1
|
Karam M, Ortega-Gascó A, Tornero D. Emerging Insights into Brain Inflammation: Stem-Cell-Based Approaches for Regenerative Medicine. Int J Mol Sci 2025; 26:3275. [PMID: 40244116 PMCID: PMC11989304 DOI: 10.3390/ijms26073275] [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: 02/28/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Neuroinflammation is a complex immune response triggered by brain injury or pathological stimuli, and is highly exacerbated in neurodegenerative diseases. It plays a dual role in the central nervous system, promoting repair in acute stages while aggravating disease progression by contributing to neuronal loss, synaptic dysfunction, and glial dysregulation in chronic phases. Inflammatory responses are mainly orchestrated by microglia and infiltrated monocytes, which, when dysregulated, not only harm existing neurons, but also impair the survival and differentiation of neural stem and progenitor cells in the affected brain regions. Modulating neuroinflammation is crucial for harnessing its protective functions while minimizing its detrimental effects. Current therapeutic strategies focus on fine-tuning inflammatory responses through pharmacological agents, bioactive molecules, and stem cell-based therapies. These approaches aim to restore immune homeostasis, support neuroprotection, and promote regeneration in various neurological disorders. However, animal models sometimes fail to reproduce human-specific inflammatory responses in the brain. In this context, stem-cell-derived models provide a powerful tool to study neuroinflammatory mechanisms in a patient-specific and physiologically relevant context. These models facilitate high-throughput screening, personalized medicine, and the development of targeted therapies while addressing the limitations of traditional animal models, paving the way for more targeted and effective treatments.
Collapse
Affiliation(s)
- Marie Karam
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Alba Ortega-Gascó
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Daniel Tornero
- Laboratory of Neural Stem Cells and Brain Damage, Department of Biomedical Sciences, Institute of Neurosciences, University of Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
2
|
Li M, Jiang X, Gai X, Dai M, Li M, Wang Y, Wang H. CiteSpace-based visual analysis on transcutaneous electrical acupoint stimulation of clinical randomized controlled trial studies and its mechanism on perioperative disorders. Medicine (Baltimore) 2024; 103:e39893. [PMID: 39465871 PMCID: PMC11479488 DOI: 10.1097/md.0000000000039893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/11/2024] [Indexed: 10/29/2024] Open
Abstract
To systematically present an overview of randomized controlled trials on transcutaneous electrical acupoint stimulation (TEAS) using bibliometric methods, and describe the role and mechanisms of TEAS in most prevalent diseases. Relevant literature was searched in China National Knowledge Infrastructure, Wanfang Data, VIP, SinoMed, PubMed, and Web of Science. The literature was imported and screened into NoteExpress, screened according to inclusion and exclusion criteria, and analyzed using Excel and CiteSpace 6.3R1 software. A total of 1296 documents were included. The number of publications increased annually after 2012. Junlu Wang was the most prolific author. The main research institutions were Peking University, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Shuguang Hospital, and Tongde Hospital of Zhejiang Province. The research hotspots in this field include perioperative care, cancer, pain management, and stroke, primarily focusing on analgesia, immune enhancement, antihypertension, and reduction of gastrointestinal disorders. The main regulatory mechanisms of TEAS include the control of inflammation, oxidative stress, and regulation of the autonomic nervous system. TEAS is most widely used in the elderly, with PC6, ST36, and LI4 being the most frequently studied acupoints in clinical randomized controlled trials. The concept of accelerated rehabilitation is gradually being applied to TEAS, representing an emerging trend for future development. Clinical research on TEAS is rapidly developing, with a focus on applications in cancer and perioperative care. Future research should expand collaboration and conduct high-level clinical and mechanistic studies, which will contribute to the development of standardized protocols and clinical practice.
Collapse
Affiliation(s)
- Mengqi Li
- College of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaobo Jiang
- College of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiangmu Gai
- College of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Mengyao Dai
- College of Acupuncture and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Mengyuan Li
- Northeast Asia Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Yanxin Wang
- Department of Cardiovascular Rehabilitation, The Third Clinical Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hongfeng Wang
- Northeast Asia Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
3
|
Li Z, Wang H, Yin Y. Peripheral inflammation is a potential etiological factor in Alzheimer's disease. Rev Neurosci 2024; 35:99-120. [PMID: 37602685 DOI: 10.1515/revneuro-2023-0049] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023]
Abstract
Peripheral inflammation could constitute a risk factor for AD. This review summarizes the research related to peripheral inflammation that appears to have a relationship with Alzheimer's disease. We find there are significant associations between AD and peripheral infection induced by various pathogens, including herpes simplex virus type 1, cytomegalovirus, Epstein-Barr virus, human immunodeficiency virus, severe acute respiratory syndrome coronavirus 2, Porphyromonas gingivalis, Helicobacter pylori, and Toxoplasma gondii. Chronic inflammatory diseases are also reported to contribute to the pathophysiology of AD. The mechanisms by which peripheral inflammation affects the pathophysiology of AD are complex. Pathogen-derived neurotoxic molecule composition, disrupted BBB, and dysfunctional neurogenesis may all play a role in peripheral inflammation, promoting the development of AD. Anti-pathogenic medications and anti-inflammatory treatments are reported to decrease the risk of AD. Studies that could improve understanding the associations between AD and peripheral inflammation are needed. If our assumption is correct, early intervention against inflammation may be a potential method of preventing and treating AD.
