induced liver injury protocol with water containing 5% alcohol for 90 days. Then, the animals were treated with G-CSF (100 μg/kg/d) for 3 days before or after liver irradiation (18 Gy). At days 7, 30, and 60 post-radiation, non-invasive liver images were acquired by ultrasonography, magnetic resonance, and computed tomography. Biochemical and histological evaluations were performed to verify whether G-CSF could prevent liver tissue damage or reverse the acute liver injury. Our data showed that the treatment with G-CSF before irradiation effectively improved morphofunctional parameters caused by RILD, restoring histological arrangement, promoting liver regeneration, preserving normal organelles distribution, and glycogen granules. The amount of OV-6 and F4/80-positive cells increased, and α-SMA positive cells’ presence was normalized. Additionally, prior G-CSF administration preserved serum biochemical parameters and increased the survival rates (100%). On the other hand, after irradiation, the treatment showed a slight improvement in survival rates (79%) and did not ameliorate RILD. Overall, our data suggest that G-CSF administration before radiation might be an immunotherapeutic alternative to radiotherapy planning to avoid RILD.
Collapse
Affiliation(s)
- Isalira Peroba Rezende Ramos
- Centro Nacional de Biologia Estrutural e Bioimagem-CENABIO, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Marlon Lemos Dias
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-REGENERA, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | | | | | - Sergio Augusto Lopes Souza
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bianca Gutfilen
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thiago Barboza
- Departamento de Radiologia, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cibele Ferreira Pimentel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-REGENERA, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.,Laboratório de Terapia e Fisiologia Celular e Molecular-LTFCM, Centro Universitário Estadual da Zona Oeste-UEZO, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Biomedicina Translacional-BIOTRANS (UEZO-UNIGRANRIO-InMETRO), Duque de Caxias, Brazil
| | - Cintia Marina Paz Batista
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Tais Hanae Kasai-Brunswick
- Centro Nacional de Biologia Estrutural e Bioimagem-CENABIO, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.,Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-REGENERA, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Fabio Da Silva De Azevedo Fortes
- Laboratório de Terapia e Fisiologia Celular e Molecular-LTFCM, Centro Universitário Estadual da Zona Oeste-UEZO, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Biomedicina Translacional-BIOTRANS (UEZO-UNIGRANRIO-InMETRO), Duque de Caxias, Brazil
| | - Cherley Borba Vieira De Andrade
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.,Departmento de Histologia e Embriologia, Universidade do Estado do Rio de Janeiro, UERJ, Rio de Janeiro, Brazil
| | - Regina Coeli Dos Santos Goldenberg
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-REGENERA, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Dark Side of Cancer Therapy: Cancer Treatment-Induced Cardiopulmonary Inflammation, Fibrosis, and Immune Modulation. Int J Mol Sci 2021; 22:ijms221810126. [PMID: 34576287 PMCID: PMC8465322 DOI: 10.3390/ijms221810126] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 12/15/2022] Open
Abstract
Advancements in cancer therapy increased the cancer free survival rates and reduced the malignant related deaths. Therapeutic options for patients with thoracic cancers include surgical intervention and the application of chemotherapy with ionizing radiation. Despite these advances, cancer therapy-related cardiopulmonary dysfunction (CTRCPD) is one of the most undesirable side effects of cancer therapy and leads to limitations to cancer treatment. Chemoradiation therapy or immunotherapy promote acute and chronic cardiopulmonary damage by inducing reactive oxygen species, DNA damage, inflammation, fibrosis, deregulation of cellular immunity, cardiopulmonary failure, and non-malignant related deaths among cancer-free patients who received cancer therapy. CTRCPD is a complex entity with multiple factors involved in this pathogenesis. Although the mechanisms of cancer therapy-induced toxicities are multifactorial, damage to the cardiac and pulmonary tissue as well as subsequent fibrosis and organ failure seem to be the underlying events. The available biomarkers and treatment options are not sufficient and efficient to detect cancer therapy-induced early asymptomatic cell fate cardiopulmonary toxicity. Therefore, application of cutting-edge multi-omics technology, such us whole-exome sequencing, DNA methylation, whole-genome sequencing, metabolomics, protein mass spectrometry and single cell transcriptomics, and 10 X spatial genomics, are warranted to identify early and late toxicity, inflammation-induced carcinogenesis response biomarkers, and cancer relapse response biomarkers. In this review, we summarize the current state of knowledge on cancer therapy-induced cardiopulmonary complications and our current understanding of the pathological and molecular consequences of cancer therapy-induced cardiopulmonary fibrosis, inflammation, immune suppression, and tumor recurrence, and possible treatment options for cancer therapy-induced cardiopulmonary toxicity.
Collapse
|
22
|
Li X, Wang Q, Yu S, Zhang M, Liu X, Deng G, Liu Y, Wu S. Multifunctional MnO 2-based nanoplatform-induced ferroptosis and apoptosis for synergetic chemoradiotherapy. Nanomedicine (Lond) 2021; 16:2343-2361. [PMID: 34523352 DOI: 10.2217/nnm-2021-0286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Background: Radiosensitizers that can effectively consume glutathione provide broad prospects for enhancing the efficacy and reducing the side effects of radiotherapy. Aim: To explore the potential role of CuS@mSiO2@MnO2 nanocomposites in synergetic chemoradiotherapy. Methods: Nanocomposites were characterized by transmission electron microscopy, UV-Vis spectrometry and dynamic light scattering and were loaded with doxorubicin (DOX). The uptake and biodistribution of nanocomposites were observed by CCK8 assay, MRI and confocal laser scanning microscopy. The radiosensitization effect of nanocomposites and nanocomposites/DOX was assessed both in vitro and in vivo. Results: In vitro application of nanocomposites, with an average diameter of 30 nm and ζ-potential of 13.2 ± 0.4 mV, in combination with radiotherapy, depleted glutathione and induced ferroptosis and apoptosis. Nanocomposites/DOX exhibited tumor cell damage in vivo. Conclusion: We propose that this glutathione-depleting nanosystem could be a radiosensitizer as well as a drug transporter.
Collapse
Affiliation(s)
- Xi Li
- Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Qi Wang
- Department of Orthopedics, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200333, China.,Trauma Center, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Sihui Yu
- Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Minyi Zhang
- College of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Xijian Liu
- College of Chemistry & Chemical Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Guoying Deng
- Trauma Center, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Yuan Liu
- Reproductive Medicine Center, Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Sufang Wu
- Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 650 Xin Songjiang Road, Shanghai, 201620, China
| |
Collapse
|
23
|
Camprodon G, Huguet F. Unrecognized digestive toxicities of radiation therapy. Cancer Radiother 2021; 25:723-728. [PMID: 34391649 DOI: 10.1016/j.canrad.2021.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 12/16/2022]
Abstract
The aim of this article is to review unrecognized toxicities resulting from radiation therapy of digestive neoplasms. Due to their precocious occurrence, acute toxicities are well-known by radiation oncologist, and their treatment well-established. Thus, acute toxicities will not be described in this review. We will focus on incidence, diagnosis, and management of late and uncommon toxicities occurring in the digestive tract and digestive organs. Prevention, by respecting healthy tissues constraints, is the main tool to reduce incidence of those rare complications. Nonetheless, once installed, late toxicities remain a major burden in terms of quality of life and can even be life threatening. Hence, information and education about their diagnosis and management is important.
