1
|
Lemche E, Hortobágyi T, Kiecker C, Turkheimer F. Neuropathological links between T2DM and LOAD: systematic review and meta-analysis. Physiol Rev 2025; 105:1429-1486. [PMID: 40062731 DOI: 10.1152/physrev.00040.2024] [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: 10/01/2024] [Revised: 02/01/2025] [Accepted: 02/22/2025] [Indexed: 04/16/2025] Open
Abstract
Recent decades have described parallel neuropathological mechanisms increasing the risk for developing late-onset Alzheimer's dementia (LOAD) in type 2 diabetes mellitus (T2DM); however, still little is known of the role of diabetic encephalopathy and brain atrophy in LOAD. The aim of this systematic review is to provide a comprehensive view on diabetic encephalopathy/cerebral atrophy, taking into account neuroimaging data, neuropathology, metabolic and endocrine mechanisms, amyloid formation, brain perfusion impairments, neuroimmunology, and inflammasome activation. Key switches were identified, to further meta-analyze genomic candidate loci and epigenetic modifications. For the qualitative meta-analysis of genomic bases extracted, human linkage studies were examined; for epigenetic mechanisms, data from both human and animal studies are described. For the systematic review of pathophysiological mechanisms, 1,259 publications were evaluated and 93 gene loci extracted for candidate risk linkages. Sixty-six publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight the insulin signaling system, vascular markers, inflammation and inflammasome pathways, amylin interactions, and glycosylation mechanisms. The protocol was registered with PROSPERO (ID: CRD42023440535).
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Clemens Kiecker
- Department for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
2
|
Lin Y, Chen J, Tan J, Yu Z, Pi R, Xiong J, Ding Y, Chen M, Bai X. Pericytes in the Pulmonary Microenvironment: Guardians or Adversaries? Lung 2025; 203:65. [PMID: 40448710 DOI: 10.1007/s00408-025-00820-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 05/11/2025] [Indexed: 06/02/2025]
Abstract
Pericytes, specialized mural cells residing within the basement membrane of pulmonary microvessels, participate in various biological processes, including vascular homeostasis, immunomodulation, and tissue repair. However, these beneficial physiological roles can be detrimental under pathological conditions. Numerous pulmonary fibrosis models have demonstrated pericyte differentiation into scar-forming myofibroblasts, leading to collagen deposition and matrix remodeling, thereby contributing to tissue fibrosis. Similarly, pericytes play crucial roles in inflammatory diseases. This review aims to explore the dual roles of pericytes in the lung and the underlying mechanisms of their role conversion, providing insights for developing therapeutic strategies targeting these cells.
Collapse
Affiliation(s)
- Yan Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiaqi Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiale Tan
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zihang Yu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ruozheng Pi
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingrong Xiong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Minfeng Chen
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China.
| | - Xue Bai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Salinas MD, Martínez CM, Roca FJ, García-Bernal D, Martínez-Morga M, Rodríguez-Madoz JR, Prósper F, Zapata AG, Moraleda JM, Martínez S, Valdor R. Chaperone-mediated autophagy sustains pericyte stemness necessary for brain tissue homeostasis. J Adv Res 2025:S2090-1232(25)00259-0. [PMID: 40286844 DOI: 10.1016/j.jare.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Pericytes (PCs) are mural cells exhibiting some mesenchymal stem cell (MSC) properties and contribute to tissue regeneration after injury. We have previously shown that glioblastoma cancer cells induce in PCs, a pathogenic upregulation of chaperone-mediated autophagy (CMA) which modulates immune functions and MSC-like properties to support tumor growth. OBJECTIVES The aim of the study was to interrogate the role of CMA-regulated MSC properties in PCs in the context of tissue repair during inflammation triggered by a demyelinating injury. METHODS Studies of RNA-seq were done PCs with (WT) and without (LAMP-2A KO) CMA. Cell characterization related to stemness, lineage and morphology was done in WT and KO PCs. Secretome analysis and cell differentiation assay using the supernatants from CMA-efficient and deficient PCs cultures was done in mesenchymal cells. Inflammatory response of brain cells was assessed with WT and KO PCs secretome. To corroborate in vitro results, CMA modulation in response to inflammation in PCs and tissue repair markers were measured in the lesion areas of a demyelination mouse model and correlated with the tissue reparation after intravenous PC administration. An inflammatory mediator was used to study effects on PC-CMA activity. RESULTS We found that inflammatory mediators such as IFNγ downregulate CMA in PCs, suppressing PC stemness and promoting a pro-inflammatory secretome. Restoration of PC CMA activity during inflammation maintains PC MSC properties and induces an MSC-like proteome which decreases inflammation and promotes tissue repair. We identified secreted proteins involved in regenerative and protective processes, and therefore, necessary to restore brain tissue homeostasis after inflammation induced by a demyelinating injury. CONCLUSION we show that manipulation of CMA activity in host PCs could be a useful therapeutical approach in the context of brain inflammation, which might be extended to other diseases where the pericyte has a key role in response to inflammation.
Collapse
Affiliation(s)
- María Dolores Salinas
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain
| | | | - Francisco J Roca
- Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Unit of Infectious Disease Pathology, Clinical Microbiology and Tropical Medicine, IMIB, 30120 Murcia, Spain
| | - David García-Bernal
- Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain; Virgen de la Arrixaca University Hospital, Hematopoietic Transplant Group, IMIB, 30120 Murcia, Spain
| | - Marta Martínez-Morga
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain
| | - Juan R Rodríguez-Madoz
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Navarra, Spain; Centro de investigación Biomédica en Red de Cancer (CIBERONC), Madrid, Spain
| | - Felipe Prósper
- Hemato-Oncology Program, Cima Universidad de Navarra, IdiSNA, 31008 Pamplona, Navarra, Spain; Centro de investigación Biomédica en Red de Cancer (CIBERONC), Madrid, Spain; Department of Dermatology and Cell Therapy, Clinica Universidad de Navarra (CUN), IdiSNA, 31008 Pamplona Navarra, Spain; Cancer Center Clinica Universidad de Navarra (CCUN), 31008 Pamplona, Navarra, Spain
| | - Agustín G Zapata
- Department of Cell Biology, Faculty of Biology, Complutense University, 28040 Madrid, Spain
| | - Jose María Moraleda
- Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain; Virgen de la Arrixaca University Hospital, Hematopoietic Transplant Group, IMIB, 30120 Murcia, Spain
| | - Salvador Martínez
- Instituto de Neurociencias-Miguel Hernández University (UMH-CSIC), 03550, San Juan de Alicante, ISABIAL, CIBERSAM, Alicante, Spain
| | - Rut Valdor
- Unit of Autophagy, Immune Response and Tolerance in Pathologic Processes, Biomedical Research Institute of Murcia-Pascual Parrilla (IMIB), 30120 Murcia, Spain; Department of Biochemistry and Molecular Biology B, and Immunology, University of Murcia (UMU), 30120 Murcia, Spain; Cell Therapy and Hematopoietic Transplant Group, Faculty of Medicine, UMU, 30120 Murcia, Spain.
| |
Collapse
|
4
|
Stevenson TJ, Lee K, Li S, Montgomery JM, Lee KY, Dragunow M. Heterogeneity in pericyte inflammatory responses across age and species highlight the importance of human cell models. Mol Brain 2025; 18:37. [PMID: 40251678 PMCID: PMC12007128 DOI: 10.1186/s13041-025-01209-7] [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: 12/13/2024] [Accepted: 04/10/2025] [Indexed: 04/20/2025] Open
Abstract
Pericytes in the central nervous system are essential for maintaining blood-brain barrier function, regulating blood flow, modulating immune responses, and interacting closely with surrounding cells of the neurovascular unit to support brain homeostasis. Increasing evidence has highlighted their involvement in age-related neuroinflammation, where their dysfunction may contribute to sustained inflammatory states associated with neurodegenerative disorders. Here, we compared inflammatory responses to lipopolysaccharide (LPS) in primary cerebral pericytes from neonatal and adult mice and adult humans. Our findings indicate that neonatal mouse pericytes display heightened inflammatory activation, with elevated levels of ICAM-1 and several cytokines, compared to adult mouse pericytes reflecting a more reactive phenotype. In contrast, adult mouse pericytes exhibited a significantly reduced cytokine release profile, suggesting lower responsiveness. Notably, while cytokine secretion patterns in adult human pericytes, in part, mirrored those in neonatal mouse pericytes, nitric oxide production, which was observed in mouse pericytes, was absent in the human cells. These results underscore species- and age-dependent variations in cellular behavior, emphasizing the importance of utilizing human brain cell systems when conducting research on neuroinflammation. Understanding these distinctions is vital for designing accurate studies and developing targeted therapies for neuroinflammatory conditions.
Collapse
Affiliation(s)
- Taylor J Stevenson
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Kahee Lee
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Susan Li
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Johanna M Montgomery
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Kevin Y Lee
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.
- Department of Physiology, University of Auckland, Auckland, New Zealand.
- Department of Neurological Surgery, University of California San Francisco, San Francisco, United States of America.
| | - Michael Dragunow
- Department of Pharmacology, University of Auckland, Auckland, New Zealand.
- Centre for Brain Research, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
5
|
Slowikowski E, Willems C, Lemes RMR, Schuermans S, Berghmans N, Rocha RPF, Martens E, Proost P, Delang L, Marques RE, Filho JCA, Marques PE. A central role for CCR2 in monocyte recruitment and blood-brain barrier disruption during Usutu virus encephalitis. J Neuroinflammation 2025; 22:107. [PMID: 40241134 PMCID: PMC12004732 DOI: 10.1186/s12974-025-03435-1] [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: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Usutu virus (USUV) is an emerging neurotropic flavivirus capable of causing encephalitis in humans. Here, our main goal was to characterize the innate immune response in the brain during USUV encephalitis and to identify strategies to control disease severity. Using an immunocompetent mouse model of USUV encephalitis, we showed that microglia activation, blood-brain barrier (BBB) disruption and inflammatory monocyte recruitment are hallmarks of disease 6 days post infection. Activated microglia were in close association to USUV-infected cells, concomitantly with elevated levels of IL-6, IFN-γ, CCL2, CCL5, CXCL10 and CXCL1 in the brain. Monocyte recruitment was CCR2-dependent and driven by IFN-γ and CCL2 production beneath the brain vasculature. Moreover, CCR2 deficiency inhibited microglia activation and BBB disruption, showing the central role of CCR2 in USUV encephalitis. Accordingly, treatment with dexamethasone prevented pro-inflammatory mediator production and reduced leukocyte recruitment significantly, restraining encephalitis severity. Concluding, USUV encephalitis is driven by CCR2-mediated monocyte recruitment and BBB disruption, and blocked therapeutically by glucocorticoids.