Collapse
Affiliation(s)
- Ziyuan Li
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| | - Yafu Yin
- Department of Nuclear Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Kongjiang Road 1665, Yangpu District, Shanghai 200092, China
| |
Collapse
|
4
|
Zhao Y, Ma J, Ding G, Wang Y, Yu H, Cheng X. Astragalus polysaccharides promote neural stem cells-derived oligodendrogenesis through attenuating CD8 +T cell infiltration in experimental autoimmune encephalomyelitis. Int Immunopharmacol 2024; 126:111303. [PMID: 38043269 DOI: 10.1016/j.intimp.2023.111303] [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: 07/25/2023] [Revised: 10/31/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Endogenous neural stem cells (NSCs) have the potential to generate remyelinating oligodendrocytes, which play an important role in multiple sclerosis (MS). However, the differentiation of NSCs into oligodendrocytes is insufficient, which is considered a major cause of remyelination failure. Our previous work reported that Astragalus polysaccharides (APS) had a neuroprotective effect on experimental autoimmune encephalomyelitis (EAE) mice. However, it remains unclear whether APS regulate NSCs differentiation in EAE mice. In this study, our data illustrated that APS administration could promote NSCs in the subventricular zone (SVZ) to differentiate into oligodendrocytes. Furthermore, we found that APS significantly improved neuroinflammation and inhibited CD8+T cell infiltration into SVZ of EAE mice. We also found that MOG35-55-specific CD8+T cells suppressed NSCs differentiation into oligodendrocytes by secreting IFN-γ, and APS facilitated the differentiation of NSCs into oligodendrocytes which was related to decreased IFN-γ secretion. In addition, APS treatment did not show a better effect on the NSCs-derived oligodendrogenesis after CD8+T cell depletion. This present study demonstrated that APS alleviated neuroinflammation and CD8+T cell infiltration into SVZ to induce oligodendroglial differentiation, and thus exerted neuroprotective effect. Our findings revealed that reducing the infiltration of CD8+T cells might contribute to enhancing NSCs-derived neurogenesis. And APS might be a promising drug candidate to treat MS.
Collapse
Affiliation(s)
- Yan Zhao
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jinyun Ma
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Guiqing Ding
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yuanhua Wang
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Hua Yu
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiaodong Cheng
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| |
Collapse
|
5
|
Chien SC, Chen CP. Genetic Counseling of Fetal Microcephaly. J Med Ultrasound 2024; 32:1-7. [PMID: 38665355 PMCID: PMC11040482 DOI: 10.4103/jmu.jmu_18_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/10/2023] [Indexed: 04/28/2024] Open
Abstract
Fetal microcephaly is a small head with various losses of cerebral cortical volume. The affected cases may suffer from a wide range in severity of impaired cerebral development from slight to severe mental retardation. It can be an isolated finding or with other anomalies depending on the heterogeneous causes including genetic mutations, chromosomal abnormalities, congenital infectious diseases, maternal alcohol consumption, and metabolic disorders during pregnancy. It is often a lifelong and incurable condition. Thus, early detection of fetal microcephaly and identification of the underlying causes are important for clinical staff to provide appropriate genetic counseling to the parents and accurate management.
Collapse
Affiliation(s)
| | - Chih-Ping Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Institute of Clinical and Community Health Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
6
|
Chandwani MN, Kamte YS, Singh VR, Hemerson ME, Michaels AC, Leak RK, O'Donnell LA. The anti-viral immune response of the adult host robustly modulates neural stem cell activity in spatial, temporal, and sex-specific manners. Brain Behav Immun 2023; 114:61-77. [PMID: 37516388 DOI: 10.1016/j.bbi.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/20/2023] [Accepted: 07/14/2023] [Indexed: 07/31/2023] Open
Abstract
Viruses induce a wide range of neurological sequelae through the dysfunction and death of infected cells and persistent inflammation in the brain. Neural stem cells (NSCs) are often disturbed during viral infections. Although some viruses directly infect and kill NSCs, the antiviral immune response may also indirectly affect NSCs. To better understand how NSCs are influenced by a productive immune response, where the virus is successfully resolved and the host survives, we used the CD46+ mouse model of neuron-restricted measles virus (MeV) infection. As NSCs are spared from direct infection in this model, they serve as bystanders to the antiviral immune response initiated by selective infection of mature neurons. MeV-infected mice showed distinct regional and temporal changes in NSCs in the primary neurogenic niches of the brain, the hippocampus and subventricular zone (SVZ). Hippocampal NSCs increased throughout the infection (7 and 60 days post-infection; dpi), while mature neurons transiently declined at 7 dpi and then rebounded to basal levels by 60 dpi. In the SVZ, NSC numbers were unchanged, but mature neurons declined even after the infection was controlled at 60 dpi. Further analyses demonstrated sex, temporal, and region-specific changes in NSC proliferation and neurogenesis throughout the infection. A relatively long-term increase in NSC proliferation and neurogenesis was observed in the hippocampus; however, neurogenesis was reduced in the SVZ. This decline in SVZ neurogenesis was associated with increased immature neurons in the olfactory bulb in female, but not male mice, suggesting potential migration of newly-made neurons out of the female SVZ. These sex differences in SVZ neurogenesis were accompanied by higher infiltration of B cells and greater expression of interferon-gamma and interleukin-6 in female mice. Learning, memory, and olfaction tests revealed no overt behavioral changes after the acute infection subsided. These results indicate that antiviral immunity modulates NSC activity in adult mice without inducing gross behavioral deficits among those tested, suggestive of mechanisms to restore neurons and maintain adaptive behavior, but also revealing the potential for robust NSC disruption in subclinical infections.
Collapse
Affiliation(s)
- Manisha N Chandwani
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Yashika S Kamte
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Vivek R Singh
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Marlo E Hemerson
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Alexa C Michaels
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Rehana K Leak
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Lauren A O'Donnell
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Kamte YS, Chandwani MN, London NM, Potosnak CE, Leak RK, O'Donnell LA. Perturbations in neural stem cell function during a neurotropic viral infection in juvenile mice. J Neurochem 2023; 166:809-829. [PMID: 37530081 DOI: 10.1111/jnc.15914] [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: 03/17/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 08/03/2023]
Abstract
Viral infections of the central nervous system (CNS) often cause worse neurological outcomes in younger hosts. Throughout childhood, the brain undergoes extensive development and refinement to produce functional neural networks. Network function is maintained partly with the help of neural stem cells (NSCs) that replace neuronal and glia subtypes in the two neurogenic niches of the brain (the hippocampus and subventricular zone). Accumulating evidence suggests that viruses disrupt NSC function in adulthood and infancy, but the in vivo impact of childhood infections on acute and long-term NSC function is unknown. Using a juvenile mouse model of measles virus (MeV) infection, where only mature neurons in the brain are infected, we defined the effects of the antiviral immune response on NSCs from juvenile to adult stages of life. We found that (a) virus persists in the brains of survivors despite an anti-viral immune response; (b) NSC numbers decrease dramatically during early infection, but ultimately stabilize in adult survivors; (c) infection is associated with mild apoptosis throughout the juvenile brain, but NSC proliferation is unchanged; (d) the loss of NSC numbers is dependent upon the stage of NSC differentiation; and (e) immature neurons increase early during infection, concurrent with depletion of NSC pools. Collectively, we show that NSCs are exquisitely sensitive to the inflammatory microenvironment created during neuron-restricted MeV infection in juveniles, responding with an early loss of NSCs but increased neurogenesis. These studies provide insight into potential cellular mechanisms associated with long-term neurological deficits in survivors of childhood CNS infections.