Collapse
Affiliation(s)
- G Camprodon
- Service d'Oncologie Médicale, Institut Gustave Roussy, 114, rue Edouard Vaillant, 94805 Villejuif, France.
| | - F Huguet
- Service d'Oncologie Radiothérapie, Hôpital Tenon, Sorbonne Université, AP-HP, Paris, France
| |
Collapse
|
24
|
Yang SJ, Wang XQ, Jia YH, Wang R, Cao K, Zhang X, Zhong J, Tan DM, Tan Y. Human umbilical cord mesenchymal stem cell transplantation restores hematopoiesis in acute radiation disease. Am J Transl Res 2021; 13:8670-8682. [PMID: 34539986 PMCID: PMC8430114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/27/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE Nuclear technology has been widely used in military and civilian fields, and radiotherapy is an effective and common form of treatment for cancer. However, acute radiation disease caused by high doses of radiation is a serious complication. The aim of this study was to investigate the chance of mitigating radiation-triggered hematopoiesis failure using human umbilical cord mesenchymal stem cell (HUCMSC) transplantation. METHODS Umbilical cords were obtained from three full-term female neonatus through cesarean section at Xinqiao Hospital. Bone marrow mesenchymal stem cells (BMSCs) were cultivated as depicted before. Briefly, monocytes were collected from bone marrow blood by means of density separation columns. An acute radiation disease mouse model was established to compare the restoration effect of HUCMSCs and BMSCs transplanted via the tail vein. The hematopoietic stem cell transplantation (HSCT) mouse model was obtained through bone marrow cell transplantation (BMCT) from C57BL/6 mice (H-2b, donor) to female CB6F1 mice (H-2b×d, recipient) after irradiation. The mice were divided into five groups, including control (saline), irradiated (radiation), bone marrow (HSCT, transplanted 1×106 BM cells), HUCMSC (transplanted a mixture of 1×106 HUCMSCs and 1×106 BM cells), and BMSC group (transplanted a mixture of 1×106 BMSCs and 1×106 BM cells). The blood condition results were used to test the radiation-induced inflammatory reaction, and bone marrow pathological staining (H&E) was used to determine the radiation-induced bone marrow hematopoiesis failure. RESULTS After radiation, HUCMSC transplantation significantly improved the survival rate. By analyzing the blood condition test, colony formation, and bone marrow pathology, it was found that the HUCMSC group demonstrated significant functional improvements in terms of the recovery from hematopoiesis failure and reduction of inflammatory reaction. CONCLUSIONS HUCMSCs have more advantages over BMSCs in restoring and promoting the recovery of radiation-induced hematopoietic damage, thus having a new therapeutic potential for patients with acute radiation disease.
Collapse
Affiliation(s)
- Shi-Jie Yang
- Laboratory Animal Center, Chongqing Medical UniversityChongqing 400016, China
- Medical Center of Hematology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Xiao-Qi Wang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Yan-Hui Jia
- Medical Center of Hematology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Rui Wang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Ke Cao
- Laboratory Animal Center, Chongqing Medical UniversityChongqing 400016, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
| | - Jiangfan Zhong
- Medical Center of Hematology, Xinqiao Hospital, Army Medical UniversityChongqing 400037, China
- Department of Pathology, University of Southern California, Keck School of MedicineLos Angeles, CA 90033, USA
| | - Dong-Mei Tan
- Laboratory Animal Center, Chongqing Medical UniversityChongqing 400016, China
| | - Yi Tan
- Laboratory Animal Center, Chongqing Medical UniversityChongqing 400016, China
| |
Collapse
|
25
|
Nanduri LSY, Duddempudi PK, Yang WL, Tamarat R, Guha C. Extracellular Vesicles for the Treatment of Radiation Injuries. Front Pharmacol 2021; 12:662437. [PMID: 34084138 PMCID: PMC8167064 DOI: 10.3389/fphar.2021.662437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/04/2021] [Indexed: 01/02/2023] Open
Abstract
Normal tissue injury from accidental or therapeutic exposure to high-dose radiation can cause severe acute and delayed toxicities, which result in mortality and chronic morbidity. Exposure to single high-dose radiation leads to a multi-organ failure, known as acute radiation syndrome, which is caused by radiation-induced oxidative stress and DNA damage to tissue stem cells. The radiation exposure results in acute cell loss, cell cycle arrest, senescence, and early damage to bone marrow and intestine with high mortality from sepsis. There is an urgent need for developing medical countermeasures against radiation injury for normal tissue toxicity. In this review, we discuss the potential of applying secretory extracellular vesicles derived from mesenchymal stromal/stem cells, endothelial cells, and macrophages for promoting repair and regeneration of organs after radiation injury.
Collapse
Affiliation(s)
- Lalitha Sarad Yamini Nanduri
- Department of Radiation Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, United States
| | - Phaneendra K. Duddempudi
- Department of Biochemistry, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, United States
| | - Weng-Lang Yang
- Department of Radiation Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, United States
| | - Radia Tamarat
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Fontenay-aux-Roses, France
| | - Chandan Guha
- Department of Radiation Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, United States
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, United States
- Department of Urology, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, United States
- Institute for Onco-Physics, Albert Einstein College of Medicine, Montefiore Medical Center, New York, NY, United States
| |
Collapse
|
26
|
Montay-Gruel P, Zhu Y, Petit B, Leavitt R, Warn M, Giedzinski E, Ollivier J, Sinclair DA, Vozenin MC, Limoli CL. Extracellular Vesicles for the Treatment of Radiation-Induced Normal Tissue Toxicity in the Lung. Front Oncol 2021; 10:602763. [PMID: 33738245 PMCID: PMC7962869 DOI: 10.3389/fonc.2020.602763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022] Open
Abstract
Human stem cell-derived extracellular vesicles (EV) provide many advantages over cell-based therapies for the treatment of functionally compromised tissue beds and organ sites. Here we sought to determine whether human embryonic stem cell (hESC)-derived EV could resolve in part, the adverse late normal tissue complications associated with exposure of the lung to ionizing radiation. The hESC-derived EV were systemically administered to the mice via the retro-orbital sinus to explore the potential therapeutic benefits following exposure to high thoracic doses of radiation (14 Gy). Data demonstrated that hESC-derived EV treatment significantly improved overall survival of the irradiated cohorts (P < 0.001). Increased survival was also associated with significant reductions in lung fibrosis as quantified by CBCT imaging (P < 0.01, 2 weeks post-irradiation). Qualitative histological analyses revealed reduced indications of radiation induced pulmonary injury in animals treated with EV. EV were then subjected to a rigorous proteomic analysis to ascertain the potential bioactive cargo that may prove beneficial in ameliorating radiation-induced normal tissue toxicities in the lung. Proteomics validated several consensus exosome markers (e.g., CD68) and identified major classes of proteins involved in nuclear pore complexes, epigenetics, cell cycle, growth and proliferation, DNA repair, antioxidant function, and cellular metabolism (TCA cycle and oxidative phosphorylation, OXYPHOS). Interestingly, EV were also found to contain mitochondrial components (mtDNA, OXYPHOS protein subunits), which may contribute to the metabolic reprograming and recovery of radiation-injured pulmonary tissue. To evaluate the safety of EV treatments in the context of the radiotherapeutic management of tumors, mice harboring TC1 tumor xenografts were subjected to the same EV treatments shown to forestall lung fibrosis. Data indicated that over the course of one month, no change in the growth of flank tumors between treated and control cohorts was observed. In conclusion, present findings demonstrate that systemic delivery of hESC-derived EV could ameliorate radiation-induced normal tissue complications in the lung, through a variety of potential mechanisms based on EV cargo analysis.