Collapse
Affiliation(s)
- Emily Slowikowski
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Céleste Willems
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Robertha Mariana Rodrigues Lemes
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sara Schuermans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Nele Berghmans
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Rebeca Paiva Fróes Rocha
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Erik Martens
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Leen Delang
- Virus-Host Interactions and Therapeutic Approaches (VITA) Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - José Carlos Alves Filho
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Babić A, Bonifačić D, Komen V, Kovačić S, Mamić M, Vuletić V. Blood Biomarkers in Ischemic Stroke Diagnostics and Treatment-Future Perspectives. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:514. [PMID: 40142325 PMCID: PMC11943631 DOI: 10.3390/medicina61030514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025]
Abstract
Stroke is a leading cause of disability and the second most common cause of death worldwide, with its incidence increasing due to an aging population. Early diagnosis is crucial for timely medical intervention. Biomarkers serve as objective indicators to predict outcomes, monitor treatment responses, and assess prognosis. This review examines the evolving landscape of stroke biomarkers, highlighting their potential clinical applications and the challenges hindering their widespread use. Blood biomarkers are readily accessible and provide insight into the pathophysiological processes underlying stroke. This review focuses on neuronal and glial biomarkers, as well as those associated with inflammation, thrombosis, excitotoxicity, and neuroprotection. Also, it focuses on genetic biomarkers. The timing of biomarker measurement is particularly critical in the early stages of stroke, when rapid decision-making is essential, and it requires systematic investigation. Although numerous molecules have been proposed as stroke biomarkers in recent years, none have yet been integrated into routine clinical practice. Stroke biomarkers hold great promise for enhancing diagnosis, risk stratification, and personalized treatment strategies. However, well-designed studies and rigorous validation are necessary to bridge the gap between research findings and clinical implementation. Integrating biomarkers with existing diagnostic tools could revolutionize stroke management and improve patient outcomes. Continued research into blood biomarkers and their clinical utility remains imperative for advancing stroke care.
Collapse
Affiliation(s)
- Anja Babić
- Department of Neurology, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - David Bonifačić
- Department of Neurology, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Vita Komen
- Department of Neurology, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Slavica Kovačić
- Department of Diagnostic and Interventional Radiology, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia
- Department of Radiology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Melani Mamić
- Department of Neurology, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Vladimira Vuletić
- Department of Neurology, Clinical Hospital Centre Rijeka, 51000 Rijeka, Croatia
- Department of Neurology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
7
|
Cembran A, Eisenbaum M, Paris D, Mullan M, Crawford F, Ferguson S, Bachmeier C. Contribution of brain pericytes to neuroinflammation following repetitive head trauma. J Inflamm (Lond) 2025; 22:11. [PMID: 40033399 DOI: 10.1186/s12950-025-00439-9] [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: 12/16/2024] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Neuroinflammation is a prominent pathological hallmark of traumatic brain injury (TBI) and glia cells have been widely characterized in the onset or progression of brain inflammation. While an effect of inflammation on cerebrovascular breakdown has been observed, little is known about the specific contribution of brain pericytes to the inflammatory response in TBI. Here, we focused on studying the pericyte response to inflammatory stimuli commonly found in the brain following TBI. METHODS Mouse brain vascular pericytes were exposed to IL-1β, TNF-α and IFN-γ for 2 h and 24 h and probed for markers of pericyte health and a panel of inflammatory mediators. As the platelet-derived growth factor (PDGF) pathway is critical to pericyte function, we also assessed the effect of PDGF-BB stimulation on the inflammatory response in pericytes. Cultured pericytes were treated with PDGF-BB (10 ng/mL) prior to, simultaneously, and following inflammatory insult. To further investigate their role in brain immunosurveillance, we analyzed the cytokine secretome in mouse pericyte cultures treated with PDGF-BB, as well as in brain vascular pericytes isolated from repetitive mild TBI (r-mTBI) mice that were fed phenytoin-enriched chow, an inducer of PDGF-BB secretion. RESULTS Cytokine stimulation with TNF-α, IL-1β, and IFN-γ for 2 and 24 h led to significant upregulation of PDGFRβ in cultured pericytes, with an 8-fold increase after 24 h. MTT assays showed no significant change in cell viability, indicating that cytokine treatment did not induce cytotoxicity. Further, elevated levels of pro-inflammatory markers STAT1 and p-NFkB were observed in response to cytokine exposure, with a concurrent increase in VCAM1 and MMP9 expression. PDGF-BB treatment significantly attenuated the inflammatory response in pericytes, reducing PDGFRβ levels and the activation of inflammatory pathways, including STAT1 and NFkB. Cytokine secretion profiles also revealed that PDGF-BB, when administered post-inflammatory insult, selectively reduced pro-inflammatory cytokines such as IL-1β and IFN-γ. Additionally, phenytoin treatment in r-mTBI mice decreased IL-1β, TNF-α, IL-5, and KC/GRO levels in isolated brain pericytes, while IL-2, IL-4, and IL-6 levels were unchanged compared to untreated r-mTBI animals. CONCLUSIONS Our results indicate an immunoreactive role for brain pericytes in the propagation of neuroinflammation. Moreover, following brain insults, we found PDGF-BB stimulation can normalize pericyte function and reduce cerebrovascular inflammation, a key factor in secondary brain injury. Targeting brain pericytes may provide novel therapeutic opportunities to improve cerebrovascular health and reduce brain inflammation in the aftermath of TBI.
Collapse
Affiliation(s)
| | | | | | | | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, USA
- James A. Haley Veterans' Hospital, Tampa, FL, USA
| | | | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA.
- Bay Pines VA Healthcare System, Bay Pines, FL, USA.
| |
Collapse
|
8
|
Atak D, Yıldız E, Özkan E, Yousefi M, Özkan A, Yılmaz AB, Kızılırmak AB, Alnajjar IA, Kanar Ç, Caan ZL, Zeybek ŞÜ, Küçükali Cİ, Tüzün E, Gürsoy‐Özdemir Y, Vural A. Longitudinal Investigation of Brain and Spinal Cord Pericytes After Inducible PDGFRβ + Cell Ablation in Adult Mice. J Neurochem 2025; 169:e70035. [PMID: 40066845 PMCID: PMC11894923 DOI: 10.1111/jnc.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
Central nervous system (CNS) pericytes play crucial roles in vascular development and blood-brain barrier maturation during prenatal development, as well as in regulating cerebral blood flow in adults. They have also been implicated in the pathogenesis of numerous neurological disorders. However, the behavior of pericytes in the adult brain after injury remains poorly understood, partly due to limitations in existing pericyte ablation models. To investigate pericyte responses following acute ablation and characterize a novel rodent model for pericyte research, we developed a tamoxifen-inducible PDGFRβ+ cell ablation model by crossing PDGFRβ-P2A-CreERT2 and Rosa26-DTA176 transgenic mouse lines. Using this model, we studied the effects of different tamoxifen doses and conducted histological examinations 15 and 60 days post-injection to assess the impacts of PDGFRβ+ cell ablation in both acute and chronic phases, respectively. Our results demonstrate that a low dose of tamoxifen effectively ablates PDGFRβ+ cells of the CNS in mice without reducing survival or causing significant systemic side effects, such as weight loss. Additionally, we found that the extent of PDGFRβ+ cell depletion varies between the cortex and the spinal cord, as well as between the gray and white matter regions of the spinal cord. Importantly, we observed that both pericyte coverage and numbers increased in the weeks following acute ablation, indicating the regenerative capacity of CNS pericytes in vivo. This study offers a valuable tool for future studies on the role of pericytes in neurological disorders by overcoming the limitations of constitutive pericyte ablation models and providing its longitudinal characterization in the CNS.
Collapse
Affiliation(s)
- Dila Atak
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Erdost Yıldız
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Esra Özkan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| | - Mohammadreza Yousefi
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Ayşe Özkan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of PhysiologyBakırçay UniversityİzmirTürkiye
| | - Aysu Bilge Yılmaz
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Ali Burak Kızılırmak
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | | | - Çiçek Kanar
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Zeynep Lal Caan
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
| | - Şakir Ümit Zeybek
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Cem İsmail Küçükali
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicineİstanbul UniversityIstanbulTürkiye
| | - Yasemin Gürsoy‐Özdemir
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| | - Atay Vural
- Koç University Research Center for Translational Medicine (KUTTAM)Koç UniversityİstanbulTürkiye
- Department of NeurologyKoç UniversityİstanbulTürkiye
| |
Collapse
|
9
|
Zhang ZS, Yang A, Luo X, Zhou HN, Liu YY, Bao DQ, Zhang J, Zang JT, Li QH, Li T, Liu LM. Pericyte-derived extracellular vesicles improve vascular barrier function in sepsis via the Angpt1/PI3K/AKT pathway and pericyte recruitment: an in vivo and in vitro study. Stem Cell Res Ther 2025; 16:70. [PMID: 39940043 PMCID: PMC11823130 DOI: 10.1186/s13287-025-04201-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: 08/30/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Extracellular vesicles derived from pericytes (PCEVs) have been shown to improve vascular permeability, with their therapeutic effects attributed to the presence of pro-regenerative molecules. We hypothesized that angiopoietin 1 (Angpt1) carried by PCEVs contributes to their therapeutic effects after sepsis. METHODS A cecal ligation and puncture (CLP)-induced sepsis rat model was used in vivo, and the effects of PCEVs on vascular endothelial cells were studied in vitro. First, proteomic and Gene Ontology enrichment analyses were performed to analyze the therapeutic mechanism of PCEVs, revealing that the angiogenesis-related protein Angpt1 was highly expressed in PCEVs. We then down-regulated Angpt1 in PCEVs. The role of PCEV-carried Angpt1 on intestinal barrier function, PCs recruitment, and inflammatory cytokines was measured by using septic Sprague-Dawley rats and platelet-derived growth factor receptor beta (PDGFR-β)-Cre + mT/mG transgenic mice. RESULTS PCEVs significantly improved vascular permeability, proliferation, and angiogenesis in CLP-induced gut barrier injury both in vivo and in vitro. Further studies have shown that PCEVs exert a protective effect on intestinal barrier function and PC recruitment. Additionally, PCEVs reduced serum inflammatory factor levels. Our data also demonstrated that the protein levels of phospho-PI3K and phospho-Akt both increased after PCEVs administration, whereas knocking out Angpt1 suppressed the protective effects of PCEVs through decreased activation of PI3K/Akt signaling. CONCLUSION PCEVs protect against sepsis by regulating the vascular endothelial barrier, promoting PC recruitment, protecting intestinal function, and restoring properties via activation of the Angpt1/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Zi-Sen Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ao Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xi Luo
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - He-Nan Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yi-Yan Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dai-Qin Bao
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jie Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jia-Tao Zang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qing-Hui Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Shock and Transfusion, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
10
|
Grootaert MOJ, Pasut A, Raman J, Simmonds SJ, Callewaert B, Col Ü, Dewerchin M, Carmeliet P, Heymans S, Jones EAV. Mural cell dysfunction contributes to diastolic heart failure by promoting endothelial dysfunction and vessel remodelling. Cardiovasc Diabetol 2025; 24:62. [PMID: 39920783 PMCID: PMC11806843 DOI: 10.1186/s12933-025-02623-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is a complex cardiovascular disease associated with metabolic comorbidities. Microvascular dysfunction has been proposed to drive HFpEF, likely via endothelial cell (EC) dysfunction, yet the role of the mural cells herein has never been explored. METHODS We used the diabetic db/db mouse given 1% salt as a new model of HFpEF and crossed then with PDGFRβtg/tg-CreERT2-EYFPtg/tg mice to label the mural cells. We combined single-cell RNA sequencing, NichetNet analysis and histology to determine the role of mural cell dysfunction in HFpEF. RESULTS Db/db mice given 1% salt for 8 weeks developed diastolic dysfunction preceded by capillary density loss, pericyte loss and vessel regression. At 4 weeks of salt, hearts of db/db mice already showed EC dysfunction associated with an anti-angiogenic signature, and an increase in pericyte-EC intracellular space. Db/db + salt hearts were further characterised by increased ACTA2 expression, arteriole wall thickening and vessel enlargement. NicheNet analysis on the single cell transcriptomic data revealed little signalling from the ECs to the mural cells; instead, mural cells signalled strongly to ECs. Mechanistically, pericyte dysfunction induces an EC growth arrest via TNFα-dependent paracrine signalling and downstream signalling through STAT1. CONCLUSION Mural cell dysfunction contributes to HFpEF by inducing coronary vessel remodelling, at least in part by reducing EC proliferation and inducing EC inflammation through TNFα-dependent paracrine signalling.
