1
|
Achón Buil B, Rentsch NH, Weber RZ, Rickenbach C, Halliday SJ, Hotta A, Tackenberg C, Rust R. Beneath the radar: immune-evasive cell sources for stroke therapy. Trends Mol Med 2024; 30:223-238. [PMID: 38272713 DOI: 10.1016/j.molmed.2023.12.004] [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: 10/16/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 01/27/2024]
Abstract
Stem cell therapy is an emerging treatment paradigm for stroke patients with remaining neurological deficits. While allogeneic cell transplants overcome the manufacturing constraints of autologous grafts, they can be rejected by the recipient's immune system, which identifies foreign cells through the human leukocyte antigen (HLA) system. The heterogeneity of HLA molecules in the human population would require a very high number of cell lines, which may still be inadequate for patients with rare genetic HLAs. Here, we outline key progress in genetic HLA engineering in pluripotent stem and derived cells to evade the host's immune system, reducing the number of allogeneic cell lines required, and examine safety measures explored in both preclinical studies and upcoming clinical trials.
Collapse
Affiliation(s)
- Beatriz Achón Buil
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Nora H Rentsch
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Rebecca Z Weber
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Chiara Rickenbach
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Stefanie J Halliday
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Akitsu Hotta
- Center for iPS cell Research and Application, Kyoto University, Kyoto, Japan
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Ruslan Rust
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland; Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo St, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Duchniewicz M, Lee JYW, Menon DK, Needham EJ. Candidate Genetic and Molecular Drivers of Dysregulated Adaptive Immune Responses After Traumatic Brain Injury. J Neurotrauma 2024; 41:3-12. [PMID: 37376743 DOI: 10.1089/neu.2023.0187] [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: 06/29/2023] Open
Abstract
Abstract Neuroinflammation is a significant and modifiable cause of secondary injury after traumatic brain injury (TBI), driven by both central and peripheral immune responses. A substantial proportion of outcome after TBI is genetically mediated, with an estimated heritability effect of around 26%, but because of the comparatively small datasets currently available, the individual drivers of this genetic effect have not been well delineated. A hypothesis-driven approach to analyzing genome-wide association study (GWAS) datasets reduces the burden of multiplicity testing and allows variants with a high prior biological probability of effect to be identified where sample size is insufficient to withstand data-driven approaches. Adaptive immune responses show substantial genetically mediated heterogeneity and are well established as a genetic source of risk for numerous disease states; importantly, HLA class II has been specifically identified as a locus of interest in the largest TBI GWAS study to date, highlighting the importance of genetic variance in adaptive immune responses after TBI. In this review article we identify and discuss adaptive immune system genes that are known to confer strong risk effects for human disease, with the dual intentions of drawing attention to this area of immunobiology, which, despite its importance to the field, remains under-investigated in TBI and presenting high-yield testable hypotheses for application to TBI GWAS datasets.