Collapse
Affiliation(s)
- Pierre Montay-Gruel
- Department of Radiation Oncology, University of California, Irvine, CA, United States
| | - Yafeng Zhu
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging Research, Harvard Medical School, Boston, MA, United States
| | - Benoit Petit
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ron Leavitt
- Department of Radiation Oncology, University of California, Irvine, CA, United States
| | - Mike Warn
- Department of Radiation Oncology, University of California, Irvine, CA, United States
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, CA, United States
| | - Jonathan Ollivier
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - David A Sinclair
- Department of Genetics, Blavatnik Institute, Paul F. Glenn Center for the Biology of Aging Research, Harvard Medical School, Boston, MA, United States
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, United States
| |
Collapse
|
27
|
Serrano Martinez P, Giuranno L, Vooijs M, Coppes RP. The Radiation-Induced Regenerative Response of Adult Tissue-Specific Stem Cells: Models and Signaling Pathways. Cancers (Basel) 2021; 13:cancers13040855. [PMID: 33670536 PMCID: PMC7921940 DOI: 10.3390/cancers13040855] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/10/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy is involved in the treatment of many cancers, but damage induced to the surrounding normal tissue is often inevitable. Evidence suggests that the maintenance of homeostasis and regeneration of the normal tissue is driven by specific adult tissue stem/progenitor cells. These tasks involve the input from several signaling pathways. Irradiation also targets these stem/progenitor cells, triggering a cellular response aimed at achieving tissue regeneration. Here we discuss the currently used in vitro and in vivo models and the involved specific tissue stem/progenitor cell signaling pathways to study the response to irradiation. The combination of the use of complex in vitro models that offer high in vivo resemblance and lineage tracing models, which address organ complexity constitute potential tools for the study of the stem/progenitor cellular response post-irradiation. The Notch, Wnt, Hippo, Hedgehog, and autophagy signaling pathways have been found as crucial for driving stem/progenitor radiation-induced tissue regeneration. We review how these signaling pathways drive the response of solid tissue-specific stem/progenitor cells to radiotherapy and the used models to address this.
Collapse
Affiliation(s)
- Paola Serrano Martinez
- Department of Biomedical Sciences of Cells and Systems-Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands;
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Lorena Giuranno
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
| | - Marc Vooijs
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P.O. Box 616, 6200 MD Maastricht, The Netherlands;
- Correspondence: (M.V.); (R.P.C.)
| | - Robert P. Coppes
- Department of Biomedical Sciences of Cells and Systems-Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands;
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
- Correspondence: (M.V.); (R.P.C.)
| |
Collapse
|
28
|
Therapeutic Potential of Mesenchymal Stromal Cells and Extracellular Vesicles in the Treatment of Radiation Lesions-A Review. Cells 2021; 10:cells10020427. [PMID: 33670501 PMCID: PMC7922519 DOI: 10.3390/cells10020427] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/08/2021] [Accepted: 02/13/2021] [Indexed: 12/14/2022] Open
Abstract
Ionising radiation-induced normal tissue damage is a major concern in clinic and public health. It is the most limiting factor in radiotherapy treatment of malignant diseases. It can also cause a serious harm to populations exposed to accidental radiation exposure or nuclear warfare. With regard to the clinical use of radiation, there has been a number of modalities used in the field of radiotherapy. These includes physical modalities such modified collimators or fractionation schedules in radiotherapy. In addition, there are a number of pharmacological agents such as essential fatty acids, vasoactive drugs, enzyme inhibitors, antioxidants, and growth factors for the prevention or treatment of radiation lesions in general. However, at present, there is no standard procedure for the treatment of radiation-induced normal tissue lesions. Stem cells and their role in tissue regeneration have been known to biologists, in particular to radiobiologists, for many years. It was only recently that the potential of stem cells was studied in the treatment of radiation lesions. Stem cells, immediately after their successful isolation from a variety of animal and human tissues, demonstrated their likely application in the treatment of various diseases. This paper describes the types and origin of stem cells, their characteristics, current research, and reviews their potential in the treatment and regeneration of radiation induced normal tissue lesions. Adult stem cells, among those mesenchymal stem cells (MSCs), are the most extensively studied of stem cells. This review focuses on the effects of MSCs in the treatment of radiation lesions.
Collapse
|
29
|
Rocchi C, Barazzuol L, Coppes RP. The evolving definition of salivary gland stem cells. NPJ Regen Med 2021; 6:4. [PMID: 33526786 PMCID: PMC7851389 DOI: 10.1038/s41536-020-00115-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Dysfunction of the salivary gland and irreversible hyposalivation are the main side effects of radiotherapy treatment for head and neck cancer leading to a drastic decrease of the quality of life of the patients. Approaches aimed at regenerating damaged salivary glands have been proposed as means to provide long-term restoration of tissue function in the affected patients. In studies to elucidate salivary gland regenerative mechanisms, more and more evidence suggests that salivary gland stem/progenitor cell behavior, like many other adult tissues, does not follow that of the hard-wired professional stem cells of the hematopoietic system. In this review, we provide evidence showing that several cell types within the salivary gland epithelium can serve as stem/progenitor-like cells. While these cell populations seem to function mostly as lineage-restricted progenitors during homeostasis, we indicate that upon damage specific plasticity mechanisms might be activated to take part in regeneration of the tissue. In light of these insights, we provide an overview of how recent developments in the adult stem cell research field are changing our thinking of the definition of salivary gland stem cells and their potential plasticity upon damage. These new perspectives may have important implications on the development of new therapeutic approaches to rescue radiation-induced hyposalivation.
Collapse
Affiliation(s)
- Cecilia Rocchi
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands. .,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands. .,Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| | - Lara Barazzuol
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Rob P Coppes
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
30
|
Kim IG, Cho H, Shin J, Cho JH, Cho SW, Chung EJ. Regeneration of irradiation-damaged esophagus by local delivery of mesenchymal stem-cell spheroids encapsulated in a hyaluronic-acid-based hydrogel. Biomater Sci 2021; 9:2197-2208. [PMID: 33506817 DOI: 10.1039/d0bm01655a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Radiation therapy (RT) is a typical treatment for head and neck cancers. Generally, prolonged irradiation of the esophagus causes esophageal fibrosis due to increased reactive oxygen species and proinflammatory cytokines. This study was designed to determine whether catechol-functionalized hyaluronic acid (HA-CA) hydrogel-encapsulated human mesenchymal stem-cell spheroids (MSC-SPs) could ameliorate damage to the esophagus in a mouse model of radiation-induced esophageal fibrosis. MSC-SPs were cultured in concave microwells 600 μm in diameter at a cell density of 1 × 106 cells per mL. Most cells formed spheroids with a 100-300 μm size distribution in concave microwells. MSC-SPs were well maintained in the HA gel, and live-dead staining confirmed that most cells survived. The HA gel containing the MSC-SPs was then injected into the damaged esophageal layer. Inflammatory signs or adverse tissue reactions were not observed after esophageal injection of HA-gel-encapsulated MSC-SPs. Based on Masson's trichrome staining at 4 and 12 weeks postinjection, the inner esophageal layer (IEL) was significantly thinner in the MSC-SP + HA gel group compared to those in the other experimental groups. While the saline and HA gel treatments made the esophageal muscles loose and thick, the MSC-SP + HA gel group showed bundles of tightly packed esophageal muscles, as assayed by desmin immunostaining. qPCR analysis showed that epithelial genes tended to increase over time in the MSC-SP + HA gel group, and the expression of most fibrosis-related genes decreased. This study proposes the potential of using HA-CA-hydrogel-encapsulated MSC-SPs as a promising therapy against radiation-induced esophageal fibrosis.