Collapse
MESH Headings
- Animals
- Pericytes/metabolism
- Pericytes/pathology
- Disease Models, Animal
- Vascular Remodeling
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Heart Failure, Diastolic/physiopathology
- Heart Failure, Diastolic/metabolism
- Heart Failure, Diastolic/pathology
- Heart Failure, Diastolic/genetics
- Heart Failure, Diastolic/etiology
- Coronary Vessels/metabolism
- Coronary Vessels/physiopathology
- Coronary Vessels/pathology
- Signal Transduction
- Ventricular Function, Left
- Male
- Mice, Transgenic
- Mice, Inbred C57BL
- Stroke Volume
- Mice
- Receptor, Platelet-Derived Growth Factor beta/genetics
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Paracrine Communication
- Actins
Collapse
Affiliation(s)
- Mandy O J Grootaert
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium.
- Endocrinology, Diabetes and Nutrition, UCLouvain, Avenue Hippocrate, box 55, Brussels, 1200, Belgium.
| | - Alessandra Pasut
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
| | - Jana Raman
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Steven J Simmonds
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Bram Callewaert
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Ümare Col
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology (CCB), KU Leuven, Leuven, Belgium
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Stephane Heymans
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands
| | - Elizabeth A V Jones
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Herestraat 49, box 911, Leuven, 3000, Belgium.
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, Maastricht, 6229 ER, The Netherlands.
| |
Collapse
|
11
|
Orian JM. A New Perspective on Mechanisms of Neurodegeneration in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis: the Early and Critical Role of Platelets in Neuro/Axonal Loss. J Neuroimmune Pharmacol 2025; 20:14. [PMID: 39904925 PMCID: PMC11794395 DOI: 10.1007/s11481-025-10182-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: 05/22/2024] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) autoimmune disorder, with limited treatment options. This disease is characterized by differential pathophysiology between grey matter (GM) and white matter (WM). The predominant WM hallmark is the perivascular plaque, associated with blood brain barrier (BBB) loss of function, lymphocytic infiltration, microglial reactivity, demyelination and axonal injury and is adequately addressed with immunomodulatory drugs. By contrast, mechanisms underlying GM damage remain obscure, with consequences for neuroprotective strategies. Cortical GM pathology is already significant in early MS and characterized by reduced BBB disruption and lymphocytic infiltration relative to WM, but a highly inflammatory environment, microglial reactivity, demyelination and neuro/axonal loss. There is no satisfactory explanation for the occurrence of neurodegeneration without large-scale inflammatory cell influx in cortical GM. A candidate mechanism suggests that it results from soluble factors originating from meningeal inflammatory cell aggregates, which diffuse into the underlying cortical tissue and trigger microglial activation. However, the recent literature highlights the central role of platelets in inflammation, together with the relationship between coagulation factors, particularly fibrinogen, and tissue damage in MS. Using the experimental autoimmune encephalomyelitis (EAE) model, we identified platelets as drivers of neuroinflammation and platelet-neuron associations from the pre-symptomatic stage. We propose that fibrinogen leakage across the BBB is a signal for platelet infiltration and that platelets represent a major and early participant in neurodegeneration. This concept is compatible with the new appreciation of platelets as immune cells and of neuronal damage driven by inflammatory cells sequestered in the meninges.
Collapse
Affiliation(s)
- Jacqueline Monique Orian
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, Vic. 3086, Australia.
| |
Collapse
|
12
|
Wu Y, Korobeynyk VI, Zamboni M, Waern F, Cole JD, Mundt S, Greter M, Frisén J, Llorens-Bobadilla E, Jessberger S. Multimodal transcriptomics reveal neurogenic aging trajectories and age-related regional inflammation in the dentate gyrus. Nat Neurosci 2025; 28:415-430. [PMID: 39762661 PMCID: PMC11802457 DOI: 10.1038/s41593-024-01848-4] [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: 07/06/2023] [Accepted: 11/07/2024] [Indexed: 02/08/2025]
Abstract
The mammalian dentate gyrus (DG) is involved in certain forms of learning and memory, and DG dysfunction has been implicated in age-related diseases. Although neurogenic potential is maintained throughout life in the DG as neural stem cells (NSCs) continue to generate new neurons, neurogenesis decreases with advancing age, with implications for age-related cognitive decline and disease. In this study, we used single-cell RNA sequencing to characterize transcriptomic signatures of neurogenic cells and their surrounding DG niche, identifying molecular changes associated with neurogenic aging from the activation of quiescent NSCs to the maturation of fate-committed progeny. By integrating spatial transcriptomics data, we identified the regional invasion of inflammatory cells into the hippocampus with age and show here that early-onset neuroinflammation decreases neurogenic activity. Our data reveal the lifelong molecular dynamics of NSCs and their surrounding neurogenic DG niche with age and provide a powerful resource to understand age-related molecular alterations in the aging hippocampus.
Collapse
Affiliation(s)
- Yicheng Wu
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Vladyslav I Korobeynyk
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Margherita Zamboni
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Felix Waern
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - John Darby Cole
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | | | - Sebastian Jessberger
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
13
|
Kim S, Jung UJ, Kim SR. The Crucial Role of the Blood-Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications. J Clin Med 2025; 14:386. [PMID: 39860392 PMCID: PMC11765772 DOI: 10.3390/jcm14020386] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The blood-brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer's and Parkinson's disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
14
|
Morton L, Garza AP, Debska‐Vielhaber G, Villafuerte LE, Henneicke S, Arndt P, Meuth SG, Schreiber S, Dunay IR. Pericytes and Extracellular Vesicle Interactions in Neurovascular Adaptation to Chronic Arterial Hypertension. J Am Heart Assoc 2025; 14:e038457. [PMID: 39719419 PMCID: PMC12054408 DOI: 10.1161/jaha.124.038457] [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: 08/25/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Chronic arterial hypertension restructures the vascular architecture of the brain, leading to a series of pathological responses that culminate in cerebral small-vessel disease. Pericytes respond dynamically to vascular challenges; however, how they manifest under the continuous strain of hypertension has not been elucidated. METHODS AND RESULTS In this study, we characterized pericyte behavior alongside hypertensive states in the spontaneously hypertensive stroke-prone rat model, focusing on their phenotypic and metabolic transformation. Flow cytometry was used to characterize pericytes by their expression of platelet-derived growth factor receptor β, neuroglial antigen 2, cluster of differentiation 13-alanyl aminopeptidase, and antigen Kiel 67. Microvessels were isolated for gene expression profiling and in vitro pericyte expansion. Immunofluorescence validated the cell culture model. Plasma-derived extracellular vesicles from hypertensive rodents were applied as a treatment to assess their effects on pericyte function and detailed metabolic assessments on enriched pericytes measured oxidative phosphorylation and glycolysis. Our results reveal a shift in platelet-derived growth factor receptor β+ pericytes toward increased neuroglial antigen 2 and cluster of differentiation 13-alanyl aminopeptidase coexpression, indicative of their critical role in vascular stabilization and inflammatory responses within the hypertensive milieu. Significant alterations were found within key pathways including angiogenesis, blood-brain barrier integrity, hypoxia, and inflammation. Circulating extracellular vesicles from hypertensive rodents distinctly influenced pericyte mitochondrial function, evidencing their dual role as carriers of disease pathology and potential therapeutic agents. Furthermore, a shift toward glycolytic metabolism in hypertensive pericytes was confirmed, coupled with ATP production dysregulation. CONCLUSIONS Our findings demonstrate that cerebral pericytes undergo phenotypic and metabolic reprogramming in response to hypertension, with hypertensive-derived plasma-derived extracellular vesicles impairing their mitochondrial function. Importantly, plasma-derived extracellular vesicles from normotensive controls restore this function, suggesting their potential as both therapeutic agents and precision biomarkers for hypertensive vascular complications. Further investigation into plasma-derived extracellular vesicle cargo is essential to further explore their therapeutic potential in vascular health.
Collapse
Affiliation(s)
- Lorena Morton
- Medical Faculty, Institute of Inflammation and NeurodegenerationOtto‐von‐Guericke University MagdeburgMagdeburgGermany
| | - Alejandra P. Garza
- Medical Faculty, Institute of Inflammation and NeurodegenerationOtto‐von‐Guericke University MagdeburgMagdeburgGermany
| | | | - Luis E. Villafuerte
- Medical Faculty, Institute of Inflammation and NeurodegenerationOtto‐von‐Guericke University MagdeburgMagdeburgGermany
| | - Solveig Henneicke
- Department of NeurologyOtto von Guericke University MagdeburgMagdeburgGermany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, MagdeburgMagdeburgGermany
| | - Philipp Arndt
- Department of NeurologyOtto von Guericke University MagdeburgMagdeburgGermany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, MagdeburgMagdeburgGermany
| | - Sven G. Meuth
- Department of NeurologyHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Stefanie Schreiber
- Department of NeurologyOtto von Guericke University MagdeburgMagdeburgGermany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, MagdeburgMagdeburgGermany
- Center for Behavioral Brain Sciences (CBBS)MagdeburgGermany
- German Center for Mental Health (DZPG)Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C‐I‐R‐C)Halle‐Jena‐MagdeburgGermany
| | - Ildiko R. Dunay
- Medical Faculty, Institute of Inflammation and NeurodegenerationOtto‐von‐Guericke University MagdeburgMagdeburgGermany
- Center for Behavioral Brain Sciences (CBBS)MagdeburgGermany
- German Center for Mental Health (DZPG)Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C‐I‐R‐C)Halle‐Jena‐MagdeburgGermany
| |
Collapse
|
15
|
Leonard-Duke J, Agro SMJ, Csordas DJ, Bruce AC, Eggertsen TG, Tavakol TN, Comlekoglu T, Barker TH, Bonham CA, Saucerman JJ, Taite LJ, Peirce SM. Multiscale computational model predicts how environmental changes and treatments affect microvascular remodeling in fibrotic disease. PNAS NEXUS 2025; 4:pgae551. [PMID: 39720203 PMCID: PMC11667245 DOI: 10.1093/pnasnexus/pgae551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/13/2024] [Indexed: 12/26/2024]
Abstract
Investigating the molecular, cellular, and tissue-level changes caused by disease, and the effects of pharmacological treatments across these biological scales, necessitates the use of multiscale computational modeling in combination with experimentation. Many diseases dynamically alter the tissue microenvironment in ways that trigger microvascular network remodeling, which leads to the expansion or regression of microvessel networks. When microvessels undergo remodeling in idiopathic pulmonary fibrosis (IPF), functional gas exchange is impaired and lung function declines. We integrated a multiscale computational model with independent experiments to investigate how combinations of biomechanical and biochemical cues in IPF alter cell fate decisions leading to microvascular remodeling. Our computational model predicted that extracellular matrix (ECM) stiffening reduced microvessel area, which was accompanied by physical uncoupling of endothelial cell (EC) and pericytes, the cells that comprise microvessels. Nintedanib, an Food and Drug Administration-approved drug for treating IPF, was predicted to further potentiate microvessel regression by decreasing the percentage of quiescent pericytes while increasing the percentage of pericytes undergoing pericyte-myofibroblast transition in high ECM stiffnesses. Importantly, the model suggested that YAP/TAZ inhibition may overcome the deleterious effects of nintedanib by promoting EC-pericyte coupling and maintaining microvessel homeostasis. Overall, our combination of computational and experimental modeling can predict and explain how cell decisions affect tissue changes during disease and in response to treatments.