Collapse
Affiliation(s)
- Michał Duchniewicz
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - John Y W Lee
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - David K Menon
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Edward J Needham
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Liu J, van Beusekom H, Bu X, Chen G, Henrique Rosado de Castro P, Chen X, Chen X, Clarkson AN, Farr TD, Fu Y, Jia J, Jolkkonen J, Kim WS, Korhonen P, Li S, Liang Y, Liu G, Liu G, Liu Y, Malm T, Mao X, Oliveira JM, Modo MM, Ramos‐Cabrer P, Ruscher K, Song W, Wang J, Wang X, Wang Y, Wu H, Xiong L, Yang Y, Ye K, Yu J, Zhou X, Zille M, Masters CL, Walczak P, Boltze J, Ji X, Wang Y. Preserving cognitive function in patients with Alzheimer's disease: The Alzheimer's disease neuroprotection research initiative (ADNRI). NEUROPROTECTION 2023; 1:84-98. [PMID: 38223913 PMCID: PMC10783281 DOI: 10.1002/nep3.23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 01/16/2024]
Abstract
The global trend toward aging populations has resulted in an increase in the occurrence of Alzheimer's disease (AD) and associated socioeconomic burdens. Abnormal metabolism of amyloid-β (Aβ) has been proposed as a significant pathomechanism in AD, supported by results of recent clinical trials using anti-Aβ antibodies. Nonetheless, the cognitive benefits of the current treatments are limited. The etiology of AD is multifactorial, encompassing Aβ and tau accumulation, neuroinflammation, demyelination, vascular dysfunction, and comorbidities, which collectively lead to widespread neurodegeneration in the brain and cognitive impairment. Hence, solely removing Aβ from the brain may be insufficient to combat neurodegeneration and preserve cognition. To attain effective treatment for AD, it is necessary to (1) conduct extensive research on various mechanisms that cause neurodegeneration, including advances in neuroimaging techniques for earlier detection and a more precise characterization of molecular events at scales ranging from cellular to the full system level; (2) identify neuroprotective intervention targets against different neurodegeneration mechanisms; and (3) discover novel and optimal combinations of neuroprotective intervention strategies to maintain cognitive function in AD patients. The Alzheimer's Disease Neuroprotection Research Initiative's objective is to facilitate coordinated, multidisciplinary efforts to develop systemic neuroprotective strategies to combat AD. The aim is to achieve mitigation of the full spectrum of pathological processes underlying AD, with the goal of halting or even reversing cognitive decline.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Heleen van Beusekom
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MCUniversity Medical CenterRotterdamThe Netherlands
| | - Xian‐Le Bu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Gong Chen
- Guangdong‐HongKong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouGuangdongChina
| | | | - Xiaochun Chen
- Fujian Key Laboratory of Molecular Neurology, Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Institute of NeuroscienceFujian Medical UniversityFuzhouFujianChina
| | - Xiaowei Chen
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
- Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| | - Andrew N. Clarkson
- Department of Anatomy, Brain Health Research Centre and Brain Research New ZealandUniversity of OtagoDunedinNew Zealand
| | - Tracy D. Farr
- School of Life SciencesUniversity of NottinghamNottinghamUK
| | - Yuhong Fu
- Brain and Mind Centre & School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Clinical Research Center for Geriatric DiseasesCapital Medical UniversityBeijingChina
| | - Jukka Jolkkonen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Woojin Scott Kim
- Brain and Mind Centre & School of Medical SciencesThe University of SydneySydneyNew South WalesAustralia
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Guang‐Hui Liu
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Membrane Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Yu‐Hui Liu
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopioFinland
| | - Xiaobo Mao
- Institute for Cell Engineering, Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative MedicineUniversity of MinhoGuimarãesPortugal
- ICVS/3B's—PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Mike M. Modo
- Department of Bioengineering, McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Department of Radiology, McGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Pedro Ramos‐Cabrer
- Magnetic Resonance Imaging LaboratoryCIC BiomaGUNE Research Center, Basque Research and Technology Alliance (BRTA)Donostia‐San SebastianSpain
| | - Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical SciencesLund UniversityLundSweden
| | - Weihong Song
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province. Zhejiang Clinical Research Center for Mental Disorders, School of Mental Health and The Affiliated Kangning Hospital, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou Medical UniversityZhejiangChina
| | - Jun Wang
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
| | - Xuanyue Wang
- School of Optometry and Vision ScienceUniversity of New South WalesSydneyNew South WalesAustralia
| | - Yun Wang
- Neuroscience Research Institute, Department of Neurobiology, School of Basic, Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National, Health Commission and State Key Laboratory of Natural and Biomimetic DrugsPeking UniversityBeijingChina
- PKU‐IDG/McGovern Institute for Brain ResearchPeking UniversityBeijingChina
| | - Haitao Wu
- Department of NeurobiologyBeijing Institute of Basic Medical SciencesBeijingChina
| | - Lize Xiong
- Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain‐Like Intelligence, Shanghai Fourth People's Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yi Yang
