1
|
Li D, Huo X, Shen L, Qian M, Wang J, Mao S, Chen W, Li R, Zhu T, Zhang B, Liu K, Wu F, Bai Y. Astrocyte heterogeneity in ischemic stroke: Molecular mechanisms and therapeutic targets. Neurobiol Dis 2025; 209:106885. [PMID: 40139279 DOI: 10.1016/j.nbd.2025.106885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Ischemic stroke is one of the major causes of death and disability in adults, bringing a significant economic burden to the society and families. Despite significant advancements in stroke treatment, focusing solely on neurons is insufficient for improving disease progression and prognosis. Astrocytes are the most ubiquitous cells in the brain, and they undergo morphological and functional changes after brain insults, which has been known as astrocyte reactivity. Transcriptomics have shown that reactive astrocytes (RA) are heterogeneous, and they can be roughly classified into neurotoxic and neuroprotective types, thereby affecting the development of central nervous system (CNS) diseases. However, the relationship between stroke and reactive astrocyte heterogeneity has not been fully elucidated, and regulating the heterogeneity of astrocytes to play a neuroprotective role may provide a new perspective for the treatment of stroke. Here we systematically review current advancements in astrocyte heterogeneity following ischemic stroke, elucidate the molecular mechanisms underlying their activation, and further summarize promising therapeutic agents and molecular targets capable of modulating astrocyte heterogeneity.
Collapse
Affiliation(s)
- Daxing Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xinchen Huo
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ling Shen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Minjie Qian
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jindou Wang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Shijie Mao
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wenjing Chen
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Runheng Li
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Tianhao Zhu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Beicheng Zhang
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Kunxuan Liu
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China
| | - Feifei Wu
- Laboratory for Human Anatomy, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Ying Bai
- Department of Pharmacology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
2
|
Miyata M, Kamagata K, Saito Y, Uchida W, Takabayashi K, Moriguchi S, Momota Y, Ichihashi M, Kurose S, Endo H, Tagai K, Kataoka Y, Mimura M, Aoki S, Higuchi M, Takahata K. Diffusion Tensor Image Analysis Along the Perivascular Space in Former Professional Athletes with Repetitive Mild Traumatic Brain Injury History. Acad Radiol 2025:S1076-6332(25)00379-4. [PMID: 40318971 DOI: 10.1016/j.acra.2025.04.028] [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: 02/03/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/07/2025]
Abstract
RATIONALE AND OBJECTIVES The long-term changes in the glymphatic system of former professional athletes exposed to repetitive mild traumatic brain injuries remain poorly understood. This study aimed to use diffusion tensor image analysis along the perivascular space (DTI-ALPS) to evaluate the glymphatic system activity and correlate the ALPS index with neuropsychiatric symptoms in former professional athletes. MATERIALS AND METHODS 30 former professional athletes and 24 age- and sex-matched controls underwent DTI with 3 T magnetic resonance imaging, and neuropsychiatric tests were performed in the athlete group. RESULTS The ALPS index (mean, right, and left) in the athlete group was compared to that in controls, and correlations with clinical variables were analyzed. The mean, right, and left ALPS indices in the athlete group were significantly lower than those of the control group (mean: 1.49±0.12 vs. 1.61±0.16, cohen's d=0.847, p<0.01; right: 1.51±0.12 vs. 1.61±0.16, cohen's d=0.722, p=0.01; and left: 1.47±0.15 vs. 1.60±0.20, cohen's d=0.765, p<0.01). The mean and right ALPS indices were positively correlated with the Wisconsin Card Sorting Test performance in the athlete group (mean: r=0.41, p=0.04; right: r=0.43, p=0.03; not significant after Bonferroni correction). CONCLUSION A lower ALPS index in former professional athletes may be associated with impairments in cognitive function, reflected in glymphatic dysfunction.
Collapse
Affiliation(s)
- Mari Miyata
- Department of Applied MRI Research, Institute of Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan; Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuya Saito
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Wataru Uchida
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kaito Takabayashi
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sho Moriguchi
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Yuki Momota
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Masanori Ichihashi
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Shin Kurose
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Hironobu Endo
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Kenji Tagai
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Yuko Kataoka
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Masaru Mimura
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Makoto Higuchi
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Keisuke Takahata
- Advanced Neuroimaging Center, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology (QST), Chiba, Japan.
| |
Collapse
|
3
|
Hicks AJ, Carrington H, Bura L, Yang A, Pesce R, Yew B, Dams-O'Connor K. Blood-Based Protein Biomarkers in the Chronic Phase of Traumatic Brain Injury: A Systematic Review. J Neurotrauma 2025; 42:759-797. [PMID: 40176450 DOI: 10.1089/neu.2024.0294] [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] [Indexed: 04/04/2025] Open
Abstract
There has been limited exploration of blood-based biomarkers in the chronic period following traumatic brain injury (TBI). Our objective was to conduct a systematic review of studies examining blood-based protein biomarkers with at least one sample collected 12 months post-TBI in adults (≥16 years). Database searches were conducted in Embase, MEDLINE, and Science Citation Index-Expanded on July 24, 2023. Risk of bias was assessed using modified Joanna Briggs Institute critical appraisal tools. Only 30 of 12,523 articles met inclusion criteria, with samples drawn from 12 months to 48 years. Higher quality evidence (low risk of bias; large samples) identified promising inflammatory biomarkers at 12 months post-injury in both moderate-severe TBI (GFAP) and mild TBI (eotaxin-1, IFN-y, IL-8, IL-9, IL-17A, MCP-1, MIP-1β, FGF-basic, and TNF-α). Studies with low risk of bias but smaller samples also suggest NSE, MME, and CRP may be informative, alongside protein variants for α-syn (10H, D5), amyloid-β (A4, C6T), TDP-43 (AD-TDP 1;2;3;9;11), and tau (D11C). Findings for NfL were inconclusive. Longitudinal data were mostly available for acute samples followed until 12 months post-injury, with limited evaluation of changes beyond 12 months. Associations of some blood-based biomarkers with cognitive, sleep, and functional outcomes were reported. The overall strength of the evidence in this review was limited by the risk of bias and small sample sizes. Replication is required within prospective longitudinal studies that move beyond 12 months post-injury. Novel efforts should be guided by promising neurodegenerative-disease markers and use panels to model polypathology.
Collapse
Affiliation(s)
- Amelia J Hicks
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Holly Carrington
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lisa Bura
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alicia Yang
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rico Pesce
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Belinda Yew
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | |
Collapse
|
4
|
Liao Y, Wen L, Zheng R, Shen Y, Ha TA, Lin M, Cheng R, Gao Y, Shang P. Novel Perspectives Focused on the Relationship Between Herpesvirus Encephalitis and Anti-GFAP-Antibody-Positive Astrocytopathy. Mol Neurobiol 2025; 62:6179-6194. [PMID: 39731639 DOI: 10.1007/s12035-024-04660-0] [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: 06/03/2024] [Accepted: 12/02/2024] [Indexed: 12/30/2024]
Abstract
Virus encephalitis (VE), recognized as one of the common kinds of central nervous system (CNS) diseases after virus infection, has a surprising correlation with autoimmune encephalitis (AE) when autoimmune antibodies emerge in cerebrospinal fluid (CSF) or serum. Herpes simplex virus and Epstein-Barr virus are the most critical agents worldwide. By molecular mimicry, herpes viruses can invade the brain directly or indirectly. As a type-III intermediate filament, glial fibrillary acidic protein (GFAP) can be seen in both the central and peripheral nervous system and is regarded as a marker of astrocyte activation. Autoimmune glial fibrillary acidic protein astrocytopathy (GFAP-A), an autoimmune inflammatory CNS disorder with unearthed pathogenic mechanism yet, is correlated with CD8 + T cells and AQP4 astrocytopathy in TNF signaling. It brings a new concept of VE and GFAP coexisting, which has been documented in several case reports. Considering the infectious role of herpes viruses in CNS, EBV contributes to GFAP-IgG significantly and may result in GFAP-A. Coincidently, the existence of GFAP-IgG in patients with infection of herpes viruses has been documented as well. There exist multiple diagnoses of VE, ranging from traditional diagnostic criteria, such as CSF examination and electronic techniques, to a novel approach, according to case reports, the detection of GFAP-lgG. In terms of treatment, except for (IVIG), the explorations for new curative targets and optimal diagnostic time are of great necessity. In conclusion, emphasis given to the CNS autoimmune effect brought by the virus infection is highly worthy.
Collapse
Affiliation(s)
- Yuqiao Liao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Linxin Wen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruoyi Zheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yinan Shen
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Teng-Ai Ha
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingkai Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Stomatology, Southern Medical University, Guangzhou, China
| | - Ruogu Cheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Gao
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pei Shang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
5
|
Capasso G, Franssen CFM, Perna AF, Massy ZA, Menzies RI, Zoccali C, Tessitore A, Nedergaard M, Okusa MD, Ortiz A, Wagner CA, Unwin RJ. Drivers and mechanisms of cognitive decline in chronic kidney disease. Nat Rev Nephrol 2025:10.1038/s41581-025-00963-0. [PMID: 40281076 DOI: 10.1038/s41581-025-00963-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2025] [Indexed: 04/29/2025]
Abstract
Cognitive impairment is highly prevalent among individuals with chronic kidney disease (CKD). Despite its high prevalence, the contributing factors and mechanisms underlying brain-kidney dysfunction in CKD remain poorly understood. However, advances in neuroscience, including novel imaging techniques and cognitive assessment methods, have begun to clarify this complex relationship. Several factors contribute directly to cognitive decline in people with CKD, including accumulation of uraemic toxins, microvascular damage, malnutrition, chronic inflammation and disruptions in key neuroprotective pathways, such as those involving Klotho and the glymphatic system. These factors are also linked to the accelerated ageing observed in people with CKD, a key contributor to cognitive decline. However, most studies on cognition in people with CKD have been cross-sectional and associative, offering limited insight into causation. Research advances, such as studies on the effect of uraemic toxins on the blood-brain barrier and the role of the endothelial glycocalyx in vascular damage, offer promising new directions. Emerging data from longitudinal cohort studies are also enhancing our understanding of these processes, with potential implications for both the treatment of CKD-related cognitive decline and the broader issue of cognitive dysfunction in ageing populations. Here, we examine key mechanisms linking CKD to cognitive decline and consider potential therapeutic interventions.
Collapse
Affiliation(s)
- Giovambattista Capasso
- Biogem, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy.
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Naples, Italy.
| | - Casper F M Franssen
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alessandra F Perna
- Department of Translational Medical Science, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ziad A Massy
- AURA (Association pour l'Utilization du Rein Artificiel dans la Region Parisienne) Paris, Department of Nephrology, CHU Ambroise Paré, AP-HP, Paris, France
- Centre for Research in Epidemiology and Population Health (CESP), University Paris-Saclay, University Versailles-Saint Quentin, Inserm UMRS 1018, Clinical Epidemiology Team, Villejuif, France
| | - Robert I Menzies
- Edinburgh Kidney, British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Carmine Zoccali
- Biogem, Institute of Molecular Biology and Genetics, Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio, Italy
| | - Alessandro Tessitore
- Department of Advanced Surgical and Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maiken Nedergaard
- Center for Basic and Translational Neuroscience, University of Copenhagen, Copenhagen, Denmark
| | - Mark D Okusa
- Division of Nephrology, Center for Immunity Inflammation and Regenerative Medicine University of Virginia, Charlottesville, VA, USA
| | - Alberto Ortiz
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD UAM), Madrid, Spain
| | - Carsten A Wagner
- Institute of Physiology and Zurich Kidney Center University of Zurich, Zurich, Switzerland
| | - Robert J Unwin
- UCL Centre for Kidney and Bladder Health, University College London, London, UK
| |
Collapse
|
6
|
Shen Y, Zhang L, Ding G, Boyd E, Kaur J, Li Q, Haacke EM, Hu J, Jiang Q. Vascular Contribution to Cerebral Waste Clearance Affected by Aging or Diabetes. Diagnostics (Basel) 2025; 15:1019. [PMID: 40310437 PMCID: PMC12026099 DOI: 10.3390/diagnostics15081019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
Background: The brain's vascular system has recently been shown to provide an important efflux pathway for cerebral waste clearance (CWC). However, little is known about the influence of aging or diabetes on the CWC. The aim of the current study is to investigate the vasculature contribution to CWC under aging and diabetic conditions. Methods: Male Wistar rats under aging and diabetic conditions were evaluated using dynamic intra-cisterna superparamagnetic iron oxide-enhanced susceptibility-weighted imaging (SPIO-SWI). Theoretical analysis of the expected signal intensity using SPIO-SWI was compared with the corresponding dynamic in vivo images. Quantitative susceptibility mapping (QSM) was used to evaluate the iron-based tracer concentration in the venous system. Results: Our data demonstrated that the theoretical analysis predicted the dynamic changes in the signal intensity after SPIO infusion. The distinct hyperintense signals due to the lower concentration of the SPIO over time in cerebrospinal fluid (CSF) and meningeal lymphatic (ML) vessels likely represented the CWC through various efflux pathways, including cerebral vascular and ML vessels. The QSM analysis further revealed reduced CWC from the vasculature in both the aged and diabetic groups compared to the younger group. Conclusions: Our results demonstrated that SPIO-SWI can quantitatively evaluate the CWC efflux contributions from cerebral vascular vessels under aging or diabetic conditions.
Collapse
Affiliation(s)
- Yimin Shen
- Department of Radiology, Wayne State University, Detroit, MI 48202, USA; (Y.S.); (E.M.H.); (J.H.)
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (L.Z.); (G.D.); (E.B.); (J.K.); (Q.L.)
- Department of Neurology, Michigan State University, East Lansing, MI 48824, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (L.Z.); (G.D.); (E.B.); (J.K.); (Q.L.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Edward Boyd
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (L.Z.); (G.D.); (E.B.); (J.K.); (Q.L.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Jasleen Kaur
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (L.Z.); (G.D.); (E.B.); (J.K.); (Q.L.)
