1
|
Krishnan B, Marcatti M, Fracassi A, Zhang WR, Guptarak J, Johnson K, Grant A, Kayed R, Taglialatela G, Micci MA. Hippocampal Neural Stem Cell Exosomes Promote Brain Resilience against the Impact of Tau Oligomers. J Neurosci 2025; 45:e1664242025. [PMID: 40050115 PMCID: PMC12005371 DOI: 10.1523/jneurosci.1664-24.2025] [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: 09/27/2024] [Revised: 12/20/2024] [Accepted: 01/28/2025] [Indexed: 04/19/2025] Open
Abstract
A promising therapeutic intervention for preventing the onset and progression of Alzheimer's disease is to protect and improve synaptic resilience, a well-established early vulnerability associated with the toxic effects of oligomers of amyloid β (AβO) and Tau (TauO). We have previously reported that exosomes from hippocampal neural stem cells (NSCs) protect synapses against AβO. Here, we demonstrate how exosomes can also shield against TauO toxicity in adult mice synapses, potentially benefiting primary and secondary tauopathies. Exosomes from hippocampal NSCs (NSCexo) or mature neurons (MNexo) were delivered intracerebroventricularly to adult wild-type male mice (C57Bl6/J). After 24 h, TauO were administered to suppress long-term potentiation (LTP) and memory, measured by electrophysiology and contextual memory deficits measured using novel object recognition test. We also assessed TauO binding to synapses using isolated synaptosomes and cultured hippocampal neurons. Furthermore, mimics of select miRNAs present in NSCexo were delivered intracerebroventricularly to mice prior to assessment of TauO-induced suppression of hippocampal LTP. Our results showed that NSC-, not MN-, derived exosomes, prevented TauO-induced memory impairment, LTP suppression, and reduced Tau accumulation and TauO internalization in synaptosomes. These findings suggest that NSC-derived exosomes can protect against synaptic dysfunction and memory deficits induced by both AβO and TauO, offering a novel therapeutic strategy for multiple neurodegenerative states.
Collapse
Affiliation(s)
- Balaji Krishnan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Michela Marcatti
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Anna Fracassi
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Wen-Ru Zhang
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Jutatip Guptarak
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Kathia Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Auston Grant
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Giulio Taglialatela
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas 77555
| |
Collapse
|
2
|
Dickstein DL, Zhang R, Ru N, Vozenin MC, Perry BC, Wang J, Baulch JE, Acharya MM, Limoli CL. Structural plasticity of pyramidal cell neurons measured after FLASH and conventional dose-rate irradiation. Brain Struct Funct 2025; 230:41. [PMID: 40024988 PMCID: PMC11872753 DOI: 10.1007/s00429-025-02902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Evidence shows that ultra-high dose-rate FLASH-radiotherapy (FLASH-RT) provides relative protection against normal tissue complications and functional decrements in the irradiated brain. Past work has shown that radiation-induced cognitive impairment, neuroinflammation and reduced structural complexity ofgranule cell neurons were not observed to the same extent after FLASH-RT (> MGy/s) compared to conventional dose-rate (CONV, 0.1 Gy/s) delivery. In this study, we explored the sensitivity of hippocampal CA1 and medial prefrontal cortex (mPFC) pyramidal neurons to cranial irradiation and dose-rate modulation using electron and confocal microscopy. Neuron ultrastructural analyses by electron microscopy after 10 Gy FLASH- or CONV-RT exposures indicated that irradiation had little impact on dendritic complexity and synapse density in the CA1, but did increase the length and head diameter of smaller non-perforated synapses. Similarly, irradiation caused no change in mPFC prelimbic/infralimbic axospinous synapse density, but reductions in non-perforated synapse diameters. While irradiation resulted in thinner myelin sheaths compared to controls, none of these metrics were dose-rate sensitive. Analysis of fluorescently labeled CA1 neurons revealed no radiation-induced or dose-rate-dependent changes in overall dendritic complexity or spine density, in contrast to our past analysis of granule cell neurons. Super-resolution confocal microscopy following a clinical dosing paradigm (3 × 10 Gy) showed significant reductions in excitatory vesicular glutamate transporter 1 and inhibitory vesicular GABA transporter puncta density within the CA1 that were largely dose-rate independent. Collectively, these data reveal that, compared to granule cell neurons, CA1 and mPFC neurons are relatively more radioresistant irrespective of radiation dose-rate.
Collapse
Affiliation(s)
- Dara L Dickstein
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA.
| | - Richard Zhang
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Ning Ru
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Marie-Catherine Vozenin
- Secteur Radio-Oncologie et Radiobiologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Bayley C Perry
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA
| | - Juan Wang
- Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD, 20814, USA
- The Henry M. Jackson Foundation for the Advancement of Military Health Inc, Bethesda, MD, USA
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA, USA.
| |
Collapse
|
3
|
Li X, Ding Z. Cognitive dysfunction induced by cranial radiotherapy: mechanisms and therapeutic methods. Brain Res Bull 2024; 218:111106. [PMID: 39447765 DOI: 10.1016/j.brainresbull.2024.111106] [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: 05/05/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Cranial radiotherapy can damage normal brain tissues, inducing cognitive dysfunction in patients. Radiotherapy-induced cognitive dysfunction is associated with hippocampal injury, white matter damage and microvascular injury. In this study, the mechanisms of cognitive dysfunction induced by cranial radiotherapy and combined chemoradiotherapy are reviewed, and the advances in therapeutic methods for radiotherapy-induced brain injury are summarized. The mechanisms of radiotherapy-induced brain injury include a decline of neurogenesis, impairment of neurons and glial cells, vascular injury, oxidative stress and DNA damage, cell death, and inflammatory response. Disruption of the bloodbrain barrier (BBB) increases the exposure of the brain to chemotherapeutic agents, thus exacerbating radiotherapy-induced brain damage. The current methods used to prevent radiotherapy-induced brain injury mainly include precision radiotherapy, stem cell transplantation, and treatment with neuroprotective drugs. The combined application of precision radiotherapy and neuroprotective drugs, including antioxidants, anti-inflammatory agents and other drugs, might exert better neuroprotective effects. To resolve the issues of neuroprotective drugs, such as difficulty in crossing the BBB, nanoenzymes and drug delivery nano-systems could be applied in the future.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
| |
Collapse
|
4
|
Drayson OGG, Montay-Gruel P, Limoli CL. Radiomics approach for identifying radiation-induced normal tissue toxicity in the lung. Sci Rep 2024; 14:24256. [PMID: 39415029 PMCID: PMC11484882 DOI: 10.1038/s41598-024-75993-y] [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: 02/12/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024] Open
Abstract
The rapidly evolving field of radiomics has shown that radiomic features are able to capture characteristics of both tumor and normal tissue that can be used to make accurate and clinically relevant predictions. In the present study we sought to determine if radiomic features can characterize the adverse effects caused by normal tissue injury as well as identify if human embryonic stem cell (hESC) derived extracellular vesicle (EV) treatment can resolve certain adverse complications. A cohort of 72 mice (n = 12 per treatment group) were exposed to X-ray radiation to the whole lung (3 × 8 Gy) or to the apex of the right lung (3 × 12 Gy), immediately followed by retro-orbital injection of EVs. Cone-Beam Computed Tomography images were acquired before and 2 weeks after treatment. In total, 851 radiomic features were extracted from the whole lungs and < 20 features were selected to train and validate a series of random forest classification models trained to predict radiation status, EV status and treatment group. It was found that all three classification models achieved significantly high prediction accuracies on a validation subset of the dataset (AUCs of 0.91, 0.86 and 0.80 respectively). In the locally irradiated lung, a significant difference between irradiated and unirradiated groups as well as an EV sparing effect were observed in several radiomic features that were not seen in the unirradiated lung (including wavelet-LLH Kurtosis, wavelet HLL Large Area High Gray Level Emphasis, and Gray Level Non-Uniformity). Additionally, a radiation difference was not observed in a secondary comparison cohort, but there was no impact of imaging machine parameters on the radiomic signature of unirradiated mice. Our data demonstrate that radiomics has the potential to identify radiation-induced lung injury and could be applied to predict therapeutic efficacy at early timepoints.
Collapse
Affiliation(s)
- Olivia G G Drayson
- Department of Radiation Oncology, University of California, Irvine, CA, 92697-2695, USA.
- Dept. of Radiation Oncology, University of California, Irvine, CA, 92617-2695, USA.
| | - Pierre Montay-Gruel
- Department of Radiation Oncology, University of California, Irvine, CA, 92697-2695, USA
- Antwerp Research in Radiation Oncology (AReRO), Centre for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, 92697-2695, USA
| |
Collapse
|
5
|
Zhao B, Lin H, Jiang X, Li W, Gao Y, Li M, Yu Y, Chen N, Gao J. Exosome-like nanoparticles derived from fruits, vegetables, and herbs: innovative strategies of therapeutic and drug delivery. Theranostics 2024; 14:4598-4621. [PMID: 39239509 PMCID: PMC11373634 DOI: 10.7150/thno.97096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 07/19/2024] [Indexed: 09/07/2024] Open
Abstract
Over the past ten years, significant advancements have been made in exploring plant-derived exosome-like nanoparticles (PELNs) for disease therapeutics and drug delivery. PELNs, as inherent nanoscale particles comprised of proteins, lipids, nucleic acids, and secondary metabolites, exhibit the capacity for cellular uptake by human cells. This intercellular interaction transcends biological boundaries, effectively influencing biological functions in animals. PELNs have outstanding biocompatibility, low immunogenicity, enhanced safety, and environmentally friendly sustainability. This article summarized the preparation methods and characteristics of PELNs. It provided a systematic review of the varied roles of PELNs derived from fruits, vegetables, and herbs in disease therapeutics and drug delivery. The challenges in their production and application were discussed, and future prospects in this rapidly evolving field were explored.
Collapse
Affiliation(s)
- Bo Zhao
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 315016, China
| | - Hangjuan Lin
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 315016, China
| | - Xinchi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wanshu Li
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 315016, China
| | - Yuli Gao
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 315016, China
| | - Minghui Li
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 315016, China
| | - Yanan Yu
- Department of Pharmacy, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 315016, China
| | - Ninggang Chen
- Department of Dermatology Medical Cosmetology Center, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 315016, China
| | - Jianqing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
6
|
Dickstein DL, Zhang R, Ru N, Vozenin MC, Perry BC, Wang J, Baulch J, Acharya MM, Limoli CL. Structural plasticity of pyramidal cell neurons measured after FLASH and conventional dose-rate irradiation. RESEARCH SQUARE 2024:rs.3.rs-4656938. [PMID: 39108471 PMCID: PMC11302692 DOI: 10.21203/rs.3.rs-4656938/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Evidence shows that ultra-high dose-rate FLASH-radiotherapy (FLASH-RT) protects against normal tissue complications and functional decrements in the irradiated brain. Past work has shown that radiation-induced cognitive impairment, neuroinflammation and reduced structural complexity of granule cell neurons were not observed to the same extent after FLASH-RT (> MGy/s) compared to conventional dose-rate (CONV, 0.1 Gy/s) delivery. To explore the sensitivity of different neuronal populations to cranial irradiation and dose-rate modulation, hippocampal CA1 and medial prefrontal cortex (PFC) pyramidal neurons were analyzed by electron and confocal microscopy. Neuron ultrastructural analyses by electron microscopy after 10 Gy FLASH- or CONV-RT exposures indicated that irradiation had little impact on dendritic complexity and synapse density in the CA1, but did increase length and head diameter of smaller non-perforated synapses. Similarly, irradiation caused no change in PFC prelimbic/infralimbic axospinous synapse density, but reductions in non-perforated synapse diameters. While irradiation resulted in thinner myelin sheaths compared to controls, none of these metrics were dose-rate sensitive. Analysis of fluorescently labeled CA1 neurons revealed no radiation-induced or dose-rate-dependent changes in overall dendritic complexity or spine density, in contrast to our past analysis of granule cell neurons. Super-resolution confocal microscopy following a clinical dosing paradigm (3×10Gy) showed significant reductions in excitatory vesicular glutamate transporter 1 and inhibitory vesicular GABA transporter puncta density within the CA1 that were largely dose-rate independent. Collectively, these data reveal that, compared to granule cell neurons, CA1 and mPFC neurons are more radioresistant irrespective of radiation dose-rate.