Collapse
Affiliation(s)
- Yashika S Kamte
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Manisha N Chandwani
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Natalie M London
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Chloe E Potosnak
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Rehana K Leak
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| | - Lauren A O'Donnell
- School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
8
|
Garg TK, Garg S, Miousse IR, Wise SY, Carpenter AD, Fatanmi OO, van Rhee F, Singh VK, Hauer-Jensen M. Gamma-Tocotrienol Modulates Total-Body Irradiation-Induced Hematopoietic Injury in a Nonhuman Primate Model. Int J Mol Sci 2022; 23:ijms232416170. [PMID: 36555814 PMCID: PMC9784560 DOI: 10.3390/ijms232416170] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Radiation exposure causes acute damage to hematopoietic and immune cells. To date, there are no radioprotectors available to mitigate hematopoietic injury after radiation exposure. Gamma-tocotrienol (GT3) has demonstrated promising radioprotective efficacy in the mouse and nonhuman primate (NHP) models. We determined GT3-mediated hematopoietic recovery in total-body irradiated (TBI) NHPs. Sixteen rhesus macaques divided into two groups received either vehicle or GT3, 24 h prior to TBI. Four animals in each treatment group were exposed to either 4 or 5.8 Gy TBI. Flow cytometry was used to immunophenotype the bone marrow (BM) lymphoid cell populations, while clonogenic ability of hematopoietic stem cells (HSCs) was assessed by colony forming unit (CFU) assays on day 8 prior to irradiation and days 2, 7, 14, and 30 post-irradiation. Both radiation doses showed significant changes in the frequencies of B and T-cell subsets, including the self-renewable capacity of HSCs. Importantly, GT3 accelerated the recovery in CD34+ cells, increased HSC function as shown by improved recovery of CFU-granulocyte macrophages (CFU-GM) and burst-forming units erythroid (B-FUE), and aided the recovery of circulating neutrophils and platelets. These data elucidate the role of GT3 in hematopoietic recovery, which should be explored as a potential medical countermeasure to mitigate radiation-induced injury to the hematopoietic system.
Collapse
Affiliation(s)
- Tarun K. Garg
- UAMS Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sarita Garg
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Isabelle R. Miousse
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephen Y. Wise
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Alana D. Carpenter
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Oluseyi O. Fatanmi
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Frits van Rhee
- UAMS Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Vijay K. Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Correspondence: (V.K.S.); (M.H.-J.); Tel.: +1-301-295-2347 (V.K.S.); +1-501-686-7912 (M.H.-J.); Fax: +1-501-421-0022 (M.H.-J.)
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence: (V.K.S.); (M.H.-J.); Tel.: +1-301-295-2347 (V.K.S.); +1-501-686-7912 (M.H.-J.); Fax: +1-501-421-0022 (M.H.-J.)
| |
Collapse
|
9
|
Association between Brain Injury Markers and Testosterone in Critically-Ill COVID-19 Male Patients. Microorganisms 2022; 10:microorganisms10112095. [PMID: 36363686 PMCID: PMC9697553 DOI: 10.3390/microorganisms10112095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Accumulating data suggest that various neurologic manifestations are reported in critically-ill COVID-19 patients. Although low testosterone levels were associated with poor outcomes, the relationship between testosterone levels and indices of brain injury are still poorly understood. Therefore, we aimed to explore whether testosterone levels are associated with glial fibrillary acidic protein (GFAP) and ubiquitin carboxy-terminal hydrolase L1 (UCH-L1), biomarkers of brain injury, in patients with a severe form of COVID-19. The present study was conducted on 65 male patients aged 18−65 with severe COVID-19. Blood samples were collected at three time points: upon admission to ICU, 7 days after, and 14 days after. In patients with neurological sequels (n = 20), UCH-L1 serum concentrations at admission were markedly higher than in patients without them (240.0 (155.4−366.4) vs. 146.4 (92.5−243.9) pg/mL, p = 0.022). GFAP concentrations on admission did not differ between the groups (32.2 (24.2−40.1) vs. 29.8 (21.8−39.4) pg/mL, p = 0.372). Unlike GFAP, UCH-L1 serum concentrations exhibited a negative correlation with serum testosterone in all three time points (r = −0.452, p < 0.001; r = −0.430, p < 0.001 and r = −0.476, p = 0.001, respectively). The present study suggests that the traumatic brain injury biomarker UCH-L1 may be associated with neurological impairments seen in severe COVID-19. Moreover, a negative correlation between UCH-L1 and serum testosterone concentrations implies that testosterone may have a role in the development of neurological sequels in critically-ill COVID-19 patients.