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (L.Z.); (G.D.); (E.B.); (J.K.); (Q.L.)
| | - E. Mark Haacke
- Department of Radiology, Wayne State University, Detroit, MI 48202, USA; (Y.S.); (E.M.H.); (J.H.)
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI 48202, USA; (Y.S.); (E.M.H.); (J.H.)
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (L.Z.); (G.D.); (E.B.); (J.K.); (Q.L.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
7
|
Lisi I, Moro F, Mazzone E, Marklund N, Pischiutta F, Kobeissy F, Mao X, Corrigan F, Helmy A, Nasrallah F, Pietro VD, Ngwenya LB, Portela LV, Semple BD, Schneider ALC, Arrastia RD, Menon DK, Smith DH, Wellington C, Loane DJ, Wang KKW, Zanier ER. Exploiting blood-based biomarkers to align preclinical models with human traumatic brain injury. Brain 2025; 148:1062-1080. [PMID: 39514789 PMCID: PMC11967814 DOI: 10.1093/brain/awae350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/17/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
Rodent models are important research tools for studying the pathophysiology of traumatic brain injury (TBI) and developing new therapeutic interventions for this devastating neurological disorder. However, the failure rate for the translation of drugs from animal testing to human treatments for TBI is 100%. While there are several potential explanations for this, previous clinical trials have relied on extrapolation from preclinical studies for critical design considerations, including drug dose optimization, post-injury drug treatment initiation and duration. Incorporating clinically relevant biomarkers in preclinical studies may provide an opportunity to calibrate preclinical models to identical (or similar) measurements in humans, link to human TBI biomechanics and pathophysiology, and guide therapeutic decisions. To support this translational goal, we conducted a systematic literature review of preclinical TBI studies in rodents measuring blood levels of clinically used GFAP, UCH-L1, NfL, total-Tau (t-Tau) or phosphorylated-Tau (p-Tau) published in PubMed/EMBASE up to 10 April 2024. Although many factors influence clinical TBI outcomes, many of those cannot routinely be assessed in rodent studies (e.g. intracranial pressure monitoring). Thus we focused on blood biomarkers' temporal trajectories and discuss our findings in the context of the latest clinical TBI biomarker data. Of 805 original preclinical studies, 74 met the inclusion criteria, with a median quality score of 5 (25th-75th percentiles: 4-7) on the CAMARADES checklist. GFAP was measured in 43 studies, UCH-L1 in 21, NfL in 20, t-Tau in 19 and p-Tau in seven. Data from rodent models indicate that all biomarkers exhibited injury severity-dependent elevations with distinct temporal profiles. GFAP and UCH-L1 peaked within the first day after TBI (30- and 4-fold increases, respectively, in moderate-to-severe TBI versus sham), with the highest levels observed in the contusion TBI model. NfL peaked within days (18-fold increase) and remained elevated up to 6 months post-injury. GFAP and NfL show a pharmacodynamic response in 64.7% and 60%, respectively, of studies evaluating neuroprotective therapies in preclinical models. However, GFAP's rapid decline post-injury may limit its utility for understanding the response to new therapeutics beyond the hyperacute phase after experimental TBI. Furthermore, as in humans, subacute NfL levels inform on chronic white matter loss after TBI. t-Tau and p-Tau levels increased over weeks after TBI (up to 6- and 16-fold, respectively); however, their relationship with underlying neurodegeneration has yet to be addressed. Further investigation into biomarker levels in the subacute and chronic phases after TBI will be needed to fully understand the pathomechanisms underpinning blood biomarkers' trajectories and select the most suitable experimental model to optimally relate preclinical mechanistic studies to clinical observations in humans. This new approach could accelerate the translation of neuroprotective treatments from laboratory experiments to real-world clinical practices.
Collapse
Affiliation(s)
- Ilaria Lisi
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan 20156, Italy
| | - Federico Moro
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan 20156, Italy
| | - Edoardo Mazzone
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan 20156, Italy
| | - Niklas Marklund
- Department of Clinical Sciences Lund, Neurosurgery, Lund University and Skåne University Hospital, Lund 222 42, Sweden
| | - Francesca Pischiutta
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan 20156, Italy
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Xiang Mao
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Frances Corrigan
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Fatima Nasrallah
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Valentina Di Pietro
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Laura B Ngwenya
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH 670715, USA
| | - Luis V Portela
- Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul—UFRGS, Porto Alegre, RS 90040-060, Brasil
| | - Bridgette D Semple
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, VIC 3800, Australia
| | - Andrea L C Schneider
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6021, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ramon Diaz Arrastia
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David K Menon
- Division of Anaesthesia, University of Cambridge, Cambridge CB2 2QQ, UK
| | - Douglas H Smith
- Center for Brain Injury and Repair and the Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cheryl Wellington
- Department of Pathology, Djavad Mowafaghain Centre for Brain Health, International Collaboration on Repair Discoveries, School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - David J Loane
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 152-160, Ireland
| | - Kevin K W Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Elisa R Zanier
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan 20156, Italy
| |
Collapse
|
8
|
Álvarez-Sánchez L, Ferré-González L, Peña-Bautista C, Balaguer Á, Amengual JL, Baquero M, Cubas L, Casanova B, Cháfer-Pericás C. New approach to specific Alzheimer's disease diagnosis based on plasma biomarkers in a cognitive disorder cohort. Eur J Clin Invest 2025:e70034. [PMID: 40119567 DOI: 10.1111/eci.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/11/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND The validation of a combination of plasma biomarkers and demographic variables is required to establish reliable cut-offs for Alzheimer's disease diagnosis (AD). METHODS Plasma biomarkers (Aβ42/Aβ40, p-Tau181, t-Tau, NfL, GFAP), ApoE genotype, and demographic variables were obtained from a retrospective clinical cohort of cognitive disorders (n = 478). These patients were diagnosed as AD (n = 254) or non-AD (n = 224) according to cerebrospinal fluid (CSF) Aβ42/Aβ40 levels. An analysis using a Ridge logistic regression model was performed to predict the occurrence of AD. The predictive performance of the model was assessed using the observations from a training set (70% of the sample) and validated using a test set (30% of the sample) in each group. Optimum cutoffs for the model were evaluated. RESULTS The model including plasma Aβ42/Aβ40, p-Tau181, GFAP, ApoE genotype and age was optimal for predicting CSF Aβ42/Aβ40 positivity (AUC .91, sensitivity .86, specificity .82). The model including only plasma biomarkers (Aβ42/Aβ40, p-Tau181, GFAP) provided reliable results (AUC .88, sensitivity .83, specificity .78). Also, GFAP, individually, showed the best performance in discriminating between AD and non-AD groups (AUC .859). The established cut-offs in a three-range strategy performed satisfactorily for the validated predictive model (probability) and individual plasma GFAP (concentration). CONCLUSIONS The plasma GFAP levels and the validated predictive model based on plasma biomarkers represent a relevant step toward the development of a potential clinical approach for AD diagnosis, which should be assessed in further research.
Collapse
Affiliation(s)
- Lourdes Álvarez-Sánchez
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Laura Ferré-González
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Carmen Peña-Bautista
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Ángel Balaguer
- Plataforma de Big Data, IA y Bioestadística, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Julián Luis Amengual
- Plataforma de Big Data, IA y Bioestadística, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Miguel Baquero
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Division of Neurology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Laura Cubas
- Division of Neuroinmunology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Bonaventura Casanova
- Division of Neuroinmunology, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Consuelo Cháfer-Pericás
- Alzheimer's Disease Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
9
|
Uzgiris AJ, Ladic LA, Pfister SX. Advances in neurofilament light chain analysis. Adv Clin Chem 2025; 126:31-71. [PMID: 40185536 DOI: 10.1016/bs.acc.2025.01.006] [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] [Indexed: 04/07/2025]
Abstract
This chapter provides a comprehensive summary of clinical laboratory testing for neurofilament light chain (NfL) in neurologic disease. A primer on the NfL structure and function is presented with its potential use as a biomarker. The most widely utilized methods for NfL in biologic samples are highlighted and examined. Limitations of current knowledge are considered, as are outstanding questions related to dissemination and standardization of testing. Herein we focus on methods available today and those in development for clinical use. In the final section, a broad vision is presented of how NfL may be utilized in the future to improve diagnosis and treatment of neurologic diseases as well as for maintaining health.
Collapse
Affiliation(s)
- Arejas J Uzgiris
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States.
| | - Lance A Ladic
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States
| | - Sophia X Pfister
- Siemens Healthcare Diagnostics Inc., Tarrytown, NY, United States
| |
Collapse
|
10
|
Csecsei P, Acs P, Gottschal M, Imre P, Miklos E, Simon D, Erdo-Bonyar S, Berki T, Zavori L, Varnai R. The relevance of combined testing of cerebrospinal fluid glial fibrillary acidic protein and ubiquitin C-terminal hydrolase L1 in multiple sclerosis and peripheral neuropathy. Neurol Sci 2025; 46:1301-1312. [PMID: 39565457 PMCID: PMC11828760 DOI: 10.1007/s10072-024-07790-4] [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: 07/08/2024] [Accepted: 09/30/2024] [Indexed: 11/21/2024]
Abstract
INTRODUCTION This study investigates the significance of glial fibrillary acidic protein (GFAP) and ubiquitin C-terminal hydrolase L1 (UCHL-1) in cerebrospinal fluid (CSF) of patients with multiple sclerosis (MS) and peripheral neuropathy (PN). METHODS We included 41 MS patients, 35 PN patients, and 36 controls across 5 sites. MS patient data included lesion counts, disease activity, albumin quotient, and Expanded Disability Status Scale (EDSS) scores. PN patients included those with acute and chronic inflammatory demyelinating polyneuropathy and sensorimotor neuropathy based on nerve conduction studies. CSF concentrations of GFAP and UCHL-1 were measured using the MILLIPLEX Map Human Neuroscience Magnetic Bead Panel 1. RESULTS Both GFAP and UCHL-1 levels were significantly higher in the two patient groups compared to controls. In the MS group, GFAP showed a strong correlation with disease duration, EDSS score, non-enhancing lesions, and the CSF/blood albumin quotient. UCHL-1 levels were significantly higher in patients with active disease (gadolinium-enhancing lesions). The combination of UCHL-1 and GFAP improved diagnostic accuracy (AUC 0.895, 95% CI 0.780-1.000) compared to the independent measurement of either marker for indicating Gd-negative lesions. In the PN group, CSF GFAP levels were significantly lower in patients with purely demyelinating neuropathy compared to those with axonal or mixed neuropathy. CONCLUSION GFAP serves as a sensitive marker for axonal damage in PN, while UCHL-1 closely correlates with disease activity in MS patients.
Collapse
Affiliation(s)
- Peter Csecsei
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Peter Acs
- Department of Neurology, Medical School, University of Pecs, Pecs, Hungary
| | - Marianna Gottschal
- Department of Neurology, Kanizsai Dorottya Hospital, Nagykanizsa, Hungary
| | - Piroska Imre
- Department of Neurology, Veszprém County Csolnoky Ferenc Hospital, Veszprém, Hungary
| | - Egon Miklos
- Department of Neurology, Vas County Markusovszky University Teaching Hospital, Szombathely, Hungary
| | - Diana Simon
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Szigeti str. 12, Pecs, 7624, Hungary.
| | - Szabina Erdo-Bonyar
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Szigeti str. 12, Pecs, 7624, Hungary
| | - Timea Berki
- Department of Immunology and Biotechnology, Medical School, University of Pecs, Szigeti str. 12, Pecs, 7624, Hungary
| | - Laszlo Zavori
- Emergency Department, Saudi German Hospital, Dubai, United Arab Emirates
| | - Reka Varnai
- Department of Primary Health Care, Medical School University of Pecs, Pecs, Hungary
| |
Collapse
|
11
|
Nessel I, Whiley L, Dyall SC, Michael-Titus AT. A plasma lipid signature in acute human traumatic brain injury: Link with neuronal injury and inflammation markers. J Cereb Blood Flow Metab 2025; 45:443-458. [PMID: 39188133 PMCID: PMC11572080 DOI: 10.1177/0271678x241276951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 08/28/2024]
Abstract
Traumatic brain injury (TBI) leads to major membrane lipid breakdown. We investigated plasma lipids over 3 days post-TBI, to identify a signature of acute human TBI and assess its correlation with neuronal injury and inflammation. Plasma from patients with TBI (Abbreviated Injury Scale (AIS)3 - serious injury, n = 5; AIS4 - severe injury, n = 8), and controls (n = 13) was analysed for lipidomic profile, neurofilament light (NFL) and cytokines, and the omega-3 index was measured in red blood cells. A lipid signature separated TBI from controls, at 24 and 72 h. Major species driving the separation were: lysophosphatidylcholine (LPC), phosphatidylcholine (PC) and hexosylceramide (HexCer). Docosahexaenoic acid (DHA, 22:6) and LPC (0:0/22:6) decreased post-injury. NFL levels were increased at 24 and 72 h post-injury in AIS4 TBI vs. controls. Interleukin (IL-)6, IL-2 and IL-13 were elevated at 24 h in AIS4 patients vs. controls. NFL and IL-6 were negatively correlated with several lipids. The omega-3 index at admission was low in all patients (controls: 4.3 ± 1.1% and TBI: 4.0 ± 1.1%) and did not change significantly over 3 days post-injury. We have identified specific lipid changes, correlated with markers of injury and inflammation in acute TBI. These observations could inform future lipid-based therapeutic approaches.