- Department of NeurologyThe First Hospital of Jilin University, Chang ChunJilinChina
| | - Keqiang Ye
- Faculty of Life and Health SciencesBrain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced TechnologyShenzhenChina
| | - Jin‐Tai Yu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xin‐Fu Zhou
- Division of Health Sciences, School of Pharmacy and Medical Sciences and Sansom InstituteUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- Suzhou Auzone BiotechSuzhouJiangsuChina
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and ToxicologyUniversity of ViennaViennaAustria
| | - Colin L. Masters
- The Florey InstituteThe University of Melbourne, ParkvilleVictoriaAustralia
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | | | - Xunming Ji
- Department of NeurosurgeryXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Yan‐Jiang Wang
- Department of Neurology, Daping HospitalThird Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
- Guangyang Bay LaboratoryChongqing Institute for Brain and IntelligenceChongqingChina
- Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghaiChina
| |
Collapse
|
4
|
Lan XY, Kalkowski L, Chu CY, Jablonska A, Li S, Kai M, Gao Y, Janowski M, Walczak P. Unlocking the potential of ultra-high dose fractionated radiation for effective treatment of glioblastoma. RESEARCH SQUARE 2023:rs.3.rs-3500563. [PMID: 37961626 PMCID: PMC10635404 DOI: 10.21203/rs.3.rs-3500563/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Conventional radiation therapy for glioblastoma (GBM) has limited efficacy. Regenerative medicine brings hope for repairing damaged tissue, opening opportunities for elevating the maximum acceptable radiation dose. In this study, we explored the effect of ultra-high dose fractionated radiation on brain injury and tumor responses in immunocompetent mice. We also evaluated the role of the HIF-1α under radiation. Methods Naïve and hypoxia-inducible factor-1 alpha (HIF-1α)+/- heterozygous mice received a fractionated daily dose of 20 Gy for three or five consecutive days. Magnetic resonance imaging (MRI) and histology were performed to assess brain injury post-radiation. The 2×105 human GBM1 luciferase-expressing cells were transplanted with tolerance induction protocol. Fractionated radiotherapy was performed during the exponential phase of tumor growth. BLI, MRI, and immunohistochemistry staining were performed to evaluate tumor growth dynamics and radiotherapy responses. Additionally, animal lifespan was recorded. Results Fractionated radiation of 5×20 Gy induced severe brain damage, starting 3 weeks after radiation. All animals from this group died within 12 weeks. In contrast, later onset and less severe brain injury were observed starting 12 weeks after radiation of 3×20 Gy. It resulted in complete GBM eradication and survival of all treated animals. Furthermore, HIF-1α+/- mice exhibited more obvious vascular damage 63 weeks after fractionated radiation of 3×20 Gy. Conclusion Ultra-high dose fractionated 3×20 Gy radiation can eradicate the GBM cells at the cost of only mild brain injury. The HIF-1α gene is a promising target for ameliorating vascular impairment post-radiation, encouraging the implementation of neurorestorative strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yue Gao
- University of Maryland Baltimore
| | | | | |
Collapse
|
5
|
Wang F, Sun Z, Peng D, Gianchandani S, Le W, Boltze J, Li S. Cell-therapy for Parkinson's disease: a systematic review and meta-analysis. J Transl Med 2023; 21:601. [PMID: 37679754 PMCID: PMC10483810 DOI: 10.1186/s12967-023-04484-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Cell-based strategies focusing on replacement or protection of dopaminergic neurons have been considered as a potential approach to treat Parkinson's disease (PD) for decades. However, despite promising preclinical results, clinical trials on cell-therapy for PD reported mixed outcomes and a thorough synthesis of these findings is lacking. We performed a systematic review and meta-analysis to evaluate cell-therapy for PD patients. METHODS We systematically identified all clinical trials investigating cell- or tissue-based therapies for PD published before July 2023. Out of those, studies reporting transplantation of homogenous cells (containing one cell type) were included in meta-analysis. The mean difference or standardized mean difference in quantitative neurological scale scores before and after cell-therapy was analyzed to evaluate treatment effects. RESULTS The systematic literature search revealed 106 articles. Eleven studies reporting data from 11 independent trials (210 patients) were eligible for meta-analysis. Disease severity and motor function evaluation indicated beneficial effects of homogenous cell-therapy in the 'off' state at 3-, 6-, 12-, or 24-month follow-ups, and for motor function even after 36 months. Most of the patients were levodopa responders (61.6-100% in different follow-ups). Cell-therapy was also effective in improving the daily living activities in the 'off' state of PD patients. Cells from diverse sources were used and multiple transplantation modes were applied. Autografts did not improve functional outcomes, while allografts exhibited beneficial effects. Encouragingly, both transplantation into basal ganglia and to areas outside the basal ganglia were effective to reduce disease severity. Some trials reported adverse events potentially related to the surgical procedure. One confirmed and four possible cases of graft-induced dyskinesia were reported in two trials included in this meta-analysis. CONCLUSIONS This meta-analysis provides preliminary evidence for the beneficial effects of homogenous cell-therapy for PD, potentially to the levodopa responders. Allogeneic cells were superior to autologous cells, and the effective transplantation sites are not limited to the basal ganglia. PROSPERO registration number: CRD42022369760.