Collapse
|
10
|
Effect of Transcutaneous Electrical Acupoint Stimulation Combined with Transversus Abdominis Plane Block on Postoperative Recovery in Elderly Patients Undergoing Laparoscopic Gastric Cancer Surgery: A Randomized Controlled Trial. Pain Ther 2022; 11:1327-1339. [PMID: 36098938 PMCID: PMC9633915 DOI: 10.1007/s40122-022-00429-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 08/25/2022] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION This study assessed the influence of transcutaneous electrical acupoint stimulation (TEAS) combined with transversus abdominis plane block (TAPB) on the recovery of elderly patients undergoing laparoscopic gastric cancer surgery. METHODS Ninety patients (age ≥ 60 years) undergoing laparoscopic gastric cancer surgery were randomly divided into general anesthesia group (group G), TAPB group (group NG), and TEAS combined with TAPB group (group NTG). Patients in the NTG group received TEAS at PC6, LI4, and ST36 acupoints and TAPB. Patients in the NG group received TAPB. The quality of recovery (QoR) was assessed using the QoR-15 questionnaire. The percentages of T lymphocyte subsets were determined. Consumption of anesthetics, extubation time, visual analog scale (VAS) scores, time of first postoperative ambulation and flatus, and postoperative adverse events were also recorded. RESULTS QoR-15 scores on postoperative day (POD) 3 and POD 7 were higher in the NTG group than in the G and NG groups (P < 0.05). On POD 1 and POD 3, the percentages of CD3+ and CD4+ T cells and the CD4+/CD8+ ratio were higher and the percentage of CD8+ T cells was lower in the NTG group than in the G and NG groups (P < 0.05). Remifentanil consumption, and the incidence of postoperative nausea and vomiting (PONV) were lower and extubation time and time of first postoperative flatus were shorter in the NTG group than in the G and NG groups (P < 0.05). Compared with the G group, the VAS scores on POD 1 were lower in the NG group and those on POD 2 were lower in the NTG group (P < 0.05). CONCLUSION The combination of TEAS and TAPB ameliorated postoperative pain, improved immune and gastrointestinal function, reduced the incidence of PONV, and effectively promoted postoperative recovery in elderly patients undergoing laparoscopic gastric cancer surgery. TRIAL REGISTRATION Chinese Clinical Trial Registry (ChiCTR2100042119).
Collapse
|
11
|
Var SR, Shetty AV, Grande AW, Low WC, Cheeran MC. Microglia and Macrophages in Neuroprotection, Neurogenesis, and Emerging Therapies for Stroke. Cells 2021; 10:3555. [PMID: 34944064 PMCID: PMC8700390 DOI: 10.3390/cells10123555] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022] Open
Abstract
Stroke remains the number one cause of morbidity in the United States. Within weeks to months after an ischemic event, there is a resolution of inflammation and evidence of neurogenesis; however, years following a stroke, there is evidence of chronic inflammation in the central nervous system, possibly by the persistence of an autoimmune response to brain antigens as a result of ischemia. The mechanisms underlying the involvement of macrophage and microglial activation after stroke are widely acknowledged as having a role in ischemic stroke pathology; thus, modulating inflammation and neurological recovery is a hopeful strategy for treating the long-term outcomes after ischemic injury. Current treatments fail to provide neuroprotective or neurorestorative benefits after stroke; therefore, to ameliorate brain injury-induced deficits, therapies must alter both the initial response to injury and the subsequent inflammatory process. This review will address differences in macrophage and microglia nomenclature and summarize recent work in elucidating the mechanisms of macrophage and microglial participation in antigen presentation, neuroprotection, angiogenesis, neurogenesis, synaptic remodeling, and immune modulating strategies for treating the long-term outcomes after ischemic injury.
Collapse
Affiliation(s)
- Susanna R. Var
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Anala V. Shetty
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
- Department of Biological Sciences, University of Minnesota Medical School, Minneapolis, MN 55108, USA
| | - Andrew W. Grande
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Walter C. Low
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Maxim C. Cheeran
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA
| |
Collapse
|
12
|
Kamte YS, Chandwani MN, Michaels AC, O’Donnell LA. Neural Stem Cells: What Happens When They Go Viral? Viruses 2021; 13:v13081468. [PMID: 34452333 PMCID: PMC8402908 DOI: 10.3390/v13081468] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022] Open
Abstract
Viruses that infect the central nervous system (CNS) are associated with developmental abnormalities as well as neuropsychiatric and degenerative conditions. Many of these viruses such as Zika virus (ZIKV), cytomegalovirus (CMV), and herpes simplex virus (HSV) demonstrate tropism for neural stem cells (NSCs). NSCs are the multipotent progenitor cells of the brain that have the ability to form neurons, astrocytes, and oligodendrocytes. Viral infections often alter the function of NSCs, with profound impacts on the growth and repair of the brain. There are a wide spectrum of effects on NSCs, which differ by the type of virus, the model system, the cell types studied, and the age of the host. Thus, it is a challenge to predict and define the consequences of interactions between viruses and NSCs. The purpose of this review is to dissect the mechanisms by which viruses can affect survival, proliferation, and differentiation of NSCs. This review also sheds light on the contribution of key antiviral cytokines in the impairment of NSC activity during a viral infection, revealing a complex interplay between NSCs, viruses, and the immune system.