Collapse
Affiliation(s)
- Isabell Nessel
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Luke Whiley
- Health Futures Institute, Murdoch University, Murdoch, Australia
| | - Simon C Dyall
- School of Life and Health Sciences, University of Roehampton, London, UK
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
12
|
Michalaki E, Pulliam AN, Datta Roy PM, Dixon JB, LaPlaca MC. Near-Infrared Imaging of Glymphatic Clearance in a Pre-Clinical Model of Repetitive Closed Head Traumatic Brain Injury. Neurotrauma Rep 2025; 6:115-128. [PMID: 39990707 PMCID: PMC11839536 DOI: 10.1089/neur.2024.0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025] Open
Abstract
Traumatic brain injury (TBI) is a major health disorder for which there are few treatments. The glymphatic system is the brain's inbuilt lymphatic-like system that is thought to be responsible for clearing waste products from the brain to the lymph nodes. Although there is evidence that glymphatic drainage is crucial for brain homeostasis, its role in TBI pathogenesis remains elusive. Here, we investigated how glymphatic clearance is altered following TBI in rats using real-time non-invasive imaging. Twenty-four hours following repetitive closed-head TBI or sham conditions, we injected infrared dye intraventricularly and used near-infrared (NIR) imaging to quantify signal intensity, intensity over time, and appearance time of NIR dye in different brain regions. TBI yielded a lower NIR signal and lower rate of NIR dye change in the lateral ventricle and surrounding parietal cortex compared with sham conditions, indicating reduced cerebrospinal fluid perfusion. NIR dye appearance took significantly longer to reach the anterior regions of the brain, while perfusion to the posterior of the brain was faster in TBI compared with sham animals. Aquaporin-4 (AQP4) expression was reduced 24 h after TBI across all cortical regions examined in the posterior of the brain and in the ventral cortex at all coronal levels, suggesting a complex relationship between AQP4 and glymph function. Furthermore, NIR imaging revealed that NIR dye was detectable in the cervical lymph nodes (CLNs) of sham animals but not in TBI animals, yet there was evidence of blood accumulation in the CLNs of TBI animals, suggesting that TBI-related extravascular blood is removed through the glymph system. These data indicate that TBI disrupts normal brain efflux kinetics and reduces glymphatic drainage to the CLNs, demonstrating that restoring glymphatic function may be a promising therapeutic target.
Collapse
Affiliation(s)
- Eleftheria Michalaki
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Alexis N. Pulliam
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Pooja M. Datta Roy
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - J. Brandon Dixon
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Michelle C. LaPlaca
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Nepozitek J, Dusek P, Sonka K. Glymphatic system, sleep, and Parkinson's disease: interconnections, research opportunities, and potential for disease modification. Sleep 2025; 48:zsae251. [PMID: 39450429 PMCID: PMC11725518 DOI: 10.1093/sleep/zsae251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Indexed: 10/26/2024] Open
Affiliation(s)
- Jiri Nepozitek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| | - Petr Dusek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| | - Karel Sonka
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, 12000 Prague, Czech Republic
| |
Collapse
|
14
|
Zhang H, Wang J, Qu Y, Yang Y, Guo ZN. Brain injury biomarkers and applications in neurological diseases. Chin Med J (Engl) 2025; 138:5-14. [PMID: 38915214 PMCID: PMC11717530 DOI: 10.1097/cm9.0000000000003061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 06/26/2024] Open
Abstract
ABSTRACT Neurological diseases are a major health concern, and brain injury is a typical pathological process in various neurological disorders. Different biomarkers in the blood or the cerebrospinal fluid are associated with specific physiological and pathological processes. They are vital in identifying, diagnosing, and treating brain injuries. In this review, we described biomarkers for neuronal cell body injury (neuron-specific enolase, ubiquitin C-terminal hydrolase-L1, αII-spectrin), axonal injury (neurofilament proteins, tau), astrocyte injury (S100β, glial fibrillary acidic protein), demyelination (myelin basic protein), autoantibodies, and other emerging biomarkers (extracellular vesicles, microRNAs). We aimed to summarize the applications of these biomarkers and their related interests and limits in the diagnosis and prognosis for neurological diseases, including traumatic brain injury, status epilepticus, stroke, Alzheimer's disease, and infection. In addition, a reasonable outlook for brain injury biomarkers as ideal detection tools for neurological diseases is presented.
Collapse
Affiliation(s)
- Han Zhang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jing Wang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
- Neuroscience Research Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
15
|
Gogishvili D, Honey MIJ, Verberk IMW, Vermunt L, Hol EM, Teunissen CE, Abeln S. The GFAP proteoform puzzle: How to advance GFAP as a fluid biomarker in neurological diseases. J Neurochem 2025; 169:e16226. [PMID: 39289040 DOI: 10.1111/jnc.16226] [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: 06/05/2024] [Revised: 07/19/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
Glial fibrillary acidic protein (GFAP) is a well-established biomarker of reactive astrogliosis in the central nervous system because of its elevated levels following brain injury and various neurological disorders. The advent of ultra-sensitive methods for measuring low-abundant proteins has significantly enhanced our understanding of GFAP levels in the serum or plasma of patients with diverse neurological diseases. Clinical studies have demonstrated that GFAP holds promise both as a diagnostic and prognostic biomarker, including but not limited to individuals with Alzheimer's disease. GFAP exhibits diverse forms and structures, herein referred to as its proteoform complexity, encompassing conformational dynamics, isoforms and post-translational modifications (PTMs). In this review, we explore how the proteoform complexity of GFAP influences its detection, which may affect the differential diagnostic performance of GFAP in different biological fluids and can provide valuable insights into underlying biological processes. Additionally, proteoforms are often disease-specific, and our review provides suggestions and highlights areas to focus on for the development of new assays for measuring GFAP, including isoforms, PTMs, discharge mechanisms, breakdown products, higher-order species and interacting partners. By addressing the knowledge gaps highlighted in this review, we aim to support the clinical translation and interpretation of GFAP in both CSF and blood and the development of reliable, reproducible and specific prognostic and diagnostic tests. To enhance disease pathology comprehension and optimise GFAP as a biomarker, a thorough understanding of detected proteoforms in biofluids is essential.
Collapse
Affiliation(s)
- Dea Gogishvili
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| | - Madison I J Honey
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, UMC Utrecht Brain Centre, University Medical Centre Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam University Medical Centers, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, The Netherlands
| | - Sanne Abeln
- Bioinformatics, Computer Science Department, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- AI Technology for Life, Department of Computing and Information Sciences, Department of Biology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
16
|
Hol EM, Dykstra W, Chevalier J, Cuadrado E, Bugiani M, Aronica E, Verkhratsky A. Neuroglia in leukodystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:159-175. [PMID: 40148043 DOI: 10.1016/b978-0-443-19102-2.00032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Leukodystrophies are a heterogeneous group of rare genetic neurologic disorders characterized by white matter degeneration resulting from mutations affecting glial cells. This review focuses on the primary subtypes-astroglial, oligodendroglial, and microglial leukodystrophies-offering a detailed description of their neuropathologic features and clinical manifestations. It delves into key aspects of the pathogenesis, emphasizing the distinct cellular mechanisms that drive white matter damage. Advances in disease modeling, including the development of animal models with pathologic gene expressions and patient-derived iPS-cell models, have significantly enhanced our understanding of these rare disorders. Insights into the roles of different glial cell types highlight the complexity of leukodystrophies and provide a foundation for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Werner Dykstra
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Juliette Chevalier
- Department of Child Neurology and Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Eloy Cuadrado
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marianna Bugiani
- Department of Child Neurology and Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
17
|
Kim D, Tithof J. Lumped parameter simulations of cervical lymphatic vessels: dynamics of murine cerebrospinal fluid efflux from the skull. Fluids Barriers CNS 2024; 21:104. [PMID: 39702363 DOI: 10.1186/s12987-024-00605-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Growing evidence suggests that for rodents, a substantial fraction of cerebrospinal fluid (CSF) drains by crossing the cribriform plate into the nasopharyngeal lymphatics, eventually reaching the cervical lymphatic vessels (CLVs). Disruption of this drainage pathway is associated with various neurological disorders. METHODS We employ a lumped parameter method to numerically model CSF drainage across the cribriform plate to CLVs. Our model uses intracranial pressure as an inlet pressure and central venous blood pressure as an outlet pressure. The model incorporates initial lymphatic vessels (modeling those in the nasal region) that absorb the CSF and collecting lymphatic vessels (modeling CLVs) to transport the CSF against an adverse pressure gradient. To determine unknown parameters such as wall stiffness and valve properties, we utilize a Monte Carlo approach and validate our simulation against recent in vivo experimental measurements. RESULTS Our parameter analysis reveals the physical characteristics of CLVs. Our results suggest that the stiffness of the vessel wall and the closing state of the valve are crucial for maintaining the vessel size and volume flow rate observed in vivo. We find that a decreased contraction amplitude and frequency leads to a reduction in volume flow rate, and we test the effects of varying the different pressures acting on the CLVs. Finally, we provide evidence that branching of initial lymphatic vessels may deviate from Murray's law to reduce sensitivity to elevated intracranial pressure. CONCLUSIONS This is the first numerical study of CSF drainage through CLVs. Our comprehensive parameter analysis offers guidance for future numerical modeling of CLVs. This study also provides a foundation for understanding physiology of CSF drainage, helping guide future experimental studies aimed at identifying causal mechanisms of reduction in CLV transport and potential therapeutic approaches to enhance flow.
Collapse
Affiliation(s)
- Daehyun Kim
- Department of Mechanical Engineering, University of Minnesota, 111 Church St SE, Minneapolis, MN, 55455, USA
| | - Jeffrey Tithof
- Department of Mechanical Engineering, University of Minnesota, 111 Church St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
18
|
Shen H, Zhu B, Qian Y, Jin J, Zhou J, Peng G, Mo J. Advances in Research on Meningeal Lymphatic Vessels in Central Nervous System Diseases. J Craniofac Surg 2024:00001665-990000000-02238. [PMID: 39630968 DOI: 10.1097/scs.0000000000010872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 10/12/2024] [Indexed: 12/07/2024] Open
Abstract
Meningeal lymphatic vessels (mLVs), located around the dural sinuses, are considered significant participants in cerebrospinal fluid (CSF) circulation. Meningeal lymphatic vessels not only drain fluids and metabolic waste from the brain into deep cervical lymph nodes (dCLNs) but also transport immune cells from the brain to dCLNs, thus regulating the interaction between the central and peripheral immune systems. These vessels play a crucial role in maintaining normal physiological functions of the central nervous system (CNS). Meningeal lymphatic vessels are involved in the pathophysiological processes of various CNS diseases, including neurodegenerative diseases, cerebrovascular diseases, and brain tumors. In aging and various CNS diseases, damage and dysfunction of mLVs have been observed, leading to the abnormal accumulation of toxic substances and exacerbating neural damage. By transporting antigen-presenting cells that have taken up antigens within the brain to dCLNs, mLVs modulate the activation of peripheral immune cells and their migration and infiltration into brain lesions. Certain drug interventions or physical therapies can modulate the drainage function of mLVs, effectively improving the prognosis of CNS diseases. This review provides a detailed introduction to the anatomic structure, physiological roles, and research advances of mLVs in CNS diseases. In addition, we propose new strategies for targeting mLVs in the treatment of CNS diseases.
Collapse
Affiliation(s)
- Huimin Shen
- Department of Neurosurgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang
| | - Bingrui Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University
| | - Yajun Qian
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiancheng Jin
- Department of Neurosurgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang
| | - Jiankuai Zhou
- Department of Neurosurgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang
| | - Guotao Peng
- Department of Neurosurgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang
| | - Jun Mo
- Department of Neurosurgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang
| |
Collapse
|
19
|
Arena JD, Smith DH, Diaz Arrastia R, Cullen DK, Xiao R, Fan J, Harris DC, Lynch CE, Johnson VE. The neuropathological basis of elevated serum neurofilament light following experimental concussion. Acta Neuropathol Commun 2024; 12:189. [PMID: 39633506 PMCID: PMC11619522 DOI: 10.1186/s40478-024-01883-z] [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: 04/30/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Mild traumatic brain injury (mTBI) or concussion is a substantial health problem globally, with up to 15% of patients experiencing persisting symptoms that can significantly impact quality of life. Currently, the diagnosis of mTBI relies on clinical presentation with ancillary neuroimaging to exclude more severe forms of injury. However, identifying patients at risk for a poor outcome or protracted recovery is challenging, in part due to the lack of early objective tests that reflect the relevant underlying pathology. While the pathophysiology of mTBI is poorly understood, axonal damage caused by rotational forces is now recognized as an important consequence of injury. Moreover, serum measurement of the neurofilament light (NfL) protein has emerged as a potentially promising biomarker of injury. Understanding the pathological processes that determine serum NfL dynamics over time, and the ability of NfL to reflect underlying pathology will be critical for future clinical research aimed at reducing the burden of disability after mild TBI. Using a gyrencephalic model of head rotational acceleration scaled to human concussion, we demonstrate significant elevations in serum NfL, with a peak at 3 days post-injury. Moreover, increased serum NfL was detectable out to 2 weeks post-injury, with some evidence it follows a biphasic course. Subsequent quantitative histological examinations demonstrate that axonal pathology, including in the absence of neuronal somatic degeneration, was the likely source of elevated serum NfL. However, the extent of axonal pathology quantified via multiple markers did not correlate strongly with the extent of serum NfL. Interestingly, the extent of blood-brain barrier (BBB) permeability offered more robust correlations with serum NfL measured at multiple time points, suggesting BBB disruption is an important determinant of serum biomarker dynamics after mTBI. These data provide novel insights to the temporal course and pathological basis of serum NfL measurements that inform its utility as a biomarker in mTBI.
Collapse
Affiliation(s)
- John D Arena
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Douglas H Smith
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ramon Diaz Arrastia
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - D Kacy Cullen
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rui Xiao
- The Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jiaxin Fan
- The Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Danielle C Harris
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cillian E Lynch
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Victoria E Johnson
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
20
|
Wu CH, Liao WH, Chu YC, Hsiao MY, Kung Y, Wang JL, Chen WS. Very Low-Intensity Ultrasound Facilitates Glymphatic Influx and Clearance via Modulation of the TRPV4-AQP4 Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401039. [PMID: 39494466 DOI: 10.1002/advs.202401039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Recently, the glymphatic system has been proposed as a mechanism for waste clearance from the brain parenchyma. Glymphatic dysfunction has previously been shown to be associated with several neurological diseases, including Alzheimer's disease, traumatic brain injury, and stroke. As such, it may serve as an important target for therapeutic interventions. In the present study, very low-intensity ultrasound (VLIUS) (center frequency, 1 MHz; pulse repetition frequency, 1 kHz; duty factor, 1%; spatial peak temporal average intensity [Ispta] = 3.68 mW cm2; and duration, 5 min) is found to significantly enhance the influx of cerebrospinal fluid tracers into the paravascular spaces of the brain, and further facilitate interstitial substance clearance from the brain parenchyma, including exogenous β-amyloid. Notably, no evidence of brain damage is observed following VLIUS stimulation. VLIUS may enhance glymphatic influx via the transient receptor potential vanilloid-4-aquaporin-4 pathway in astrocytes. This mechanism may provide insights into VLIUS-regulated glymphatic function that modifies the natural course of central nervous system disorders related to waste clearance dysfunction.