Collapse
Affiliation(s)
- In Gul Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
31
|
Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Show Comparable Functionality to Their Autologous Origin. Cells 2020; 10:cells10010033. [PMID: 33379312 PMCID: PMC7823915 DOI: 10.3390/cells10010033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 01/12/2023] Open
Abstract
A multimodal therapeutic approach involving radiotherapy is required when treating head and neck squamous cell carcinoma. However, radiotherapy is restricted due to its high risk for damages to the surrounding healthy tissue of the treated area. Tissue regeneration and wound healing is promoted by the survival and regenerative capacities of tissue-resident or invading stem cells. Mesenchymal stem cells (MSCs) exhibit a promising therapeutic potential in the field of cell-based tissue engineering and regenerative medicine due to their immunomodulatory properties and differentiation capacity. However, the generation of MSCs for therapeutic applications is still a major challenge. We aimed to produce highly homogeneous induced pluripotent stem cell-derived mesenchymal stem cells (iP-MSCs) in an autologous manner from initially isolated human mucosa mesenchymal stem cells (mMSCs) of the upper respiratory tract. Therefore, mMSCs were reprogrammed into induced pluripotent stem cells (iPSCs) by non-integrative chromosomal technologies and differentiated into corresponding iP-MSCs. We demonstrated that mMSCs and iP-MSCs show similar cell characteristics in terms of morphology, clonogenic potential, differentiation, and surface phenotype. Moreover, iP-MSCs demonstrated related immunosuppressive capacity as mMSCs including the secretion of cytokines, and T cell inhibition. Therefore, generating iP-MSCs in an autologous manner may be a novel personalized treatment option in regenerative medicine.
Collapse
|
32
|
Fu W, Zhang X, Mei L, Zhou R, Yin W, Wang Q, Gu Z, Zhao Y. Stimuli-Responsive Small-on-Large Nanoradiosensitizer for Enhanced Tumor Penetration and Radiotherapy Sensitization. ACS NANO 2020; 14:10001-10017. [PMID: 32658453 DOI: 10.1021/acsnano.0c03094] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Development of an efficient nanoradiosensitization system that enhances the radiation doses in cancer cells to sensitize radiotherapy (RT) while sparing normal tissues is highly desirable. Here, we construct a tumor microenvironment (TME)-responsive disassembled small-on-large molybdenum disulfide/hafnium dioxide (MoS2/HfO2) dextran (M/H-D) nanoradiosensitizer. The M/H-D can degrade and release the HfO2 nanoparticles (NPs) in TME to enhance tumor penetration of the HfO2 NPs upon near-infrared (NIR) exposure, which can solve the bottleneck of insufficient internalization of the HfO2 NPs. Simultaneously, the NIR photothermal therapy increased peroxidase-like catalytic efficiency of the M/H-D nanoradiosensitizer in TME, which selectively catalyzed intratumorally overexpressed H2O2 into highly oxidized hydroxyl radicals (·OH). The heat induced by PTT also relieved the intratumoral hypoxia to sensitize RT. Consequently, this TME-responsive precise nanoradiosensitization achieved improved irradiation effectiveness, potent oxygenation in tumor, and efficient suppression to tumor, which can be real-time monitored by computed tomography and photoacoustic imaging.
Collapse
Affiliation(s)
- Wenhui Fu
- Laboratory for Micro-sized Functional Materials, Department of Chemistry and College of Elementary Education, Capital Normal University, Beijing 100048, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Xiao Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Linqiang Mei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruyi Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Qiang Wang
- Laboratory for Micro-sized Functional Materials, Department of Chemistry and College of Elementary Education, Capital Normal University, Beijing 100048, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- Center of Materials Science and Optoelectronics Engineering, College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
33
|
Qian L, Cen J. Hematopoietic Stem Cells and Mesenchymal Stromal Cells in Acute Radiation Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8340756. [PMID: 32855768 PMCID: PMC7443042 DOI: 10.1155/2020/8340756] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/02/2020] [Accepted: 07/24/2020] [Indexed: 02/08/2023]
Abstract
With the extensive utilization of radioactive materials for medical, industrial, agricultural, military, and research purposes, medical researchers are trying to identify new methods to treat acute radiation syndrome (ARS). Radiation may cause injury to different tissues and organs, but no single drug has been proven to be effective in all circumstances. Radioprotective agents are always effective if given before irradiation, but many nuclear accidents are unpredictable. Medical countermeasures that can be beneficial to different organ and tissue injuries caused by radiation are urgently needed. Cellular therapy, especially stem cell therapy, has been a promising approach in ARS. Hematopoietic stem cells (HSCs) and mesenchymal stromal cells (MSCs) are the two main kinds of stem cells which show good efficacy in ARS and have attracted great attention from researchers. There are also some limitations that need to be investigated in future studies. In recent years, there are also some novel methods of stem cells that could possibly be applied on ARS, like "drug" stem cell banks obtained from clinical grade human induced pluripotent stem cells (hiPSCs), MSC-derived products, and infusion of HSCs without preconditioning treatment, which make us confident in the future treatment of ARS. This review focuses on major scientific and clinical advances of hematopoietic stem cells and mesenchymal stromal cells on ARS.
Collapse
Affiliation(s)
- Liren Qian
- Department of Hematology, The Sixth Medical Center, Chinese PLA General Hospital, Fucheng Road #6, Beijing 100048, China
| | - Jian Cen
- Department of Hematology, The Sixth Medical Center, Chinese PLA General Hospital, Fucheng Road #6, Beijing 100048, China
| |
Collapse
|
34
|
Khodamoradi E, Hoseini-Ghahfarokhi M, Amini P, Motevaseli E, Shabeeb D, Musa AE, Najafi M, Farhood B. Targets for protection and mitigation of radiation injury. Cell Mol Life Sci 2020; 77:3129-3159. [PMID: 32072238 PMCID: PMC11104832 DOI: 10.1007/s00018-020-03479-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023]
Abstract
Protection of normal tissues against toxic effects of ionizing radiation is a critical issue in clinical and environmental radiobiology. Investigations in recent decades have suggested potential targets that are involved in the protection against radiation-induced damages to normal tissues and can be proposed for mitigation of radiation injury. Emerging evidences have been shown to be in contrast to an old dogma in radiation biology; a major amount of reactive oxygen species (ROS) production and cell toxicity occur during some hours to years after exposure to ionizing radiation. This can be attributed to upregulation of inflammatory and fibrosis mediators, epigenetic changes and disruption of the normal metabolism of oxygen. In the current review, we explain the cellular and molecular changes following exposure of normal tissues to ionizing radiation. Furthermore, we review potential targets that can be proposed for protection and mitigation of radiation toxicity.