Collapse
Affiliation(s)
| | | | - Ning Ru
- University of California, Irvine School of Medicine
| | | | | | - Juan Wang
- Uniformed Services University of Health Sciences
| | - Janet Baulch
- University of California, Irvine School of Medicine
| | | | | |
Collapse
|
7
|
Smith SM, Ranjan K, Hoover BM, Drayson OGG, Acharya MM, Kramár EA, Baulch JE, Limoli CL. Extracellular vesicles from GABAergic but not glutamatergic neurons protect against neurological dysfunction following cranial irradiation. Sci Rep 2024; 14:12274. [PMID: 38806540 PMCID: PMC11133350 DOI: 10.1038/s41598-024-62691-y] [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: 01/04/2024] [Accepted: 05/20/2024] [Indexed: 05/30/2024] Open
Abstract
Cranial irradiation used to control brain malignancies invariably leads to progressive and debilitating declines in cognition. Clinical efforts implementing hippocampal avoidance and NMDAR antagonism, have sought to minimize dose to radiosensitive neurogenic regions while normalizing excitatory/inhibitory (E/I) tone. Results of these trials have yielded only marginal benefits to cognition, prompting current studies to evaluate the potential of systemic extracellular vesicle (EV) therapy to restore neurocognitive functionality in the irradiated brain. Here we tested the hypothesis that EVs derived from inhibitory but not excitatory neuronal cultures would prove beneficial to cognition and associated pathology. Rats subjected to a clinically relevant, fractionated cranial irradiation paradigm were given multiple injections of either GABAergic- or glutamatergic-derived EV and subjected to behavioral testing. Rats treated with GABAergic but not glutamatergic EVs showed significant improvements on hippocampal- and cortical-dependent behavioral tasks. While each treatment enhanced levels of the neurotrophic factors BDNF and GDNF, only GABAergic EVs preserved granule cell neuron dendritic spine density. Additional studies conducted with GABAergic EVs, confirmed significant benefits on amygdala-dependent behavior and modest changes in synaptic plasticity as measured by long-term potentiation. These data point to a potentially more efficacious approach for resolving radiation-induced neurological deficits, possibly through a mechanism able to restore homeostatic E/I balance.
Collapse
Affiliation(s)
- Sarah M Smith
- Department of Radiation Oncology, University of California Irvine, Medical Sciences I, Room B-146B, Irvine, CA, 92697-2695, USA
| | - Kashvi Ranjan
- Department of Radiation Oncology, University of California Irvine, Medical Sciences I, Room B-146B, Irvine, CA, 92697-2695, USA
| | - Brianna M Hoover
- Department of Radiation Oncology, University of California Irvine, Medical Sciences I, Room B-146B, Irvine, CA, 92697-2695, USA
| | - Olivia G G Drayson
- Department of Radiation Oncology, University of California Irvine, Medical Sciences I, Room B-146B, Irvine, CA, 92697-2695, USA
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Eniko A Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Janet E Baulch
- Department of Radiation Oncology, University of California Irvine, Medical Sciences I, Room B-146B, Irvine, CA, 92697-2695, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California Irvine, Medical Sciences I, Room B-146B, Irvine, CA, 92697-2695, USA.
| |
Collapse
|
8
|
Ye Z, Wang J, Shi W, Zhou Z, Zhang Y, Wang J, Yang H. Reprimo (RPRM) as a Potential Preventive and Therapeutic Target for Radiation-Induced Brain Injury via Multiple Mechanisms. Int J Mol Sci 2023; 24:17055. [PMID: 38069378 PMCID: PMC10707327 DOI: 10.3390/ijms242317055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Patients receiving cranial radiotherapy for primary and metastatic brain tumors may experience radiation-induced brain injury (RIBI). Thus far, there has been a lack of effective preventive and therapeutic strategies for RIBI. Due to its complicated underlying pathogenic mechanisms, it is rather difficult to develop a single approach to target them simultaneously. We have recently reported that Reprimo (RPRM), a tumor suppressor gene, is a critical player in DNA damage repair, and RPRM deletion significantly confers radioresistance to mice. Herein, by using an RPRM knockout (KO) mouse model established in our laboratory, we found that RPRM deletion alleviated RIBI in mice via targeting its multiple underlying mechanisms. Specifically, RPRM knockout significantly reduced hippocampal DNA damage and apoptosis shortly after mice were exposed to whole-brain irradiation (WBI). For the late-delayed effect of WBI, RPRM knockout obviously ameliorated a radiation-induced decline in neurocognitive function and dramatically diminished WBI-induced neurogenesis inhibition. Moreover, RPRM KO mice exhibited a significantly lower level of acute and chronic inflammation response and microglial activation than wild-type (WT) mice post-WBI. Finally, we uncovered that RPRM knockout not only protected microglia against radiation-induced damage, thus preventing microglial activation, but also protected neurons and decreased the induction of CCL2 in neurons after irradiation, in turn attenuating the activation of microglial cells nearby through paracrine CCL2. Taken together, our results indicate that RPRM plays a crucial role in the occurrence of RIBI, suggesting that RPRM may serve as a novel potential target for the prevention and treatment of RIBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongying Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China; (Z.Y.); (J.W.); (W.S.); (Z.Z.); (Y.Z.); (J.W.)
| |
Collapse
|
9
|
Haroon J, Aboody K, Flores L, McDonald M, Mahdavi K, Zielinski M, Jordan K, Rindner E, Surya J, Venkatraman V, Go-Stevens V, Ngai G, Lara J, Hyde C, Schafer S, Schafer M, Bystritsky A, Nardi I, Kuhn T, Ross D, Jordan S. Use of transcranial low-intensity focused ultrasound for targeted delivery of stem cell-derived exosomes to the brain. Sci Rep 2023; 13:17707. [PMID: 37853206 PMCID: PMC10584845 DOI: 10.1038/s41598-023-44785-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023] Open
Abstract
The blood-brain barrier (BBB) presents a significant challenge for targeted drug delivery. A proposed method to improve drug delivery across the BBB is focused ultrasound (fUS), which delivers ultrasound waves to a targeted location in the brain and is hypothesized to open the BBB. Furthermore, stem cell-derived exosomes have been suggested as a possible anti-inflammatory molecule that may have neural benefits, if able to pass the BBB. In the present study, transcranial low-intensity focused ultrasound (LIFU), without the use of intravenous microbubbles, was assessed for both (1) its ability to influence the BBB, as well as (2) its ability to increase the localization of intravenously administered small molecules to a specific region in the brain. In vivo rat studies were conducted with a rodent-customized 2 MHz LIFU probe (peak pressure = 1.5 MPa), and injection of labeled stem cell-derived exosomes. The results suggested that LIFU (without microbubbles) did not appear to open the BBB after exposure times of 20, 40, or 60 min; instead, there appeared to be an increase in transcytosis of the dextran tracer. Furthermore, the imaging results of the exosome study showed an increase in exosome localization in the right hippocampus following 60 min of targeted LIFU.
Collapse
Affiliation(s)
- J Haroon
- The Regenesis Project, Santa Monica, CA, USA.
| | - K Aboody
- Department of Stem Cell Biology & Regenerative Medicine, and Beckman Research Institute, City of Hope, Duarte, CA, USA.
| | - L Flores
- Department of Stem Cell Biology & Regenerative Medicine, and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - M McDonald
- Department of Stem Cell Biology & Regenerative Medicine, and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - K Mahdavi
- The Regenesis Project, Santa Monica, CA, USA
| | - M Zielinski
- The Regenesis Project, Santa Monica, CA, USA
| | - K Jordan
- The Regenesis Project, Santa Monica, CA, USA
| | - E Rindner
- The Regenesis Project, Santa Monica, CA, USA
| | - J Surya
- The Regenesis Project, Santa Monica, CA, USA
| | | | - V Go-Stevens
- Department of Stem Cell Biology & Regenerative Medicine, and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - G Ngai
- Department of Stem Cell Biology & Regenerative Medicine, and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - J Lara
- Department of Stem Cell Biology & Regenerative Medicine, and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - C Hyde
- Department of Stem Cell Biology & Regenerative Medicine, and Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - S Schafer
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, USA
| | - M Schafer
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, USA
| | - A Bystritsky
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, USA
| | - I Nardi
- Kimera Labs Inc., Miramar, USA
| | - T Kuhn
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, USA
| | - D Ross
- Kimera Labs Inc., Miramar, USA
| | - S Jordan
- The Regenesis Project, Santa Monica, CA, USA
- Department of Neurology, University of California Los Angeles, Los Angeles, USA
| |
Collapse
|
10
|
Yin X, Liu X, Xiao X, Yi K, Chen W, Han C, Wang L, Li Y, Liu J. Human neural stem cells repress glioma cell progression in a paracrine manner by downregulating the Wnt/β-catenin signalling pathway. FEBS Open Bio 2023; 13:1772-1788. [PMID: 37410396 PMCID: PMC10476570 DOI: 10.1002/2211-5463.13671] [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: 04/24/2022] [Revised: 04/07/2023] [Accepted: 07/04/2023] [Indexed: 07/07/2023] Open
Abstract
Neural stem cells (NSCs) play crucial roles in neurological disorders and tissue injury repair through exerting paracrine effects. However, the effects of NSC-derived factors on glioma progression remain unclear. This study aimed to evaluate the effects of human NSC-conditioned medium (NSC-CM) on the behaviour of glioma cells using an in vitro co-culture system. Cell counting kit-8 and 5-ethynyl-2'-deoxyuridine assays revealed that NSC-CM inhibited glioma cell proliferation and growth in a fetal bovine serum (FBS)-independent manner. In addition, our wound-healing assay demonstrated that NSC-CM repressed glioma cell migration, while results from transwell and 3D spheroid invasion assays indicated that NSC-CM also reduced the invasion capacity of glioma cells. Flow cytometry showed that NSC-CM prevented cell cycle progression from the G1 to S phase and promoted apoptosis. Western blotting was used to show that the expression of Wnt/β-catenin pathway-related proteins, including β-catenin, c-Myc, cyclin D1, CD44 and Met, was remarkably decreased in NSC-CM-treated glioma cells. Furthermore, the addition of a Wnt/β-catenin pathway activator, CHIR99021, significantly induced the expression of β-catenin and Met and increased the proliferative and invasive capabilities of control medium-treated glioma cells but not those of NSC-CM-treated glioma cells. The use of enzyme-linked immunosorbent assays (ELISA) revealed the secretion of some antitumour factors in human and rat NSCs, including interferon-α and dickkopf-1. Our data suggest that NSC-CM partially inhibits glioma cell progression by downregulating Wnt/β-catenin signalling. This study may serve as a basis for developing future antiglioma therapies based on NSC derivatives.