Collapse
Affiliation(s)
- Fang Wang
- Department of Neurology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Zhengwu Sun
- Department of Clinical Pharmacy, Central Hospital of Dalian University of Technology, Dalian, China
| | - Daoyong Peng
- Department of Neurology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Shikha Gianchandani
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Weidong Le
- Institute of Neurology, Sichuan Academy of Medical Sciences, Sichuan Provincial Hospital, Chengdu, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Beijing, 100038, China.
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Foss A, Pathania M. Pediatric Glioma Models Provide Insights into Tumor Development and Future Therapeutic Strategies. Dev Neurosci 2023; 46:22-43. [PMID: 37231843 DOI: 10.1159/000531040] [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: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
In depth study of pediatric gliomas has been hampered due to difficulties in accessing patient tissue and a lack of clinically representative tumor models. Over the last decade, however, profiling of carefully curated cohorts of pediatric tumors has identified genetic drivers that molecularly segregate pediatric gliomas from adult gliomas. This information has inspired the development of a new set of powerful in vitro and in vivo tumor models that can aid in identifying pediatric-specific oncogenic mechanisms and tumor microenvironment interactions. Single-cell analyses of both human tumors and these newly developed models have revealed that pediatric gliomas arise from spatiotemporally discrete neural progenitor populations in which developmental programs have become dysregulated. Pediatric high-grade gliomas also harbor distinct sets of co-segregating genetic and epigenetic alterations, often accompanied by unique features within the tumor microenvironment. The development of these novel tools and data resources has led to insights into the biology and heterogeneity of these tumors, including identification of distinctive sets of driver mutations, developmentally restricted cells of origin, recognizable patterns of tumor progression, characteristic immune environments, and tumor hijacking of normal microenvironmental and neural programs. As concerted efforts have broadened our understanding of these tumors, new therapeutic vulnerabilities have been identified, and for the first time, promising new strategies are being evaluated in the preclinical and clinical settings. Even so, dedicated and sustained collaborative efforts are necessary to refine our knowledge and bring these new strategies into general clinical use. In this review, we will discuss the range of currently available glioma models, the way in which they have each contributed to recent developments in the field, their benefits and drawbacks for addressing specific research questions, and their future utility in advancing biological understanding and treatment of pediatric glioma.