Collapse
Affiliation(s)
- Chueh-Hung Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, 300, Taiwan
| | - Wei-Hao Liao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
| | - Ya-Cherng Chu
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Ming-Yen Hsiao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
| | - Yi Kung
- Department of Biomechatronic Engineering, National Chiayi University, Chiayi, 600, Taiwan
| | - Jaw-Lin Wang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 100, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, 350, Taiwan
| |
Collapse
|
21
|
Friberg S, Lindblad C, Zeiler FA, Zetterberg H, Granberg T, Svenningsson P, Piehl F, Thelin EP. Fluid biomarkers of chronic traumatic brain injury. Nat Rev Neurol 2024; 20:671-684. [PMID: 39363129 DOI: 10.1038/s41582-024-01024-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability across the world. Evidence for the usefulness of imaging and fluid biomarkers to predict outcomes and screen for the need to monitor complications in the acute stage is steadily increasing. Still, many people experience symptoms such as fatigue and cognitive and motor dysfunction in the chronic phase of TBI, where objective assessments for brain injury are lacking. Consensus criteria for traumatic encephalopathy syndrome, a clinical syndrome possibly associated with the neurodegenerative disease chronic traumatic encephalopathy, which is commonly associated with sports concussion, have been defined only recently. However, these criteria do not fit all individuals living with chronic consequences of TBI. The pathophysiology of chronic TBI shares many similarities with other neurodegenerative and neuroinflammatory conditions, such as Alzheimer disease. As with Alzheimer disease, advancements in fluid biomarkers represent one of the most promising paths for unravelling the chain of pathophysiological events to enable discrimination between these conditions and, with time, provide prediction modelling and therapeutic end points. This Review summarizes fluid biomarker findings in the chronic phase of TBI (≥6 months after injury) that demonstrate the involvement of inflammation, glial biology and neurodegeneration in the long-term complications of TBI. We explore how the biomarkers associate with outcome and imaging findings and aim to establish mechanistic differences in biomarker patterns between types of chronic TBI and other neurodegenerative conditions. Finally, current limitations and areas of priority for future fluid biomarker research are highlighted.
Collapse
Affiliation(s)
- Susanna Friberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Lindblad
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Department of Neurosurgery, Uppsala University Hospital, Uppsala, Sweden
| | - Frederick A Zeiler
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Section of Neurosurgery, Department of Surgery, University of Manitoba, Rady Faculty of Health Sciences, Winnipeg, Manitoba, Canada
- Department of Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, Manitoba, Canada
- Pan Am Clinic Foundation, Winnipeg, Manitoba, Canada
- Division of Anaesthesia, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute, University College London, London, UK
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London, Queen Square Institute of Neurology, London, UK
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Tobias Granberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Eric P Thelin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
22
|
Phillips WT, Schwartz JG. Nasal lymphatic obstruction of CSF drainage as a possible cause of Alzheimer's disease and dementia. Front Aging Neurosci 2024; 16:1482255. [PMID: 39497786 PMCID: PMC11532075 DOI: 10.3389/fnagi.2024.1482255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
Alzheimer's disease, the most common form of dementia among older adults, slowly destroys memory and thinking skills. In recent years, scientists have made tremendous progress in understanding Alzheimer's disease, still, they do not yet fully understand what causes the disease. This article proposes a novel etiology for Alzheimer's disease. Our hypothesis developed from a review of nuclear medicine scans, in which the authors observed a significant increase in nasal turbinate vasodilation and blood pooling in patients with hypertension, sleep apnea, diabetes and/or obesity, all risk factors for Alzheimer's disease. The authors propose that nasal turbinate vasodilation and resultant blood pooling lead to the obstruction of normal nasal lymphatic clearance of cerebrospinal fluid and its waste products from the brain. The nasal turbinate vasodilation, due to increased parasympathetic activity, occurs alongside the well-established increased sympathetic activity of the cardiovascular system as seen in patients with hypertension. The increased parasympathetic activity is likely due to an autonomic imbalance secondary to the increase in worldwide consumption of highly processed food associated with dysregulation of the glucose regulatory system. The authors' hypothesis offers a novel mechanism and a new paradigm for the etiology of Alzheimer's disease and helps explain the rapid worldwide rise in the disease and other dementias which are expected to double in the next 20 years. This new paradigm provides compelling evidence for the modulation of the parasympathetic nervous system as a novel treatment strategy for Alzheimer's disease and other degenerative brain diseases, specifically targeting nasal turbinate lymphatic flow.
Collapse
|
23
|
Du T, Raghunandan A, Mestre H, Plá V, Liu G, Ladrón-de-Guevara A, Newbold E, Tobin P, Gahn-Martinez D, Pattanayak S, Huang Q, Peng W, Nedergaard M, Kelley DH. Restoration of cervical lymphatic vessel function in aging rescues cerebrospinal fluid drainage. NATURE AGING 2024; 4:1418-1431. [PMID: 39147980 DOI: 10.1038/s43587-024-00691-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/16/2024] [Indexed: 08/17/2024]
Abstract
Cervical lymphatic vessels (cLVs) have been shown to drain solutes and cerebrospinal fluid (CSF) from the brain. However, their hydrodynamical properties have never been evaluated in vivo. Here, we developed two-photon optical imaging with particle tracking in vivo of CSF tracers (2P-OPTIC) in superficial and deep cLVs of mice, characterizing their flow and showing that the major driver is intrinsic pumping by contraction of the lymphatic vessel wall. Moreover, contraction frequency and flow velocity were reduced in aged mice, which coincided with a reduction in smooth muscle actin expression. Slowed flow in aged mice was rescued using topical application of prostaglandin F2α, a prostanoid that increases smooth muscle contractility, which restored lymphatic function in aged mice and enhanced central nervous system clearance. We show that cLVs are important regulators of CSF drainage and that restoring their function is an effective therapy for improving clearance in aging.
Collapse
Affiliation(s)
- Ting Du
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
| | - Aditya Raghunandan
- Department of Mechanical Engineering, University of Rochester, Rochester, NY, USA
- Department of Mechanical Engineering, University of Michigan-Dearborn, Dearborn, MI, USA
| | - Humberto Mestre
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Virginia Plá
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Guojun Liu
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
| | - Antonio Ladrón-de-Guevara
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Evan Newbold
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
| | - Paul Tobin
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
| | - Daniel Gahn-Martinez
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
| | - Saurav Pattanayak
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
| | - Qinwen Huang
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
| | - Weiguo Peng
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Division of Glial Disease and Therapeutics, University of Rochester, Rochester, NY, USA.
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark.
| | - Douglas H Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
24
|
Shlobin NA, Staple BL, Sclafani M, Harter DH. The Glymphatic System and Subarachnoid Lymphatic-Like Membrane: Recent Developments in Cerebrospinal Fluid Research. World Neurosurg 2024; 190:147-156. [PMID: 39002777 DOI: 10.1016/j.wneu.2024.07.062] [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: 03/08/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) circulates throughout the ventricles, cranial and spinal subarachnoid spaces, and central spinal cord canal. CSF protects the central nervous system through mechanical cushioning, regulation of intracranial pressure, regulation of metabolic homeostasis, and provision of nutrients. Recently, investigators have characterized the glial-lymphatic (glymphatic) system, the analog of the lymphatic system in the central nervous system, and described a fourth meningeal layer; the subarachnoid lymphatic-like membrane (SLYM)relevant to the CSF. METHODS A narrative review was conducted. RESULTS In this review, we summarize these advances. We describe the development of the original model, controversies, a revised model, and a new conceptual framework. We characterize the biological functions, influence of sleep-wake cycles, and effect of aging with relevance to the glymphatic system. We highlight the role of the glymphatic system in Alzheimer's disease, idiopathic normal pressure hydrocephalus, ischemic stroke, subarachnoid hemorrhage, and traumatic brain injury. Next, we characterize the structure and role of the SLYM. Finally, we explore the relevance of the glymphatic system and SLYM to neurosurgery. CONCLUSIONS This manuscript will inform clinicians and scientists regarding preclinical and translational advances in the understanding of the structure, dynamics, and function of the CSF.
Collapse
Affiliation(s)
- Nathan A Shlobin
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| | - Brandon L Staple
- College of Medicine, University of Nebraska, Omaha, Nebraska, USA
| | | | - David H Harter
- Department of Neurosurgery, NYU Langone, New York, New York, USA
| |
Collapse
|
25
|
Guo Y, Wu L, Liu J, Liu J, Sun Z. Correlation between glymphatic dysfunction and cranial defect in severe traumatic brain injury: a retrospective case-control study based on a diffusion tensor image analysis along the perivascular space (DTI-ALPS) investigation. Quant Imaging Med Surg 2024; 14:6756-6766. [PMID: 39281142 PMCID: PMC11400707 DOI: 10.21037/qims-24-348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/17/2024] [Indexed: 09/18/2024]
Abstract
Background To date, limited research has been conducted on the functionality of the glymphatic system during the recovery phase of severe traumatic brain injury (sTBI). This study aimed to use a diffusion tensor image analysis along the perivascular space (DTI-ALPS) to evaluate glymphatic system function in patients recovering from sTBI who underwent unilateral decompressive craniectomy, and to examine the correlation between the ALPS index and the size of the cranial defect. We hypothesized that assessments would reveal ongoing impairments in glymphatic system function among sTBI patients during the recovery phase. Methods A total of 23 patients with a history of sTBI who had previously undergone unilateral decompressive craniectomy at Xiangya Hospital of Central South University from January 2020 to December 2020 were enrolled in the study, along with 33 healthy control (HC) subjects. All the subjects underwent magnetic resonance imaging (MRI) with DTI scans, and the ALPS index was subsequently calculated to assess glymphatic system functionality. Additionally, the circumference and sectional area of the cranial defect were measured for each patient. An analysis of variance (ANOVA) was used to compare the ALPS index values between the sTBI patients and HC subjects, while a Pearson correlation analysis was used to examine the correlation between the ALPS index and cranial defect characteristics. Results The ALPS index values of both the craniectomy side (t=-9.08, P<0.001) and non-craniectomy side (t=-5.06, P<0.001) of the sTBI group were significantly lower than those of the HC group. However, no statistically significant differences were observed between the ALPS index values of the craniectomy and non-craniectomy sides. Additionally, no significant differences were observed in the ALPS index values of both the craniectomy and non-craniectomy sides among the early, intermediate, and late recovery phases. In the sTBI patients, a moderately strong negative correlation was found between the circumference of the cranial defect and the ALPS index of the craniectomy side (r=-0.62, P=0.002), and a moderately negative correlation was found between the sectional area of the cranial defect and the ALPS index of the craniectomy side (r=-0.56, P=0.005). Conclusions The non-invasive DTI-ALPS technique revealed significantly reduced ALPS index values during the recovery phase of sTBI, indicating persistent impairment in glymphatic system function. A significant negative correlation was found between the ALPS index value of the craniectomy side and the size of the cranial defect. These findings suggest that the ALPS index may serve as a valuable prognostic factor in the recovery phase of sTBI.
Collapse
Affiliation(s)
- Yong Guo
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Lin Wu
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jiacheng Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jinfang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhongyi Sun
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
Deng S, Hu Y, Chen S, Xue Y, Yao D, Sun Q, Nedergaard M, Wang W, Ding F. Chronic sleep fragmentation impairs brain interstitial clearance in young wildtype mice. J Cereb Blood Flow Metab 2024; 44:1515-1531. [PMID: 38639025 PMCID: PMC11418708 DOI: 10.1177/0271678x241230188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/06/2023] [Accepted: 12/27/2023] [Indexed: 04/20/2024]
Abstract
Accumulating evidence shows that most chronic neurological diseases have a link with sleep disturbances, and that patients with chronically poor sleep undergo an accelerated cognitive decline. Indeed, a single-night of sleep deprivation may increase metabolic waste levels in cerebrospinal fluid. However, it remains unknown how chronic sleep disturbances in isolation from an underlying neurological disease may affect the glymphatic system. Clearance of brain interstitial waste by the glymphatic system occurs primarily during sleep, driven by multiple oscillators including arterial pulsatility, and vasomotion. Herein, we induced sleep fragmentation in young wildtype mice and assessed the effects on glymphatic activity and cognitive functions. Chronic sleep fragmentation reduced glymphatic function and impaired cognitive functions in healthy mice. A mechanistic analysis showed that the chronic sleep fragmentation suppressed slow vasomotion, without altering cardiac-driven pulsations. Taken together, results of this study document that chronic sleep fragmentation suppresses brain metabolite clearance and impairs cognition, even in the absence of disease.
Collapse
Affiliation(s)
- Saiyue Deng
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yusi Hu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Simiao Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, 310003, China
| | - Yang Xue
- Department of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Di Yao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Sun
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Maiken Nedergaard
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, Department of Neurology, University of Rochester Medical Center, Rochester, NY, 14642, United States
- Division of Glial Disease and Therapeutics, Center for Translational Neuromedicine, University of Copenhagen, Copenhagen 2200, Denmark
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fengfei Ding
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
27
|
Pocevičiūtė D, Wennström M, Ohlsson B. Okinawa-Based Nordic Diet Decreases Plasma Glial Fibrillary Acidic Protein Levels in Type 2 Diabetes Patients. Nutrients 2024; 16:2847. [PMID: 39275164 PMCID: PMC11396978 DOI: 10.3390/nu16172847] [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: 08/08/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/16/2024] Open
Abstract
Elevated levels of glial fibrillary acidic protein (GFAP) in plasma reflect neuroinflammation and are linked to cognitive decline. Preclinical studies show that dietary change can attenuate astrocyte reactivity and neuroinflammation. In the current study, we investigate if the Okinawa-based Nordic (O-BN) diet alters plasma GFAP levels in patients with Type 2 Diabetes (T2D), a metabolic disorder associated with cognitive disturbances and an increased risk of dementia. Plasma GFAP levels were measured in T2D patients (n = 30) at baseline, after 3 months of the diet, and after a subsequent 4 months of unrestricted diets. The GFAP levels decreased significantly after 3 months of the diet (p = 0.048) but reverted to baseline levels after 4 months of unrestricted diets. At baseline, the GFAP levels correlated significantly with levels of the neurodegeneration marker neurofilament light polypeptide (r = 0.400*) and, after correcting for age, sex, and body mass index, with proinflammatory plasma cytokines (ranging from r = 0.440* to r = 0.530**) and the metabolic hormone islet amyloid polypeptide (r = 0.478*). We found no correlation with psychological well-being. These results suggest that the O-BN diet reduces neuroinflammation in T2D patients and may thus be an important preventive measure for managing T2D and reducing the risk of neurodegenerative disorders.