Collapse
Affiliation(s)
- Xiaolin Yin
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical UniversityDalian Medical UniversityChina
| | - Xiumei Liu
- Dalian Innovation Institute of Stem Cell and Precision MedicineChina
| | - Xiangyi Xiao
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical UniversityDalian Medical UniversityChina
| | - Kaiyu Yi
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical UniversityDalian Medical UniversityChina
| | - Weigong Chen
- Dalian Innovation Institute of Stem Cell and Precision MedicineChina
| | - Chao Han
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical UniversityDalian Medical UniversityChina
| | - Liang Wang
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical UniversityDalian Medical UniversityChina
| | - Ying Li
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical UniversityDalian Medical UniversityChina
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical UniversityDalian Medical UniversityChina
| |
Collapse
|
11
|
Usmani MT, Krattli RP, El-Khatib SM, Le ACD, Smith SM, Baulch JE, Ng DQ, Acharya MM, Chan A. BDNF Augmentation Using Riluzole Reverses Doxorubicin-Induced Decline in Cognitive Function and Neurogenesis. Neurotherapeutics 2023; 20:838-852. [PMID: 36720792 PMCID: PMC10275819 DOI: 10.1007/s13311-022-01339-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2022] [Indexed: 02/02/2023] Open
Abstract
Cancer-related cognitive impairment (CRCI) considerably affects the quality of life of millions of cancer survivors. Brain-derived neurotrophic factor (BDNF) has been shown to promote survival, differentiation, and maintenance of in vivo dentate neurogenesis, and chemotherapy induces a plethora of physiological and cellular alterations, including a decline in neurogenesis and increased neuroinflammation linked with cognitive impairments. In our clinical studies, breast cancer patients treated with doxorubicin (Adriamycin®, ADR) experienced a significant reduction in the blood levels of BDNF that was associated with a higher risk of CRCI. Our past rodent studies in CRCI have also shown a significant reduction in dentate neurogenesis accompanied by cognitive impairment. In this study, using a female mouse model of ADR-induced cognitive decline, we tested the impact of riluzole (RZ), an orally active BDNF-enhancing medication that is FDA-approved for amyotrophic lateral sclerosis. ADR-treated mice receiving RZ in the drinking water for 1 month showed significant improvements in hippocampal-dependent learning and memory function (spatial recognition), fear extinction memory consolidation, and reduced anxiety-like behavior. RZ prevented chemotherapy-induced reductions of BDNF levels in the hippocampus. Importantly, RZ mitigated chemotherapy-induced loss of newly born, immature neurons, dentate neurogenesis, and neuroinflammation. In conclusion, this data provides pre-clinical evidence for a translationally feasible approach to enhance the neuroprotective effects of RZ treatment to prevent CRCI.
Collapse
Affiliation(s)
- Manal T Usmani
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Robert P Krattli
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Sanad M El-Khatib
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Anh C D Le
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Sarah M Smith
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA
| | - Janet E Baulch
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA.
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA.
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, USA.
| |
Collapse
|
12
|
Drayson OGG, Vozenin MC, Limoli CL. A rigorous behavioral testing platform for the assessment of radiation-induced neurological outcomes. Methods Cell Biol 2023; 180:177-197. [PMID: 37890929 PMCID: PMC11093273 DOI: 10.1016/bs.mcb.2023.02.015] [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: 10/29/2023]
Abstract
Behavioral testing is a popular and reliable method of neurocognitive assessment of rodents but the lack of standard operating procedures has led to a high variation of protocols in use. Therefore, there exists a strong need to standardize protocols for a combined behavioral platform in order to maintain consistency across institutions and assist newcomers in the field. This paper provides details on the methodology of several behavioral tasks which have been validated in identifying radiation induced cognitive impairment as well as provide guidance on timescales and best practices. The cognitive assessments outlined here are optimized for rodent studies and either target learning and memory (open field task, object in updated location, novel object recognition, object in place, and temporal order) or mood and cognition (social interaction, elevated plus maze, light dark box, forced swim test, and fear extinction). We have utilized this platform successfully in evaluating cognitive injury induced by various radiation types, doses, fractionation schedules and also with ultra-high dose rate FLASH radiotherapy. Recommended materials and software are provided as well as advice on methods of data analysis. In this way a comprehensive behavioral platform is described with broad applicability to assess cognitive endpoints critical to therapeutic outcome.
Collapse
Affiliation(s)
- Olivia G G Drayson
- Department of Radiation Oncology, University of California at Irvine, Irvine, CA, United States
| | - Marie-Catherine Vozenin
- Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Charles L Limoli
- Department of Radiation Oncology, University of California at Irvine, Irvine, CA, United States
| |
Collapse
|
13
|
Nieland L, Mahjoum S, Grandell E, Breyne K, Breakefield XO. Engineered EVs designed to target diseases of the CNS. J Control Release 2023; 356:493-506. [PMID: 36907561 DOI: 10.1016/j.jconrel.2023.03.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/28/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
Diseases of the central nervous system (CNS) are challenging to treat, mainly due to the blood-brain barrier (BBB), which restricts drugs in circulation from entering target regions in the brain. To address this issue extracellular vesicles (EVs) have gained increasing scientific interest as carriers able to cross the BBB with multiplex cargos. EVs are secreted by virtually every cell, and their escorted biomolecules are part of an intercellular information gateway between cells within the brain and with other organs. Scientists have undertaken efforts to safeguard the inherent features of EVs as therapeutic delivery vehicles, such as protecting and transferring functional cargo, as well as loading them with therapeutic small molecules, proteins, and oligonucleotides and targeting them to specific cell types for the treatment of CNS diseases. Here, we review current emerging approaches that engineer the EV surface and cargo to improve targeting and functional responses in the brain. We summarize existing applications of engineered EVs as a therapeutic delivery platform for brain diseases, some of which have been evaluated clinically.
Collapse
Affiliation(s)
- Lisa Nieland
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA; Department of Neurosurgery, Leiden University Medical Center, Leiden 2300 RC, the Netherlands.
| | - Shadi Mahjoum
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Emily Grandell
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA; Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Koen Breyne
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Xandra O Breakefield
- Department of Neurology, Molecular Neurogenetics Unit, Massachusetts General Hospital, Boston, MA 02129, USA; Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Cheng J, Jiang J, He B, Lin WJ, Li Y, Duan J, Li H, Huang X, Cai J, Xie J, Zhang Z, Yang Y, Xu Y, Hu X, Wu M, Zhuo X, Liu Q, Shi Z, Yu P, Rong X, Ye X, Saw PE, Wu LJ, Simone CB, Chua MLK, Mai HQ, Tang Y. A phase 2 study of thalidomide for the treatment of radiation-induced blood-brain barrier injury. Sci Transl Med 2023; 15:eabm6543. [PMID: 36812346 DOI: 10.1126/scitranslmed.abm6543] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Radiation-induced brain injury (RIBI) is a debilitating sequela after radiotherapy to treat head and neck cancer, and 20 to 30% of patients with RIBI fail to respond to or have contraindications to the first-line treatments of bevacizumab and corticosteroids. Here, we reported a Simon's minmax two-stage, single-arm, phase 2 clinical trial (NCT03208413) to assess the efficacy of thalidomide in patients with RIBI who were unresponsive to or had contraindications to bevacizumab and corticosteroid therapies. The trial met its primary endpoint, with 27 of 58 patients enrolled showing ≥25% reduction in the volume of cerebral edema on fluid-attenuated inversion recovery-magnetic resonance imaging (FLAIR-MRI) after treatment (overall response rate, 46.6%; 95% CI, 33.3 to 60.1%). Twenty-five (43.1%) patients demonstrated a clinical improvement based on the Late Effects Normal Tissues-Subjective, Objective, Management, Analytic (LENT/SOMA) scale, and 36 (62.1%) experienced cognitive improvement based on the Montreal Cognitive Assessment (MoCA) scores. In a mouse model of RIBI, thalidomide restored the blood-brain barrier and cerebral perfusion, which were attributed to the functional rescue of pericytes secondary to elevation of platelet-derived growth factor receptor β (PDGFRβ) expression by thalidomide. Our data thus demonstrate the therapeutic potential of thalidomide for the treatment of radiation-induced cerebral vasculature impairment.
Collapse
Affiliation(s)
- Jinping Cheng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jingru Jiang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Baixuan He
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Yi Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jingjing Duan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Honghong Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaolong Huang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jinhua Cai
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jiatian Xie
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhan Zhang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yuhua Yang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xia Hu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Minyi Wu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaohuang Zhuo
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Qiang Liu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Pei Yu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaoming Rong
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaojing Ye
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510080, China
| | - Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA.,Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Charles B Simone
- Department of Radiation Oncology, New York Proton Center, New York, NY 10035, USA
| | - Melvin L K Chua
- Department of Head and Neck and Thoracic Cancers, Division of Radiation Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore.,Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore.,Oncology Academic Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Hai-Qiang Mai
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, China
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.,Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| |
Collapse
|
15
|
Perez WD, Perez-Torres CJ. Neurocognitive and radiological changes after cranial radiation therapy in humans and rodents: a systematic review. Int J Radiat Biol 2023; 99:119-137. [PMID: 35511499 DOI: 10.1080/09553002.2022.2074167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Radiation-induced brain injury is a common long-term side effect for brain cancer survivors, leading to a reduced quality of life. Although there is growing research pertaining to this topic, the relationship between cognitive and radiologically detected lesions of radiation-induced brain injury in humans remains unclear. Furthermore, clinically translatable similarities between rodent models and human findings are also undefined. The objective of this review is to then identify the current evidence of radiation-induced brain injury in humans and to compare these findings to current rodent models of radiation-induced brain injury. METHODS This review includes an examination of the current literature on cognitive and radiological characteristics of radiation-induced brain injury in humans and rodents. A thorough search was conducted on PubMed, Web of Science, and Scopus to identify studies that performed cognitive assessments and magnetic resonance imaging techniques on either humans or rodents after cranial radiation therapy. A qualitative synthesis of the data is herein reported. RESULTS A total of 153 studies pertaining to cognitively or radiologically detected radiation injury of the brain are included in this systematic review; 106 studies provided data on humans while 47 studies provided data on rodents. Cognitive deficits in humans manifest across multiple domains after brain irradiation. Radiological evidence in humans highlight various neuroimaging-detectable changes post-irradiation. It is unclear, however, whether these findings reflect ground truth or research interests. Additionally, rodent models do not comprehensively reproduce characteristics of cognitive and radiological injury currently identified in humans. CONCLUSION This systematic review demonstrates that associations between and within cognitive and radiological radiation-induced brain injuries often rely on the type of assessment. Well-designed studies that evaluate the spectrum of potential injury are required for a precise understanding of not only the clinical significance of radiation-induced brain injury in humans, but also how to replicate injury development in pre-clinical models.
Collapse
Affiliation(s)
- Whitney D Perez
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Carlos J Perez-Torres
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, USA.,Academy of Integrated Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.,School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
16
|
Ratushnyak MG, Zhirnik AS, Smirnova OD, Semochkina YP, Parfenova AA, Goryunov KV, Silachev DN, Moskaleva EY. The Use of Neural Stem Cells-Derived Exosomes to Prevent Late Radiation-Induced Cognitive Impairments in Mice. Bull Exp Biol Med 2023; 174:571-577. [PMID: 36894818 DOI: 10.1007/s10517-023-05749-7] [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: 10/11/2022] [Indexed: 03/11/2023]
Abstract
We studied the effect of intranasal administration of neural stem cell (NSC)-derived exosomes on behavior and cognitive functions of mice in the late period after head irradiation in a dose of 8 Gy. The used exosomes had specific markers (CD9+/CD63+, 99.5%; TSG101+, 98.4%) and mean size 105.7±8.8 nm according to dynamic light scattering data and 119.0±12.4 nm according to nanoparticle tracking analysis (NTA). Exosome suspension (2×1012 particles/ml according to NTA measurements) was administered intranasally for 4 weeks starting from 48 h after irradiation in a volume of 5 μl/nostril (2×1010 exosomes/mouse). It was shown that intranasal administration of mouse NSC-derived exosomes prevented delayed radiation-induced behavioral changes and recognition memory impairments in mice after head irradiation.