Collapse
|
13
|
Pan Y, Tian D, Wang H, Zhao Y, Zhang C, Wang S, Xie D, Zhang D, Zhu Y, Zhang Y. Inhibition of Perforin-Mediated Neurotoxicity Attenuates Neurological Deficits After Ischemic Stroke. Front Cell Neurosci 2021; 15:664312. [PMID: 34262436 PMCID: PMC8274971 DOI: 10.3389/fncel.2021.664312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
Perforin-mediated cytotoxicity plays a crucial role in microbial defense, tumor surveillance, and primary autoimmune disorders. However, the contribution of the cytolytic protein perforin to ischemia-induced secondary tissue damage in the brain has not been fully investigated. Here, we examined the kinetics and subpopulations of perforin-positive cells and then evaluated the direct effects of perforin-mediated cytotoxicity on outcomes after ischemic stroke. Using flow cytometry, we showed that perforin+CD45+ immune cells could be detected at 12 h and that the percentage of these cells increased largely until on day 3 and then significantly declined on day 7. Surprisingly, the percentage of Perforin+CD45+ cells also unexpectedly increased from day 7 to day 14 after ischemic stroke in Perforin1-EGFP transgenic mice. Our results suggested that Perforin+CD45+ cells play vital roles in the ischemic brain at early and late stages and further suggested that Perforin+CD45+ cells are a heterogeneous population. Surprisingly, in addition to CD8+ T cells, NK cells, and NKT cells, central nervous system (CNS)-resident immune microglia, which are first triggered and activated within minutes after ischemic stroke in mice, also secreted perforin during ischemic brain injury. In our study, the percentage of perforin+ microglia increased from 12 h after ischemic stroke, increased largely until on day 3 after ischemic stroke, and then moderately declined from days 3 to 7. Intriguingly, the percentage of perforin+ microglia also dramatically increased from days 7 to 14 after ischemic stroke. Furthermore, compared with wild-type littermates, Perforin 1-/- mice exhibited significant increases in the cerebral infarct volume, neurological deficits, and neurogenesis and inhibition of neurotoxic astrogliosis. Interestingly, the number of CD45+CD3+ T cells was significantly decreased in Perforin 1-/- mice compared with their wild-type littermates, especially the number of γδ T cells. In addition, Perforin 1-/- mice had lower levels of IL-17 than their wild-type littermates. Our results identified a critical function of perforin-mediated neurotoxicity in the ischemic brain, suggesting that targeting perforin-mediated neurotoxicity in brain-resident microglia and invading perforin+CD45+ immune cells may be a potential strategy for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yuhualei Pan
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Clinical Research Institute, Beijing, China
| | - Dan Tian
- Beijing Clinical Research Institute, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Huan Wang
- Beijing Clinical Research Institute, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yushang Zhao
- Beijing Clinical Research Institute, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chengjie Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Song Wang
- Beijing Clinical Research Institute, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dan Xie
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dong Zhang
- Beijing Clinical Research Institute, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanbing Zhu
- Beijing Clinical Research Institute, Beijing, China.,Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yongbo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Esgalhado AJ, Reste-Ferreira D, Albino SE, Sousa A, Amaral AP, Martinho A, Oliveira IT, Verde I, Lourenço O, Fonseca AM, Cardoso EM, Arosa FA. CD45RA, CD8β, and IFNγ Are Potential Immune Biomarkers of Human Cognitive Function. Front Immunol 2020; 11:592656. [PMID: 33324408 PMCID: PMC7723833 DOI: 10.3389/fimmu.2020.592656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/12/2020] [Indexed: 11/23/2022] Open
Abstract
There is increasing evidence that in humans the adaptive immunological system can influence cognitive functions of the brain. We have undertaken a comprehensive immunological analysis of lymphocyte and monocyte populations as well as of HLA molecules expression in a cohort of elderly volunteers (age range, 64–101) differing in their cognitive status. Hereby, we report on the identification of a novel signature in cognitively impaired elderly characterized by: (1) elevated percentages of CD8+ T effector-memory cells expressing high levels of the CD45RA phosphate receptor (Temrahi); (2) high percentages of CD8+ T cells expressing high levels of the CD8β chain (CD8βhi); (3) augmented production of IFNγ by in vitro activated CD4+ T cells. Noteworthy, CD3+CD8+ Temrahi and CD3+CD8βhi cells were associated with impaired cognition. Cytomegalovirus seroprevalence showed that all volunteers studied but one were CMV positive. Finally, we show that some of these phenotypic and functional features are associated with an increased frequency of the HLA-B8 serotype, which belongs to the ancestral haplotype HLA-A1, Cw7, B8, DR3, DQ2, among cognitively impaired volunteers. To our knowledge, this is the first proof in humans linking the amount of cell surface CD45RA and CD8β chain expressed by CD8+ Temra cells, and the amount of IFNγ produced by in vitro activated CD4+ T cells, with impaired cognitive function in the elderly.
Collapse
Affiliation(s)
- André J Esgalhado
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Débora Reste-Ferreira
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Stephanie E Albino
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Adriana Sousa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Ana Paula Amaral
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - António Martinho
- Molecular Genetics Laboratory, Coimbra Blood and Transplantation Center, Coimbra, Portugal
| | - Isabel T Oliveira
- C4-UBI, Cloud Computing Competence Centre, University of Beira Interior, Covilhã, Portugal
| | - Ignacio Verde
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Olga Lourenço
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Ana M Fonseca
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Elsa M Cardoso
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal.,IPG, Guarda Polytechnic Institute, Guarda, Portugal
| | - Fernando A Arosa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
15
|
Li D, Liu X, Liu T, Liu H, Tong L, Jia S, Wang YF. Neurochemical regulation of the expression and function of glial fibrillary acidic protein in astrocytes. Glia 2019; 68:878-897. [PMID: 31626364 DOI: 10.1002/glia.23734] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/27/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022]
Abstract
Glial fibrillary acidic protein (GFAP), a type III intermediate filament, is a marker of mature astrocytes. The expression of GFAP gene is regulated by many transcription factors (TFs), mainly Janus kinase-2/signal transducer and activator of transcription 3 cascade and nuclear factor κ-light-chain-enhancer of activated B cell signaling. GFAP expression is also modulated by protein kinase and other signaling molecules that are elicited by neuronal activity and hormones. Abnormal expression of GFAP proteins occurs in neuroinflammation, neurodegeneration, brain edema-eliciting diseases, traumatic brain injury, psychiatric disorders and others. GFAP, mainly in α-isoform, is the major component of cytoskeleton and the scaffold of astrocytes, which is essential for the maintenance of astrocytic structure and shape. GFAP also has highly morphological plasticity because of its quick changes in assembling and polymerizing states in response to environmental challenges. This plasticity and its corresponding cellular morphological changes endow astrocytes the functions of physical barrier between adjacent neurons and stabilizer of extracellular environment. Moreover, GFAP colocalizes and even molecularly associates with many functional molecules. This feature allows GFAP to function as a platform for direct interactions between different molecules. Last, GFAP involves transportation and localization of other functional proteins and thus serves as a protein transport guide in astrocytes. This guiding role of GFAP involves an elastic retraction and extension cytoskeletal network that couples with GFAP reassembling, transporting, and membrane protein recycling machinery. This paper reviews our current understanding of the expression and functions of GFAP as well as their regulation.