Collapse
Affiliation(s)
- Julie Leonard-Duke
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Samuel M J Agro
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| | - David J Csordas
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Anthony C Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Taylor G Eggertsen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Tara N Tavakol
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Tien Comlekoglu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Catherine A Bonham
- Department of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Lakeshia J Taite
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA 22903, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
16
|
Ismail FS, Faustmann PM, Corvace F, Faustmann TJ. Neuroglia in autoimmune encephalitis. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:147-157. [PMID: 40148042 DOI: 10.1016/b978-0-443-19102-2.00002-8] [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
Neuroglial cells play a crucial role in central nervous system (CNS) health and disease. Antibody-associated autoimmune encephalitis (AE) represents a group of inflammatory brain diseases with antibodies (Abs) against neuronal cell surface (e.g., anti-N-methyl-d-aspartate receptor (NMDAR), anti-leucine-rich glioma-inactivated 1 (LGI1), γ-aminobutyric acid (GABA) type A or B receptor (GABAA/BR)) or intracellular neuronal proteins. AE with Abs against glial antigens, e.g., myelin oligodendrocyte glycoprotein (MOG), glial fibrillary acidic protein (GFAP) are also described. Besides the known pathomechanisms with direct pathogenic effects of primary neuronal Abs and activation of innate (dendritic cells) and adaptive (B and T cells) immune systems, research findings suggest the involvement of glial cells including astrocytes, microglia, oligodendrocytes in the pathogenesis of Ab-associated AE, but only a limited number of studies is available. Neuropathologic findings showed reactive astrogliosis and microgliosis with microglial activation/proliferation, e.g., in anti-NMDAR and anti-LGI1 encephalitis. Direct effects of the GABAAR and NMDAR Abs on astrocytic receptors are discussed. Because of the primary involvement of B and T cells in the pathogenesis of Ab-associated AE it can be assumed that astrocytic and microglial activation is largely a response to the primary changes, but additional direct effects of Abs on astrocytic receptors are possible. Further research in this field is required to explore the exact role of glial cells in Ab-associated AE.
Collapse
Affiliation(s)
- Fatme Seval Ismail
- Department of Neurology, Klinikum Vest, Academic Teaching Hospital of the Ruhr University Bochum, Recklinghausen, Germany
| | - Pedro M Faustmann
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany.
| | - Franco Corvace
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| | - Timo Jendrik Faustmann
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
17
|
Cashion JM, Brown LS, Morris GP, Fortune AJ, Courtney JM, Makowiecki K, Premilovac D, Cullen CL, Young KM, Sutherland BA. Pericyte ablation causes hypoactivity and reactive gliosis in adult mice. Brain Behav Immun 2025; 123:681-696. [PMID: 39406266 DOI: 10.1016/j.bbi.2024.10.014] [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: 10/23/2023] [Revised: 09/16/2024] [Accepted: 10/11/2024] [Indexed: 10/22/2024] Open
Abstract
Capillary pericytes are important regulators of cerebral blood flow, blood-brain barrier integrity and neuroinflammation, but can become lost or dysfunctional in disease. The consequences of pericyte loss or dysfunction is extremely difficult to discern when it forms one component of a complex disease process. To evaluate this directly, we examined the effect of adult pericyte loss on mouse voluntary movement and motor function, and physiological responses such as hypoxia, blood-brain barrier (BBB) integrity and glial reactivity. Tamoxifen delivery to Pdgfrβ-CreERT2:: Rosa26-DTA transgenic mice was titrated to produce a dose-dependent ablation of pericytes in vivo. 100mg/kg of tamoxifen ablated approximately half of all brain pericytes, while two consecutive daily doses of 300mg/kg tamoxifen ablated >80% of brain pericytes. In the open field test, mice with ∼50% pericyte loss spent more time immobile and travelled half the distance of control mice. Mice with >80% pericyte ablation also slipped more frequently while performing the beam walk task. Our histopathological analyses of the brain revealed that blood vessel density was unchanged, but vessel lumen width was increased. Pericyte-ablated mice also exhibited: mild BBB disruption; increased neuronal hypoxia; astrogliosis and increased IBA1+ immunoreactivity, suggestive of microgliosis and/or macrophage infiltration. Our results highlight the importance of pericytes in the brain, as pericyte loss can directly compromise brain health and induce behavioural alterations in mice.
Collapse
Affiliation(s)
- Jake M Cashion
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Lachlan S Brown
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Gary P Morris
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Alastair J Fortune
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Jo-Maree Courtney
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kalina Makowiecki
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Dino Premilovac
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
18
|
Feske S, Colucci F, Coetzee WA. Do K ATP channels have a role in immunity? Front Immunol 2024; 15:1484971. [PMID: 39669557 PMCID: PMC11634800 DOI: 10.3389/fimmu.2024.1484971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/30/2024] [Indexed: 12/14/2024] Open
Abstract
Ion channels, exchangers and pumps are expressed ubiquitously in cells from all phyla of life. In mammals, their role is best described in excitable cells, where they regulate the initiation and propagation of action potentials. There are over 70 different types of K+ channels subunits that contribute to these processes. In non-excitable cells, K+ channels set the resting membrane potential, which in turn drives the activity of other translocators. K+ channels also help maintain cell volume, influence cell proliferation and apoptosis and regulate Ca2+ signaling, which in turn is crucial for many cellular processes, including metabolism, secretion, and gene expression. K+ channels play crucial roles in the activation, proliferation and a variety of other functions in cells of the innate and adaptive immune system. The ATP-sensitive K+ (KATP) channel has an established role in diverse cells, but its presence and function in immunity is scantly described. Public gene expression databases show that KATP channel subunits are highly expressed in NKT and NK cells, and that they are significantly upregulated after infection in CD8+ T cells and macrophages. We discuss these findings in the light of the available literature and propose a role for KATP channels in cytotoxicity of cells that are primed for a rapid immune response. Possible underlying molecular mechanisms are discussed.
Collapse
Affiliation(s)
- Stefan Feske
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
| | - Francesco Colucci
- Department of Obstetrics and Gynecology, University of Cambridge, Cambridge, United Kingdom
| | - William A. Coetzee
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Physiology & Neuroscience, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
19
|
Kim WS, Prasongyuenyong K, Ko A, Debnath R, Chen Z, Zhou JX, Shaaf E, Ko KI. ICAM1 + gingival fibroblasts modulate periodontal inflammation to mitigate bone loss. Front Immunol 2024; 15:1484483. [PMID: 39650645 PMCID: PMC11621011 DOI: 10.3389/fimmu.2024.1484483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Tissue-resident fibroblasts are heterogeneous and provide an endogenous source of cytokines that regulate immunologic events in many osteolytic diseases. Identifying distinct inflammatory fibroblast subsets and conducting mechanistic in vivo studies are critical for understanding disease pathogenesis and precision therapeutics, which is poorly explored in periodontitis. Here, we surveyed published single-cell datasets for fibroblast-specific analysis and show that Intercellular Adhesion Molecule-1 (ICAM1) expression selectively defines a fibroblast subset that exhibits an inflammatory transcriptional profile associated with nuclear factor-κB (NF-κB) pathway. ICAM1+ fibroblasts expand in both human periodontitis and murine ligature-induced periodontitis model, which have upregulated expression of CCL2 and CXCL1 compared to other fibroblast populations. Using a mouse model to selectively target gingival stromal cells, we further show that disruption of an inflammatory pathway by inhibiting transcriptional activity of NF-κB in these cells accelerated periodontal bone loss. Mechanistically, this was linked to a reduction of CCL2 expression by the ICAM1+ fibroblasts, leading to impaired macrophage recruitment and efferocytosis that was associated with persistent neutrophilic inflammation. These results may have a significant therapeutic implication as ICAM1+ gingival fibroblasts exert a protective response by regulating innate immune responses that are needed for the controlled inflammatory events in early stages of periodontitis.
Collapse
Affiliation(s)
- William S. Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kawintip Prasongyuenyong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Annette Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rahul Debnath
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhaoxu Chen
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jonathan X. Zhou
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emon Shaaf
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kang I. Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
20
|
Shen H, Ouyang Y, Zhang L, Li J, Wang S. Blood Cell Membrane-Coated Nanomaterials as a Versatile Biomimetic Nanoplatform for Antitumor Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1757. [PMID: 39513837 PMCID: PMC11548044 DOI: 10.3390/nano14211757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
The application of nanomaterials in tumor therapy is increasingly widespread, offering more possibilities for enhanced tumor therapy. However, the unclear biological distribution and metabolism of nanomaterials may lead to immune rejection or inflammatory reactions, posing numerous challenges to their clinical translation. The rich diversity and multifaceted functions of blood cells offer promising biological avenues for enhancing the application of nanoparticles in cancer therapy. Blood cell membranes, being made of naturally found components in the body, exhibit significant biocompatibility, which can reduce the body's immune rejection response, extend the drug's residence time in the bloodstream, and enhance its bioavailability. Integrating blood cell membranes with nanomaterials enhances tumor therapy by improving targeted delivery, prolonging circulation time, and evading immune responses. This review summarizes recent advancements in the application of blood cell membrane-coated nanomaterials for antitumor therapy, with a particular focus on their use in photodynamic and photothermal treatments. Additionally, it explores their potential for synergistic effects when combined with other therapeutic modalities.
Collapse
Affiliation(s)
| | | | | | - Jing Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; (H.S.); (L.Z.)