Collapse
Affiliation(s)
- Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Hoseini-Ghahfarokhi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
- Misan Radiotherapy Center, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
35
|
Wang B, Wang H, Zhang M, Ji R, Wei J, Xin Y, Jiang X. Radiation-induced myocardial fibrosis: Mechanisms underlying its pathogenesis and therapeutic strategies. J Cell Mol Med 2020; 24:7717-7729. [PMID: 32536032 PMCID: PMC7348163 DOI: 10.1111/jcmm.15479] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/18/2020] [Accepted: 05/24/2020] [Indexed: 12/24/2022] Open
Abstract
Radiation-induced myocardial fibrosis (RIMF) is a potentially lethal clinical complication of chest radiotherapy (RT) and a final stage of radiation-induced heart disease (RIHD). RIMF is characterized by decreased ventricular elasticity and distensibility, which can result in decreased ejection fraction, heart failure and even sudden cardiac death. Together, these conditions impair the long-term health of post-RT survivors and limit the dose and intensity of RT required to effectively kill tumour cells. Although the exact mechanisms involving in RIMF are unclear, increasing evidence indicates that the occurrence of RIMF is related to various cells, regulatory molecules and cytokines. However, accurately diagnosing and identifying patients who may progress to RIMF has been challenging. Despite the urgent need for an effective treatment, there is currently no medical therapy for RIMF approved for routine clinical application. In this review, we investigated the underlying pathophysiology involved in the initiation and progression of RIMF before outlining potential preventative and therapeutic strategies to counter this toxicity.
Collapse
Affiliation(s)
- Bin Wang
- Department of Radiation OncologyThe First Hospital of Jilin UniversityChangchunChina
- Jilin Provincial Key Laboratory of Radiation Oncology & TherapyThe First Hospital of Jilin UniversityChangchunChina
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunChina
| | - Huanhuan Wang
- Department of Radiation OncologyThe First Hospital of Jilin UniversityChangchunChina
- Jilin Provincial Key Laboratory of Radiation Oncology & TherapyThe First Hospital of Jilin UniversityChangchunChina
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunChina
| | - Mengmeng Zhang
- Phase I Clinical Research CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Rui Ji
- Department of BiologyValencia CollegeOrlandoFLUSA
| | - Jinlong Wei
- Department of Radiation OncologyThe First Hospital of Jilin UniversityChangchunChina
| | - Ying Xin
- Key Laboratory of PathobiologyMinistry of EducationJilin UniversityChangchunChina
| | - Xin Jiang
- Department of Radiation OncologyThe First Hospital of Jilin UniversityChangchunChina
- Jilin Provincial Key Laboratory of Radiation Oncology & TherapyThe First Hospital of Jilin UniversityChangchunChina
- NHC Key Laboratory of RadiobiologySchool of Public HealthJilin UniversityChangchunChina
| |
Collapse
|
36
|
Barazzuol L, Coppes RP, van Luijk P. Prevention and treatment of radiotherapy-induced side effects. Mol Oncol 2020; 14:1538-1554. [PMID: 32521079 PMCID: PMC7332214 DOI: 10.1002/1878-0261.12750] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 01/10/2023] Open
Abstract
Radiotherapy remains a mainstay of cancer treatment, being used in roughly 50% of patients. The precision with which the radiation dose can be delivered is rapidly improving. This precision allows the more accurate targeting of radiation dose to the tumor and reduces the amount of surrounding normal tissue exposed. Although this often reduces the unwanted side effects of radiotherapy, we still need to further improve patients' quality of life and to escalate radiation doses to tumors when necessary. High-precision radiotherapy forces one to choose which organ or functional organ substructures should be spared. To be able to make such choices, we urgently need to better understand the molecular and physiological mechanisms of normal tissue responses to radiotherapy. Currently, oversimplified approaches using constraints on mean doses, and irradiated volumes of normal tissues are used to plan treatments with minimized risk of radiation side effects. In this review, we discuss the responses of three different normal tissues to radiotherapy: the salivary glands, cardiopulmonary system, and brain. We show that although they may share very similar local cellular processes, they respond very differently through organ-specific, nonlocal mechanisms. We also discuss how a better knowledge of these mechanisms can be used to treat or to prevent the effects of radiotherapy on normal tissue and to optimize radiotherapy delivery.
Collapse
Affiliation(s)
- Lara Barazzuol
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Rob P. Coppes
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Peter van Luijk
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
37
|
Ioannides P, Giedzinski E, Limoli CL. Evaluating different routes of extracellular vesicle administration for cranial therapies. ACTA ACUST UNITED AC 2020; 6. [PMID: 34277952 PMCID: PMC8281946 DOI: 10.20517/2394-4722.2020.22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Aim: Human stem cell-derived extracellular vesicles (EV) provide many advantages over cell-based therapies for the treatment of functionally compromised tissue beds and organ sites. Here we aimed to highlight multiple administration routes for the potential treatment of various forms of brain injury. Methods: Human neural stem cell-derived EV were isolated from conditioned media and administered via three distinct routes: intrahippocampal transplantation, retro-orbital vein injection, and intranasal. EV were administered after which brains were evaluated to determine the capability of EV to translocate into normal tissue. Results: Data showed no significant differences in the amount of EV able to translocate across the brain, indicating the functional equivalence of each administration route to effectively deliver EV to the brain parenchyma. Conclusion: Findings show that both systemic administration routes (retro-orbital vein or intranasal delivery) afforded effective penetrance and perfusion of EV throughout the brain in a minimally invasive manner, and point to a translationally tractable option for treating certain neurological disorders including those resulting from cranial irradiation procedures.
Collapse
Affiliation(s)
- Pericles Ioannides
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| |
Collapse
|
38
|
Sun H, Wang X, Zhai S. The Rational Design and Biological Mechanisms of Nanoradiosensitizers. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E504. [PMID: 32168899 PMCID: PMC7153263 DOI: 10.3390/nano10030504] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 01/01/2023]
Abstract
Radiotherapy (RT) has been widely used for cancer treatment. However, the intrinsic drawbacks of RT, such as radiotoxicity in normal tissues and tumor radioresistance, promoted the development of radiosensitizers. To date, various kinds of nanoparticles have been found to act as radiosensitizers in cancer radiotherapy. This review focuses on the current state of nanoradiosensitizers, especially the related biological mechanisms, and the key design strategies for generating nanoradiosensitizers. The regulation of oxidative stress, DNA damage, the cell cycle, autophagy and apoptosis by nanoradiosensitizers in vitro and in vivo is highlighted, which may guide the rational design of therapeutics for tumor radiosensitization.