Collapse
Affiliation(s)
- Dongyang Li
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Xiaoyu Liu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Tianming Liu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Haitao Liu
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Li Tong
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Shuwei Jia
- Department of Physiology, Harbin Medical University, Harbin, China
| | - Yu-Feng Wang
- Department of Physiology, Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Wang XB, Wu DJ, Chen WP, Liu J, Ju YJ. Impact of radiotherapy on immunological parameters, levels of inflammatory factors, and clinical prognosis in patients with esophageal cancer. JOURNAL OF RADIATION RESEARCH 2019; 60:353-363. [PMID: 31034571 PMCID: PMC6530619 DOI: 10.1093/jrr/rrz006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 11/05/2018] [Indexed: 05/10/2023]
Abstract
The aim of this study was to observe dynamic changes in immunological parameters and levels of inflammatory factors from pre-radiotherapy to post-radiotherapy in patients with esophageal cancer, and to evaluate the related clinical prognosis. In all, 110 patients with esophageal cancer who underwent radiotherapy were enrolled. Before radiotherapy, post-radiotherapy, and 3 months after radiotherapy, the percentages of T lymphocyte subsets and natural killer (NK) cells in peripheral blood were detected using flow cytometry. The levels of serum inflammatory factors were measured with the enzyme-linked immunosorbent assay (ELISA). Thirty peripheral blood samples from healthy people were similarly analysed as the control. Before radiotherapy, the percentages of CD4+ and CD8+ T cells and NK cells, and the CD4+/CD8+ rate in esophageal cancer patients were significantly different from those in the healthy control group (P < 0.001); the levels of inflammatory factors were increased significantly (P < 0.001). The percentages of the above cells and the levels of inflammatory factors also differed statistically significantly between pre- and post-radiotherapy (P < 0.001) in the esophageal cancer patients. Three months after radiotherapy, the percentages of CD3+ (P = 0.453), CD4+ (P = 0.108), and CD8+ T cells (P = 0.163) and NK cells (P = 0.103) had recovered to the level before radiotherapy; and the levels of TNF-α (P = 0.101), IL-6 (P = 0.302) and IL-8 (P = 0.250) were also restored. After radiotherapy, alterations in immunological parameters were associated with the irradiation volume and the myelosuppression condition. Patients with recovered immunological parameters showed a longer median survival time than those with poor recovery of immunological parameters. For esophageal cancer patients who were immunosuppressive and had an activated inflammatory response before radiotherapy, radiotherapy aggravated these symptoms, and this aggravation was positively associated with myelosuppression and irradiation volume. In addition, recovery of the immunological parameters indicated better prognosis.
Collapse
Affiliation(s)
- Xiao-Bo Wang
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
| | - Di-Jun Wu
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
| | - Wei-Ping Chen
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
| | - Jian Liu
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
| | - Yong-Jian Ju
- Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong, P.R. China
- Corresponding author: Department of Radiotherapy, The First People’s Hospital of Nantong, No. 6 Hai’er Xiang North Road, Nantong 226001, P.R. China. Tel/Fax: +86-0513-85061155.
| |
Collapse
|
17
|
Chandwani MN, Creisher PS, O'Donnell LA. Understanding the Role of Antiviral Cytokines and Chemokines on Neural Stem/Progenitor Cell Activity and Survival. Viral Immunol 2018; 32:15-24. [PMID: 30307795 DOI: 10.1089/vim.2018.0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Viral infections of the central nervous system are accompanied by the expression of cytokines and chemokines that can be critical for the control of viral replication in the brain. The outcomes of cytokine/chemokine signaling in neural cells vary widely, with cell-specific effects on cellular activity, proliferation, and survival. Neural stem/progenitor cells (NSPCs) are often altered during viral infections, through direct infection by the virus or by the influence of immune cell activity or cytokine/chemokine signaling. However, it has been challenging to dissect the contribution of the virus and specific inflammatory mediators during an infection. In addition to initiating an antiviral program in infected NSPCs, cytokines/chemokines can induce multiple changes in NSPC behavior that can perturb NSPC numbers, differentiation into other neural cells, and migration to sites of injury, and ultimately brain development and repair. The focus of this review was to dissect the effects of common antiviral cytokines and chemokines on NSPC activity, and to consider the subsequent pathological consequences for the host from changes in NSPC function.
Collapse
Affiliation(s)
- Manisha N Chandwani
- Department of Pharmaceutical, Administrative, and Social Sciences, Graduate School of Pharmaceutical Sciences, Duquesne University School of Pharmacy , Pittsburgh, Pennsylvania
| | - Patrick S Creisher
- Department of Pharmaceutical, Administrative, and Social Sciences, Graduate School of Pharmaceutical Sciences, Duquesne University School of Pharmacy , Pittsburgh, Pennsylvania
| | - Lauren A O'Donnell
- Department of Pharmaceutical, Administrative, and Social Sciences, Graduate School of Pharmaceutical Sciences, Duquesne University School of Pharmacy , Pittsburgh, Pennsylvania
| |
Collapse
|
18
|
Effects of Acyclovir and IVIG on Behavioral Outcomes after HSV1 CNS Infection. Behav Neurol 2017; 2017:5238402. [PMID: 29358844 PMCID: PMC5735307 DOI: 10.1155/2017/5238402] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/06/2017] [Accepted: 09/16/2017] [Indexed: 12/27/2022] Open
Abstract
Herpes simplex virus 1 (HSV) encephalitis (HSE) has serious neurological complications, involving behavioral and cognitive impairments that cause significant morbidity and a reduced quality of life. We showed that HSE results from dysregulated central nervous system (CNS) inflammatory responses. We hypothesized that CNS inflammation is casually involved in behavioral abnormalities after HSE and that treatment with ACV and pooled human immunoglobulin (IVIG), an immunomodulatory drug, would improve outcomes compared to mice treated with phosphate buffered saline (PBS) or ACV alone. Anxiety levels were high in HSV-infected PBS and ACV-treated mice compared to mice treated with ACV + IVIG, consistent with reports implicating inflammation in anxiety induced by lipopolysaccharide (LPS) or stress. Female, but not male, PBS-treated mice were cognitively impaired, and unexpectedly, ACV was protective, while the inclusion of IVIG surprisingly antagonized ACV's beneficial effects. Distinct serum proteomic profiles were observed for male and female mice, and the antagonistic effects of ACV and IVIG on behavior were paralleled by similar changes in the serum proteome of ACV- and ACV + IVIG-treated mice. We conclude that inflammation and other factors mediate HSV-induced behavioral impairments and that the effects of ACV and IVIG on behavior involve novel mechanisms.