Collapse
Affiliation(s)
- Amelia Foss
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Manav Pathania
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Gao Y, Li D, Lin J, Thomas AM, Miao J, Chen D, Li S, Chu C. Cerebral small vessel disease: Pathological mechanisms and potential therapeutic targets. Front Aging Neurosci 2022; 14:961661. [PMID: 36034144 PMCID: PMC9412755 DOI: 10.3389/fnagi.2022.961661] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) represents a diverse cluster of cerebrovascular diseases primarily affecting small arteries, capillaries, arterioles and venules. The diagnosis of CSVD relies on the identification of small subcortical infarcts, lacunes, white matter hyperintensities, perivascular spaces, and microbleeds using neuroimaging. CSVD is observed in 25% of strokes worldwide and is the most common pathology of cognitive decline and dementia in the elderly. Still, due to the poor understanding of pathophysiology in CSVD, there is not an effective preventative or therapeutic approach for CSVD. The most widely accepted approach to CSVD treatment is to mitigate vascular risk factors and adopt a healthier lifestyle. Thus, a deeper understanding of pathogenesis may foster more specific therapies. Here, we review the underlying mechanisms of pathological characteristics in CSVD development, with a focus on endothelial dysfunction, blood-brain barrier impairment and white matter change. We also describe inflammation in CSVD, whose role in contributing to CSVD pathology is gaining interest. Finally, we update the current treatments and preventative measures of CSVD, as well as discuss potential targets and novel strategies for CSVD treatment.
Collapse
Affiliation(s)
- Yue Gao
- Department of Neurointervention and Neurological Intensive Care, Dalian Municipal Central Hospital, Dalian, China
| | - Di Li
- Department of Neurointervention and Neurological Intensive Care, Dalian Municipal Central Hospital, Dalian, China
| | - Jianwen Lin
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Aline M. Thomas
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, United States
| | - Jianyu Miao
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Dong Chen
- Department of Neurosurgery, Dalian Municipal Central Hospital, Dalian, China
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chengyan Chu
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
- *Correspondence: Chengyan Chu,
| |
Collapse
|
8
|
Li S, Rao JH, Lan XY, Li X, Chu CY, Liang Y, Janowski M, Zhang HT, Walczak P. White matter demyelination predates axonal injury after ischemic stroke in cynomolgus monkeys. Exp Neurol 2021; 340:113655. [PMID: 33617887 DOI: 10.1016/j.expneurol.2021.113655] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/11/2021] [Accepted: 02/16/2021] [Indexed: 02/02/2023]
Abstract
Unraveling the pathology of stroke is a prerequisite for designing therapeutic strategies. It was reported that myelin injury exceeded axonal loss in the peri-infarct region of rodent white matter stroke. An in-depth investigation of the post-stroke white matter damage in higher-order species might innovate stroke intervention. In this study, adult male cynomolgus monkeys received surgical middle cerebral artery occlusion (MCAO), and serial magnetic resonance scans to non-invasively assess brain damage. Spontaneous movements were recorded to evaluate post-stroke behavior. The axon and myelin loss, as well as immune cell infiltration were examined using immunohistochemistry. Magnetic resonance imaging revealed cerebral infarcts and white matter injury after MCAO in monkeys, which were confirmed by neurological deficits. Immunostaining of white matter fibers showed substantial demyelination whilst retention of axons in the infarcts 8 days post MCAO, while a progressive loss of myelin and axons was observed after one month. Gliosis, microglia activation, and leukocyte infiltration were identified in the lesions. These results demonstrate that demyelination predates axonal injury in non-human primate ischemic stroke, which provides a time window for stroke intervention focusing on prevention of progressive axonal loss through myelin regeneration.
Collapse
Affiliation(s)
- Shen Li
- Department of Neurology, Dalian Municipal Central Hospital affiliated with Dalian Medical University, Dalian, China
| | - Jun-Hua Rao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangdong, China
| | - Xiao-Yan Lan
- Department of Neurology, Dalian Municipal Central Hospital affiliated with Dalian Medical University, Dalian, China; Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Xu Li
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Cheng-Yan Chu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Hong-Tian Zhang
- Bayi Brain Hospital affiliated with the 7th Medical Center of PLA General Hospital, Beijing, China.