Collapse
Affiliation(s)
- Dovilė Pocevičiūtė
- Cognitive Disorder Research Unit, Department of Clinical Sciences Malmö, Lund University, 214 28 Malmö, Sweden;
| | - Malin Wennström
- Cognitive Disorder Research Unit, Department of Clinical Sciences Malmö, Lund University, 214 28 Malmö, Sweden;
| | - Bodil Ohlsson
- Department of Internal Medicine, Lund University, Skåne University Hospital, 214 28 Malmö, Sweden;
| |
Collapse
|
28
|
Phillips WT, Schwartz JG. Nasal turbinate lymphatic obstruction: a proposed new paradigm in the etiology of essential hypertension. Front Med (Lausanne) 2024; 11:1380632. [PMID: 39219790 PMCID: PMC11362006 DOI: 10.3389/fmed.2024.1380632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Hypertension affects an estimated 1.3 billion people worldwide and is considered the number one contributor to mortality via stroke, heart failure, renal failure, and dementia. Although the physiologic mechanisms leading to the development of essential hypertension are poorly understood, the regulation of cerebral perfusion has been proposed as a primary cause. This article proposes a novel etiology for essential hypertension. Our hypothesis developed from a review of nuclear medicine scans, where the authors observed a significantly abnormal increase in nasal turbinate vasodilation in hypertensive patients using quantitative region of interest analysis. The authors propose that nasal turbinate vasodilation and resultant blood pooling obstruct the flow of cerebrospinal fluid passing through nasal turbinate lymphatics, thereby increasing intracranial pressure. The authors discuss the glymphatic/lymphatic clearance system which is impaired with age, and at which time hypertension also develops. The increased intracranial pressure leads to compensatory hypertension via Cushing's mechanism, i.e., the selfish brain hypothesis. The nasal turbinate vasodilation, due to increased parasympathetic activity, occurs simultaneously along with the well-established increased sympathetic activity of the cardiovascular system. The increased parasympathetic activity is likely due to an autonomic imbalance secondary to the increase in worldwide consumption of processed food. This hypothesis explains the rapid worldwide rise in essential hypertension in the last 50 years and offers a novel mechanism and a new paradigm for the etiology of essential hypertension. This new paradigm offers compelling evidence for the modulation of parasympathetic nervous system activity as a novel treatment strategy, specifically targeting nasal turbinate regulation, to treat diseases such as hypertension, idiopathic intracranial hypertension, and degenerative brain diseases. The proposed mechanism of essential hypertension presented in this paper is a working hypothesis and confirmatory studies will be needed.
Collapse
|
29
|
Wei YC, Kung YC, Lin C, Yeh CH, Chen PY, Huang WY, Shyu YC, Lin CP, Chen CK. Differential neuropsychiatric associations of plasma biomarkers in older adults with major depression and subjective cognitive decline. Transl Psychiatry 2024; 14:333. [PMID: 39152102 PMCID: PMC11329686 DOI: 10.1038/s41398-024-03049-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 06/23/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024] Open
Abstract
Older adults with major depressive disorder (MDD) or early cognitive decline during the subjective cognitive decline (SCD) stage may exhibit neuropsychiatric symptoms such as anxiety, depression, and subtle cognitive impairment. The clinicopathological features and biological mechanisms of MDD differ from those of SCD among older adults; these conditions thus require different treatment strategies. This study enrolled 82 participants above 50 years old with normal cognitive levels from the communities to examine biomarker-behavior correlations between MDD (n = 23) and SCD (n = 23) relative to a normal control (NC) group (n = 36). Multidomain assessments were performed for all participants, including immunomagnetic reduction tests to detect plasma beta-amyloid (Aβ), total tau (Tau), phosphorylated tau-181 (p-Tau181), neurofilament light chain, and glial fibrillary acidic protein (GFAP). This study observed that depressive symptoms in MDD were associated with amyloid pathology (plasma Aβ40 vs. HADS-D: R = 0.45, p = 0.031; Aβ42/Aβ40 vs. HADS-D: R = -0.47, p = 0.024), which was not observed in the NC (group difference p < 0.05). Moreover, cognitive decline in MDD was distinguished by a mixed neurodegenerative process involving amyloid (plasma Aβ42 vs. facial memory test: R = 0.48, p = 0.025), tau (Tau/Aβ42 vs. digit symbol substitution test (DSST): R = -0.53, p = 0.01), and astrocytic injury (plasma GFAP vs. Montreal cognitive assessment score: R = -0.44, p = 0.038; plasma GFAP vs. DSST: R = -0.52, p = 0.014), findings that did not apply to the NC (group difference p < 0.05). Moreover, this study revealed different biomarker-behavior correlations between individuals with SCD and the NC. Compared with the NC, cognitive decline in the SCD group might be unrelated to amyloid pathology and instead might be early manifestations of tau pathology. This study underscores the difference in clinicopathological features between MDD and SCD among older adults, which differ from those of the NC. These findings enhance our understanding of the mechanisms underlying MDD and SCD in older individuals.
Collapse
Affiliation(s)
- Yi-Chia Wei
- Department of Neurology, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yi-Chia Kung
- Department of Radiology, Tri-Service General Hospital, Taipei, 114, Taiwan
| | - Chemin Lin
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Chun-Hung Yeh
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Pin-Yuan Chen
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Wen-Yi Huang
- Department of Neurology, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yu-Chiau Shyu
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, 333, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.
- Department of Education and Research, Taipei City Hospital, Taipei, 103, Taiwan.
| | - Chih-Ken Chen
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Psychiatry, Chang Gung Memorial Hospital, Keelung, 204, Taiwan.
| |
Collapse
|
30
|
Liu X, Barisano G, Shao X, Jann K, Ringman JM, Lu H, Arfanakis K, Caprihan A, DeCarli C, Gold BT, Maillard P, Satizabal CL, Fadaee E, Habes M, Stables L, Singh H, Fischl B, van der Kouwe A, Schwab K, Helmer KG, Greenberg SM, Wang DJ. Cross-Vendor Test-Retest Validation of Diffusion Tensor Image Analysis along the Perivascular Space (DTI-ALPS) for Evaluating Glymphatic System Function. Aging Dis 2024; 15:1885-1898. [PMID: 37307817 PMCID: PMC11272201 DOI: 10.14336/ad.2023.0321-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/21/2023] [Indexed: 06/14/2023] Open
Abstract
The diffusion tensor image analysis along the perivascular space (DTI-ALPS) method was proposed to evaluate glymphatic system (GS) function. However, few studies have validated its reliability and reproducibility. Fifty participants' DTI data from the MarkVCID consortium were included in this study. Two pipelines by using DSI studio and FSL software were developed for data processing and ALPS index calculation. The ALPS index was obtained by the average of bilateral ALPS index and was used for testing the cross-vendor, inter-rater and test-retest reliability by using R studio software. The ALPS index demonstrated favorable inter-scanner reproducibility (ICC=0.77 to 0.95, P< 0.001), inter-rater reliability (ICC=0.96 to 1, P< 0.001) and test-retest repeatability (ICC=0.89 to 0.95, P< 0.001), offering a potential biomarker for in vivo evaluation of GS function.
Collapse
Affiliation(s)
- Xiaodan Liu
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.
| | | | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | - Kay Jann
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | - John M Ringman
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA.
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University, Chicago, IL, USA.
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA.
| | | | - Charles DeCarli
- Department of Neurology, University of California, Davis, Davis, CA, USA.
| | - Brian T Gold
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
| | - Pauline Maillard
- Department of Neurology, University of California, Davis, Davis, CA, USA.
| | - Claudia L Satizabal
- Population Health Sciences and Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Elyas Fadaee
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| | - Lara Stables
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Herpreet Singh
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Bruce Fischl
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Computer Science and AI Lab, Cambridge, Massachusetts, USA.
| | - Andre van der Kouwe
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Kristin Schwab
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Karl G Helmer
- Department of Radiology, Harvard Medical School, Boston, MA, USA.
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA.
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
| | - Danny J.J Wang
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| | | |
Collapse
|
31
|
Walter AE, Savalia K, Yoon J, Morrison J, Schneider ALC, Diaz-Arrastia R, Sandsmark DK. Change in Enlarged Perivascular Spaces over Time and Associations with Outcomes After Traumatic Brain Injury. Neurotrauma Rep 2024; 5:738-748. [PMID: 39144451 PMCID: PMC11319858 DOI: 10.1089/neur.2024.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024] Open
Abstract
Enlarged perivascular spaces (EPVs) can be seen on magnetic resonance imaging (MRI) scans in various neurological diseases, including traumatic brain injury (TBI). EPVs have been associated with cognitive dysfunction and sleep disturbances; however, their clinical significance remains unclear. The goal of this study was to identify MRI burden of EPVs over time following TBI and to explore their relationship with postinjury outcomes. Individuals with TBI underwent postinjury data collection at Day 1 (blood), 2 weeks (blood, MRI, outcomes), and 6 months (blood, MRI, outcomes). EPV burden was assessed using T1 and FLAIR sequences on representative slices in the centrum semiovale, basal ganglia, and midbrain. Serum blood was assayed to measure concentrations of neurofilament light (NfL) and glial fibrillary acidic protein (GFAP). Thirty-two participants with TBI were included (mean age 36.8 years, 78% male, 50% White). Total EPVs count did not significantly change from 2 weeks (23.5 [95% confidence interval or CI = 22.0-32.0]) to 6 months (26.0 [95% CI = 22.0-30.0], p = 0.16). For self-reported measures of sleep, there were no significant associations between EPVs count and Insomnia Severity Index (2 weeks: β = -0.004; 95% CI = -0.094, 0.086; 6 months: β = 0.002; 95% CI = -0.122, 0.125) or the subset of sleep questions on the Rivermead Post-Concussion Symptoms Questionnaire (2 weeks: β = -0.005; 95% CI = -0.049, 0.039; 6 months: β = -0.019; 95% CI = -0.079, 0.042). Functional outcome, determined by 6 months incomplete recovery (Glasgow Outcome Scale-Extended [GOS-E < 8]) versus complete recovery (GOS-E = 8), was significantly associated with a higher number of EPVs at 2 weeks (odds ratio = 0.94, 95% CI = 0.88-0.99). Spearman correlations showed no significant relationship between EPVs count and GFAP or NfL. This study used commonly acquired MRI sequences to quantify EPVs and investigated their utility as a potential imaging biomarker in TBI. Given the minimal change in EPVs over time, this period may not be long enough for potential recovery or may indicate that EPVs are structural findings that do not significantly change over time.
Collapse
Affiliation(s)
- Alexa E. Walter
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Krupa Savalia
- Departments of Neurology and Neurological Surgery, University of California Davis, Davis, California, USA
| | - Jason Yoon
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Justin Morrison
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrea L. C. Schneider
- Departments of Neurology and Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Danielle K. Sandsmark
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
32
|
Conti F, McCue JJ, DiTuro P, Galpin AJ, Wood TR. Mitigating Traumatic Brain Injury: A Narrative Review of Supplementation and Dietary Protocols. Nutrients 2024; 16:2430. [PMID: 39125311 PMCID: PMC11314487 DOI: 10.3390/nu16152430] [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: 07/03/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Traumatic brain injuries (TBIs) constitute a significant public health issue and a major source of disability and death in the United States and worldwide. TBIs are strongly associated with high morbidity and mortality rates, resulting in a host of negative health outcomes and long-term complications and placing a heavy financial burden on healthcare systems. One promising avenue for the prevention and treatment of brain injuries is the design of TBI-specific supplementation and dietary protocols centred around nutraceuticals and biochemical compounds whose mechanisms of action have been shown to interfere with, and potentially alleviate, some of the neurophysiological processes triggered by TBI. For example, evidence suggests that creatine monohydrate and omega-3 fatty acids (DHA and EPA) help decrease inflammation, reduce neural damage and maintain adequate energy supply to the brain following injury. Similarly, melatonin supplementation may improve some of the sleep disturbances often experienced post-TBI. The scope of this narrative review is to summarise the available literature on the neuroprotective effects of selected nutrients in the context of TBI-related outcomes and provide an evidence-based overview of supplementation and dietary protocols that may be considered in individuals affected by-or at high risk for-concussion and more severe head traumas. Prophylactic and/or therapeutic compounds under investigation include creatine monohydrate, omega-3 fatty acids, BCAAs, riboflavin, choline, magnesium, berry anthocyanins, Boswellia serrata, enzogenol, N-Acetylcysteine and melatonin. Results from this analysis are also placed in the context of assessing and addressing important health-related and physiological parameters in the peri-impact period such as premorbid nutrient and metabolic health status, blood glucose regulation and thermoregulation following injury, caffeine consumption and sleep behaviours. As clinical evidence in this research field is rapidly emerging, a comprehensive approach including appropriate nutritional interventions has the potential to mitigate some of the physical, neurological, and emotional damage inflicted by TBIs, promote timely and effective recovery, and inform policymakers in the development of prevention strategies.