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China; (H.S.); (L.Z.)
| |
Collapse
|
21
|
Raj S, Sarangi P, Goyal D, Kumar H. The Hidden Hand in White Matter: Pericytes and the Puzzle of Demyelination. ACS Pharmacol Transl Sci 2024; 7:2912-2923. [PMID: 39421660 PMCID: PMC11480894 DOI: 10.1021/acsptsci.4c00192] [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: 04/03/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Disruption of myelin, the fatty sheath-insulating nerve fibers in the white matter, blocks or slows the rapid transmission of electrical signals along nerve cells and contributes to several neurodegenerative diseases such as multiple sclerosis. Traditionally, research has focused on neuronal dysfunction as the primary factor, including autoimmunity, infections, inflammation, and genetic disorders causing demyelination. However, recent insights emphasize the critical role of pericytes, non-neuronal cells that regulate blood flow and maintain the health of blood vessels within white matter. This Perspective explores the principal mechanisms through which pericyte dysfunction contributes to damage and demyelination, including impaired communication with neurons (neurovascular uncoupling), excessive formation of scar tissue (fibrosis), and the infiltration of detrimental substances from the bloodstream. Understanding these mechanisms of pericyte-driven demyelination may lead to the creation of new therapeutic strategies for tackling a range of neurodegenerative conditions.
Collapse
Affiliation(s)
- Siddharth Raj
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Priyabrata Sarangi
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Divya Goyal
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| |
Collapse
|
22
|
Jurcau MC, Jurcau A, Cristian A, Hogea VO, Diaconu RG, Nunkoo VS. Inflammaging and Brain Aging. Int J Mol Sci 2024; 25:10535. [PMID: 39408862 PMCID: PMC11476611 DOI: 10.3390/ijms251910535] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Progress made by the medical community in increasing lifespans comes with the costs of increasing the incidence and prevalence of age-related diseases, neurodegenerative ones included. Aging is associated with a series of morphological changes at the tissue and cellular levels in the brain, as well as impairments in signaling pathways and gene transcription, which lead to synaptic dysfunction and cognitive decline. Although we are not able to pinpoint the exact differences between healthy aging and neurodegeneration, research increasingly highlights the involvement of neuroinflammation and chronic systemic inflammation (inflammaging) in the development of age-associated impairments via a series of pathogenic cascades, triggered by dysfunctions of the circadian clock, gut dysbiosis, immunosenescence, or impaired cholinergic signaling. In addition, gender differences in the susceptibility and course of neurodegeneration that appear to be mediated by glial cells emphasize the need for future research in this area and an individualized therapeutic approach. Although rejuvenation research is still in its very early infancy, accumulated knowledge on the various signaling pathways involved in promoting cellular senescence opens the perspective of interfering with these pathways and preventing or delaying senescence.
Collapse
Affiliation(s)
| | - Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Alexander Cristian
- Department of Psycho-Neurosciences and Rehabilitation, University of Oradea, 410087 Oradea, Romania
| | - Vlad Octavian Hogea
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | | | | |
Collapse
|
23
|
Zhang Y, Mu BR, Ran Z, Zhu T, Huang X, Yang X, Wang DM, Ma QH, Lu MH. Pericytes in Alzheimer's disease: Key players and therapeutic targets. Exp Neurol 2024; 379:114825. [PMID: 38777251 DOI: 10.1016/j.expneurol.2024.114825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/09/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that leads to progressive cognitive decline and neuropathological changes. Pericytes, which are vessel mural cells on the basement membrane of capillaries, play a crucial role in regulating cerebrovascular functions and maintaining neurovascular unit integrity. Emerging research substantiates the involvement of pericytes in AD. This review provides a comprehensive overview of pericytes, including their structure, origin, and markers and various functions within the central nervous system. Emphatically, the review explores the intricate mechanisms through which pericytes contribute to AD, including their interactions with amyloid beta and apolipoprotein E, as well as various signaling pathways. The review also highlights potential for targeted pericyte therapy for AD, with a focus on stem cell therapy and drug treatments. Future research directions include the classification of pericyte subtypes, studies related to aging, and the role of pericytes in exosome-related mechanisms in AD pathology. In conclusion, this review consolidates current knowledge on the pivotal roles of pericytes in AD and their potential as therapeutic targets, providing valuable insights for future research and clinical interventions aimed at addressing the impact of AD on patients' lives.
Collapse
Affiliation(s)
- Yu Zhang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ben-Rong Mu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhao Ran
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tao Zhu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xin Huang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215021, China
| | - Xiong Yang
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dong-Mei Wang
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou 215021, China.
| | - Mei-Hong Lu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
24
|
Ma YZ, Cao JX, Zhang YS, Su XM, Jing YH, Gao LP. T Cells Trafficking into the Brain in Aging and Alzheimer's Disease. J Neuroimmune Pharmacol 2024; 19:47. [PMID: 39180590 DOI: 10.1007/s11481-024-10147-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
The meninges, choroid plexus (CP) and blood-brain barrier (BBB) are recognized as important gateways for peripheral immune cell trafficking into the central nervous system (CNS). Accumulation of peripheral immune cells in brain parenchyma can be observed during aging and Alzheimer's disease (AD). However, the mechanisms by which peripheral immune cells enter the CNS through these three pathways and how they interact with resident cells within the CNS to cause brain injury are not fully understood. In this paper, we review recent research on T cells recruitment in the brain during aging and AD. This review focuses on the possible pathways through which T cells infiltrate the brain, the evidence that T cells are recruited to the brain, and how infiltrating T cells interact with the resident cells in the CNS during aging and AD. Unraveling these issues will contribute to a better understanding of the mechanisms of aging and AD from the perspective of immunity, and hopefully develop new therapeutic strategies for brain aging and AD.
Collapse
Affiliation(s)
- Yue-Zhang Ma
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yi-Shu Zhang
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Mei Su
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
25
|
Frangogiannis NG. The fate and role of the pericytes in myocardial diseases. Eur J Clin Invest 2024; 54:e14204. [PMID: 38586936 DOI: 10.1111/eci.14204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
The adult mammalian heart contains a large population of pericytes that play important roles in homeostasis and disease. In the normal heart, pericytes regulate microvascular permeability and flow. Myocardial diseases are associated with marked alterations in pericyte phenotype and function. This review manuscript discusses the role of pericytes in cardiac homeostasis and disease. Following myocardial infarction (MI), cardiac pericytes participate in all phases of cardiac repair. During the inflammatory phase, pericytes may secrete cytokines and chemokines and may regulate leukocyte trafficking, through formation of intercellular gaps that serve as exit points for inflammatory cells. Moreover, pericyte contraction induces microvascular constriction, contributing to the pathogenesis of 'no-reflow' in ischemia and reperfusion. During the proliferative phase, pericytes are activated by growth factors, such as transforming growth factor (TGF)-β and contribute to fibrosis, predominantly through secretion of fibrogenic mediators. A fraction of pericytes acquires fibroblast identity but contributes only to a small percentage of infarct fibroblasts and myofibroblasts. As the scar matures, pericytes form a coat around infarct neovessels, promoting stabilization of the vasculature. Pericytes may also be involved in the pathogenesis of chronic heart failure, by regulating inflammation, fibrosis, angiogenesis and myocardial perfusion. Pericytes are also important targets of viral infections (such as SARS-CoV2) and may be implicated in the pathogenesis of cardiac complications of COVID19. Considering their role in myocardial inflammation, fibrosis and angiogenesis, pericytes may be promising therapeutic targets in myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
26
|
Hu Y, Hu XD, He ZQ, Liu Y, Gui YK, Zhu SH, Da X, Liu YN, Liu LX, Shen QY, Xu GH. Anesthesia/surgery activate MMP9 leading to blood-brain barrier disruption, triggering neuroinflammation and POD-like behavior in aged mice. Int Immunopharmacol 2024; 135:112290. [PMID: 38796964 DOI: 10.1016/j.intimp.2024.112290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
Anesthesia and surgery activate matrix metalloproteinase 9 (MMP9), leading to blood-brain barrier (BBB) disruption and postoperative delirium (POD)-like behavior, especially in the elderly. Aged mice received intraperitoneal injections of either the MMP9 inhibitor SB-3CT, melatonin, or solvent, and underwent laparotomy under 3 % sevoflurane anesthesia(anesthesia/surgery). Behavioral tests were performed 24 h pre- and post-operatively. Serum and cortical tissue levels of interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α) were measured using ELISA. Levels of PDGFRβ, MMP9, tight junction, Mfsd2a, caveolin-1, synaptophysin, and postsynaptic densin (PSD)-95 proteins in the prefrontal cortex were assayed using Western blotting. BBB permeability was assessed by detecting IgG in the prefrontal cortex and serum S100β levels. Anesthesia/surgery-induced peripheral inflammation activated MMP9, which in turn injured pericytes and tight junctions and increased transcytosis, thereby disrupting the BBB. Impaired BBB allowed the migration of peripheral inflammation into the central nervous system (CNS), thereby inducing neuroinflammation, synaptic dysfunction, and POD-like behaviors. However, MMP9 inhibition reduced pericyte and tight junction injury and transcytosis, thereby preserving BBB function and preventing the migration of peripheral inflammation into the CNS, thus attenuating synaptic dysfunction and POD-like behavior. In addition, to further validate the above findings, we showed that melatonin exerted similar effects through inhibition of MMP9. The present study shows that after anesthesia/surgery, inflammatory cytokines upregulation is involved in regulating BBB permeability in aged mice through activation of MMP9, suggesting that MMP9 may be a potential target for the prevention of POD.
Collapse
Affiliation(s)
- Yun Hu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Xu-Dong Hu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Zi-Qing He
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Yang Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Yong-Kang Gui
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Si-Hui Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Xin Da
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Yi-Nuo Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Li-Xia Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China
| | - Qi-Ying Shen
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China.
| | - Guang-Hong Xu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei, PR China.
| |
Collapse
|
27
|
Tan IL, Modderman R, Stachurska A, Almeida R, de Vries R, Heersema DJ, Gacesa R, Wijmenga C, Jonkers IH, Meilof JF, Withoff S. Potential biomarkers for multiple sclerosis stage from targeted proteomics and microRNA sequencing. Brain Commun 2024; 6:fcae209. [PMID: 38978729 PMCID: PMC11229703 DOI: 10.1093/braincomms/fcae209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/08/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
Multiple sclerosis is a chronic demyelinating disease of the central nervous system. There is a need for new circulating biomarkers for multiple sclerosis, in particular, markers that differentiate multiple sclerosis subtypes (relapsing-remitting, secondary progressive and primary progressive multiple sclerosis), as this can help in making treatment decisions. In this study, we explore two classes of potential multiple sclerosis biomarkers-proteins and microRNAs-circulating in the cerebrospinal fluid and serum. Targeted medium-throughput proteomics (92 proteins) and microRNA sequencing were performed on serum samples collected in a cross-sectional case-control cohort (cohort I, controls n = 30, multiple sclerosis n = 75) and a prospective multiple sclerosis cohort (cohort II, n = 93). For cohort I, we also made these measurements in paired cerebrospinal fluid samples. In the cohort I cerebrospinal fluid, we observed differences between multiple sclerosis and controls for 13 proteins, including some previously described to be markers for multiple sclerosis [e.g. CD27, C-X-C motif chemokine 13 (CXCL13) and interleukin-7 (IL7)]. No microRNAs were significantly differentially expressed between multiple sclerosis and controls in the cerebrospinal fluid. In serum, 10 proteins, including angiopoietin-1 receptor (TIE2), and 16 microRNAs were significantly different between relapsing-remitting multiple sclerosis and secondary progressive multiple sclerosis after performing a meta-analysis combining both cohorts. In the prospective part of the study, participants with relapsing-remitting multiple sclerosis were followed for around 3 years, during which time 12 participants converted to secondary progressive multiple sclerosis. In these longitudinally collected serum samples, we observed a peak in granzyme B, A and H proteins around the time of conversion. Single-sample enrichment analysis of serum microRNA profiles revealed that the peak in granzyme B levels around conversion coincides with enrichment for microRNAs that are enriched in CD4+, CD8+ and natural killer cells (e.g. miRNA-150). We identified several proteins and microRNAs in serum that represent potential biomarkers for relapsing-remitting and secondary progressive multiple sclerosis. Conversion to secondary progressive disease is marked by a peak in granzyme B levels and enrichment for immune-related microRNAs. This indicates that specific immune cell-driven processes may contribute to the conversion of relapsing-remitting multiple sclerosis to secondary progressive multiple sclerosis.