Collapse
Affiliation(s)
- M G Ratushnyak
- National Research Center "Kurchatov Institute", Moscow, Russia.
| | - A S Zhirnik
- National Research Center "Kurchatov Institute", Moscow, Russia
| | - O D Smirnova
- National Research Center "Kurchatov Institute", Moscow, Russia
| | - Yu P Semochkina
- National Research Center "Kurchatov Institute", Moscow, Russia
| | - A A Parfenova
- National Research Center "Kurchatov Institute", Moscow, Russia
| | - K V Goryunov
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - D N Silachev
- V. I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E Yu Moskaleva
- National Research Center "Kurchatov Institute", Moscow, Russia
| |
Collapse
|
17
|
Pan L, Fan X, Wang L, Wang Y, Li Y, Cui Y, Zheng H, Yi Q, Wu K. Prophylactic cranial irradiation for limited-stage small-cell lung cancer in the magnetic resonance imaging era. Cancer Med 2023; 12:2484-2492. [PMID: 35894822 PMCID: PMC9939136 DOI: 10.1002/cam4.5082] [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: 05/08/2022] [Revised: 06/17/2022] [Accepted: 07/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND We investigated the role of prophylactic cranial irradiation (PCI) in limited-stage small-cell lung cancer (LS-SCLC) according to tumor response in the magnetic resonance imaging (MRI) era. METHODS We retrospectively evaluated patients with LS-SCLC without brain metastases (BMs) on MRI who achieved either complete response (CR) or partial response (PR) after initial chemoradiotherapy at our center from 2006 to 2017. RESULTS This study comprised 116 patients (median age, 58 years; men, 92; women, 24). After initial chemoradiotherapy, 53 patients achieved CR, while 63 patients achieved PR. Eighty-three patients received PCI. Patients who received PCI had better overall survival (OS, 5-year: 52.5% vs. 35.1%; p = 0.012) and progression-free survival (PFS, 5-year: 45.0% vs. 28.2%; p = 0.001) and a lower incidence of BMs (5-year: 18.3% vs. 39.4%; p = 0.010). In the subgroup analysis, PCI improved OS (5-year: 67.8% vs. 46.7%, p = 0.005) and PFS (5-year: 65.2% vs. 35.0%, p = 0.021) and decreased BM risk (5-year: 12.1% vs. 52.4%, p = 0.002) for patients with CR. However, PCI had no benefit (5-year OS: 40.5% vs. 35.6%, p = 0.763; 5-year BMs: 24.6% vs. 31.9%, p = 0.561) for patients with PR. CONCLUSIONS Tumor response remained an important factor for selecting patients for PCI in the MRI era. PCI should be recommended for patients with LS-SCLC who achieve CR after initial thoracic chemoradiotherapy.
Collapse
Affiliation(s)
- Lihua Pan
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Xingwen Fan
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Radiation OncologyShanghaiChina
| | - Lifang Wang
- Department of OncologyAffiliated Hospital of Jining Medical UniversityJiningShandongChina
| | - Yihua Wang
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Radiation OncologyShanghaiChina
| | - Yaqi Li
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Radiation OncologyShanghaiChina
| | - Yingshan Cui
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Radiation OncologyShanghaiChina
| | - Hong Zheng
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Radiation OncologyShanghaiChina
| | - Qiong Yi
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Radiation OncologyShanghaiChina
| | - Kailiang Wu
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Shanghai Key Laboratory of Radiation OncologyShanghaiChina
| |
Collapse
|
18
|
Ni W, Ramalingam M, Li Y, Park JH, Dashnyam K, Lee JH, Bloise N, Fassina L, Visai L, De Angelis MGC, Pedraz JL, Kim HW, Hu J. Immunomodulatory and Anti-inflammatory effect of Neural Stem/Progenitor Cells in the Central Nervous System. Stem Cell Rev Rep 2023; 19:866-885. [PMID: 36650367 DOI: 10.1007/s12015-022-10501-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 01/19/2023]
Abstract
Neuroinflammation is a critical event that responds to disturbed homeostasis and governs various neurological diseases in the central nervous system (CNS). The excessive inflammatory microenvironment in the CNS can adversely affect endogenous neural stem cells, thereby impeding neural self-repair. Therapies with neural stem/progenitor cells (NSPCs) have shown significant inhibitory effects on inflammation, which is mainly achieved through intercellular contact and paracrine signalings. The intercellular contact between NSPCs and immune cells, the activated CNS- resident microglia, and astrocyte plays a critical role in the therapeutic NSPCs homing and immunomodulatory effects. Moreover, the paracrine effect mainly regulates infiltrating innate and adaptive immune cells, activated microglia, and astrocyte through the secretion of bioactive molecules and extracellular vesicles. However, the molecular mechanism involved in the immunomodulatory effect of NSPCs is not well discussed. This article provides a systematic analysis of the immunomodulatory mechanism of NSPCs, discusses efficient ways to enhance its immunomodulatory ability, and gives suggestions on clinical therapy.
Collapse
Affiliation(s)
- Wei Ni
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China.,Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea. .,School of Basic Medical Sciences, Chengdu University, Chengdu, 610106, People's Republic of China.
| | - Yumeng Li
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeong-Hui Park
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Khandmaa Dashnyam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Nora Bloise
- Department of Molecular Medicine, Centre for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, 27100, Pavia, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100, Pavia, Italy
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, 27100, Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine, Centre for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, 27100, Pavia, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100, Pavia, Italy
| | | | - Jose Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain.,Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, 28029, Madrid, Spain
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Jiabo Hu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
19
|
Ebrahim N, El-Halim HEA, Helal OK, El-Azab NEE, Badr OAM, Hassouna A, Saihati HAA, Aborayah NH, Emam HT, El-Wakeel HS, Aljasir M, El-Sherbiny M, Sarg NAS, Shaker GA, Mostafa O, Sabry D, Fouly MAK, Forsyth NR, Elsherbiny NM, Salim RF. Effect of bone marrow mesenchymal stem cells-derived exosomes on diabetes-induced retinal injury: Implication of Wnt/ b-catenin signaling pathway. Biomed Pharmacother 2022; 154:113554. [PMID: 35987163 DOI: 10.1016/j.biopha.2022.113554] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/01/2022] [Accepted: 08/14/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a serious microvascular complication of diabetes mellitus. Mesenchymal stem cells are currently studied as therapeutic strategy for management of DR. Exosomes, considered as a promising cell-free therapy option, display biological functions similar to those of their parent cells. In retinal development, Wnt/b-catenin signaling provides key cues for functional progression. The present study aimed to evaluate the potential efficacy of bone marrow-derived mesenchymal stem cell-derived exosomes (BM-MSCs-Ex) in diabetes-induced retinal injury via modulation of the Wnt/ b-catenin signaling pathway. METHODS Eighty-one rats were allocated into 6 groups (control, DR, DR + DKK1, DR + exosomes, DR + Wnt3a and DR + exosomes+Wnt3a). Evaluation of each group was via histopathological examination, assessment of gene and/or protein expression concerned with oxidative stress (SOD1, SOD2, Nox2, Nox4, iNOS), inflammation (TNF-α, ICAM-1, NF-κB) and angiogenesis (VEGF, VE-cadherin). RESULTS Results demonstrated that exosomes blocked the wnt/b-catenin pathway in diabetic retina concomitant with significant reduction of features of DR as shown by downregulation of retinal oxidants, upregulation of antioxidant enzymes, suppression of retinal inflammatory and angiogenic markers. These results were further confirmed by histopathological results, fundus examination and optical coherence tomography. Additionally, exosomes ameliorative effects abrogated wnt3a-triggered retinal injury in DR. CONCLUSION Collectively, these data demonstrated that exosomes ameliorated diabetes-induced retinal injury via suppressing Wnt/ b-catenin signaling with subsequent reduction of oxidative stress, inflammation and angiogenesis.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt; Stem Cell Unit, Faculty of Medicine, Benha University, Egypt.
| | | | - Omayma Kamel Helal
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt
| | | | - Omnia A M Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Egypt.
| | - Amira Hassouna
- School of Interprofessional Health Studies, Faculty of Health and Environmental Sciences, AUT University, Auckland, New Zealand.
| | - Hajir A Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Saudi Arabia.
| | | | - Hanan Tawfeek Emam
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Egypt.
| | - Hend S El-Wakeel
- Department of Physiology, Faculty of Medicine, Benha University, Egypt.
| | - Mohammad Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia.
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia; Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Egypt.
| | - Naglaa A S Sarg
- Department of Anatomy, Benha Faculty of Medicine, Benha University, Egypt.
| | - Gehan Ahmed Shaker
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Ola Mostafa
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Egypt.
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt; Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Badr University, Cairo 11562, Egypt.
| | | | - Nicholas Robert Forsyth
- Guy Hilton Research Laboratories, School of Pharmacy and Bioengineering, Faculty of Medicine and Health Sciences, Keele University, Newcastle ST5 5BG, UK.
| | - Nehal M Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia; Biochemistry department, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| | - Rabab F Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Egypt.
| |
Collapse
|
20
|
Stem Cell Exosomes Improve Survival of Neural Stem Cells after Radiation Exposure. Bull Exp Biol Med 2022; 173:544-552. [DOI: 10.1007/s10517-022-05587-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Indexed: 10/14/2022]
|
21
|
Liu Q, Huang Y, Duan M, Yang Q, Ren B, Tang F. Microglia as Therapeutic Target for Radiation-Induced Brain Injury. Int J Mol Sci 2022; 23:8286. [PMID: 35955439 PMCID: PMC9368164 DOI: 10.3390/ijms23158286] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Radiation-induced brain injury (RIBI) after radiotherapy has become an increasingly important factor affecting the prognosis of patients with head and neck tumor. With the delivery of high doses of radiation to brain tissue, microglia rapidly transit to a pro-inflammatory phenotype, upregulate phagocytic machinery, and reduce the release of neurotrophic factors. Persistently activated microglia mediate the progression of chronic neuroinflammation, which may inhibit brain neurogenesis leading to the occurrence of neurocognitive disorders at the advanced stage of RIBI. Fully understanding the microglial pathophysiology and cellular and molecular mechanisms after irradiation may facilitate the development of novel therapy by targeting microglia to prevent RIBI and subsequent neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Qun Liu
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Yan Huang
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Mengyun Duan
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China; (M.D.); (Q.Y.)
| | - Qun Yang
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China; (M.D.); (Q.Y.)
| | - Boxu Ren
- The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China; (Q.L.); (Y.H.)
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
22
|
Extracellular Vesicles and Cancer Therapy: Insights into the Role of Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11061194. [PMID: 35740091 PMCID: PMC9228181 DOI: 10.3390/antiox11061194] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023] Open
Abstract
Oxidative stress plays a significant role in cancer development and cancer therapy, and is a major contributor to normal tissue injury. The unique characteristics of extracellular vesicles (EVs) have made them potentially useful as a diagnostic tool in that their molecular content indicates their cell of origin and their lipid membrane protects the content from enzymatic degradation. In addition to their possible use as a diagnostic tool, their role in how normal and diseased cells communicate is of high research interest. The most exciting area is the association of EVs, oxidative stress, and pathogenesis of numerous diseases. However, the relationship between oxidative stress and oxidative modifications of EVs is still unclear, which limits full understanding of the clinical potential of EVs. Here, we discuss how EVs, oxidative stress, and cancer therapy relate to one another; how oxidative stress can contribute to the generation of EVs; and how EVs’ contents reveal the presence of oxidative stress. We also point out the potential promise and limitations of using oxidatively modified EVs as biomarkers of cancer and tissue injury with a focus on pediatric oncology patients.