Collapse
Affiliation(s)
- Federica Conti
- School of Physics, University of Sydney, Sydney, NSW 2050, Australia;
| | - Jackson J. McCue
- School of Medicine, University of Washington, Seattle, WA 98195, USA;
| | - Paul DiTuro
- Department of Exercise Science, University of South Carolina, Columbia, SC 29208, USA
| | - Andrew J. Galpin
- Center for Sport Performance, California State University, Fullerton, CA 92831, USA;
| | - Thomas R. Wood
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Institute for Human and Machine Cognition, Pensacola, FL 32502, USA
| |
Collapse
|
33
|
Qvarlander S, Sundström N, Malm J, Eklund A. CSF formation rate-a potential glymphatic flow parameter in hydrocephalus? Fluids Barriers CNS 2024; 21:55. [PMID: 38987813 PMCID: PMC11234690 DOI: 10.1186/s12987-024-00560-6] [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: 01/30/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Studies indicate that brain clearance via the glymphatic system is impaired in idiopathic normal pressure hydrocephalus (INPH). This has been suggested to result from reduced cerebrospinal fluid (CSF) turnover, which could be caused by a reduced CSF formation rate. The aim of this study was to determine the formation rate of CSF in a cohort of patients investigated for INPH and compare this to a historical control cohort. METHODS CSF formation rate was estimated in 135 (75 ± 6 years old, 64/71 men/women) patients undergoing investigation for INPH. A semiautomatic CSF infusion investigation (via lumbar puncture) was performed. CSF formation rate was assessed by downregulating and steadily maintaining CSF pressure at a zero level. During the last 10 min, the required outflow to maintain zero pressure, i.e., CSF formation rate, was continuously measured. The values were compared to those of a historical reference cohort from a study by Ekstedt in 1978. RESULTS Mean CSF formation rate was 0.45 ± 0.15 ml/min (N = 135), equivalent to 27 ± 9 ml/hour. There was no difference in the mean (p = 0.362) or variance (p = 0.498) of CSF formation rate between the subjects that were diagnosed as INPH (N = 86) and those who were not (N = 43). The CSF formation rate in INPH was statistically higher than in the reference cohort (0.46 ± 0.15 vs. 0.40 ± 0.08 ml/min, p = 0.005), but the small difference was probably not physiologically relevant. There was no correlation between CSF formation rate and baseline CSF pressure (r = 0.136, p = 0.115, N = 135) or age (-0.02, p = 0.803, N = 135). CONCLUSIONS The average CSF formation rate in INPH was not decreased compared to the healthy reference cohort, which does not support reduced CSF turnover. This emphasizes the need to further investigate the source and routes of the flow in the glymphatic system and the cause of the suggested impaired glymphatic clearance in INPH.
Collapse
Affiliation(s)
- Sara Qvarlander
- Department of Diagnostics and Intervention, Biomedical Engineering and Radiation Physics, Umeå University, Umeå, Sweden.
| | - Nina Sundström
- Department of Diagnostics and Intervention, Biomedical Engineering and Radiation Physics, Umeå University, Umeå, Sweden
| | - Jan Malm
- Department of Clinical Science, Neurosciences, Umeå University, Umeå, Sweden
| | - Anders Eklund
- Department of Diagnostics and Intervention, Biomedical Engineering and Radiation Physics, Umeå University, Umeå, Sweden
| |
Collapse
|
34
|
Braun M, Sevao M, Keil SA, Gino E, Wang MX, Lee J, Haveliwala MA, Klein E, Agarwal S, Pedersen T, Rhodes CH, Jansson D, Cook D, Peskind E, Perl DP, Piantino J, Schindler AG, Iliff JJ. Macroscopic changes in aquaporin-4 underlie blast traumatic brain injury-related impairment in glymphatic function. Brain 2024; 147:2214-2229. [PMID: 38802114 PMCID: PMC11146423 DOI: 10.1093/brain/awae065] [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: 09/22/2023] [Revised: 01/10/2024] [Accepted: 02/06/2024] [Indexed: 05/29/2024] Open
Abstract
Mild traumatic brain injury (mTBI) has emerged as a potential risk factor for the development of neurodegenerative conditions such as Alzheimer's disease and chronic traumatic encephalopathy. Blast mTBI, caused by exposure to a pressure wave from an explosion, is predominantly experienced by military personnel and has increased in prevalence and severity in recent decades. Yet the underlying pathology of blast mTBI is largely unknown. We examined the expression and localization of AQP4 in human post-mortem frontal cortex and observed distinct laminar differences in AQP4 expression following blast exposure. We also observed similar laminar changes in AQP4 expression and localization and delayed impairment of glymphatic function that emerged 28 days following blast injury in a mouse model of repetitive blast mTBI. In a cohort of veterans with blast mTBI, we observed that blast exposure was associated with an increased burden of frontal cortical MRI-visible perivascular spaces, a putative neuroimaging marker of glymphatic perivascular dysfunction. These findings suggest that changes in AQP4 and delayed glymphatic impairment following blast injury may render the post-traumatic brain vulnerable to post-concussive symptoms and chronic neurodegeneration.
Collapse
Affiliation(s)
- Molly Braun
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Mathew Sevao
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Samantha A Keil
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elizabeth Gino
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Marie X Wang
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Janet Lee
- VISN 20 Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Mariya A Haveliwala
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Emily Klein
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Sanjana Agarwal
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Taylor Pedersen
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - C Harker Rhodes
- Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, MD 20817, USA
- Department of Pathology, F Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- DoD/USU Brain Tissue Repository, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Deidre Jansson
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - David Cook
- VISN 20 Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elaine Peskind
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Daniel P Perl
- Department of Pathology, F Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- DoD/USU Brain Tissue Repository, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Juan Piantino
- Division of Child Neurology, Department of Pediatrics, Doernbecher Children’s Hospital, Oregon Health & Science University, Portland, OR 97239, USA
| | - Abigail G Schindler
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
- VISN 20 Geriatric Research, Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Medicine, Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jeffrey J Iliff
- VISN 20 Northwest Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
35
|
Xiong Y, Yu Q, Zhi H, Peng H, Xie M, Li R, Li K, Ma Y, Sun P. Advances in the study of the glymphatic system and aging. CNS Neurosci Ther 2024; 30:e14803. [PMID: 38887168 PMCID: PMC11183173 DOI: 10.1111/cns.14803] [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: 10/27/2023] [Revised: 04/26/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
The glymphatic system is cerebrospinal fluid-brain tissue fluid exchange flow mediated by aquaporin-4 (AQP4) on the end feet of astrocytes for a system, which is capable of rapidly removing brain metabolites and thus maintaining brain homeostasis, and is known as the central immune system. Dysfunction of the glymphatic system causes accumulation of misfolded and highly phosphorylated proteins (amyloid-β and Tau proteins), which destabilizes the proteins, and the body's neuroinflammatory factors are altered causing aging of the immune system and leading to neurodegenerative diseases. Damage to the glymphatic system and aging share common manifestations, as well as unstudied biological mechanisms that are also linked, such as mitochondria, oxidative stress, chronic inflammation, and sleep. In this paper, we first summarize the structure, function, and research methods of the glymphatic system and the relationship between the glymphatic system and the peripheral immune system, and second, sort out and summarize the factors of the glymphatic system in removing metabolites and resolving aging-related diseases and factors affecting aging, to explore its related biological mechanisms, and moreover, to provide a new way of thinking for treating or intervening aging-related diseases.
Collapse
Affiliation(s)
- Ying Xiong
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Qingying Yu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical SciencesGuangzhou University of Chinese MedicineGuangzhouChina
| | - Haimei Zhi
- Qilu Hospital of Shandong UniversityJinanChina
| | - Huiyuan Peng
- Department of RehabilitationZhongshan Hospital of Traditional Chinese MedicineZhongshanChina
| | - Mingjun Xie
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Renjun Li
- Department of PsychiatryJinan Mental Health CenterJinanChina
| | - Kejian Li
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| | - Yuexiang Ma
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Peng Sun
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
36
|
Thipani Madhu M, Balaji O, Kandi V, Ca J, Harikrishna GV, Metta N, Mudamanchu VK, Sanjay BG, Bhupathiraju P. Role of the Glymphatic System in Alzheimer's Disease and Treatment Approaches: A Narrative Review. Cureus 2024; 16:e63448. [PMID: 39077280 PMCID: PMC11285013 DOI: 10.7759/cureus.63448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2024] [Indexed: 07/31/2024] Open
Abstract
Currently, there is unavailability of disease-modifying medication for Alzheimer's disease (AD), a debilitating neurological disorder. The pathogenesis of AD appears to be complex and could be influenced by the glymphatic system present in the central nervous system (CNS). Amyloid-beta (Aβ) and other metabolic wastes are eliminated from the brain interstitium by the glymphatic system, which encompasses perivascular channels and astroglial cells. Dysfunction of the glymphatic system, which could occur due to decreased aquaporin 4 (AQP4) expression, aging-related alterations in the human brain, and sleep disruptions, may contribute to the pathogenesis of AD and also accelerate the development of AD by causing a buildup of harmful proteins like Aβ. Promising approaches have been examined for reducing AD pathology, including non-pharmacological therapies that target glymphatic function, like exercise and sleep regulation. In addition, preclinical research has also demonstrated the therapeutic potential of pharmaceutical approaches targeted at augmenting AQP4-mediated glymphatic flow. To identify the precise processes driving glymphatic dysfunction in AD and to find new treatment targets, more research is required. Innovative diagnostic and treatment approaches for AD could be made possible by techniques such as dynamic contrast-enhanced MRI, which promises to evaluate glymphatic function in neurodegenerative diseases. Treatment options for AD and other neurodegenerative diseases may be improved by comprehending and utilizing the glymphatic system's function in preserving brain homeostasis and targeting the mechanisms involved in glymphatic functioning. This review intends to enhance the understanding of the complex link between AD and the glymphatic system and focuses on the function of AQP4 channels in promoting waste clearance and fluid exchange.
Collapse
Affiliation(s)
- Mansi Thipani Madhu
- Internal Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Ojas Balaji
- Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Venkataramana Kandi
- Clinical Microbiology, Prathima Institute of Medical Sciences, Karimnagar, IND
| | - Jayashankar Ca
- Internal Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | | | - Nirosha Metta
- Neurology, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | | | - Bhangdiya G Sanjay
- Internal Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Praful Bhupathiraju
- Internal Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| |
Collapse
|
37
|
Singh AK, Asif S, Pandey DK, Chaudhary A, Kapoor V, Verma PK. Biomarkers in Acute Traumatic Brain Injury: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e63020. [PMID: 39050316 PMCID: PMC11268976 DOI: 10.7759/cureus.63020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Traumatic brain injury (TBI) stands as a significant contributor to traumatic death and disability worldwide. In recent years, researchers have identified biomarkers to gauge useful outcomes in TBI patients. However, the enigma of timely sample collection to measure the biomarkers remains a controversial point in the case of TBI, unlike other degenerative diseases like Alzheimer's disease and Parkinson's disease, where we can collect the sample at any point in time. The purpose of this study is to evaluate the sensitivity of biomarkers in TBI concerning time of injury by analyzing recent available data on biomarkers in the medical literature. A total of 2,256 studies were initially retrieved from the search engine. After an initial screening, only 1,750 unique articles remained. After excluding review articles, animal studies, meta-analysis, and studies with children (screened by title and abstract), 30 kinds of literature were found relevant to search the required variables. Further 16 studies were excluded due to the nonavailability of complete variables or data. Finally, 14 studies remained and were included in the analysis. This study has analyzed the four most commonly described biomarkers for TBI in the literature: glial fibrillary acidic protein (GFAP), S100 calcium-binding protein B, ubiquitin carboxy-terminal hydrolase L1, and Tau. According to this statistical analysis, all biomarkers included in the study have shown their serum levels after trauma. So, all these biomarkers can be used for further study in the outcome prediction and diagnosis of TBI patients. The meta-analysis suggests that the best biomarker for TBI is Tau in cases where sample collection is done within 24 hours, while GFAP is the best biomarker to be studied for TBI if sample collection is done 24 hours after trauma.
Collapse
Affiliation(s)
- Adarsh Kumar Singh
- Department of Biotechnology, Centre of BioMedical Research (CBMR) Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Shafaque Asif
- Department of Molecular Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Deepika Kumari Pandey
- Department of Neurosurgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Akash Chaudhary
- Department of Neurosurgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Vishwas Kapoor
- Department of Biostatistics and Health Informatics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| | - Pawan Kumar Verma
- Department of Neurosurgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, IND
| |
Collapse
|
38
|
Khalil M, Teunissen CE, Lehmann S, Otto M, Piehl F, Ziemssen T, Bittner S, Sormani MP, Gattringer T, Abu-Rumeileh S, Thebault S, Abdelhak A, Green A, Benkert P, Kappos L, Comabella M, Tumani H, Freedman MS, Petzold A, Blennow K, Zetterberg H, Leppert D, Kuhle J. Neurofilaments as biomarkers in neurological disorders - towards clinical application. Nat Rev Neurol 2024; 20:269-287. [PMID: 38609644 DOI: 10.1038/s41582-024-00955-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2024] [Indexed: 04/14/2024]
Abstract
Neurofilament proteins have been validated as specific body fluid biomarkers of neuro-axonal injury. The advent of highly sensitive analytical platforms that enable reliable quantification of neurofilaments in blood samples and simplify longitudinal follow-up has paved the way for the development of neurofilaments as a biomarker in clinical practice. Potential applications include assessment of disease activity, monitoring of treatment responses, and determining prognosis in many acute and chronic neurological disorders as well as their use as an outcome measure in trials of novel therapies. Progress has now moved the measurement of neurofilaments to the doorstep of routine clinical practice for the evaluation of individuals. In this Review, we first outline current knowledge on the structure and function of neurofilaments. We then discuss analytical and statistical approaches and challenges in determining neurofilament levels in different clinical contexts and assess the implications of neurofilament light chain (NfL) levels in normal ageing and the confounding factors that need to be considered when interpreting NfL measures. In addition, we summarize the current value and potential clinical applications of neurofilaments as a biomarker of neuro-axonal damage in a range of neurological disorders, including multiple sclerosis, Alzheimer disease, frontotemporal dementia, amyotrophic lateral sclerosis, stroke and cerebrovascular disease, traumatic brain injury, and Parkinson disease. We also consider the steps needed to complete the translation of neurofilaments from the laboratory to the management of neurological diseases in clinical practice.