Collapse
Affiliation(s)
- Ineke L Tan
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Rutger Modderman
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Anna Stachurska
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Rodrigo Almeida
- Telespazio Belgium S.R.L. for the European Space Agency (ESA), 2200AG Noordwijk, The Netherlands
| | - Riemer de Vries
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Dorothea J Heersema
- Department of Neurology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- MS Center Noord Nederland, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ranko Gacesa
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Iris H Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Jan F Meilof
- MS Center Noord Nederland, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
28
|
Yang P, Li Y, Qian K, Zhou L, Cheng Y, Wu J, Xu M, Wang T, Yang X, Mu Y, Liu X, Zhang Q. Precise Modulation of Pericyte Dysfunction by a Multifunctional Nanoprodrug to Ameliorate Alzheimer's Disease. ACS NANO 2024; 18:14348-14366. [PMID: 38768086 DOI: 10.1021/acsnano.4c00480] [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: 05/22/2024]
Abstract
Pericyte dysfunction severely undermines cerebrovascular integrity and exacerbates neurodegeneration in Alzheimer's disease (AD). However, pericyte-targeted therapy is a yet-untapped frontier for AD. Inspired by the elevation of vascular cell adhesion molecule-1 (VCAM-1) and reactive oxygen species (ROS) levels in pericyte lesions, we fabricated a multifunctional nanoprodrug by conjugating the hybrid peptide VLC, a fusion of the VCAM-1 high-affinity peptide VHS and the neuroprotective apolipoprotein mimetic peptide COG1410, to curcumin (Cur) through phenylboronic ester bond (VLC@Cur-NPs) to alleviate complex pericyte-related pathological changes. Importantly, VLC@Cur-NPs effectively homed to pericyte lesions via VLC and released their contents upon ROS stimulation to maximize their regulatory effects. Consequently, VLC@Cur-NPs markedly increased pericyte regeneration to form a positive feedback loop and thus improved neurovascular function and ultimately alleviated memory defects in APP/PS1 transgenic mice. We present a promising therapeutic strategy for AD that can precisely modulate pericytes and has the potential to treat other cerebrovascular diseases.
Collapse
Affiliation(s)
- Peng Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yixian Li
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Kang Qian
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Lingling Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yunlong Cheng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Jing Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Minjun Xu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Tianying Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Xiyu Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Yongkang Mu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Xuan Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, & State Key Laboratory of Molecular Engineering of Polymers, School of Pharmacy, Fudan University, Shanghai 201203, People's Republic of China
| |
Collapse
|
29
|
Kim HW, Wu KLK, Tam KW, Chan YS, Shum DKY. Pericyte derivation and transplantation for blood-CNS barrier reconstitution in CNS disorders. IBRO Neurosci Rep 2024; 16:147-154. [PMID: 39007089 PMCID: PMC11240299 DOI: 10.1016/j.ibneur.2023.12.007] [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: 03/04/2023] [Revised: 12/09/2023] [Accepted: 12/29/2023] [Indexed: 07/16/2024] Open
Abstract
Disruption of the blood-central nervous system barrier (BCB) is increasingly recognized as a pathological factor in diseases and trauma of the central nervous system. Despite the neuropathological impact, current treatment modalities do not target the BCB; strategies to reconstitute the impaired BCB have been restricted to nutritional and dietary remedies. As an integral cell type in the neurovascular unit, pericytes are crucial to the development, maintenance, and repair of the BCB. As such, pericytes are well poised as cellular agents for reconstitution of the impaired BCB. Here, we summarize recent revelations regarding the role of BCB disruption in diseases and trauma of the central nervous system and highlight how pericytes are harnessed to provide targeted therapeutic effect in each case. This review will also address how recent advances in pericyte derivation strategies can serve to overcome practical hurdles in the clinical use of pericytes.
Collapse
Affiliation(s)
- Hyun Woo Kim
- Department of Orthopedics and Traumatology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kenneth Lap Kei Wu
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kin-Wai Tam
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ying-Shing Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory for Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Daisy Kwok-Yan Shum
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory for Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
30
|
Cao Y, Xu W, Liu Q. Alterations of the blood-brain barrier during aging. J Cereb Blood Flow Metab 2024; 44:881-895. [PMID: 38513138 PMCID: PMC11318406 DOI: 10.1177/0271678x241240843] [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: 10/21/2023] [Revised: 02/19/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic interface that regulates the exchange of molecules and cells between the blood and the central nervous system. It undergoes structural and functional changes during aging, which may compromise its integrity and contribute to the pathogenesis of neurodegenerative diseases. In recent years, advances in microscopy and high-throughput bioinformatics have allowed a more in-depth investigation of the aging mechanisms of BBB. This review summarizes age-related alterations of the BBB structure and function from six perspectives: endothelial cells, astrocytes, pericytes, basement membrane, microglia and perivascular macrophages, and fibroblasts, ranging from the molecular level to the human multi-system level. These basic components are essential for the proper functioning of the BBB. Recent imaging methods of BBB were also reviewed. Elucidation of age-associated BBB changes may offer insights into BBB homeostasis and may provide effective therapeutic strategies to protect it during aging.
Collapse
Affiliation(s)
- Yufan Cao
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weihai Xu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Leonard-Duke J, Agro SMJ, Csordas DJ, Bruce AC, Eggertsen TG, Tavakol TN, Barker TH, Bonham CA, Saucerman JJ, Taite LJ, Peirce SM. Multiscale computational model predicts how environmental changes and drug treatments affect microvascular remodeling in fibrotic disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585249. [PMID: 38559112 PMCID: PMC10979947 DOI: 10.1101/2024.03.15.585249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Investigating the molecular, cellular, and tissue-level changes caused by disease, and the effects of pharmacological treatments across these biological scales, necessitates the use of multiscale computational modeling in combination with experimentation. Many diseases dynamically alter the tissue microenvironment in ways that trigger microvascular network remodeling, which leads to the expansion or regression of microvessel networks. When microvessels undergo remodeling in idiopathic pulmonary fibrosis (IPF), functional gas exchange is impaired due to loss of alveolar structures and lung function declines. Here, we integrated a multiscale computational model with independent experiments to investigate how combinations of biomechanical and biochemical cues in IPF alter cell fate decisions leading to microvascular remodeling. Our computational model predicted that extracellular matrix (ECM) stiffening reduced microvessel area, which was accompanied by physical uncoupling of endothelial cell (ECs) and pericytes, the cells that comprise microvessels. Nintedanib, an FDA-approved drug for treating IPF, was predicted to further potentiate microvessel regression by decreasing the percentage of quiescent pericytes while increasing the percentage of pericytes undergoing pericyte-myofibroblast transition (PMT) in high ECM stiffnesses. Importantly, the model suggested that YAP/TAZ inhibition may overcome the deleterious effects of nintedanib by promoting EC-pericyte coupling and maintaining microvessel homeostasis. Overall, our combination of computational and experimental modeling can explain how cell decisions affect tissue changes during disease and in response to treatments.
Collapse
Affiliation(s)
- Julie Leonard-Duke
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Samuel M. J. Agro
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - David J. Csordas
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Anthony C. Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Taylor G. Eggertsen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Tara N. Tavakol
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Thomas H. Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Catherine A. Bonham
- Department of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jeffery J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Lakeshia J. Taite
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
32
|
Carvalho TP, Toledo FAO, Bautista DFA, Silva MF, Oliveira JBS, Lima PA, Costa FB, Ribeiro NQ, Lee JY, Birbrair A, Paixão TA, Tsolis RM, Santos RL. Pericytes modulate endothelial inflammatory response during bacterial infection. mBio 2024; 15:e0325223. [PMID: 38289074 PMCID: PMC10936204 DOI: 10.1128/mbio.03252-23] [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: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 03/14/2024] Open
Abstract
Pericytes are located around blood vessels, in close contact with endothelial cells. We discovered that pericytes dampen pro-inflammatory endothelial cell responses. Endothelial cells co-cultured with pericytes had markedly reduced expression of adhesion molecules (PECAM-1 and ICAM-1) and proinflammatory cytokines (CCL-2 and IL-6) in response to bacterial stimuli (Brucella ovis, Listeria monocytogenes, or Escherichia coli lipopolysaccharide). Pericyte-depleted mice intraperitoneally inoculated with either B. ovis, a stealthy pathogen that does not trigger detectable inflammation, or Listeria monocytogenes, developed peritonitis. Further, during Citrobacter rodentium infection, pericyte-depleted mice developed severe intestinal inflammation, which was not evident in control mice. The anti-inflammatory effect of pericytes required connexin 43, as either chemical inhibition or silencing of connexin 43 abrogated pericyte-mediated suppression of endothelial inflammatory responses. Our results define a mechanism by which pericytes modulate inflammation during infection, which shifts our understanding of pericyte biology: from a structural cell to a pro-active player in modulating inflammation. IMPORTANCE A previously unknown mechanism by which pericytes modulate inflammation was discovered. The absence of pericytes or blocking interaction between pericytes and endothelium through connexin 43 results in stronger inflammation, which shifts our understanding of pericyte biology, from a structural cell to a player in controlling inflammation.
Collapse
Affiliation(s)
- Thaynara P. Carvalho
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Frank A. O. Toledo
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Diego F. A. Bautista
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Monique F. Silva
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jefferson B. S. Oliveira
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pâmela A. Lima
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fabíola B. Costa
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Noelly Q. Ribeiro
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jee-Yon Lee
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Alexander Birbrair
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tatiane A. Paixão
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Reneé M. Tsolis
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Renato L. Santos
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| |
Collapse
|
33
|
Nie X, Yuan T, Yu T, Yun Z, Yu T, Liu Q. Non-stem cell-derived exosomes: a novel therapeutics for neurotrauma. J Nanobiotechnology 2024; 22:108. [PMID: 38475766 DOI: 10.1186/s12951-024-02380-0] [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: 10/23/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Neurotrauma, encompassing traumatic brain injuries (TBI) and spinal cord injuries (SCI) impacts a significant portion of the global population. While spontaneous recovery post-TBI or SCI is possible, recent advancements in cell-based therapies aim to bolster these natural reparative mechanisms. Emerging research indicates that the beneficial outcomes of such therapies might be largely mediated by exosomes secreted from the administered cells. While stem cells have garnered much attention, exosomes derived from non-stem cells, including neurons, Schwann cells, microglia, and vascular endothelial cells, have shown notable therapeutic potential. These exosomes contribute to angiogenesis, neurogenesis, and axon remodeling, and display anti-inflammatory properties, marking them as promising agents for neurorestorative treatments. This review provides an in-depth exploration of the current methodologies, challenges, and future directions regarding the therapeutic role of non-stem cell-derived exosomes in neurotrauma.