Collapse
Affiliation(s)
- Hainan Sun
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
- Shandong Vocational College of Light Industry, Zibo 255300, Shandong, China
| | - Xiaoling Wang
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| | - Shumei Zhai
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, Shandong, China; (H.S.); (X.W.)
| |
Collapse
|
39
|
Smith SM, Giedzinski E, Angulo MC, Lui T, Lu C, Park AL, Tang S, Martirosian V, Ru N, Chmielewski NN, Liang Y, Baulch JE, Acharya MM, Limoli CL. Functional equivalence of stem cell and stem cell-derived extracellular vesicle transplantation to repair the irradiated brain. Stem Cells Transl Med 2020; 9:93-105. [PMID: 31568685 PMCID: PMC6954724 DOI: 10.1002/sctm.18-0227] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 06/17/2019] [Indexed: 01/22/2023] Open
Abstract
Cranial radiotherapy, although beneficial for the treatment of brain tumors, inevitably leads to normal tissue damage that can induce unintended neurocognitive complications that are progressive and debilitating. Ionizing radiation exposure has also been shown to compromise the structural integrity of mature neurons throughout the brain, an effect believed to be at least in part responsible for the deterioration of cognitive health. Past work has shown that cranially transplanted human neural stem cells (hNSCs) or their extracellular vesicles (EVs) afforded long-term beneficial effects on many of these cognitive decrements. To provide additional insight into the potential neuroprotective mechanisms of cell-based regenerative strategies, we have analyzed hippocampal neurons for changes in structural integrity and synaptic remodeling after unilateral and bilateral transplantation of hNSCs or EVs derived from those same cells. Interestingly, hNSCs and EVs similarly afforded protection to host neurons, ameliorating the impact of irradiation on dendritic complexity and spine density for neurons present in both the ipsilateral and contralateral hippocampi 1 month following irradiation and transplantation. These morphometric improvements were accompanied by increased levels of glial cell-derived growth factor and significant attenuation of radiation-induced increases in postsynaptic density protein 95 and activated microglia were found ipsi- and contra-lateral to the transplantation sites of the irradiated hippocampus treated with hNSCs or hNSC-derived EVs. These findings document potent far-reaching neuroprotective effects mediated by grafted stem cells or EVs adjacent and distal to the site of transplantation and support their potential as therapeutic agents to counteract the adverse effects of cranial irradiation.
Collapse
Affiliation(s)
- Sarah M. Smith
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Erich Giedzinski
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Maria C. Angulo
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Tiffany Lui
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Celine Lu
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Audrey L. Park
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Sharon Tang
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Vahan Martirosian
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Ning Ru
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | | | - Yaxuan Liang
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Janet E. Baulch
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Munjal M. Acharya
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Charles L. Limoli
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| |
Collapse
|
40
|
Rong X, Zhang G, Yang Y, Gao C, Chu W, Sun H, Wang Y, Li C. Transplanted Antler Stem Cells Stimulated Regenerative Healing of Radiation-induced Cutaneous Wounds in Rats. Cell Transplant 2020; 29:963689720951549. [PMID: 32907381 PMCID: PMC7784515 DOI: 10.1177/0963689720951549] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/18/2020] [Accepted: 07/29/2020] [Indexed: 12/29/2022] Open
Abstract
Radiation-induced cutaneous injury is the main side effect of radiotherapy. The injury is difficult to cure and the pathogenesis is complex. Mesenchymal stem cells (MSCs) serve as a promising candidate for cell-based therapy for the treatment of cutaneous wounds. The aim of the present study was to investigate whether antler stem cells (AnSCs) have better therapeutic effects on radiation-induced cutaneous injury than currently available ones. In this study, a rat model of cutaneous wound injury from Sr-90 radiation was used. AnSCs (1 × 106/500 μl) were injected through the tail vein on the first day of irradiation. Our results showed that compared to the control group, AnSC-treated rats exhibited a delayed onset (14 days versus 7 days), shorter recovery time (51 days versus 84 days), faster healing rate (100% versus 70% on day 71), and higher healing quality with more cutaneous appendages regenerated (21:10:7/per given area compared to those of rat and human MSCs, respectively). More importantly, AnSCs promoted much higher quality of healing compared to other types of stem cells, with negligible scar formation. AnSC lineage tracing results showed that the injected-dye-stained AnSCs were substantially engrafted in the wound healing tissue, indicating that the therapeutic effects of AnSCs on wound healing at least partially through direct participation in the wound healing. Expression profiling of the wound-healing-related genes in the healing tissue of AnSC group more resembled a fetal wound healing. Revealing the mechanism underlying this higher quality of wound healing by using AnSC treatment would help to devise more effective cell-based therapeutics for radiation-induced wound healing in clinics.
Collapse
Affiliation(s)
- Xiaoli Rong
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
- The Third Hospital of Jilin University, Changchun, Jilin, China
| | - Guokun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| | - Yanyan Yang
- The Third Hospital of Jilin University, Changchun, Jilin, China
| | - Chenmao Gao
- The Third Hospital of Jilin University, Changchun, Jilin, China
| | - Wenhui Chu
- School of Life Science, Taizhou University, Taizhou, China
| | - Hongmei Sun
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences (CAAS), Changchun, Jilin, China
| | - Yimin Wang
- The Third Hospital of Jilin University, Changchun, Jilin, China
| | - Chunyi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun, Jilin, China
| |
Collapse
|
41
|
Giuranno L, Ient J, De Ruysscher D, Vooijs MA. Radiation-Induced Lung Injury (RILI). Front Oncol 2019; 9:877. [PMID: 31555602 PMCID: PMC6743286 DOI: 10.3389/fonc.2019.00877] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022] Open
Abstract
Radiation pneumonitis (RP) and radiation fibrosis (RF) are two dose-limiting toxicities of radiotherapy (RT), especially for lung, and esophageal cancer. It occurs in 5-20% of patients and limits the maximum dose that can be delivered, reducing tumor control probability (TCP) and may lead to dyspnea, lung fibrosis, and impaired quality of life. Both physical and biological factors determine the normal tissue complication probability (NTCP) by Radiotherapy. A better understanding of the pathophysiological sequence of radiation-induced lung injury (RILI) and the intrinsic, environmental and treatment-related factors may aid in the prevention, and better management of radiation-induced lung damage. In this review, we summarize our current understanding of the pathological and molecular consequences of lung exposure to ionizing radiation, and pharmaceutical interventions that may be beneficial in the prevention or curtailment of RILI, and therefore enable a more durable therapeutic tumor response.
Collapse
Affiliation(s)
- Lorena Giuranno
- Department of Radiotherapy, GROW School for Oncology Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jonathan Ient
- Department of Radiotherapy, GROW School for Oncology Maastricht University Medical Centre, Maastricht, Netherlands
| | - Dirk De Ruysscher
- Department of Radiotherapy, GROW School for Oncology Maastricht University Medical Centre, Maastricht, Netherlands
| | - Marc A Vooijs
- Department of Radiotherapy, GROW School for Oncology Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
42
|
Xiang H, Chen Y. Energy-Converting Nanomedicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1805339. [PMID: 30773837 DOI: 10.1002/smll.201805339] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/22/2019] [Indexed: 05/12/2023]
Abstract
Serious side effects to surrounding normal tissues and unsatisfactory therapeutic efficacy hamper the further clinic applications of conventional cancer-therapeutic strategies, such as chemotherapy and surgery. The fast development of nanotechnology provides unprecedented superiorities for cancer therapeutics. Externally activatable therapeutic modalities mediated by nanomaterials, relying on highly effective energy transformation to release therapeutic elements/effects (cytotoxic reactive oxygen species, thermal effect, photoelectric effect, Compton effect, cavitation effect, mechanical effect or chemotherapeutic drug) for cancer therapies, categorized and termed as "energy-converting nanomedicine," have arouse considerable concern due to their noninvasiveness, desirable tissue-penetration depth, and accurate modulation of therapeutic dose. This review summarizes the recent advances in the engineering of intelligent functional nanotherapeutics for energy-converting nanomedicine, including photo-based, radiation-based, ultrasound-based, magnetic field-based, microwave-based, electric field-based, and radiofrequency-based nanomedicines, which are enabled by external stimuli (light, radiation, ultrasound, magnetic field, microwave, electric field, and radiofrequency). Furthermore, biosafety issues of energy-converting nanomedicine related to future clinical translation are also addressed. Finally, the potential challenges and prospects of energy-converting nanomedicine for future clinical translation are discussed.