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA.
| |
Collapse
|
9
|
Kuddannaya S, Zhu W, Chu C, Singh A, Walczak P, Bulte JWM. In Vivo Imaging of Allografted Glial-Restricted Progenitor Cell Survival and Hydrogel Scaffold Biodegradation. ACS APPLIED MATERIALS & INTERFACES 2021; 13:23423-23437. [PMID: 33978398 PMCID: PMC9440547 DOI: 10.1021/acsami.1c03415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Transplanted glial-restricted progenitor (GRP) cells have potential to focally replace defunct astrocytes and produce remyelinating oligodendrocytes to avert neuronal death and dysfunction. However, most central nervous system cell therapeutic paradigms are hampered by high initial cell death and a host anti-graft immune response. We show here that composite hyaluronic acid-based hydrogels of tunable mechanical strengths can significantly improve transplanted GRP survival and differentiation. Allogeneic GRPs expressing green fluorescent protein and firefly luciferase were scaffolded in optimized hydrogel formulations and transplanted intracerebrally into immunocompetent BALB/c mice followed by serial in vivo bioluminescent imaging and chemical exchange saturation transfer magnetic resonance imaging (CEST MRI). We demonstrate that gelatin-sensitive CEST MRI can be exploited to monitor hydrogel scaffold degradation in vivo for ∼5 weeks post transplantation without necessitating exogenous labeling. Hydrogel scaffolding of GRPs resulted in a 4.5-fold increase in transplanted cell survival at day 32 post transplantation compared to naked cells. Histological analysis showed significant enhancement of cell proliferation as well as Olig2+ and GFAP+ cell differentiation for scaffolded cells compared to naked cells, with reduced host immunoreactivity. Hence, hydrogel scaffolding of transplanted GRPs in conjunction with serial in vivo imaging of cell survival and hydrogel degradation has potential for further advances in glial cell therapy.
Collapse
Affiliation(s)
- Shreyas Kuddannaya
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Wei Zhu
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Chengyan Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Anirudha Singh
- Department of Urology, the James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, Maryland 21287, United States
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Piotr Walczak
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| | - Jeff W M Bulte
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Chemical & Biomolecular Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
10
|
Wang R, Chu C, Wei Z, Chen L, Xu J, Liang Y, Janowski M, Stevens RD, Walczak P. Traumatic brain injury does not disrupt costimulatory blockade-induced immunological tolerance to glial-restricted progenitor allografts. J Neuroinflammation 2021; 18:104. [PMID: 33931070 PMCID: PMC8088005 DOI: 10.1186/s12974-021-02152-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 04/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell transplantation-based treatments for neurological disease are promising, yet graft rejection remains a major barrier to successful regenerative therapies. Our group and others have shown that long-lasting tolerance of transplanted stem cells can be achieved in the brain with systemic application of monoclonal antibodies blocking co-stimulation signaling. However, it is unknown if subsequent injury and the blood-brain barrier breach could expose the transplanted cells to systemic immune system spurring fulminant rejection and fatal encephalitis. Therefore, we investigated whether delayed traumatic brain injury (TBI) could trigger graft rejection. METHODS Glial-restricted precursor cells (GRPs) were intracerebroventricularly transplanted in immunocompetent neonatal mice and co-stimulation blockade (CoB) was applied 0, 2, 4, and 6 days post-grafting. Bioluminescence imaging (BLI) was performed to monitor the grafted cell survival. Mice were subjected to TBI 12 weeks post-transplantation. MRI and open-field test were performed to assess the brain damage and behavioral change, respectively. The animals were decapitated at week 16 post-transplantation, and the brains were harvested. The survival and distribution of grafted cells were verified from brain sections. Hematoxylin and eosin staining (HE) was performed to observe TBI-induced brain legion, and neuroinflammation was evaluated immunohistochemically. RESULTS BLI showed that grafted GRPs were rejected within 4 weeks after transplantation without CoB, while CoB administration resulted in long-term survival of allografts. BLI signal had a steep rise following TBI and subsequently declined but remained higher than the preinjury level. Open-field test showed TBI-induced anxiety for all animals but neither CoB nor GRP transplantation intensified the symptom. HE and MRI demonstrated a reduction in TBI-induced lesion volume in GRP-transplanted mice compared with non-transplanted mice. Brain sections further validated the survival of grafted GRPs and showed more GRPs surrounding the injured tissue. Furthermore, the brains of post-TBI shiverer mice had increased activation of microglia and astrocytes compared to post-TBI wildtype mice, but infiltration of CD45+ leukocytes remained low. CONCLUSIONS CoB induces sustained immunological tolerance towards allografted cerebral GRPs which is not disrupted following TBI, and unexpectedly TBI may enhance GRPs engraftment and contribute to post-injury brain tissue repair.