Collapse
Affiliation(s)
- Xinyu Nie
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Tianyang Yuan
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Tong Yu
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Zhihe Yun
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Tao Yu
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China
| | - Qinyi Liu
- Department of Orthopaedic, The second hospital of Jilin University, Changchun, China.
| |
Collapse
|
34
|
Bejarano L, Kauzlaric A, Lamprou E, Lourenco J, Fournier N, Ballabio M, Colotti R, Maas R, Galland S, Massara M, Soukup K, Lilja J, Brouland JP, Hottinger AF, Daniel RT, Hegi ME, Joyce JA. Interrogation of endothelial and mural cells in brain metastasis reveals key immune-regulatory mechanisms. Cancer Cell 2024; 42:378-395.e10. [PMID: 38242126 DOI: 10.1016/j.ccell.2023.12.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/11/2023] [Accepted: 12/20/2023] [Indexed: 01/21/2024]
Abstract
Brain metastasis (BrM) is a common malignancy, predominantly originating from lung, melanoma, and breast cancers. The vasculature is a key component of the BrM tumor microenvironment with critical roles in regulating metastatic seeding and progression. However, the heterogeneity of the major BrM vascular components, namely endothelial and mural cells, is still poorly understood. We perform single-cell and bulk RNA-sequencing of sorted vascular cell types and detect multiple subtypes enriched specifically in BrM compared to non-tumor brain, including previously unrecognized immune regulatory subtypes. We integrate the human data with mouse models, creating a platform to interrogate vascular targets for the treatment of BrM. We find that the CD276 immune checkpoint molecule is significantly upregulated in the BrM vasculature, and anti-CD276 blocking antibodies prolonged survival in preclinical trials. This study provides important insights into the complex interactions between the vasculature, immune cells, and cancer cells, with translational relevance for designing therapeutic interventions.
Collapse
Affiliation(s)
- Leire Bejarano
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Annamaria Kauzlaric
- Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Eleni Lamprou
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Joao Lourenco
- Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Nadine Fournier
- Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Translational Data Science Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Michelle Ballabio
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland
| | - Roberto Colotti
- In Vivo Imaging Facility (IVIF), University of Lausanne, Lausanne, Switzerland
| | - Roeltje Maas
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Sabine Galland
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Matteo Massara
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Klara Soukup
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland
| | - Johanna Lilja
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland
| | - Jean-Philippe Brouland
- Department of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Andreas F Hottinger
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Roy T Daniel
- Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Monika E Hegi
- Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Department of Neurosurgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland; Neuroscience Research Center, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland; Agora Cancer Research Centre Lausanne, Lausanne, Switzerland; Lundin Family Brain Tumor Research Center, Departments of Oncology and Clinical Neurosciences, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.
| |
Collapse
|
35
|
Chew V, Chuang CH, Hsu C. Translational research on drug development and biomarker discovery for hepatocellular carcinoma. J Biomed Sci 2024; 31:22. [PMID: 38368324 PMCID: PMC10874078 DOI: 10.1186/s12929-024-01011-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 02/10/2024] [Indexed: 02/19/2024] Open
Abstract
Translational research plays a key role in drug development and biomarker discovery for hepatocellular carcinoma (HCC). However, unique challenges exist in this field because of the limited availability of human tumor samples from surgery, the lack of homogenous oncogenic driver mutations, and the paucity of adequate experimental models. In this review, we provide insights into these challenges and review recent advancements, with a particular focus on the two main agents currently used as mainstream therapies for HCC: anti-angiogenic agents and immunotherapy. First, we examine the pre-clinical and clinical studies to highlight the challenges of determining the optimal therapeutic combinations with biologically effective dosage for HCC. Second, we discuss biomarker studies focusing on anti-PD1/anti-PD-L1-based combination therapy. Finally, we discuss the progress made in our collective understanding of tumor immunology and in multi-omics analysis technology, which enhance our understanding of the mechanisms underlying immunotherapy, characterize different patient subgroups, and facilitate the development of novel combination approaches to improve treatment efficacy. In summary, this review provides a comprehensive overview of efforts in translational research aiming at advancing our understanding of and improving the treatment of HCC.
Collapse
Affiliation(s)
- Valerie Chew
- Translational Immunology Institute, SingHealth-DukeNUS Academic Medical Centre, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Chien-Huai Chuang
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Chiun Hsu
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
36
|
Min H, O’Neil SM, Xu L, Moseman EA, Kurtzberg J, Filiano AJ. Mural cells interact with macrophages in the dura mater to regulate CNS immune surveillance. J Exp Med 2024; 221:e20230326. [PMID: 38193859 PMCID: PMC10783178 DOI: 10.1084/jem.20230326] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 09/07/2023] [Accepted: 12/13/2023] [Indexed: 01/10/2024] Open
Abstract
The central nervous system (CNS) tightly regulates access of circulating immune cells. Immunosurveillance is therefore managed in the meninges at the borders of the CNS. Here, we demonstrated that mural cells, which include pericytes and smooth muscle cells, decreased coverage around blood vessels in the dura, the outermost layer of the meninges, and upregulated gene pathways involved in leukocyte migration in presymptomatic experimental autoimmune encephalomyelitis (EAE). Partially depleting mural cells promoted the trafficking of CNS antigen-specific T cells to the dura in a process that depended on resident antigen-presenting cells, thereby increasing susceptibility to passive EAE. Mechanistically, mural cells physically contacted macrophages in the dura and transferred cytoplasmic components, including processing bodies (RNA granules shown to reprogram transcriptomes), which were critical to suppress antigen-dependent T helper (TH) cell activation and TH17 differentiation. Our study revealed a mechanism by which mural cell-macrophage interactions regulate the trafficking of CNS antigen-specific T cells to the dura.
Collapse
Affiliation(s)
- Hyunjung Min
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Shane M. O’Neil
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
| | - Li Xu
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
| | - E. Ashley Moseman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Anthony J. Filiano
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
37
|
Lummis NC, Gastfriend BD, Daneman R. Dural mural cells paint an anti-inflammatory picture. J Exp Med 2024; 221:e20232263. [PMID: 38270593 PMCID: PMC10818063 DOI: 10.1084/jem.20232263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024] Open
Abstract
Mural cells directly contact macrophages in the dural layer of the meninges to suppress pro-inflammatory phenotypes, including antigen presentation and lymphocyte differentiation. These mechanisms represent new targets for modulating CNS immune surveillance and pathological inflammation (Min et al. 2024. J. Exp. Med.https://doi.org/10.1084/jem.20230326).
Collapse
Affiliation(s)
- Nicole C. Lummis
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Benjamin D. Gastfriend
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Richard Daneman
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
38
|
van Splunder H, Villacampa P, Martínez-Romero A, Graupera M. Pericytes in the disease spotlight. Trends Cell Biol 2024; 34:58-71. [PMID: 37474376 PMCID: PMC10777571 DOI: 10.1016/j.tcb.2023.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 07/22/2023]
Abstract
Pericytes are known as the mural cells in small-caliber vessels that interact closely with the endothelium. Pericytes play a key role in vasculature formation and homeostasis, and when dysfunctional contribute to vasculature-related diseases such as diabetic retinopathy and neurodegenerative conditions. In addition, significant extravascular roles of pathological pericytes are being discovered with relevant implications for cancer and fibrosis. Pericyte research is challenged by the lack of consistent molecular markers and clear discrimination criteria versus other (mural) cells. However, advances in single-cell approaches are uncovering and clarifying mural cell identities, biological functions, and ontogeny across organs. We discuss the latest developments in pericyte pathobiology to inform future research directions and potential outcomes.
Collapse
Affiliation(s)
- Hielke van Splunder
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Pilar Villacampa
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona and Bellvitge Biomedical Research Institute (IDIBELL), Carrer de la Feixa Llarga s/n, 08907 l'Hospitalet de Llobregat, Barcelona, Spain
| | - Anabel Martínez-Romero
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Mariona Graupera
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain; Institución Catalana de Investigación y Estudios Avanzados (ICREA), Passeig de Lluís Companys 23, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Avenida de Monforte de Lemos 5, 28029 Madrid, Spain.
| |
Collapse
|
39
|
Wilk CM, Cathomas F, Török O, Le Berichel J, Park MD, Bigenwald C, Heaton GR, Hamon P, Troncoso L, Scull BP, Dangoor D, Silvin A, Fleischmann R, Belabed M, Lin H, Merad Taouli E, Boettcher S, Li L, Aubry A, Manz MG, Kofler JK, Yue Z, Lira SA, Ginhoux F, Crary JF, McClain KL, Picarsic JL, Russo SJ, Allen CE, Merad M. Circulating senescent myeloid cells infiltrate the brain and cause neurodegeneration in histiocytic disorders. Immunity 2023; 56:2790-2802.e6. [PMID: 38091952 PMCID: PMC11587932 DOI: 10.1016/j.immuni.2023.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/05/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Neurodegenerative diseases (ND) are characterized by progressive loss of neuronal function. Mechanisms of ND pathogenesis are incompletely understood, hampering the development of effective therapies. Langerhans cell histiocytosis (LCH) is an inflammatory neoplastic disorder caused by hematopoietic progenitors expressing mitogen-activated protein kinase (MAPK)-activating mutations that differentiate into senescent myeloid cells that drive lesion formation. Some individuals with LCH subsequently develop progressive and incurable neurodegeneration (LCH-ND). Here, we showed that LCH-ND was caused by myeloid cells that were clonal with peripheral LCH cells. Circulating BRAFV600E+ myeloid cells caused the breakdown of the blood-brain barrier (BBB), enhancing migration into the brain parenchyma where they differentiated into senescent, inflammatory CD11a+ macrophages that accumulated in the brainstem and cerebellum. Blocking MAPK activity and senescence programs reduced peripheral inflammation, brain parenchymal infiltration, neuroinflammation, neuronal damage and improved neurological outcome in preclinical LCH-ND. MAPK activation and senescence programs in circulating myeloid cells represent targetable mechanisms of LCH-ND.