Collapse
|
23
|
Sukati S, Ho J, Chaiswing L, Sompol P, Pandit H, Wei W, Izumi T, Chen Q, Weiss H, Noel T, Bondada S, Allan Butterfield D, St. Clair DK. Extracellular vesicles released after cranial radiation: An insight into an early mechanism of brain injury. Brain Res 2022; 1782:147840. [PMID: 35183524 PMCID: PMC8918058 DOI: 10.1016/j.brainres.2022.147840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 12/16/2022]
Abstract
Cranial radiation is important for treating both primary brain tumors and brain metastases. A potential delayed side effect of cranial radiation is neurocognitive function decline. Early detection of CNS injury might prevent further neuronal damage. Extracellular vesicles (EVs) have emerged as a potential diagnostic tool because of their unique membranous characteristics and cargos. We investigated whether EVs can be an early indicator of CNS injury by giving C57BJ/6 mice 10 Gy cranial IR. EVs were isolated from sera to quantify: 1) number of EVs using nanoparticle tracking analysis (NTA); 2) Glial fibrillary acidic protein (GFAP), an astrocyte marker; and 3) protein-bound 4-hydroxy-2-nonenal (HNE) adducts, an oxidative damage marker. Brain tissues were prepared for immunohistochemistry staining and protein immunoblotting. The results demonstrate: 1) increased GFAP levels (p < 0.05) in EVs, but not brain tissue, in the IR group; and 2) increased HNE-bound protein adduction levels (p < 0.05). The results support using EVs as an early indicator of cancer therapy-induced neuronal injury.
Collapse
|
24
|
Pancholi S, Tripathi A, Bhan A, Acharya MM, Pillai P. Emerging Concepts on the Role of Extracellular Vesicles and Its Cargo Contents in Glioblastoma-Microglial Crosstalk. Mol Neurobiol 2022; 59:2822-2837. [PMID: 35212938 PMCID: PMC10058057 DOI: 10.1007/s12035-022-02752-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023]
Abstract
Glioblastoma multiforme is the most common, highly aggressive malignant brain tumor which is marked by highest inter- and intra-tumoral heterogeneity. Despite, immunotherapy, and combination therapies developed; the clinical trials often result into large number of failures. Often cancer cells are known to communicate with surrounding cells in tumor microenvironment (TME). Extracellular vesicles (EVs) consisting of diverse cargo mediates this intercellular communication and is believed to modulate the immune function against GBM. Tumor-associated microglia (TAM), though being the resident innate immune cell of CNS, is known to attain pro-tumorigenic M2 phenotype, and this immunomodulation is aided by extracellular vesicle-mediated transfer of oncogenic, immunomodulatory molecules. Besides, oncogenic proteins, long non-coding RNAs (lncRNAs), are believed to carry oncogenic potential, and therefore, understanding the mechanism leading to microglial dysregulation mediated by GBM-derived extracellular vesicle (GDEV) lncRNAs becomes crucial. This review focuses on current understanding of role of GDEV and lncRNA in microglial dysfunction and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Sangati Pancholi
- Division of Neurobiology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Ashutosh Tripathi
- Louis A. Faillace, MD, Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Centre at Houston (UT Health), Houston, TX, USA
| | - Arunoday Bhan
- Department of Surgery, City of Hope Medical Centre, Duarte, CA, USA
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
- Department of Radiation Oncology, University of California, Irvine, CA, USA.
| | - Prakash Pillai
- Division of Neurobiology, Department of Zoology, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India.
| |
Collapse
|
25
|
Kovalchuk A, Mychasiuk R, Muhammad A, Hossain S, Ghose A, Kirkby C, Ghasroddashti E, Kovalchuk O, Kolb B. Complex housing partially mitigates low dose radiation-induced changes in brain and behavior in rats. Restor Neurol Neurosci 2022; 40:109-124. [DOI: 10.3233/rnn-211216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Purpose: In recent years, much effort has been focused on developing new strategies for the prevention and mitigation of adverse radiation effects on healthy tissues and organs, including the brain. The brain is very sensitive to radiation effects, albeit as it is highly plastic. Hence, deleterious radiation effects may be potentially reversible. Because radiation exposure affects dendritic space, reduces the brain’s ability to produce new neurons, and alters behavior, mitigation efforts should focus on restoring these parameters. To that effect, environmental enrichment through complex housing (CH) and exercise may provide a plausible avenue for exploration of protection from brain irradiation. CH is a much broader concept than exercise alone, and constitutes exposure of animals to positive physical and social stimulation that is superior to their routine housing and care conditions. We hypothesized that CHs may lessen harmful neuroanatomical and behavioural effects of low dose radiation exposure. Methods: We analyzed and compared cerebral morphology in animals exposed to low dose head, bystander (liver), and scatter irradiation on rats housed in either the environmental enrichment condos or standard housing. Results: Enriched condo conditions ameliorated radiation-induced neuroanatomical changes. Moreover, irradiated animals that were kept in enriched CH condos displayed fewer radiation-induced behavioural deficits than those housed in standard conditions. Conclusions: Animal model-based environmental enrichment strategies, such as CH, are excellent surrogate models for occupational and exercise therapy in humans, and consequently have significant translational possibility. Our study may thus serve as a roadmap for the development of new, easy, safe and cost-effective methods to prevent and mitigate low-dose radiation effects on the brain.
Collapse
Affiliation(s)
- Anna. Kovalchuk
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | | | - Arif. Muhammad
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Shakhawat. Hossain
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Abhijit. Ghose
- Jack Ady Cancer Center, Alberta Health Services, Lethbridge, AB, Canada
| | - Charles. Kirkby
- Jack Ady Cancer Center, Alberta Health Services, Lethbridge, AB, Canada
- Department of Physics and Astronomy and Department of Oncology, University of Calgary, AB, Canada
| | - Esmaeel. Ghasroddashti
- Jack Ady Cancer Center, Alberta Health Services, Lethbridge, AB, Canada
- Department of Physics and Astronomy and Department of Oncology, University of Calgary, AB, Canada
| | - Olga. Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, Canada
| | - Bryan. Kolb
- Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
26
|
Wang R, Wang X, Zhang Y, Zhao H, Cui J, Li J, Di L. Emerging prospects of extracellular vesicles for brain disease theranostics. J Control Release 2022; 341:844-868. [DOI: 10.1016/j.jconrel.2021.12.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022]
|
27
|
Zheng Z, Wang B, Zhao Q, Zhang Y, Wei J, Meng L, Xin Y, Jiang X. Research progress on mechanism and imaging of temporal lobe injury induced by radiotherapy for head and neck cancer. Eur Radiol 2021; 32:319-330. [PMID: 34327577 DOI: 10.1007/s00330-021-08164-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/07/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022]
Abstract
Radiotherapy (RT) is an effective treatment for head and neck cancer (HNC). Radiation-induced temporal lobe injury (TLI) is a serious complication of RT. Late symptoms of radiation-induced TLI are irreversible and manifest as memory loss, cognitive impairment, and even temporal lobe necrosis (TLN). It is currently believed that the mechanism of radiation-induced TLI involves microvascular injury, neuron and neural stem cell injury, glial cell damage, inflammation, and the production of free radicals. Significant RT-related structural changes and dose-dependent changes in gray matter (GM) and white matter (WM) volume and morphology were observed through computed tomography (CT) and magnetic resonance imaging (MRI) which were common imaging assessment tools. Diffusion tensor imaging (DTI), dispersion kurtosis imaging (DKI), susceptibility-weighted imaging (SWI), resting-state functional magnetic resonance (rs-fMRI), magnetic resonance spectroscopy (MRS), and positron emission tomography (PET) can be used for early diagnosis and prognosis evaluation according to functional, molecular, and cellular processes of TLI. Early diagnosis of TLI is helpful to reduce the incidence of TLN and its related complications. This review summarizes the clinical features, mechanisms, and imaging of radiation-induced TLI in HNC patients. KEY POINTS: • Radiation-induced temporal lobe injury (TLI) is a clinical complication and its symptoms mainly include memory impairment, headache, and cognitive impairment. • The mechanisms of TLI include microvascular injury, cell injury, and inflammatory and free radical injury. Significant RT-related structural changes and dose-dependent changes in TL volume and morphology were observed through CT and MRI. • SWI, MRS, DTI, and DKI and other imaging examinations can detect anatomical and functional, molecular, and cellular changes of TLI.
Collapse
Affiliation(s)
- Zhuangzhuang Zheng
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology& Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Bin Wang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology& Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Qin Zhao
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology& Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yuyu Zhang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology& Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Jinlong Wei
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology& Therapy, The First Hospital of Jilin University, Changchun, 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, China.
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China. .,Jilin Provincial Key Laboratory of Radiation Oncology& Therapy, The First Hospital of Jilin University, Changchun, 130021, China. .,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
28
|
Boerma M, Davis CM, Jackson IL, Schaue D, Williams JP. All for one, though not one for all: team players in normal tissue radiobiology. Int J Radiat Biol 2021; 98:346-366. [PMID: 34129427 PMCID: PMC8781287 DOI: 10.1080/09553002.2021.1941383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE As part of the special issue on 'Women in Science', this review offers a perspective on past and ongoing work in the field of normal (non-cancer) tissue radiation biology, highlighting the work of many of the leading contributors to this field of research. We discuss some of the hypotheses that have guided investigations, with a focus on some of the critical organs considered dose-limiting with respect to radiation therapy, and speculate on where the field needs to go in the future. CONCLUSIONS The scope of work that makes up normal tissue radiation biology has and continues to play a pivotal role in the radiation sciences, ensuring the most effective application of radiation in imaging and therapy, as well as contributing to radiation protection efforts. However, despite the proven historical value of preclinical findings, recent decades have seen clinical practice move ahead with altered fractionation scheduling based on empirical observations, with little to no (or even negative) supporting scientific data. Given our current appreciation of the complexity of normal tissue radiation responses and their temporal variability, with tissue- and/or organ-specific mechanisms that include intra-, inter- and extracellular messaging, as well as contributions from systemic compartments, such as the immune system, the need to maintain a positive therapeutic ratio has never been more urgent. Importantly, mitigation and treatment strategies, whether for the clinic, emergency use following accidental or deliberate releases, or reducing occupational risk, will likely require multi-targeted approaches that involve both local and systemic intervention. From our personal perspective as five 'Women in Science', we would like to acknowledge and applaud the role that many female scientists have played in this field. We stand on the shoulders of those who have gone before, some of whom are fellow contributors to this special issue.