Collapse
|
19
|
Devakumar D, Bamford A, Ferreira MU, Broad J, Rosch RE, Groce N, Breuer J, Cardoso MA, Copp AJ, Alexandre P, Rodrigues LC, Abubakar I. Infectious causes of microcephaly: epidemiology, pathogenesis, diagnosis, and management. THE LANCET. INFECTIOUS DISEASES 2017; 18:e1-e13. [PMID: 28844634 DOI: 10.1016/s1473-3099(17)30398-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 06/02/2017] [Accepted: 06/15/2017] [Indexed: 01/08/2023]
Abstract
Microcephaly is an important sign of neurological malformation and a predictor of future disability. The 2015-16 outbreak of Zika virus and congenital Zika infection brought the world's attention to links between Zika infection and microcephaly. However, Zika virus is only one of the infectious causes of microcephaly and, although the contexts in which they occur vary greatly, all are of concern. In this Review, we summarise important aspects of major congenital infections that can cause microcephaly, and describe the epidemiology, transmission, clinical features, pathogenesis, management, and long-term consequences of these infections. We include infections that cause substantial impairment: cytomegalovirus, herpes simplex virus, rubella virus, Toxoplasma gondii, and Zika virus. We highlight potential issues with classification of microcephaly and show how some infants affected by congenital infection might be missed or incorrectly diagnosed. Although Zika virus has brought the attention of the world to the problem of microcephaly, prevention of all infectious causes of microcephaly and appropriately managing its consequences remain important global public health priorities.
Collapse
Affiliation(s)
- Delan Devakumar
- Institute for Global Health, University College London, London, UK.
| | - Alasdair Bamford
- Infectious Diseases Department, Great Ormond Street Hospital, London, UK; Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Marcelo U Ferreira
- Department of Parasitology, Institute for Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jonathan Broad
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Richard E Rosch
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Nora Groce
- Leonard Cheshire Disability and Inclusive Development Centre, University College London, London, UK
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, UK
| | - Marly A Cardoso
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Andrew J Copp
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Paula Alexandre
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Laura C Rodrigues
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Ibrahim Abubakar
- Institute for Global Health, University College London, London, UK
| |
Collapse
|
20
|
Kulkarni A, Scully TJ, O'Donnell LA. The antiviral cytokine interferon-gamma restricts neural stem/progenitor cell proliferation through activation of STAT1 and modulation of retinoblastoma protein phosphorylation. J Neurosci Res 2016; 95:1582-1601. [PMID: 27862183 PMCID: PMC5432422 DOI: 10.1002/jnr.23987] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/18/2016] [Accepted: 10/14/2016] [Indexed: 12/20/2022]
Abstract
Neural stem/progenitor cells (NPSCs) express receptors for many inflammatory cytokines, with varying effects on differentiation and proliferation depending on the stage of development and the milieu of inflammatory mediators. In primary neurons and astrocytes, we recently showed that interferon gamma (IFNγ), a potent antiviral cytokine that is required for the control and clearance of many central nervous system (CNS) infections, could differentially affect cell survival and cell cycle progression depending upon the cell type and the profile of activated intracellular signaling molecules. Here, we show that IFNγ inhibits proliferation of primary NSPCs through dephosphorylation of the tumor suppressor Retinoblastoma protein (pRb), which is dependent on activation of signal transducers and activators of transcription‐1 (STAT1) signaling pathways. Our results show i) IFNγ inhibits neurosphere growth and proliferation rate in a dose‐dependent manner; ii) IFNγ blocks cell cycle progression through a late‐stage G1/S phase restriction; iii) IFNγ induces phosphorylation and expression of STAT1 and STAT3; iv) IFNγ decreases cyclin E/cdk2 expression and reduces phosphorylation of cyclin D1 and pRb on serine residue 795; and v) the effects of IFNγ on NSPC proliferation, cell cycle protein expression, and pRb phosphorylation are STAT1‐dependent. These data define a mechanism by which IFNγ could contribute to a reduction in NSPC proliferation in inflammatory conditions. Further delineation of the effects of inflammatory cytokines on NSPC growth could improve our understanding of how CNS infections and other inflammatory events disrupt brain development and NSPC function. © 2016 The Authors. Journal of Neuroscience Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Apurva Kulkarni
- Duquesne University, Mylan School of Pharmacy, 600 Forbes Avenue, Pittsburgh, PA, 15282
| | - Taylor J Scully
- Duquesne University, Mylan School of Pharmacy, 600 Forbes Avenue, Pittsburgh, PA, 15282
| | - Lauren A O'Donnell
- Duquesne University, Mylan School of Pharmacy, 600 Forbes Avenue, Pittsburgh, PA, 15282
| |
Collapse
|
21
|
Kulkarni A, Ganesan P, O'Donnell LA. Interferon Gamma: Influence on Neural Stem Cell Function in Neurodegenerative and Neuroinflammatory Disease. Clin Med Insights Pathol 2016; 9:9-19. [PMID: 27774000 PMCID: PMC5065109 DOI: 10.4137/cpath.s40497] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 01/05/2023] Open
Abstract
Interferon-gamma (IFNγ), a pleiotropic cytokine, is expressed in diverse neurodegenerative and neuroinflammatory conditions. Its protective mechanisms are well documented during viral infections in the brain, where IFNγ mediates non-cytolytic viral control in infected neurons. However, IFNγ also plays both protective and pathological roles in other central nervous system (CNS) diseases. Of the many neural cells that respond to IFNγ, neural stem/progenitor cells (NSPCs), the only pluripotent cells in the developing and adult brain, are often altered during CNS insults. Recent studies highlight the complex effects of IFNγ on NSPC activity in neurodegenerative diseases. However, the mechanisms that mediate these effects, and the eventual outcomes for the host, are still being explored. Here, we review the effects of IFNγ on NSPC activity during different pathological insults. An improved understanding of the role of IFNγ would provide insight into the impact of immune responses on the progression and resolution of neurodegenerative diseases.