Collapse
Affiliation(s)
- Huijing Xiang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
43
|
Tsoutsou P, Montay-Gruel P, Vozenin MC. The Era of Modern Radiation Therapy: Innovations to Spare Normal Tissues. Radiat Oncol 2019. [DOI: 10.1007/978-3-319-52619-5_70-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
44
|
Xie J, Gong L, Zhu S, Yong Y, Gu Z, Zhao Y. Emerging Strategies of Nanomaterial-Mediated Tumor Radiosensitization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1802244. [PMID: 30156333 DOI: 10.1002/adma.201802244] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/08/2018] [Indexed: 05/23/2023]
Abstract
Nano-radiosensitization has been a hot concept for the past ten years, and the nanomaterial-mediated tumor radiosensitization method is mainly focused on increasing intracellular radiation deposition by high atomic number (high Z) nanomaterials, particularly gold (Au)-mediated radiation enhancement. Recently, various new nanomaterial-mediated radiosensitive approaches have been successively reported, such as catalyzing reactive oxygen species (ROS) generation, consuming intracellular reduced glutathione (GSH), overcoming tumor hypoxia, and various synergistic radiotherapy ways. These strategies may open a new avenue for enhancing the radiotherapeutic effect and avoiding its side effects. Nevertheless, reviews systematically summarizing these newly emerging methods and their radiosensitive mechanisms are still rare. Therefore, the general strategies of nanomaterial-mediated tumor radiosensitization are comprehensively summarized, particularly aiming at introducing the emerging radiosensitive methods. The strategies are divided into three general parts. First, methods on account of the intrinsic radiosensitive properties of nanoradiosensitizers for radiosensitization are highlighted. Then, newly developed synergistic strategies based on multifunctional nanomaterials for enhancing radiotherapy efficacy are emphasized. Third, nanomaterial-mediated radioprotection approaches for increasing the radiotherapeutic ratio are discussed. Importantly, the clinical translation of nanomaterial-mediated tumor radiosensitization is also covered. Finally, further challenges and outlooks in this field are discussed.
Collapse
Affiliation(s)
- Jiani Xie
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Linji Gong
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Zhu
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuan Yong
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhanjun Gu
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuliang Zhao
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
45
|
Yan X, Shu Y, He J, Zhao J, Jia L, Xie J, Sun Y, Zhao Z, Peng S. Therapeutic Effects of Human Umbilical Cord Mesenchymal Stromal Cells in Sprague-Dawley Rats with Percutaneous Exposure to Sulfur Mustard. Stem Cells Dev 2018; 28:69-80. [PMID: 30343632 DOI: 10.1089/scd.2018.0143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Sulfur mustard (SM) exposure, whose symptoms are similar to radiation exposure, can lead to acute injury. Because mesenchymal stromal cells (MSCs) have been used to experimentally and clinically treat acute radiation syndrome, in this study, MSCs were intravenously injected into rats after percutaneous SM exposure. Then, we examined sternum and spleen samples by histopathological and immunohistochemical methods to observe pathological changes. Furthermore, blood samples were taken to test the white blood cell (WBC) count, blood platelet count (BPC), red blood cell count, and the levels of cytokines in the serum. The number of bone marrow karyocytes and the WBC in the MSC + SM group were higher than those in the SM group, and the levels of granulocyte colony-stimulating factor, granulocyte-macrophage colony stimulating factor, monocyte chemoattractant protein-1, interleukin (IL)-1α, IL-5, and interferon-γ in the MSC + SM group remained high at different time points after SM exposure. In addition, the BPC, the level of erythropoietin and the relative weight of the spleen in the MSC + SM group were significantly higher than those in the SM group. Meanwhile, spleens in the MSC + SM group were more hyperplastic and hematopoietic, and had fewer apoptotic cells than in the SM group. Furthermore, rat body weight and locomotion ability in the MSC + SM group were higher than in the SM group. This evidence supports the potential ability of MSCs in immunoregulation and functional improvements to the hemopoietic microenvironment. Intravenous injection of MSCs exerted significant therapeutic effects in rats with percutaneous exposure to SM.
Collapse
Affiliation(s)
- Xiabei Yan
- 1 Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.,2 Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention of PLA, Beijing, China
| | - Yulei Shu
- 2 Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention of PLA, Beijing, China
| | - Jun He
- 2 Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention of PLA, Beijing, China
| | - Jun Zhao
- 2 Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention of PLA, Beijing, China
| | - Li Jia
- 2 Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention of PLA, Beijing, China
| | - Jianwei Xie
- 3 Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, China
| | - Yansong Sun
- 4 Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zengming Zhao
- 2 Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention of PLA, Beijing, China
| | - Shuangqing Peng
- 2 Evaluation and Research Center for Toxicology, Institute of Disease Control and Prevention of PLA, Beijing, China
| |
Collapse
|
46
|
François S, Usunier B, Forgue-Lafitte ME, L'Homme B, Benderitter M, Douay L, Gorin NC, Larsen AK, Chapel A. Mesenchymal Stem Cell Administration Attenuates Colon Cancer Progression by Modulating the Immune Component within the Colorectal Tumor Microenvironment. Stem Cells Transl Med 2018; 8:285-300. [PMID: 3045139 PMCID: PMC6392393 DOI: 10.1002/sctm.18-0117] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/07/2018] [Indexed: 12/17/2022] Open
Abstract
We here determine the influence of mesenchymal stem cell (MSC) therapy on the progression of solid tumors. The influence of MSCs was investigated in human colorectal cancer cells as well as in an immunocompetent rat model of colorectal carcinogenesis representative of the human pathology. Treatment with bone marrow (BM)‐derived MSCs significantly reduced both cancer initiation and cancer progression by increasing the number of tumor‐free animals as well as decreasing the number and the size of the tumors by half, thereby extending their lifespan. The attenuation of cancer progression was mediated by the capacity of the MSCs to modulate the immune component. Specifically, in the adenocarcinomas (ADKs) of MSC‐treated rats, the infiltration of CD68+ monocytes/macrophages was 50% less while the presence of CD3+ lymphocytes increased almost twofold. The MSCs reprogrammed the macrophages to become regulatory cells involved in phagocytosis thereby inhibiting the production of proinflammatory cytokines. Furthermore, the MSCs decreased NK (Natural Killer) and rTh17 cell activities, Treg recruitment, the presence of CD8+ lymphocytes and endothelial cells while restoring Th17 cell activity. The expression of miR‐150 and miR‐7 increased up to fivefold indicating a likely role for these miRNAs in the modulation of tumor growth. Importantly, MSC administration limited the damage of healthy tissues and attenuated tumor growth following radiotherapy. Taken together, we here show that that MSCs have durable action on colon cancer development by modulating the immune component of the tumor microenvironment. In addition, we identify two miRNAs associated with the capacity of MSCs to attenuate cancer growth. stem cells translational medicine2019;8:285&300
Collapse
Affiliation(s)
- Sabine François
- Radiobiology of Medical Exposure Laboratory (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France.,Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France
| | - Benoit Usunier
- Radiobiology of Medical Exposure Laboratory (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Marie-Elisabeth Forgue-Lafitte