Collapse
Affiliation(s)
- Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Catherine M. Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Isabel L. Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
29
|
Zheng X, Wang J, Bi F, Li Y, Xiao J, Chai Z, Li Y, Miao Z, Wang Y. Protective effects of Lycium barbarum polysaccharide on ovariectomy‑induced cognition reduction in aging mice. Int J Mol Med 2021; 48:121. [PMID: 33955518 PMCID: PMC8121556 DOI: 10.3892/ijmm.2021.4954] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022] Open
Abstract
Women experience cognitive decline as they age due to the decrease in estrogen levels following menopause. Currently, effective pharmaceutical treatments for age‑related cognitive decline are lacking; however, several Traditional Chinese medicines have shown promising effects. Lycium barbarum polysaccharides (LBPs) were found to exert a wide variety of biological activities, including anti‑inflammatory, antioxidant and anti‑aging effects. However, to the best of our knowledge, the neuroprotective actions of LBP on cognitive impairment induced by decreased levels of estrogen have not yet been determined. To evaluate the effects of LBP on learning and memory impairment in an animal model of menopause, 45 female ICR mice were randomly divided into the following three groups: i) Sham; ii) ovariectomy (OVX); and iii) OVX + LBP treatment. The results of open‑field and novel object recognition tests revealed that mice in the OVX group had learning and memory impairments, and lacked the ability to recognize and remember new objects. Notably, these deficits were attenuated following LBP treatment. Immunohistochemical staining confirmed the protective effects of LBP on hippocampal neurons following OVX. To further investigate the underlying mechanism of OVX in mice, mRNA sequencing of the hippocampal tissue was performed, which revealed that the Toll‑like receptor 4 (TLR4) inflammatory signaling pathway was significantly upregulated in the OVX group. Moreover, reverse transcription‑quantitative PCR and immunohistochemical staining demonstrated that OVX induced hippocampal injury, upregulated the expression levels of TLR4, myeloid differentiation factor 88 and NF‑κB, and increased the expression of TNF‑α, IL‑6 and IL‑1β inflammatory factors. Conversely, LBP treatment downregulated the expression levels of mRNAs and proteins associated with the TLR4/NF‑κB signaling pathway, decreased the inflammatory response and reduced neuronal injury in mice that underwent OVX. In conclusion, the findings of the present study indicated that oral LBP treatment may alleviate OVX‑induced cognitive impairments by downregulating the expression levels of mRNAs and proteins associated with the TLR4/NF‑κB signaling pathway, thereby reducing neuroinflammation and damage to the hippocampal neurons. Thus, LBP may represent a potential agent for the prevention of learning and memory impairments in patients with accelerated aging caused by estrogen deficiency.
Collapse
Affiliation(s)
- Xiaomin Zheng
- Department of Pediatrics, General Hospital of Ningxia Medical University, 750004, P.R. China
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Junyan Wang
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Fengchen Bi
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yilu Li
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Jingjing Xiao
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Zhi Chai
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yunhong Li
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Zhenhua Miao
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yin Wang
- Department of Physiology and Neurobiology, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
30
|
Chu C, Gao Y, Lan X, Lin J, Thomas AM, Li S. Stem-Cell Therapy as a Potential Strategy for Radiation-Induced Brain Injury. Stem Cell Rev Rep 2021; 16:639-649. [PMID: 32418118 DOI: 10.1007/s12015-020-09984-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Radiation therapy is a standard and effective non-surgical treatment for primary brain tumors and metastases. However, this strategy inevitably results in damage of normal brain tissue, causing severe complications, especially the late-delayed cognitive impairment. Due to the multifactorial and complex pathological effects of radiation, there is a lack of effective preventative and restorative treatments for the irradiated brain. Stem-cell therapy has held considerable promise for decades in the treatment of central nervous system (CNS) disorders because of its unique capacity for tissue repair and functional integrity. Currently, there is growing interest in using stem cells as a novel option to attenuate the adverse effects of irradiation. In the present review, we discuss recent studies evaluating stem-cell therapies for the irradiated brain and their therapeutic effects on ameliorating radiation-related brain injury as well as their potential challenges in clinical applications. We discuss these works in context of the pathogenesis of radiation-induced injury to CNS tissue in an attempt to elucidate the potential mechanisms of engrafted stem cells to reverse radiation-induced degenerative processes.
Collapse
Affiliation(s)
- Chengyan Chu
- Department of Neurology, Dalian Municipal Central Hospital Affiliated with Dalian Medical University, No. 826 Xinan Road, Shahekou District Dalian, Dalian, Liaoning, 116033, China
| | - Yue Gao
- Department of Neurology, Dalian Municipal Central Hospital Affiliated with Dalian Medical University, No. 826 Xinan Road, Shahekou District Dalian, Dalian, Liaoning, 116033, China
| | - Xiaoyan Lan
- Department of Neurology, Dalian Municipal Central Hospital Affiliated with Dalian Medical University, No. 826 Xinan Road, Shahekou District Dalian, Dalian, Liaoning, 116033, China
| | - Jianwen Lin
- Department of Neurology, Dalian Municipal Central Hospital Affiliated with Dalian Medical University, No. 826 Xinan Road, Shahekou District Dalian, Dalian, Liaoning, 116033, China
| | - Aline M Thomas
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shen Li
- Department of Neurology, Dalian Municipal Central Hospital Affiliated with Dalian Medical University, No. 826 Xinan Road, Shahekou District Dalian, Dalian, Liaoning, 116033, China.
| |
Collapse
|
31
|
Dutta D, Khan N, Wu J, Jay SM. Extracellular Vesicles as an Emerging Frontier in Spinal Cord Injury Pathobiology and Therapy. Trends Neurosci 2021; 44:492-506. [PMID: 33581883 PMCID: PMC8159852 DOI: 10.1016/j.tins.2021.01.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/28/2020] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are membrane-delimited particles that are secreted by nearly all cell types. EVs mediate crucial physiological functions and pathophysiological processes in the CNS. As carriers of diverse bioactive cargoes (e.g., proteins, lipids, and nucleic acids) that can be modified in response to external stimuli, EVs have emerged as pathological mediators following neurotrauma such as spinal cord injury (SCI). We discuss the roles of endogenous EVs in the CNS as well as crosstalk with peripheral EVs in relation to neurotrauma, with a particular focus on SCI. We then summarize the status of EV-based therapeutic advances in preclinical animal models for these conditions. Finally, we discuss new bioengineering strategies that are poised to enhance CNS-specific therapeutic capabilities of EVs.
Collapse
Affiliation(s)
- Dipankar Dutta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Niaz Khan
- Department of Anesthesiology, and Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Junfang Wu
- Department of Anesthesiology, and Center for Shock, Trauma, and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA.
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
32
|
Brunel A, Bégaud G, Auger C, Durand S, Battu S, Bessette B, Verdier M. Autophagy and Extracellular Vesicles, Connected to rabGTPase Family, Support Aggressiveness in Cancer Stem Cells. Cells 2021; 10:1330. [PMID: 34072080 PMCID: PMC8227744 DOI: 10.3390/cells10061330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/22/2022] Open
Abstract
Even though cancers have been widely studied and real advances in therapeutic care have been made in the last few decades, relapses are still frequently observed, often due to therapeutic resistance. Cancer Stem Cells (CSCs) are, in part, responsible for this resistance. They are able to survive harsh conditions such as hypoxia or nutrient deprivation. Autophagy and Extracellular Vesicles (EVs) secretion are cellular processes that help CSC survival. Autophagy is a recycling process and EVs secretion is essential for cell-to-cell communication. Their roles in stemness maintenance have been well described. A common pathway involved in these processes is vesicular trafficking, and subsequently, regulation by Rab GTPases. In this review, we analyze the role played by Rab GTPases in stemness status, either directly or through their regulation of autophagy and EVs secretion.
Collapse
|
33
|
Koh YQ, Ng DQ, Ng CC, Boey A, Wei M, Sze SK, Ho HK, Acharya M, Limoli CL, Chan A. Extracellular Vesicle Proteome of Breast Cancer Patients with and Without Cognitive Impairment Following Anthracycline-based Chemotherapy: An Exploratory Study. Biomark Insights 2021; 16:11772719211018204. [PMID: 34103887 PMCID: PMC8150437 DOI: 10.1177/11772719211018204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
Cognitive impairment due to cancer and its therapy is a major concern among cancer patients and survivors. Extracellular vesicle (EVs) composition altered by cancer and chemotherapy may affect neurological processes such as neuroplasticity, potentially impacting the cognitive abilities of cancer patients and survivors. We investigated the EV proteome of breast cancer patients with and without cognitive impairment following anthracycline-based chemotherapy from longitudinally collected plasma. EVs were cup-shaped and positive for Flotillin-1 and TSG-101. We identified 517 differentially expressed EV proteins between the cognitive impaired and non-impaired groups during and post-chemotherapy. The observed decreased expression of p2X purinoceptor, cofilin-1, ADAM 10, and dynamin-1 in the plasma EVs of the cognitive impaired group may suggest alterations in the mechanisms underlying synaptic plasticity. The reduced expression of tight junction proteins among cognitive-impaired patients may imply weakening of the blood-brain barrier. These EV protein signatures may serve as a fingerprint that underscores the mechanisms underlying cognitive impairment in cancer patients and survivors.
Collapse
Affiliation(s)
- Yong Qin Koh
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, University of California, Irvine, CA, USA
| | - Chiu Chin Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Adrian Boey
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Meng Wei
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Munjal Acharya
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, University of California, Irvine, CA, USA.,Department of Oncology Pharmacy, National Cancer Centre Singapore, Singapore
| |
Collapse
|
34
|
Bhat K, Medina P, He L, Zhang L, Saki M, Ioannidis A, Nguyen NT, Sodhi SS, Sung D, Magyar CE, Liau LM, Kornblum HI, Pajonk F. 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine treatment after brain irradiation preserves cognitive function in mice. Neuro Oncol 2021; 22:1484-1494. [PMID: 32291451 DOI: 10.1093/neuonc/noaa095] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Normal tissue toxicity is an inevitable consequence of primary or secondary brain tumor radiotherapy. Cranial irradiation commonly leads to neurocognitive deficits that manifest months or years after treatment. Mechanistically, radiation-induced loss of neural stem/progenitor cells, neuroinflammation, and demyelination are contributing factors that lead to progressive cognitive decline. METHODS The effects of 1-[(4-nitrophenyl)sulfonyl]-4-phenylpiperazine (NSPP) on irradiated murine neurospheres, microglia cells, and patient-derived gliomaspheres were assessed by sphere-formation assays, flow cytometry, and interleukin (IL)-6 enzyme-linked immunosorbent assay. Activation of the hedgehog pathway was studied by quantitative reverse transcription PCR. The in vivo effects of NSPP were analyzed using flow cytometry, sphere-formation assays, immunohistochemistry, behavioral testing, and an intracranial mouse model of glioblastoma. RESULTS We report that NSPP mitigates radiation-induced normal tissue toxicity in the brains of mice. NSPP treatment significantly increased the number of neural stem/progenitor cells after brain irradiation in female animals, and inhibited radiation-induced microglia activation and expression of the pro-inflammatory cytokine IL-6. Behavioral testing revealed that treatment with NSPP after radiotherapy was able to successfully mitigate radiation-induced decline in memory function of the brain. In mouse models of glioblastoma, NSPP showed no toxicity and did not interfere with the growth-delaying effects of radiation. CONCLUSIONS We conclude that NSPP has the potential to mitigate cognitive decline in patients undergoing partial or whole brain irradiation without promoting tumor growth and that the use of this compound as a radiation mitigator of radiation late effects on the central nervous system warrants further investigation.
Collapse
Affiliation(s)
- Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Paul Medina
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Ling He
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Le Zhang
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Mohammad Saki
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Nhan T Nguyen
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Sirajbir S Sodhi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - David Sung
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Clara E Magyar
- Translational Pathology Core Laboratory, Image Analysis/Virtual Microscopy, Department of Pathology and Laboratory Medicine, Los Angeles, California
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
| | - Harley I Kornblum
- NPI-Semel Institute for Neuroscience and Human Behavior at UCLA, Los Angeles, California.,Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
| |
Collapse
|
35
|
Therapeutic Applications of Stem Cells and Extracellular Vesicles in Emergency Care: Futuristic Perspectives. Stem Cell Rev Rep 2021; 17:390-410. [PMID: 32839921 PMCID: PMC7444453 DOI: 10.1007/s12015-020-10029-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regenerative medicine (RM) is an interdisciplinary field that aims to repair, replace or regenerate damaged or missing tissue or organs to function as close as possible to its physiological architecture and functions. Stem cells, which are undifferentiated cells retaining self-renewal potential, excessive proliferation and differentiation capacity into offspring or daughter cells that form different lineage cells of an organism, are considered as an important part of the RM approaches. They have been widely investigated in preclinical and clinical studies for therapeutic purposes. Extracellular vesicles (EVs) are the vital mediators that regulate the therapeutic effects of stem cells. Besides, they carry various types of cargo between cells which make them a significant contributor of intercellular communication. Given their role in physiological and pathological conditions in living cells, EVs are considered as a new therapeutic alternative solution for a variety of diseases in which there is a high unmet clinical need. This review aims to summarize and identify therapeutic potential of stem cells and EVs in diseases requiring acute emergency care such as trauma, heart diseases, stroke, acute respiratory distress syndrome and burn injury. Diseases that affect militaries or societies including acute radiation syndrome, sepsis and viral pandemics such as novel coronavirus disease 2019 are also discussed. Additionally, featuring and problematic issues that hamper clinical translation of stem cells and EVs are debated in a comparative manner with a futuristic perspective. Graphical Abstract.