Collapse
Affiliation(s)
- Apurva Kulkarni
- Mylan School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Priya Ganesan
- Mylan School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Lauren A O'Donnell
- Mylan School of Pharmacy and the Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| |
Collapse
|
22
|
Fantetti KN, Gray EL, Ganesan P, Kulkarni A, O'Donnell LA. Interferon gamma protects neonatal neural stem/progenitor cells during measles virus infection of the brain. J Neuroinflammation 2016; 13:107. [PMID: 27178303 PMCID: PMC4867982 DOI: 10.1186/s12974-016-0571-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 05/06/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND In the developing brain, self-renewing neural stem/progenitor cells (NSPC) give rise to neuronal and glial lineages. NSPC survival and differentiation can be altered by neurotropic viruses and by the anti-viral immune response. Several neurotropic viruses specifically target and infect NSPCs, in addition to inducing neuronal loss, which makes it difficult to distinguish between effects on NSPCs that are due to direct viral infection or due to the anti-viral immune response. METHODS We have investigated the impact of anti-viral immunity on NSPCs in measles virus (MV)-infected neonates. A neuron-restricted viral infection model was used, where NSPCs remain uninfected. Thus, an anti-viral immune response was induced without the confounding issue of NSPC infection. Two-transgenic mouse lines were used: CD46+ mice express the human isoform of CD46, the MV entry receptor, under the control of the neuron-specific enolase promoter; CD46+/IFNγ-KO mice lack the key anti-viral cytokine IFNγ. Multi-color flow cytometry and Western Blot analysis were used to quantify effects on NSPC, neuronal, and glial cell number, and quantify effects on IFNγ-mediated signaling and cell markers, respectively. RESULTS Flow cytometric analysis revealed that NSPCs were reduced in CD46+/IFNγ-KO mice at 3, 7, and 10 days post-infection (dpi), but were unaffected in CD46+ mice. Early neurons showed the greatest cell loss at 7 dpi in both genotypes, with no effect on mature neurons and glial cells. Thus, IFNγ protected against NSPC loss, but did not protect young neurons. Western Blot analyses on hippocampal explants showed reduced nestin expression in the absence of IFNγ, and reduced doublecortin and βIII-tubulin in both genotypes. Phosphorylation of STAT1 and STAT2 occurred independently of IFNγ in the hippocampus, albeit with distinct regulation of activation. CONCLUSIONS This is the first study to demonstrate bystander effects of anti-viral immunity on NSPC function. Our results show IFNγ protects the NSPC population during a neonatal viral CNS infection. Significant loss of NSPCs in CD46+/IFNγ-KO neonates suggests that the adaptive immune response is detrimental to NSPCs in the absence of IFNγ. These results reveal the importance and contribution of the anti-viral immune response to neuropathology and may be relevant to other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Kristen N Fantetti
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA
| | - Erica L Gray
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA
| | - Priya Ganesan
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA
| | - Apurva Kulkarni
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA
| | - Lauren A O'Donnell
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, 600 Forbes Ave, Pittsburgh, PA, 15282, USA.
| |
Collapse
|
23
|
Sankowski R, Mader S, Valdés-Ferrer SI. Systemic inflammation and the brain: novel roles of genetic, molecular, and environmental cues as drivers of neurodegeneration. Front Cell Neurosci 2015; 9:28. [PMID: 25698933 PMCID: PMC4313590 DOI: 10.3389/fncel.2015.00028] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 01/15/2015] [Indexed: 12/20/2022] Open
Abstract
The nervous and immune systems have evolved in parallel from the early bilaterians, in which innate immunity and a central nervous system (CNS) coexisted for the first time, to jawed vertebrates and the appearance of adaptive immunity. The CNS feeds from, and integrates efferent signals in response to, somatic and autonomic sensory information. The CNS receives input also from the periphery about inflammation and infection. Cytokines, chemokines, and damage-associated soluble mediators of systemic inflammation can also gain access to the CNS via blood flow. In response to systemic inflammation, those soluble mediators can access directly through the circumventricular organs, as well as open the blood–brain barrier. The resulting translocation of inflammatory mediators can interfere with neuronal and glial well-being, leading to a break of balance in brain homeostasis. This in turn results in cognitive and behavioral manifestations commonly present during acute infections – including anorexia, malaise, depression, and decreased physical activity – collectively known as the sickness behavior (SB). While SB manifestations are transient and self-limited, under states of persistent systemic inflammatory response the cognitive and behavioral changes can become permanent. For example, cognitive decline is almost universal in sepsis survivors, and a common finding in patients with systemic lupus erythematosus. Here, we review recent genetic evidence suggesting an association between neurodegenerative disorders and persistent immune activation; clinical and experimental evidence indicating previously unidentified immune-mediated pathways of neurodegeneration; and novel immunomodulatory targets and their potential relevance for neurodegenerative disorders.
Collapse
Affiliation(s)
- Roman Sankowski
- Elmezzi Graduate School of Molecular Medicine , Manhasset, NY , USA ; Feinstein Institute for Medical Research , Manhasset, NY , USA
| | - Simone Mader
- Feinstein Institute for Medical Research , Manhasset, NY , USA
| | - Sergio Iván Valdés-Ferrer
- Elmezzi Graduate School of Molecular Medicine , Manhasset, NY , USA ; Feinstein Institute for Medical Research , Manhasset, NY , USA ; Department of Neurology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán , México City , Mexico
| |
Collapse
|