Collapse
Affiliation(s)
- Michael Khalil
- Department of Neurology, Medical University of Graz, Graz, Austria.
| | - Charlotte E Teunissen
- Neurochemistry Laboratory Department of Laboratory Medicine, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Sylvain Lehmann
- LBPC-PPC, Université de Montpellier, INM INSERM, IRMB CHU de Montpellier, Montpellier, France
| | - Markus Otto
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tjalf Ziemssen
- Center of Clinical Neuroscience, Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN), and Immunotherapy (FZI), Rhine-Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Maria Pia Sormani
- Department of Health Sciences, University of Genova, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Thomas Gattringer
- Department of Neurology, Medical University of Graz, Graz, Austria
- Division of Neuroradiology, Vascular and Interventional Radiology, Department of Radiology, Medical University of Graz, Graz, Austria
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Simon Thebault
- Multiple Sclerosis Division, Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdelhak
- Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Ari Green
- Weill Institute for Neurosciences, Department of Neurology, University of California at San Francisco, San Francisco, CA, USA
| | - Pascal Benkert
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Ludwig Kappos
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Manuel Comabella
- Neurology Department, Multiple Sclerosis Centre of Catalonia, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Hayrettin Tumani
- Department of Neurology, CSF Laboratory, Ulm University Hospital, Ulm, Germany
| | - Mark S Freedman
- Department of Medicine, University of Ottawa, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Axel Petzold
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Neurology, MS Centre and Neuro-ophthalmology Expertise Centre Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
- Moorfields Eye Hospital, The National Hospital for Neurology and Neurosurgery and the Queen Square Institute of Neurology, UCL, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P. R. China
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - David Leppert
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Multiple Sclerosis Centre and Research Center for Clinical Neuroimmunology and Neuroscience (RC2NB), Departments of Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland.
- Department of Neurology, University Hospital and University of Basel, Basel, Switzerland.
| |
Collapse
|
39
|
Werner JK, Albrecht J, Capaldi VF, Jain S, Sun X, Mukherjee P, Williams SG, Collen J, Diaz-Arrastia R, Manley GT, Krystal AD, Wickwire E. Association of Biomarkers of Neuronal Injury and Inflammation With Insomnia Trajectories After Traumatic Brain Injury: A TRACK-TBI Study. Neurology 2024; 102:e209269. [PMID: 38547447 PMCID: PMC11210587 DOI: 10.1212/wnl.0000000000209269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/05/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Insomnia affects about one-third of patients with traumatic brain injury and is associated with worsened outcomes after injury. We hypothesized that higher levels of plasma neuroinflammation biomarkers at the time of TBI would be associated with worse 12-month insomnia trajectories. METHODS Participants were prospectively enrolled from 18 level-1 trauma centers participating in the Transforming Research and Clinical Knowledge in Traumatic Brain Injury study from February 26, 2014, to August 8, 2018. Plasma glial fibrillary acidic protein (GFAP), high-sensitivity C-reactive protein (hsCRP), S100b, neuron-specific enolase (NSE), and ubiquitin carboxyl-terminal hydrolase-L1 (UCH-L1) were collected on days 1 (D1) and 14 (D14) after TBI. The insomnia severity index was collected at 2 weeks, 3, 6, and 12 months postinjury. Participants were classified into insomnia trajectory classes based on a latent class model. We assessed the association of biomarkers with insomnia trajectories, controlling for medical and psychological comorbidities and demographics. RESULTS Two thousand twenty-two individuals with TBI were studied. Elevations in D1 hsCRP were associated with persistent insomnia (severe, odds ratio [OR] = 1.33 [1.11, 1.59], p = 0.002; mild, OR = 1.10 [1.02, 1.19], p = 0.011). Similarly, D14 hsCRP elevations were associated with persistent insomnia (severe, OR = 1.27 [1.02, 1.59], p = 0.03). Of interest, D1 GFAP was lower in persistent severe insomnia (median [Q1, Q3]: 154 [19, 445] pg/mL) compared with resolving mild (491 [154, 1,423], p < 0.001) and persistent mild (344 [79, 1,287], p < 0.001). D14 GFAP was similarly lower in persistent (11.8 [6.4, 19.4], p = 0.001) and resolving (13.9 [10.3, 20.7], p = 0.011) severe insomnia compared with resolving mild (20.6 [12.4, 39.6]. Accordingly, increases in D1 GFAP were associated with reduced likelihood of having persistent severe (OR = 0.76 [95% CI 0.63-0.92], p = 0.004) and persistent mild (OR = 0.88 [0.81, 0.96], p = 0.003) compared with mild resolving insomnia. No differences were found with other biomarkers. DISCUSSION Elevated plasma hsCRP and, surprisingly, lower GFAP were associated with adverse insomnia trajectories after TBI. Results support future prospective studies to examine their utility in guiding insomnia care after TBI. Further work is needed to explore potential mechanistic connections between GFAP levels and the adverse insomnia trajectories.
Collapse
Affiliation(s)
- J Kent Werner
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Jennifer Albrecht
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Vincent F Capaldi
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Sonia Jain
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Xiaoying Sun
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Pratik Mukherjee
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Scott G Williams
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Jacob Collen
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Ramon Diaz-Arrastia
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Geoffrey T Manley
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Andrew D Krystal
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| | - Emerson Wickwire
- From the Department of Neurology (J.K.W.); Center for Neuroscience and Regenerative Medicine (J.K.W.), Uniformed Services University; Sleep Disorders Center (J.K.W., J.C.), Department of Medicine, Walter Reed National Military Medical Center, Bethesda; Department of Epidemiology and Public Health (J.A.), University of Maryland School of Medicine, Baltimore; Center for Military Psychiatry and Neuroscience (V.F.C., S.G.W.), Walter Reed Army Institute of Research, Silver Spring; Department of Medicine (V.F.C., J.C.), Uniformed Services University of the Health Sciences, Bethesda, MD; Biostatistics Research Center (V.F.C., S.G.W.), Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego; Department of Radiology (S.J., X.S.), School of Medicine, University of California San Francisco; Department of Medicine (P.M.), Alexander T. Augusta Military Medical Center, Fort Belvoir, VA; Department of Psychiatry (S.G.W.), Uniformed Services University of the Health Sciences, Bethesda, MD; Department of Neurology (R.D.-A.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Brain and Spinal Injury Center (G.T.M.); Department of Neurosurgery (G.T.M.); Department of Psychiatry and Behavioral Sciences (A.D.K.); Weill Institute for Neurosciences (A.D.K.), University of California, San Francisco; Sleep Disorders Center (E.W.), Division of Pulmonary and Critical Care Medicine, Department of Medicine; and Department of Psychiatry (E.W.), University of Maryland School of Medicine, Baltimore
| |
Collapse
|
40
|
Zapata-Acevedo JF, Mantilla-Galindo A, Vargas-Sánchez K, González-Reyes RE. Blood-brain barrier biomarkers. Adv Clin Chem 2024; 121:1-88. [PMID: 38797540 DOI: 10.1016/bs.acc.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The blood-brain barrier (BBB) is a dynamic interface that regulates the exchange of molecules and cells between the brain parenchyma and the peripheral blood. The BBB is mainly composed of endothelial cells, astrocytes and pericytes. The integrity of this structure is essential for maintaining brain and spinal cord homeostasis and protection from injury or disease. However, in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, and multiple sclerosis, the BBB can become compromised thus allowing passage of molecules and cells in and out of the central nervous system parenchyma. These agents, however, can serve as biomarkers of BBB permeability and neuronal damage, and provide valuable information for diagnosis, prognosis and treatment. Herein, we provide an overview of the BBB and changes due to aging, and summarize current knowledge on biomarkers of BBB disruption and neurodegeneration, including permeability, cellular, molecular and imaging biomarkers. We also discuss the challenges and opportunities for developing a biomarker toolkit that can reliably assess the BBB in physiologic and pathophysiologic states.
Collapse
Affiliation(s)
- Juan F Zapata-Acevedo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Alejandra Mantilla-Galindo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
41
|
Kaur J, Boyd ED, Ding G, Zhang L, Luo H, Li Q, Li L, Wei M, Landschoot-Ward J, Chopp M, Zhang Z, Jiang Q. The Association between Glymphatic System and Perivascular Macrophages in Brain Waste Clearance. Diagnostics (Basel) 2024; 14:731. [PMID: 38611644 PMCID: PMC11011895 DOI: 10.3390/diagnostics14070731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
The glymphatic system suggests the convective bulk flow of cerebrospinal fluid (CSF) through perivascular spaces and the interstitial spaces of the brain parenchyma for the rapid removal of toxic waste solutes from the brain. However, the presence of convective bulk flow within the brain interstitial spaces is still under debate. We first addressed this argument to determine the involvement of the glymphatic system in brain waste clearance utilizing contrast-enhanced 3D T1-weighted imaging (T1WI), diffusion tensor imaging (DTI), and confocal microscopy imaging. Furthermore, perivascular macrophages (PVMs), which are immune cells located within perivascular spaces, have not been thoroughly explored for their association with the glymphatic system. Therefore, we investigated tracer uptake by PVMs in the perivascular spaces of both the arteries/arterioles and veins/venules and the potential association of PVMs in assisting the glymphatic system for interstitial waste clearance. Our findings demonstrated that both convective bulk flow and diffusion are responsible for the clearance of interstitial waste solutes from the brain parenchyma. Furthermore, our results suggested that PVMs may play an important function in glymphatic system-mediated interstitial waste clearance. The glymphatic system and PVMs could be targeted to enhance interstitial waste clearance in patients with waste-associated neurological conditions and aging.
Collapse
Affiliation(s)
- Jasleen Kaur
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Edward D. Boyd
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Hao Luo
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Lian Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Min Wei
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Julie Landschoot-Ward
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
42
|
Eide PK, Ringstad G. Functional analysis of the human perivascular subarachnoid space. Nat Commun 2024; 15:2001. [PMID: 38443374 PMCID: PMC10914778 DOI: 10.1038/s41467-024-46329-1] [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: 04/26/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
The human subarachnoid space harbors the cerebrospinal fluid, which flows within a landscape of blood vessels and trabeculae. Functional implications of subarachnoid space anatomy remain far less understood. This study of 75 patients utilizes a cerebrospinal fluid tracer (gadobutrol) and consecutive magnetic resonance imaging to investigate features of early (i.e. within 2-3 h after injection) tracer propagation within the subarachnoid space. There is a time-dependent perivascular pattern of enrichment antegrade along the major cerebral artery trunks; the anterior-, middle-, and posterior cerebral arteries. The correlation between time of first enrichment around arteries and early enrichment in nearby cerebral cortex is significant. These observations suggest the existence of a compartmentalized subarachnoid space, where perivascular ensheathment of arteries facilitates antegrade tracer passage towards brain tissue. Periarterial transport is impaired in subjects with reduced intracranial pressure-volume reserve capacity and in idiopathic normal pressure hydrocephalus patients who also show increased perivascular space size.
Collapse
Affiliation(s)
- Per Kristian Eide
- Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Pb 4950 Nydalen, N-0424, Oslo, Norway.
- KG Jebsen Centre for Brain Fluid Research, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, PB 1072 Blindern, N-0316, Oslo, Norway.
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital- Rikshospitalet, Pb 4950 Nydalen, N-0424, Oslo, Norway
- Department of Geriatrics and Internal medicine, Sorlandet Hospital, 4838, Arendal, Arendal, Norway
| |
Collapse
|
43
|
Feng D, Liu T, Zhang X, Xiang T, Su W, Quan W, Jiang R. Fingolimod improves diffuse brain injury by promoting AQP4 polarization and functional recovery of the glymphatic system. CNS Neurosci Ther 2024; 30:e14669. [PMID: 38459666 PMCID: PMC10924110 DOI: 10.1111/cns.14669] [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: 10/23/2023] [Revised: 01/26/2024] [Accepted: 02/17/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Diffuse brain injury (DBI) models are characterized by intense global brain inflammation and edema, which characterize the most severe form of TBI. In a previous experiment, we found that fingolimod promoted recovery after controlled cortical impact injury (CCI) by modulating inflammation around brain lesions. However, it remains unclear whether fingolimod can also attenuate DBI because of its different injury mechanisms. Furthermore, whether fingolimod has additional underlying effects on repairing DBI is unknown. METHODS The impact acceleration model of DBI was established in adult Sprague-Dawley rats. Fingolimod (0.5 mg/kg) was administered 0.5, 24, and 48 h after injury for 3 consecutive days. Immunohistochemistry, immunofluorescence analysis, cytokine array, and western blotting were used to evaluate inflammatory cells, inflammatory factors, AQP4 polarization, apoptosis in brain cells, and the accumulation of APP after DBI in rats. To evaluate the function of the glymphatic system (GS), a fluorescent tracer was injected into the cistern. The neural function of rats with DBI was evaluated using various tests, including the modified neurological severity score (mNSS), horizontal ladder-crossing test, beam walking test, and tape sensing and removal test. Brain water content was also measured. RESULTS Fingolimod administration for 3 consecutive days could reduce the levels of inflammatory cytokines, neutrophil recruitment, microglia, and astrocyte activation in the brain following DBI. Moreover, fingolimod reduced apoptotic protein expression, brain cell apoptosis, brain edema, and APP accumulation. Additionally, fingolimod inhibited the loss of AQP4 polarization, improved lymphatic system function, and reduced damage to nervous system function. Notably, inhibiting the GS weakened the therapeutic effect of fingolimod on the neurological function of rats with DBI and increased the accumulation of APP in the brain. CONCLUSIONS In brief, these findings suggest that fingolimod alleviates whole-brain inflammation and GS system damage after DBI and that inhibiting the GS could weaken the positive effect of fingolimod on nerve function in rats with DBI. Thus, inhibiting inflammation and regulating the GS may be critical for the therapeutic effect of fingolimod on DBI.