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Paris, France.,Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, Paris, France
| | - Bruno L'Homme
- Radiobiology of Medical Exposure Laboratory (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Marc Benderitter
- Radiobiology of Medical Exposure Laboratory (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Luc Douay
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.,Université Pierre et Marie Curie (UPMC), Sorbonne Universités, Paris, France.,Service d'Hématologie Biologique, Hôpital Saint-Antoine/Armand Trousseau, AP-HP, Paris, France.,Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Norbert-Claude Gorin
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.,Université Pierre et Marie Curie (UPMC), Sorbonne Universités, Paris, France.,Service d'Hématologie Biologique, Hôpital Saint-Antoine/Armand Trousseau, AP-HP, Paris, France.,Service d'Hématologie Clinique et Thérapie Cellulaire, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Annette K Larsen
- Cancer Biology and Therapeutics, Centre de Recherche Saint-Antoine (CRSA), Institut National de la Santé et de la Recherche Médicale (INSERM) U938, Paris, France.,Institut Universitaire de Cancérologie (IUC), Faculté de Médecine, Sorbonne Université, Paris, France
| | - Alain Chapel
- Radiobiology of Medical Exposure Laboratory (LRMed), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France.,Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.,Université Pierre et Marie Curie (UPMC), Sorbonne Universités, Paris, France
| |
Collapse
|
47
|
Nasal administration of mesenchymal stem cells restores cisplatin-induced cognitive impairment and brain damage in mice. Oncotarget 2018; 9:35581-35597. [PMID: 30473752 PMCID: PMC6238972 DOI: 10.18632/oncotarget.26272] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 10/06/2018] [Indexed: 12/13/2022] Open
Abstract
Cognitive impairments are a common side effect of chemotherapy that often persists long after treatment completion. There are no FDA-approved interventions to treat these cognitive deficits also called ‘chemobrain’. We hypothesized that nasal administration of mesenchymal stem cells (MSC) reverses chemobrain. To test this hypothesis, we used a mouse model of cognitive deficits induced by cisplatin that we recently developed. Mice were treated with two cycles of cisplatin followed by nasal administration of MSC. Cisplatin treatment induced deficits in the puzzle box, novel object/place recognition and Y-maze tests, indicating cognitive impairment. Nasal MSC treatment fully reversed these cognitive deficits in males and females. MSC also reversed the cisplatin-induced damage to cortical myelin. Resting state functional MRI and connectome analysis revealed a decrease in characteristic path length after cisplatin, while MSC treatment increased path length in cisplatin-treated mice. MSCs enter the brain but did not survive longer than 12-72 hrs, indicating that they do not replace damaged tissue. RNA-sequencing analysis identified mitochondrial oxidative phosphorylation as a top pathway activated by MSC administration to cisplatin-treated mice. Consistently, MSC treatment restored the cisplatin-induced mitochondrial dysfunction and structural abnormalities in brain synaptosomes. Nasal administration of MSC did not interfere with the peripheral anti-tumor effect of cisplatin. In conclusion, nasal administration of MSC may represent a powerful, non-invasive, and safe regenerative treatment for resolution of chemobrain.
Collapse
|
48
|
Sun J, Zhang Y, Song X, Zhu J, Zhu Q. The Healing Effects of Conditioned Medium Derived from Mesenchymal Stem Cells on Radiation-Induced Skin Wounds in Rats. Cell Transplant 2018; 28:105-115. [PMID: 30350716 PMCID: PMC6322144 DOI: 10.1177/0963689718807410] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Radioactive dermatitis is caused by the exposure of skin and mucous membranes to radiation fields. The pathogenesis of radioactive dermatitis is complex and difficult to cure. Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) may serve as a promising candidate for the therapy of cutaneous wounds. The aim of this study was to investigate whether a WJ-MSC-derived conditioned medium (MSC-CM) could be used to treat radiation-induced skin wounds in rats using a radiation-induced cutaneous injury model. The present study was designed to examine MSC-CM therapy in the recovery of radiation-induced skin wounds in vitro and in vivo. Firstly, we prepared the MSC-CM and tested the effects of the MSC-CM on human umbilical vein endothelial cell proliferation in vitro. After that, we used a β-ray beam to make skin wounds in rats and tested the effects of MSC-CM on cutaneous wound healing in vivo. Our results indicated that MSC-CM secreted factors that promoted HUVEC proliferation, regeneration of sebaceous glands, and angiogenesis. Importantly, MSC-CM promoted wound healing in excess of the positive control (epidermal growth factor), with no, or smaller, scar formation. In conclusion, MSC-CM significantly accelerated wound closure and enhanced the wound healing quality. MSC-CM has a beneficial therapeutic effect on radiation-induced cutaneous injury skin in rats and in this way MSC-CM may serve as a basis of a novel cell-free therapeutic approach for radiation dermatitis.
Collapse
Affiliation(s)
- JiaYang Sun
- 1 Department of Orthopedics, China-Japan Union of Jilin University, Changchun, Jilin, China
| | - YunFeng Zhang
- 1 Department of Orthopedics, China-Japan Union of Jilin University, Changchun, Jilin, China
| | - XianJi Song
- 1 Department of Orthopedics, China-Japan Union of Jilin University, Changchun, Jilin, China
| | - Jiajing Zhu
- 2 Department of Radiology, China-Japan Union of Jilin University, Changchun, Jilin, China
| | - QingSan Zhu
- 1 Department of Orthopedics, China-Japan Union of Jilin University, Changchun, Jilin, China
| |
Collapse
|
49
|
Appraisal of mechanisms of radioprotection and therapeutic approaches of radiation countermeasures. Biomed Pharmacother 2018; 106:610-617. [DOI: 10.1016/j.biopha.2018.06.150] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/24/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022] Open
|
50
|
Leavitt RJ, Limoli CL, Baulch JE. miRNA-based therapeutic potential of stem cell-derived extracellular vesicles: a safe cell-free treatment to ameliorate radiation-induced brain injury. Int J Radiat Biol 2018; 95:427-435. [PMID: 30252569 DOI: 10.1080/09553002.2018.1522012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE This review compiles what is known about extracellular vesicles (EVs), their bioactive cargo, and how they might be used to treat radiation-induced brain injury. Radiotherapy (RT) is effective in cancer treatment, but can cause substantial damage to normal central nervous system tissue. Stem cell therapy has been shown to be effective in treating cognitive dysfunction arising from RT, but there remain safety concerns when grafting foreign stem cells into the brain (i.e. immunogenicity, teratoma). These limitations prompted the search for cell-free alternatives, and pointed to EVs that have been shown to have similar ameliorating effects in other tissues and injury models. CONCLUSIONS EVs are nano-scale and lipid-bound vesicles that readily pass the blood-brain barrier. Arguably the most important bioactive cargo within EVs are RNAs, in particular microRNAs (miRNA). A single miRNA can modulate entire gene networks and signalling within the recipient cell. Determining functionally relevant miRNA could lead to therapeutic treatments where synthetically-derived EVs are used as delivery vectors for miRNA. Stem cell-derived EVs can be effective in treating brain injury including radiation-induced cognitive deficits. Of particular interest are systemic modes of administration which obviate the need for invasive procedures.
Collapse
Affiliation(s)
- Ron J Leavitt
- a Department of Radiation Oncology , University of California Irvine , Irvine , CA , USA
| | - Charles L Limoli
- a Department of Radiation Oncology , University of California Irvine , Irvine , CA , USA
| | - Janet E Baulch
- a Department of Radiation Oncology , University of California Irvine , Irvine , CA , USA
| |
Collapse
|