Collapse
Affiliation(s)
- Rui Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,Departments of Anesthesiology and Critical Care Medicine, Neurology, Neurosurgery, Johns Hopkins University, Baltimore, MD, 21287, USA.,Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110006, Liaoning, China
| | - Chengyan Chu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, 670 W. Baltimore St., HSF III rm 1176, Baltimore, MD, 21201, USA
| | - Zhiliang Wei
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, 21205, USA
| | - Lin Chen
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, 21205, USA
| | - Jiadi Xu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, 21205, USA
| | - Yajie Liang
- Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, 670 W. Baltimore St., HSF III rm 1176, Baltimore, MD, 21201, USA
| | - Miroslaw Janowski
- Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, 670 W. Baltimore St., HSF III rm 1176, Baltimore, MD, 21201, USA
| | - Robert D Stevens
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,Departments of Anesthesiology and Critical Care Medicine, Neurology, Neurosurgery, Johns Hopkins University, Baltimore, MD, 21287, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, 21205, USA
| | - Piotr Walczak
- Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, 670 W. Baltimore St., HSF III rm 1176, Baltimore, MD, 21201, USA.
| |
Collapse
|
11
|
Forbes LH, Andrews MR. Advances in human stem cell therapies: pre-clinical studies and the outlook for central nervous system regeneration. Neural Regen Res 2021; 16:614-617. [PMID: 33063709 PMCID: PMC8067917 DOI: 10.4103/1673-5374.295287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cell transplantation has come to the forefront of regenerative medicine alongside the discovery and application of stem cells in both research and clinical settings. There are several types of stem cells currently being used for pre-clinical regenerative therapies, each with unique characteristics, benefits and limitations. This brief review will focus on recent basic science advancements made with embryonic stem cells and induced pluripotent stem cells. Both embryonic stem cells and induced pluripotent stem cells provide platforms for new neurons to replace dead and/or dying cells following injury. Due to their capacity for reprogramming and differentiation into any neuronal type, research in preclinical rodent models has shown that embryonic stem cells and induced pluripotent stem cells can integrate, survive and form connections in the nervous system similar to de novo cells. Going forward however, there are some limitations to consider with the use of either stem cell type. Ethically, embryonic stem cells are not an ideal source of cells, genetically, induced pluripotent stem cells are not ideal in terms of personalized treatment for those with certain genetic diseases the latter of which may guide regenerative medicine away from personalized stem cell based therapies and into optimized stem cell banks. Nonetheless, the potential of these stem cells in central nervous system regenerative therapy is only beginning to be appreciated. For example, through genetic modification, stem cells serve as ideal platforms to reintroduce missing or downregulated molecules into the nervous system to further induce regenerative growth. In this review, we highlight the limitations of stem cell based therapies whilst discussing some of the means of overcoming these limitations.
Collapse
Affiliation(s)
- Lindsey H Forbes
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Melissa R Andrews
- School of Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
12
|
Mozafari S, Baron-Van Evercooren A. Human stem cell-derived oligodendrocytes: From humanized animal models to cell therapy in myelin diseases. Semin Cell Dev Biol 2020; 116:53-61. [PMID: 33082116 DOI: 10.1016/j.semcdb.2020.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes are main targets in demyelinating and dysmyelinating diseases of the central nervous system (CNS), but are also involved in accidental, neurodegenerative and psychiatric disorders. The underlying pathology of these diseases is not fully understood and treatments are still lacking. The recent discovery of the induced pluripotent stem cell (iPSC) technology has open the possibility to address the biology of human oligodendroglial cells both in the dish and in vivo via engraftment in animal models, and paves the way for the development of treatment for myelin disorders. In this review, we make a short overview of the different sources human oligodendroglial cells, and animal models available for pre-clinical cell therapy. We discuss the anatomical and functional benefit of grafted iPSC-progenitors over their brain counterparts, their use in disease modeling and the missing gaps that still prevent to study their biology in the most integrated way, and to translate iPSC-stem cell based therapy to the clinic.