Collapse
|
36
|
Apodaca LA, Baddour AAD, Garcia C, Alikhani L, Giedzinski E, Ru N, Agrawal A, Acharya MM, Baulch JE. Human neural stem cell-derived extracellular vesicles mitigate hallmarks of Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2021; 13:57. [PMID: 33676561 PMCID: PMC7937214 DOI: 10.1186/s13195-021-00791-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/16/2021] [Indexed: 02/07/2023]
Abstract
Background Regenerative therapies to mitigate Alzheimer’s disease (AD) neuropathology have shown very limited success. In the recent era, extracellular vesicles (EVs) derived from multipotent and pluripotent stem cells have shown considerable promise for the treatment of dementia and many neurodegenerative conditions. Methods Using the 5xFAD accelerated transgenic mouse model of AD, we now show the regenerative potential of human neural stem cell (hNSC)-derived EVs on the neurocognitive and neuropathologic hallmarks in the AD brain. Two- or 6-month-old 5xFAD mice received single or two intra-venous (retro-orbital vein, RO) injections of hNSC-derived EVs, respectively. Results RO treatment using hNSC-derived EVs restored fear extinction memory consolidation and reduced anxiety-related behaviors 4–6 weeks post-injection. EV treatment also significantly reduced dense core amyloid-beta plaque accumulation and microglial activation in both age groups. These results correlated with partial restoration of homeostatic levels of circulating pro-inflammatory cytokines in the AD mice. Importantly, EV treatment protected against synaptic loss in the AD brain that paralleled improved cognition. MiRNA analysis of the EV cargo revealed promising candidates targeting neuroinflammation and synaptic function. Conclusions Collectively, these data demonstrate the neuroprotective effects of systemic administration of stem cell-derived EVs for remediation of behavioral and molecular AD neuropathologies. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00791-x.
Collapse
Affiliation(s)
- Lauren A Apodaca
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA
| | - Al Anoud D Baddour
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA
| | - Camilo Garcia
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA
| | - Leila Alikhani
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA
| | - Ning Ru
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA
| | - Anshu Agrawal
- Department of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA.
| | - Janet E Baulch
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
37
|
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
|
38
|
Yang X, Ma L, Ye Z, Shi W, Zhang L, Wang J, Yang H. Radiation-induced bystander effects may contribute to radiation-induced cognitive impairment. Int J Radiat Biol 2021; 97:329-340. [PMID: 33332177 DOI: 10.1080/09553002.2021.1864498] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE Despite being a major treatment modality for brain cancer due to its efficiency in achieving cancer control, radiotherapy has long been known to cause long-term side effects, including radiation-induced cognitive impairment (RICI). Neurogenesis inhibition due to radiation-induced damage in neural stem cells (NSCs) has been demonstrated to be an important mechanism underlying RICI. Radiation-induced bystander effects (RIBEs) denote the biological responses in non-targeted cells after their neighboring cells are irradiated. We have previously demonstrated that RIBEs could play an important role in the skin wound healing process. Therefore, we aimed to investigate whether RIBEs contribute to RICI in this study. MATERIALS AND METHODS The transwell co-culture method was used to investigate bystander effects in mouse NSCs induced by irradiated GL261 mouse glioma cells in vitro. The proliferation, neurosphere-forming capacity and differentiation potential of NSCs were determined as the bystander endpoints. The exosomes were extracted from the media used to culture GL261 cells and were injected into the hippocampus of C57BL/6 mice. Two months later, the neurogenesis of mice was assessed using BrdU incorporation and immunofluorescence microscopy, and cognitive function was evaluated by the Morris Water Maze. RESULTS After co-culture with GL261 glioma cells, mouse NSCs displayed inhibited proliferation and reduced neurosphere-forming capacity and differentiation potential. The irradiated GL261 cells caused greater inhibition and reduction in NSCs than unirradiated GL261 cells. Moreover, adding the exosomes secreted by GL261 cells into the culture of NSCs inhibited NSC proliferation, suggesting that the cancer cell-derived exosomes may be critical intercellular signals. Furthermore, injection of the exosomes from GL261 cells into the hippocampus of mice caused significant neurogenesis inhibition and cognitive impairment two month later, and the exosomes from irradiated GL261 cells induced greater inhibitory effects. CONCLUSION RIBEs mediated by the exosomes from irradiated cancer cells could contribute to RICI and, therefore, could be a novel mechanism underlying RICI.
Collapse
Affiliation(s)
- Xuejiao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, PR China
| | - Linlin Ma
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, PR China
| | - Zhujing Ye
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, PR China
| | - Wenyu Shi
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, PR China.,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Liyuan Zhang
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, PR China.,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Second Affiliated Hospital of Soochow University, Suzhou, PR China.,Institute of Radiotherapy & Oncology of Soochow University, Suzhou, PR China
| | - Jingdong Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, PR China
| | - Hongying Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University/Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, PR China.,Institute of Radiotherapy & Oncology of Soochow University, Suzhou, PR China
| |
Collapse
|
39
|
Limoli C. Can a comparison of clinical and deep space irradiation scenarios shed light on the radiation response of the brain? Br J Radiol 2020; 93:20200245. [PMID: 32970457 DOI: 10.1259/bjr.20200245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Not surprisingly, our knowledge of the impact of radiation on the brain has evolved considerably. Decades of work have struggled with identifying the critical cellular targets in the brain, the latency of functional change and understanding how irradiation alters the balance between excitatory and inhibitory circuits. Radiation-induced cell kill following clinical fractionation paradigms pointed to both stromal and parenchymal targets but also defined an exquisite sensitivity of neurogenic populations of newly born cells in the brain. It became more and more apparent too, that acute (days) events transpiring after exposure were poorly prognostic of the late (months-years) waves of radiation injury believed to underlie neurocognitive deficits. Much of these gaps in knowledge persisted as NASA became interested in how exposure to much different radiation types, doses and dose rates that characterize the space radiation environment might impair central nervous system functionality, with possibly negative implications for deep space travel. Now emerging evidence from researchers engaged in clinical, translational and environmental radiation sciences have begun to fill these gaps and have uncovered some surprising similarities in the response of the brain to seemingly disparate exposure scenarios. This article highlights many of the commonalities between the vastly different irradiation paradigms that distinguish clinical treatments from occupational exposures in deep space.
Collapse
Affiliation(s)
- Charles Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, United States
| |
Collapse
|
40
|
Pariset E, Malkani S, Cekanaviciute E, Costes SV. Ionizing radiation-induced risks to the central nervous system and countermeasures in cellular and rodent models. Int J Radiat Biol 2020; 97:S132-S150. [PMID: 32946305 DOI: 10.1080/09553002.2020.1820598] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Harmful effects of ionizing radiation on the Central Nervous System (CNS) are a concerning outcome in the field of cancer radiotherapy and form a major risk for deep space exploration. Both acute and chronic CNS irradiation induce a complex network of molecular and cellular alterations including DNA damage, oxidative stress, cell death and systemic inflammation, leading to changes in neuronal structure and synaptic plasticity with behavioral and cognitive consequences in animal models. Due to this complexity, countermeasure or therapeutic approaches to reduce the harmful effects of ionizing radiation include a wide range of protective and mitigative strategies, which merit a thorough comparative analysis. MATERIALS AND METHODS We reviewed current approaches for developing countermeasures to both targeted and non-targeted effects of ionizing radiation on the CNS from the molecular and cellular to the behavioral level. RESULTS We focus on countermeasures that aim to mitigate the four main detrimental actions of radiation on CNS: DNA damage, free radical formation and oxidative stress, cell death, and harmful systemic responses including tissue death and neuroinflammation. We propose a comprehensive review of CNS radiation countermeasures reported for the full range of irradiation types (photons and particles, low and high linear energy transfer) and doses (from a fraction of gray to several tens of gray, fractionated and unfractionated), with a particular interest for exposure conditions relevant to deep-space environment and radiotherapy. Our review reveals the importance of combined strategies that increase DNA protection and repair, reduce free radical formation and increase their elimination, limit inflammation and improve cell viability, limit tissue damage and increase repair and plasticity. CONCLUSIONS The majority of therapeutic approaches to protect the CNS from ionizing radiation have been limited to acute high dose and high dose rate gamma irradiation, and few are translatable from animal models to potential human application due to harmful side effects and lack of blood-brain barrier permeability that precludes peripheral administration. Therefore, a promising research direction would be to focus on practical applicability and effectiveness in a wider range of irradiation paradigms, from fractionated therapeutic to deep space radiation. In addition to discovering novel therapeutics, it would be worth maximizing the benefits and reducing side effects of those that already exist. Finally, we suggest that novel cellular and tissue models for developing and testing countermeasures in the context of other impairments might also be applied to the field of CNS responses to ionizing radiation.
Collapse
Affiliation(s)
- Eloise Pariset
- Universities Space Research Association, Columbia, MD, USA.,Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Sherina Malkani
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA.,Young Scientist Program, Blue Marble Space Institute of Science, Moffett Field, CA, USA
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
| |
Collapse
|
41
|
Leavitt RJ, Acharya MM, Baulch JE, Limoli CL. Extracellular Vesicle-Derived miR-124 Resolves Radiation-Induced Brain Injury. Cancer Res 2020; 80:4266-4277. [PMID: 32816912 PMCID: PMC7541572 DOI: 10.1158/0008-5472.can-20-1599] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/08/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Radiation-induced cognitive dysfunction (RICD) is a progressive and debilitating health issue facing patients following cranial radiotherapy to control central nervous system cancers. There has been some success treating RICD in rodents using human neural stem cell (hNSC) transplantation, but the procedure is invasive, requires immunosuppression, and could cause other complications such as teratoma formation. Extracellular vesicles (EV) are nanoscale membrane-bound structures that contain biological contents including mRNA, miRNA, proteins, and lipids that can be readily isolated from conditioned culture media. It has been previously shown that hNSC-derived EV resolves RICD following cranial irradiation using an immunocompromised rodent model. Here, we use immunocompetent wild-type mice to show that hNSC-derived EV treatment administered either intravenously via retro-orbital vein injection or via intracranial transplantation can ameliorate cognitive deficits following 9 Gy head-only irradiation. Cognitive function assessed on the novel place recognition, novel object recognition, and temporal order tasks was not only improved at early (5 weeks) but also at delayed (6 months) postirradiation times with just a single EV treatment. Improved behavioral outcomes were also associated with reduced neuroinflammation as measured by a reduction in activated microglia. To identify the mechanism of action, analysis of EV cargo implicated miRNA (miR-124) as a potential candidate in the mitigation of RICD. Furthermore, viral vector-mediated overexpression of miR-124 in the irradiated brain ameliorated RICD and reduced microglial activation. Our findings demonstrate for the first time that systemic administration of hNSC-derived EV abrogates RICD and neuroinflammation in cranially irradiated wild-type rodents through a mechanism involving miR-124. SIGNIFICANCE: Radiation-induced neurocognitive decrements in immunocompetent mice can be resolved by systemic delivery of hNSC-derived EVs involving a mechanism dependent on expression of miR-124.