Collapse
Affiliation(s)
- Dongyi Feng
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Tao Liu
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Xinjie Zhang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Tangtang Xiang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Wanqiang Su
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Wei Quan
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Rongcai Jiang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| |
Collapse
|
44
|
Alghanimy A, Work LM, Holmes WM. The glymphatic system and multiple sclerosis: An evolving connection. Mult Scler Relat Disord 2024; 83:105456. [PMID: 38266608 DOI: 10.1016/j.msard.2024.105456] [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: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder that affects the central nervous system, resulting in demyelination and an array of neurological manifestations. Recently, there has been significant scientific interest in the glymphatic system, which operates as a waste-clearance system for the brain. This article reviews the existing literature, and explores potential links between the glymphatic system and MS, shedding light on its evolving significance in the context of MS pathogenesis. The authors consider the pathophysiological implications of glymphatic dysfunction in MS, the impact of disrupted sleep on glymphatic function, and the bidirectional relationship between MS and sleep disturbances. By offering an understanding of the intricate interplay between the glymphatic system and MS, this review provides valuable insights which may lead to improved diagnostic techniques and more effective therapeutic interventions.
Collapse
Affiliation(s)
- Alaa Alghanimy
- School of Psychology and Neuroscience, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G61 1QH, United Kingdom; Radiological Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| | - Lorraine M Work
- School of Cardiovascular and Metabolic Health, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - William M Holmes
- School of Psychology and Neuroscience, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow G61 1QH, United Kingdom
| |
Collapse
|
45
|
Das N, Dhamija R, Sarkar S. The role of astrocytes in the glymphatic network: a narrative review. Metab Brain Dis 2024; 39:453-465. [PMID: 38008886 DOI: 10.1007/s11011-023-01327-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023]
Abstract
To date, treatment of Central Nervous System (CNS) pathology has largely focused on neuronal structure and function. Yet, revived attention towards fluid circulation within the CNS has exposed the need to further explore the role of glial cells in maintaining homeostasis within neural networks. In the past decade, discovery of the neural glymphatic network has revolutionized traditional understanding of fluid dynamics within the CNS. Advancements in neuroimaging have revealed alternative pathways of cerebrospinal fluid (CSF) generation and efflux. Here, we discuss emerging perspectives on the role of astrocytes in CSF hydrodynamics, with particular focus on the contribution of aquaporin-4 channels to the glymphatic network. Astrocytic structural features and expression patterns are detailed in relation to their function in maintaining integrity of the Blood Brain Barrier (BBB) as part of the neurovascular unit (NVU). This narrative also highlights the potential role of glial dysfunction in pathogenesis of neurodegenerative disease, hydrocephalus, intracranial hemorrhage, ischemic stroke, and traumatic brain injury. The purpose of this literature summary is to provide an update on the changing landscape of scientific theory surrounding production, flow, and absorption of cerebrospinal fluid. The overarching aim of this narrative review is to advance the conception of basic, translational, and clinical research endeavors investigating glia as therapeutic targets for neurological disease.
Collapse
Affiliation(s)
- Nikita Das
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ravi Dhamija
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, HFT-132, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
46
|
Boyd ED, Zhang L, Ding G, Li L, Lu M, Li Q, Huang R, Kaur J, Hu J, Chopp M, Zhang Z, Jiang Q. The Glymphatic Response to the Development of Type 2 Diabetes. Biomedicines 2024; 12:401. [PMID: 38398003 PMCID: PMC10886551 DOI: 10.3390/biomedicines12020401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/21/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
The glymphatic system has recently been shown to be important in neurological diseases, including diabetes. However, little is known about how the progressive onset of diabetes affects the glymphatic system. The aim of this study is to investigate the glymphatic system response to the progressive onset of diabetes in a rat model of type 2 diabetic mellitus. Male Wistar rats (n = 45) with and without diabetes were evaluated using MRI glymphatic tracer kinetics, functional tests, and brain tissue immunohistochemistry. Our data demonstrated that the contrast agent clearance impairment gradually progressed with the diabetic duration. The MRI data showed that an impairment in contrast clearance occurred prior to the cognitive deficits detected using functional tests and permitted the detection of an early DM stage compared to the immuno-histopathology and cognitive tests. Additionally, the quantitative MRI markers of brain waste clearance demonstrated region-dependent sensitivity in glymphatic impairment. The improved sensitivity of MRI markers in the olfactory bulb and the whole brain at an early DM stage may be attributed to the important role of the olfactory bulb in the parenchymal efflux pathway. MRI can provide sensitive quantitative markers of glymphatic impairment during the progression of DM and can be used as a valuable tool for the early diagnosis of DM with a potential for clinical application.
Collapse
Affiliation(s)
- Edward D. Boyd
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Lian Li
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
| | - Mei Lu
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI 48202, USA;
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
| | - Rui Huang
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
| | - Jasleen Kaur
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Jiani Hu
- Department of Radiology, Wayne State University, Detroit, MI 48202, USA;
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Neurology, Wayne State University, Detroit, MI 28202, USA
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Neurology, Wayne State University, Detroit, MI 28202, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, E&R B126, 2799 West Grand Boulevard, Detroit, MI 48202, USA; (L.Z.); (G.D.); (L.L.); (Q.L.); (J.K.); (M.C.); (Z.Z.); (Q.J.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Neurology, Wayne State University, Detroit, MI 28202, USA
| |
Collapse
|
47
|
Treble-Barna A, Petersen BA, Stec Z, Conley YP, Fink EL, Kochanek PM. Brain-Derived Neurotrophic Factor in Pediatric Acquired Brain Injury and Recovery. Biomolecules 2024; 14:191. [PMID: 38397427 PMCID: PMC10886547 DOI: 10.3390/biom14020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
We review emerging preclinical and clinical evidence regarding brain-derived neurotrophic factor (BDNF) protein, genotype, and DNA methylation (DNAm) as biomarkers of outcomes in three important etiologies of pediatric acquired brain injury (ABI), traumatic brain injury, global cerebral ischemia, and stroke. We also summarize evidence suggesting that BDNF is (1) involved in the biological embedding of the psychosocial environment, (2) responsive to rehabilitative therapies, and (3) potentially modifiable. BDNF's unique potential as a biomarker of neuroplasticity and neural repair that is reflective of and responsive to both pre- and post-injury environmental influences separates it from traditional protein biomarkers of structural brain injury with exciting potential to advance pediatric ABI management by increasing the accuracy of prognostic tools and informing clinical decision making through the monitoring of therapeutic effects.
Collapse
Affiliation(s)
- Amery Treble-Barna
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
| | - Bailey A. Petersen
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
| | - Zachary Stec
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (B.A.P.); (Z.S.)
| | - Yvette P. Conley
- Department of Health Promotion & Development, University of Pittsburgh School of Nursing, Pittsburgh, PA 15213, USA;
| | - Ericka L. Fink
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (E.L.F.); (P.M.K.)
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| |
Collapse
|
48
|
Al Masri M, Corell A, Michaëlsson I, Jakola AS, Skoglund T. The glymphatic system for neurosurgeons: a scoping review. Neurosurg Rev 2024; 47:61. [PMID: 38253938 PMCID: PMC10803566 DOI: 10.1007/s10143-024-02291-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/05/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024]
Abstract
The discovery of the glymphatic system has revolutionized our understanding of cerebrospinal fluid (CSF) circulation and interstitial waste clearance in the brain. This scoping review aims to synthesize the current literature on the glymphatic system's role in neurosurgical conditions and its potential as a therapeutic target. We conducted a comprehensive search in PubMed and Scopus databases for studies published between January 1, 2012, and October 31, 2023. Studies were selected based on their relevance to neurosurgical conditions and glymphatic function, with both animal and human studies included. Data extraction focused on the methods for quantifying glymphatic function and the main results. A total of 67 articles were included, covering conditions such as idiopathic normal pressure hydrocephalus (iNPH), idiopathic intracranial hypertension (IIH), subarachnoid hemorrhage (SAH), stroke, intracranial tumors, and traumatic brain injury (TBI). Significant glymphatic dysregulation was noted in iNPH and IIH, with evidence of impaired CSF dynamics and delayed clearance. SAH studies indicated glymphatic dysfunction with the potential therapeutic effects of nimodipine and tissue plasminogen activator. In stroke, alterations in glymphatic activity correlated with the extent of edema and neurological recovery. TBI studies highlighted the role of the glymphatic system in post-injury cognitive outcomes. Results indicate that the regulation of aquaporin-4 (AQP4) channels is a critical target for therapeutic intervention. The glymphatic system plays a critical role in the pathophysiology of various neurosurgical conditions, influencing brain edema and CSF dynamics. Targeting the regulation of AQP4 channels presents as a significant therapeutic strategy. Although promising, the translation of these findings into clinical practice requires further human studies. Future research should focus on establishing non-invasive biomarkers for glymphatic function and exploring the long-term effects of glymphatic dysfunction.
Collapse
Affiliation(s)
- Mohammad Al Masri
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alba Corell
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neurosurgery, Sahlgrenska University Hospital, Blå Stråket 5, 3 tr, SE-41345, Gothenburg, Sweden
| | - Isak Michaëlsson
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neurosurgery, Sahlgrenska University Hospital, Blå Stråket 5, 3 tr, SE-41345, Gothenburg, Sweden
| | - Asgeir S Jakola
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Neurosurgery, Sahlgrenska University Hospital, Blå Stråket 5, 3 tr, SE-41345, Gothenburg, Sweden
| | - Thomas Skoglund
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Neurosurgery, Sahlgrenska University Hospital, Blå Stråket 5, 3 tr, SE-41345, Gothenburg, Sweden.
| |
Collapse
|
49
|
Pulliam A, Gier EC, Gaul DA, Moore SG, Fernández FM, LaPlaca MC. Comparing Brain and Blood Lipidome Changes following Single and Repetitive Mild Traumatic Brain Injury in Rats. ACS Chem Neurosci 2024; 15:300-314. [PMID: 38179922 PMCID: PMC10797623 DOI: 10.1021/acschemneuro.3c00603] [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: 09/16/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
Traumatic brain injury (TBI) is a major health concern in the United States and globally, contributing to disability and long-term neurological problems. Lipid dysregulation after TBI is underexplored, and a better understanding of lipid turnover and degradation could point to novel biomarker candidates and therapeutic targets. Here, we investigated overlapping lipidome changes in the brain and blood using a data-driven discovery approach to understand lipid alterations in the brain and serum compartments acutely following mild TBI (mTBI) and the potential efflux of brain lipids to peripheral blood. The cortices and sera from male and female Sprague-Dawley rats were analyzed via ultra-high performance liquid chromatography-mass spectrometry (UHPLC-MS) in both positive and negative ion modes following single and repetitive closed head impacts. The overlapping lipids in the data sets were identified with an in-house data dictionary for investigating lipid class changes. MS-based lipid profiling revealed overall increased changes in the serum compartment, while the brain lipids primarily showed decreased changes. Interestingly, there were prominent alterations in the sphingolipid class in the brain and blood compartments after single and repetitive injury, which may suggest efflux of brain sphingolipids into the blood after TBI. Genetic algorithms were used for predictive panel selection to classify injured and control samples with high sensitivity and specificity. These overlapping lipid panels primarily mapped to the glycerophospholipid metabolism pathway with Benjamini-Hochberg adjusted q-values less than 0.05. Collectively, these results detail overlapping lipidome changes following mTBI in the brain and blood compartments, increasing our understanding of TBI-related lipid dysregulation while identifying novel biomarker candidates.
Collapse
Affiliation(s)
- Alexis
N. Pulliam
- Coulter
Department of Biomedical Engineering, Georgia
Institute of Technology/Emory University, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Eric C. Gier
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - David A. Gaul
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Samuel G. Moore
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Facundo M. Fernández
- School
of Chemistry and Biochemistry, Georgia Institute
of Technology, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Michelle C. LaPlaca
- Coulter
Department of Biomedical Engineering, Georgia
Institute of Technology/Emory University, Atlanta, GA 30332 USA
- Petit
Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
50
|
Zheng X, Yang J, Hou Y, Shi X, Liu K. Prediction of clinical progression in nervous system diseases: plasma glial fibrillary acidic protein (GFAP). Eur J Med Res 2024; 29:51. [PMID: 38216970 PMCID: PMC10785482 DOI: 10.1186/s40001-023-01631-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/29/2023] [Indexed: 01/14/2024] Open
Abstract
Glial fibrillary acidic protein (GFAP), an intracellular type III intermediate filament protein, provides structural support and maintains the mechanical integrity of astrocytes. It is predominantly found in the astrocytes which are the most abundant subtypes of glial cells in the brain and spinal cord. As a marker protein of astrocytes, GFAP may exert a variety of physiological effects in neurological diseases. For example, previous published literatures showed that autoimmune GFAP astrocytopathy is an inflammatory disease of the central nervous system (CNS). Moreover, the studies of GFAP in brain tumors mainly focus on the predictive value of tumor volume. Furthermore, using biomarkers in the early setting will lead to a simplified and standardized way to estimate the poor outcome in traumatic brain injury (TBI) and ischemic stroke. Recently, observational studies revealed that cerebrospinal fluid (CSF) GFAP, as a valuable potential diagnostic biomarker for neurosyphilis, had a sensitivity of 76.60% and specificity of 85.56%. The reason plasma GFAP could serve as a promising biomarker for diagnosis and prediction of Alzheimer's disease (AD) is that it effectively distinguished AD dementia from multiple neurodegenerative diseases and predicted the individual risk of AD progression. In addition, GFAP can be helpful in differentiating relapsing-remitting multiple sclerosis (RRMS) versus progressive MS (PMS). This review article aims to provide an overview of GFAP in the prediction of clinical progression in neuroinflammation, brain tumors, TBI, ischemic stroke, genetic disorders, neurodegeneration and other diseases in the CNS and to explore the potential therapeutic methods.
Collapse
Affiliation(s)
- Xiaoxiao Zheng
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, China
| | - Jingyao Yang
- Institute of Physiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yiwei Hou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, China
| | - Xinye Shi
- Department of Cardiology, Shanxi Yingkang Yisheng General Hospital, Renmin North Road 5188#, Yuncheng, China
| | - Kangding Liu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, China.
| |
Collapse
|