Collapse
Affiliation(s)
- Sabah Mozafari
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France.
| |
Collapse
|
13
|
Nemeth CL, Tomlinson SN, Sharma R, Sharma A, Kannan S, Kannan RM, Fatemi A. Glial restricted precursor delivery of dendrimer N-acetylcysteine promotes migration and differentiation following transplant in mouse white matter injury model. NANOSCALE 2020; 12:16063-16068. [PMID: 32724988 PMCID: PMC7448752 DOI: 10.1039/c9nr10804a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Oligodendrocyte replacement using glial restricted precursors (GRPs) is a promising avenue for the treatment of acquired or genetic white matter disorders; however, limited long-term survival of these cells post-transplant may impede maximal recovery. Nanotherapeutic approaches can facilitate stem cell delivery while simultaneously delivering factors aimed at enhancing and nourishing stem cells en route to, and at, the target site. Hydroxyl polyamidoamine (PAMAM) dendrimer nanoparticles have been used in a variety of models to deliver therapeutics in a targeted manner to injury sites at low doses. Here, survival and migration of GRPs was assessed in a mouse model of neonatal white matter injury with different methods of dendrimer nanoparticle support. Our findings demonstrate the ability of GRPs to take up nanoparticle-drug conjugates and for these conjugates to act beyond the injury site in vivo. Compared to GRPs alone, mice receiving dendrimer-drug in parallel to GRPs, or via GRPs as the delivery vector, showed improved migration and differentiation of cells 8 weeks post-transplant. These studies demonstrate that drug-conjugated nanoparticles can enhance transplanted progenitor cell survival and migration, and suggest that combination therapies may allow engraftment without overt immunosuppression.
Collapse
Affiliation(s)
- Christina L Nemeth
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Lan X, Kedziorek DA, Chu C, Jablonska A, Li S, Kai M, Liang Y, Janowski M, Walczak P. Modeling human pediatric and adult gliomas in immunocompetent mice through costimulatory blockade. Oncoimmunology 2020; 9:1776577. [PMID: 32923139 PMCID: PMC7458632 DOI: 10.1080/2162402x.2020.1776577] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Currently, human glioma tumors are mostly modeled in immunodeficient recipients; however, lack of interactions with adaptive immune system is a serious flaw, particularly in the era when immunotherapies dominate treatment strategies. Our group was the first to successfully establish the orthotopic transplantation of human glioblastoma (GBM) in immunocompetent mice by inducing immunological tolerance using a short-term, systemic costimulation blockade strategy (CTLA-4-Ig and MR1). In this study, we further validated the feasibility of this method by modeling pediatric diffuse intrinsic pontine glioma (DIPG) and two types of adult GBM (GBM1, GBM551), in mice with intact immune systems and immunodeficient mice. We found that all three glioma models were successfully established, with distinct difference in tumor growth patterns and morphologies, after orthotopic xenotransplantation in tolerance-induced immunocompetent mice. Long-lasting tolerance that is maintained for up to nearly 200 d in GBM551 confirmed the robustness of this model. Moreover, we found that tumors in immunocompetent mice displayed features more similar to the clinical pathophysiology found in glioma patients, characterized by inflammatory infiltration and strong neovascularization, as compared with tumors in immunodeficient mice. In summary, we have validated the robustness of the costimulatory blockade strategy for tumor modeling and successfully established three human glioma models including the pediatric DIPG whose preclinical study is particularly thwarted by the lack of proper animal models.
Collapse
Affiliation(s)
- Xiaoyan Lan
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Dorota A Kedziorek
- Russel H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chengyan Chu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Anna Jablonska
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Shen Li
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Mihoko Kai
- Department of Radiation Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| |
Collapse
|