Collapse
Affiliation(s)
- Ron J Leavitt
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, Irvine, California.
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, California.
| |
Collapse
|
42
|
Bálentová S, Adamkov M. Pathological changes in the central nervous system following exposure to ionizing radiation. Physiol Res 2020; 69:389-404. [PMID: 32469226 PMCID: PMC8648310 DOI: 10.33549/physiolres.934309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 03/03/2020] [Indexed: 12/19/2022] Open
Abstract
Experimental studies in animals provide relevant knowledge about pathogenesis of radiation-induced injury to the central nervous system. Radiation-induced injury can alter neuronal, glial cell population, brain vasculature and may lead to molecular, cellular and functional consequences. Regarding to its fundamental role in the formation of new memories, spatial navigation and adult neurogenesis, the majority of studies have focused on the hippocampus. Most recent findings in cranial radiotherapy revealed that hippocampal avoidance prevents radiation-induced cognitive impairment of patients with brain primary tumors and metastases. However, numerous preclinical studies have shown that this problem is more complex. Regarding the fact, that the radiation-induced cognitive impairment reflects hippocampal and non-hippocampal compartments, it is highly important to investigate molecular, cellular and functional changes in different brain regions and their integration at clinically relevant doses and schedules. Here, we provide a literature review in order support the translation of preclinical findings to clinical practice and improve the physical and mental status of patients with brain tumors.
Collapse
Affiliation(s)
- S Bálentová
- Institute of Histology and Embryology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic.
| | | |
Collapse
|
43
|
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: 117] [Impact Index Per Article: 23.4] [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
|
44
|
Neuroprotection of Radiosensitive Juvenile Mice by Ultra-High Dose Rate FLASH Irradiation. Cancers (Basel) 2020; 12:cancers12061671. [PMID: 32599789 PMCID: PMC7352849 DOI: 10.3390/cancers12061671] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/02/2020] [Accepted: 06/18/2020] [Indexed: 02/07/2023] Open
Abstract
Major advances in high precision treatment delivery and imaging have greatly improved the tolerance of radiotherapy (RT); however, the selective sparing of normal tissue and the reduction of neurocognitive side effects from radiation-induced toxicities remain significant problems for pediatric patients with brain tumors. While the overall survival of pediatric patients afflicted with medulloblastoma (MB), the most common type primary brain cancer in children, remains high (≥80%), lifelong neurotoxic side-effects are commonplace and adversely impact patients’ quality of life. To circumvent these clinical complications, we have investigated the capability of ultra-high dose rate FLASH-radiotherapy (FLASH-RT) to protect the radiosensitive juvenile mouse brain from normal tissue toxicities. Compared to conventional dose rate (CONV) irradiation, FLASH-RT was found to ameliorate radiation-induced cognitive dysfunction in multiple independent behavioral paradigms, preserve developing and mature neurons, minimize microgliosis and limit the reduction of the plasmatic level of growth hormone. The protective “FLASH effect” was pronounced, especially since a similar whole brain dose of 8 Gy delivered with CONV-RT caused marked reductions in multiple indices of behavioral performance (objects in updated location, novel object recognition, fear extinction, light-dark box, social interaction), reductions in the number of immature (doublecortin+) and mature (NeuN+) neurons and increased neuroinflammation, adverse effects that were not found with FLASH-RT. Our data point to a potentially innovative treatment modality that is able to spare, if not prevent, many of the side effects associated with long-term treatment that disrupt the long-term cognitive and emotional well-being of medulloblastoma survivors.
Collapse
|
45
|
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
|
46
|
Branscome H, Paul S, Yin D, El-Hage N, Agbottah ET, Zadeh MA, Liotta LA, Kashanchi F. Use of Stem Cell Extracellular Vesicles as a "Holistic" Approach to CNS Repair. Front Cell Dev Biol 2020; 8:455. [PMID: 32587858 PMCID: PMC7298153 DOI: 10.3389/fcell.2020.00455] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
Neurodegeneration is a hallmark of many diseases and disorders of the central nervous system (CNS). High levels of neuroinflammation are often associated with irreparable damage to CNS cells due to the dysregulation of signaling cascades that are unable to restore a homeostatic balance. Due to the inherent complexity of the CNS, development of CNS-related therapeutics has met limited success. While stem cell therapy has been evaluated in the context of CNS repair, the mechanisms responsible for their functional properties have not been clearly defined. In recent years, there has been growing interest in the use of stem cell extracellular vesicles (EVs) for the treatment of various CNS pathologies as these vesicles are believed to mediate many of the functional effects associated with their donor stem cells. The potency of stem cell EVs is believed to be largely driven by their biological cargo which includes various types of RNAs, proteins, and cytokines. In this review, we describe the characteristic properties of stem cell EVs and summarize their reported neuroprotective and immunomodulatory functions. A special emphasis is placed on the identification of specific biological cargo, including proteins and non-coding RNA molecules, that have been found to be associated with stem cell EVs. Collectively, this review highlights the potential of stem cell EVs as an alternative to traditional stem cell therapy for the repair of cellular damage associated with diverse CNS pathologies.
Collapse
Affiliation(s)
- Heather Branscome
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, United States
- American Type Culture Collection (ATCC), Manassas, VA, United States
| | - Siddhartha Paul
- American Type Culture Collection (ATCC) Cell Systems, Gaithersburg, MD, United States
| | - Dezhong Yin
- American Type Culture Collection (ATCC) Cell Systems, Gaithersburg, MD, United States
| | - Nazira El-Hage
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Emmanuel T. Agbottah
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Mohammad Asad Zadeh
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Lance A. Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, United States
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, School of Systems Biology, George Mason University, Manassas, VA, United States
| |
Collapse
|
47
|
Koh YQ, Tan CJ, Toh YL, Sze SK, Ho HK, Limoli CL, Chan A. Role of Exosomes in Cancer-Related Cognitive Impairment. Int J Mol Sci 2020; 21:ijms21082755. [PMID: 32326653 PMCID: PMC7215650 DOI: 10.3390/ijms21082755] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
A decline in cognitive function following cancer treatment is one of the most commonly reported post-treatment symptoms among patients with cancer and those in remission, and include memory, processing speed, and executive function. A clear understanding of cognitive impairment as a result of cancer and its therapy can be obtained by delineating structural and functional changes using brain imaging studies and neurocognitive assessments. There is also a need to determine the underlying mechanisms and pathways that impact the brain and affect cognitive functioning in cancer survivors. Exosomes are small cell-derived vesicles formed by the inward budding of multivesicular bodies, and are released into the extracellular environment via an exocytic pathway. Growing evidence suggests that exosomes contribute to various physiological and pathological conditions, including neurological processes such as synaptic plasticity, neuronal stress response, cell-to-cell communication, and neurogenesis. In this review, we summarize the relationship between exosomes and cancer-related cognitive impairment. Unraveling exosomes’ actions and effects on the microenvironment of the brain, which impacts cognitive functioning, is critical for the development of exosome-based therapeutics for cancer-related cognitive impairment.
Collapse
Affiliation(s)
- Yong Qin Koh
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 119077, Singapore
| | - Chia Jie Tan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 119077, Singapore
| | - Yi Long Toh
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 119077, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 119077, Singapore
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697-2695, USA
| | - Alexandre Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 119077, Singapore
- Department of Clinical Pharmacy Practice, University of California, Irvine, CA 92697, USA
- Correspondence: ; Tel.: +1-949-824-8896
| |
Collapse
|
48
|
Pazzaglia S, Briganti G, Mancuso M, Saran A. Neurocognitive Decline Following Radiotherapy: Mechanisms and Therapeutic Implications. Cancers (Basel) 2020; 12:cancers12010146. [PMID: 31936195 PMCID: PMC7017115 DOI: 10.3390/cancers12010146] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/02/2020] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
The brain undergoes ionizing radiation (IR) exposure in many clinical situations, particularly during radiotherapy for malignant brain tumors. Cranial radiation therapy is related with the hazard of long-term neurocognitive decline. The detrimental ionizing radiation effects on the brain closely correlate with age at treatment, and younger age associates with harsher deficiencies. Radiation has been shown to induce damage in several cell populations of the mouse brain. Indeed, brain exposure causes a dysfunction of the neurogenic niche due to alterations in the neuronal and supporting cell progenitor signaling environment, particularly in the hippocampus—a region of the brain critical to memory and cognition. Consequent deficiencies in rates of generation of new neurons, neural differentiation and apoptotic cell death, lead to neuronal deterioration and lasting repercussions on neurocognitive functions. Besides neural stem cells, mature neural cells and glial cells are recognized IR targets. We will review the current knowledge about radiation-induced damage in stem cells of the brain and discuss potential treatment interventions and therapy methods to prevent and mitigate radiation related cognitive decline.
Collapse
Affiliation(s)
- Simonetta Pazzaglia
- Laboratory of Biomedical Technologies, ENEA CR-Casaccia, Via Anguillarese 301, 00123 Rome, Italy;
| | - Giovanni Briganti
- Department of Radiation Physics Guglielmo Marconi University, Via Plinio 44, 00193 Rome, Italy;
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, ENEA CR-Casaccia, Via Anguillarese 301, 00123 Rome, Italy;
- Correspondence: (M.M.); (A.S.)
| | - Anna Saran
- Laboratory of Biomedical Technologies, ENEA CR-Casaccia, Via Anguillarese 301, 00123 Rome, Italy;
- Department of Radiation Physics Guglielmo Marconi University, Via Plinio 44, 00193 Rome, Italy;
- Correspondence: (M.M.); (A.S.)
| |
Collapse
|
49
|
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
|
50
|
Allen BD, Apodaca LA, Syage AR, Markarian M, Baddour AAD, Minasyan H, Alikhani L, Lu C, West BL, Giedzinski E, Baulch JE, Acharya MM. Attenuation of neuroinflammation reverses Adriamycin-induced cognitive impairments. Acta Neuropathol Commun 2019; 7:186. [PMID: 31753024 PMCID: PMC6868786 DOI: 10.1186/s40478-019-0838-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022] Open
Abstract
Numerous clinical studies have established the debilitating neurocognitive side effects of chemotherapy in the treatment of breast cancer, often referred as chemobrain. We hypothesize that cognitive impairments are associated with elevated microglial inflammation in the brain. Thus, either elimination of microglia or restoration of microglial function could ameliorate cognitive dysfunction. Using a rodent model of chronic Adriamycin (ADR) treatment, a commonly used breast cancer chemotherapy, we evaluated two strategies to ameliorate chemobrain: 1) microglia depletion using the colony stimulating factor-1 receptor (CSF1R) inhibitor PLX5622 and 2) human induced pluripotent stem cell-derived microglia (iMG)-derived extracellular vesicle (EV) treatment. In strategy 1 mice received ADR once weekly for 4 weeks and were then administered CSF1R inhibitor (PLX5622) starting 72 h post-ADR treatment. ADR-treated animals given a normal diet exhibited significant behavioral deficits and increased microglial activation 4–6 weeks later. PLX5622-treated mice exhibited no ADR-related cognitive deficits and near complete depletion of IBA-1 and CD68+ microglia in the brain. Cytokine and RNA sequencing analysis for inflammation pathways validated these findings. In strategy 2, 1 week after the last ADR treatment, mice received retro-orbital vein injections of iMG-EV (once weekly for 4 weeks) and 1 week later, mice underwent behavior testing. ADR-treated mice receiving EV showed nearly complete restoration of cognitive function and significant reductions in microglial activation as compared to untreated ADR mice. Our data demonstrate that ADR treatment elevates CNS inflammation that is linked to cognitive dysfunction and that attenuation of neuroinflammation reverses the adverse neurocognitive effects of chemotherapy.
Collapse
|