Collapse
Affiliation(s)
- C Matthias Wilk
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Flurin Cathomas
- Nash Family Department of Neuroscience, Brain & Body Research Center, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Orsolya Török
- Department of Neurology, University of Pécs, Medical School, Pécs, Hungary
| | - Jessica Le Berichel
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Camille Bigenwald
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - George R Heaton
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pauline Hamon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leanna Troncoso
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brooks P Scull
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Diana Dangoor
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Artificial Intelligence, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aymeric Silvin
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - Ryan Fleischmann
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Howard Lin
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Elias Merad Taouli
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steffen Boettcher
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Long Li
- Nash Family Department of Neuroscience, Brain & Body Research Center, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonio Aubry
- Nash Family Department of Neuroscience, Brain & Body Research Center, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Julia K Kofler
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhenyu Yue
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sergio A Lira
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florent Ginhoux
- Gustave Roussy Cancer Campus, Villejuif, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France
| | - John F Crary
- Department of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Artificial Intelligence, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuropathology Brain Bank and Research CoRE, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenneth L McClain
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer L Picarsic
- Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Brain & Body Research Center, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carl E Allen
- Texas Children's Cancer Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncology Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
40
|
Hu S, Yang B, Shu S, He X, Sang H, Fan X, Zhang H. Targeting Pericytes for Functional Recovery in Ischemic Stroke. Neuromolecular Med 2023; 25:457-470. [PMID: 37166748 DOI: 10.1007/s12017-023-08748-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: 02/24/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
Pericytes surrounding endothelial cells in the capillaries are emerging as an attractive cell resource, which can show a large variety of functions in ischemic stroke, including preservation of the blood-brain barrier, regulation of immune function, and support for cerebral vasculature. These functions have been fully elucidated in previous studies. However, in recent years, increasing evidence has shown that pericytes play an important role in neurological recovery after ischemic stroke due to their regenerative function which can be summarized in two aspects according to current discoveries, one is that pericytes are thought to be multipotential themselves, and the other is that pericytes can promote the differentiation of oligodendrocyte progenitor cells (OPCs). Considering the neuroprotective treatment for stroke has not been much progressed in recent years, new therapies targeting pericytes may be a future direction. Here, we will review the beneficial effects of pericytes in ischemic stroke from two directions: the barrier and vascular functions and the regenerative functions of pericytes.
Collapse
Affiliation(s)
- Shuqi Hu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Bingjie Yang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Song Shu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xudong He
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hongfei Sang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Xuemei Fan
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Hao Zhang
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
41
|
Teske NC, Dyckhoff-Shen S, Beckenbauer P, Bewersdorf JP, Engelen-Lee JY, Hammerschmidt S, Kälin RE, Pfister HW, Brouwer MC, Klein M, Glass R, van de Beek D, Koedel U. Pericytes are protective in experimental pneumococcal meningitis through regulating leukocyte infiltration and blood-brain barrier function. J Neuroinflammation 2023; 20:267. [PMID: 37978545 PMCID: PMC10655320 DOI: 10.1186/s12974-023-02938-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: 07/16/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Brain pericytes participate in the regulation of cerebral blood flow and the maintenance of blood-brain barrier integrity. Because of their perivascular localization, their receptor repertoire, and their potential ability to respond to inflammatory and infectious stimuli by producing various cytokines and chemokines, these cells are also thought to play an active role in the immune response to brain infections. This assumption is mainly supported by in vitro studies, investigations in in vivo disease models are largely missing. Here, we analysed the role of brain pericytes in pneumococcal meningitis, in vitro and in vivo in two animal models of pneumococcal meningitis. METHODS Primary murine and human pericytes were stimulated with increasing concentrations of different serotypes of Streptococcus pneumoniae in the presence or absence of Toll-like receptor inhibitors and their cell viability and cytokine production were monitored. To gain insight into the role of pericytes in brain infection in vivo, we performed studies in a zebrafish embryo model of pneumococcal meningitis in which pericytes were pharmacologically depleted. Furthermore, we analyzed the impact of genetically induced pericyte ablation on disease progression, intracranial complications, and brain inflammation in an adult mouse model of this disease. RESULTS Both murine and human pericytes reacted to pneumococcal exposure with the release of selected cytokines. This cytokine release is pneumolysin-dependent, TLR-dependent in murine (but not human) pericytes and can be significantly increased by macrophage-derived IL-1b. Pharmacological depletion of pericytes in zebrafish embryos resulted in increased cerebral edema and mortality due to pneumococcal meningitis. Correspondingly, in an adult mouse meningitis model, a more pronounced blood-brain barrier disruption and leukocyte infiltration, resulting in an unfavorable disease course, was observed following genetic pericyte ablation. The degree of leukocyte infiltration positively correlated with an upregulation of chemokine expression in the brains of pericyte-depleted mice. CONCLUSIONS Our findings show that pericytes play a protective role in pneumococcal meningitis by impeding leukocyte migration and preventing blood-brain barrier breaching. Thus, preserving the integrity of the pericyte population has the potential as a new therapeutic strategy in pneumococcal meningitis.
Collapse
Affiliation(s)
- Nina C Teske
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany.
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland.
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany.
| | | | - Paul Beckenbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Joo-Yeon Engelen-Lee
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Roland E Kälin
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Hans-Walter Pfister
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| | - Matthijs C Brouwer
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Matthias Klein
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Diederik van de Beek
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Uwe Koedel
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| |
Collapse
|
42
|
Yuan H, Sun D, Ji Y, Meng B, Lu B, Liu R, Xing X, Wang R, Chen J. Pericyte loss impairs the blood-brain barrier and cognitive function in aged mice after anesthesia/surgery. Brain Res Bull 2023; 204:110799. [PMID: 38867419 DOI: 10.1016/j.brainresbull.2023.110799] [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: 07/11/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 06/14/2024]
Abstract
AIMS This study was designed to investigate the role of pericytes in the pathogenesis of perioperative neurocognitive disorder (PND). METHODS In this study, we established a PND model via sevoflurane anesthesia and tibial fracture surgery in 2-month-old and 16-month-old male C57BL/6 mice. On the third postoperative day, the mice were subjected to behavioral testing or sacrificed to collect brain tissue. The progression of hippocampal blood-brain barrier (BBB) disruption and neuroinflammation was detected using transmission electron microscope and immunofluorescence. We also used western blotting to measure the levels of plasma-derived protein immunoglobulin G (IgG) and albumin in the hippocampus to assess the leakage of the BBB. RESULTS Aged mice did not experience age-related cognitive decline and BBB disruption compared with younger mice but only increased glial cell activity. Anesthesia/Surgery damaged cognitive function, reduced pericyte coverage, decreased the length of capillaries and levels of occludin and claudin-5, destroyed the structure of the BBB, exacerbated IgG and albumin accumulation in the hippocampus, and enhanced the activation of microglia and astrocytes in the hippocampus of aged mice. However, these negative effects did not occur in young mice. CONCLUSION Our study showed that the loss of pericytes led to increased BBB permeability and neuroinflammation after anesthesia/surgery in aged mice, ultimately resulting in cognitive dysfunction.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Anesthesiology, Ningbo NO. 2 Hospital, Ningbo 315010, China; Department of Pain, Ningbo NO. 2 Hospital, Ningbo 315010, China
| | - Daofan Sun
- Department of Anesthesiology, Ningbo NO. 2 Hospital, Ningbo 315010, China
| | - Yiqin Ji
- Department of Anesthesiology, The First Affiliated Hospital of Ningbo University, Ningbo 315010, China
| | - Bo Meng
- Department of Anesthesiology, Ningbo NO. 2 Hospital, Ningbo 315010, China; Department of Pain, Ningbo NO. 2 Hospital, Ningbo 315010, China
| | - Bo Lu
- Department of Anesthesiology, Ningbo NO. 2 Hospital, Ningbo 315010, China
| | - Rongjun Liu
- Department of Anesthesiology, Ningbo NO. 2 Hospital, Ningbo 315010, China
| | - Xiuzhong Xing
- Department of Anesthesiology, Ningbo NO. 2 Hospital, Ningbo 315010, China
| | - Ruichun Wang
- Department of Anesthesiology, Ningbo NO. 2 Hospital, Ningbo 315010, China
| | - Junping Chen
- Department of Anesthesiology, Ningbo NO. 2 Hospital, Ningbo 315010, China; Department of Pain, Ningbo NO. 2 Hospital, Ningbo 315010, China.
| |
Collapse
|
43
|
Li W, Niu X, Dai Y, Wu X, Li J, Sheng W. Rnf-213 Knockout Induces Pericyte Reduction and Blood-Brain Barrier Impairment in Mouse. Mol Neurobiol 2023; 60:6188-6200. [PMID: 37438553 DOI: 10.1007/s12035-023-03480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 06/30/2023] [Indexed: 07/14/2023]
Abstract
Moyamoya disease (MMD) is a rare cerebrovascular disorder characterized by progressive occlusion of the internal carotid artery and the formation of an abnormal compensatory capillary network at the base of the brain. Genomics studies identified Ring finger protein 213 (RNF213) as a common genetic factor that increases the susceptibility to MMD in East Asian people. However, the function of RNF213 and its roles in pathogenesis of MMD is unclear. Here, we showed that genetic knockout of Rnf213 in mice causes significant pericyte reduction and blood-brain barrier impairment in the cortex. These phenotypes are accompanied with microglia activation and elevated level of proinflammatory cytokines. Additionally, Rnf213-deficient mice showed reduced expression of tight junction proteins, including Occludin, Claudin-5, and ZO-1. Together, these data suggested that RNF213 might contribute to the pathogenesis of MMD through disruption of pericyte homeostasis and blood-brain barrier integrity by dysregulation of inflammatory responses and tight junction formation.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xingyang Niu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuanyuan Dai
- Department of Neurology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Xiaoxin Wu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jiaoxing Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wenli Sheng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
44
|
Wilk CM, Cathomas F, Török O, Le Berichel J, Park MD, Heaton GR, Hamon P, Troncoso L, Scull BP, Dangoor D, Silvin A, Fleischmann R, Belabed M, Lin H, Taouli EM, Boettcher S, Manz MG, Kofler JK, Yue Z, Lira SA, Ginhoux F, Crary JF, McClain KL, Picarsic JL, Russo SJ, Allen CE, Merad M. Circulating senescent myeloid cells drive blood brain barrier breakdown and neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.10.561744. [PMID: 37873371 PMCID: PMC10592746 DOI: 10.1101/2023.10.10.561744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Neurodegenerative diseases (ND) are characterized by progressive loss of neuronal function. Mechanisms of ND pathogenesis are incompletely understood, hampering the development of effective therapies. Langerhans cell histiocytosis (LCH) is an inflammatory neoplastic disorder caused by hematopoietic progenitors expressing MAPK activating mutations that differentiate into senescent myeloid cells that drive lesion formation. Some patients with LCH subsequently develop progressive and incurable neurodegeneration (LCH-ND). Here, we show that LCH-ND is caused by myeloid cells that are clonal with peripheral LCH cells. We discovered that circulating BRAF V600E + myeloid cells cause the breakdown of the blood-brain barrier (BBB), enhancing migration into the brain parenchyma where they differentiate into senescent, inflammatory CD11a + macrophages that accumulate in the brainstem and cerebellum. Blocking MAPK activity and senescence programs reduced parenchymal infiltration, neuroinflammation, neuronal damage and improved neurological outcome in preclinical LCH-ND. MAPK activation and senescence programs in circulating myeloid cells represent novel and targetable mechanisms of ND.
Collapse
|
45
|
He S, Peng Y, Chen X, Ou Y. Causality between inflammatory bowel disease and the cerebral cortex: insights from Mendelian randomization and integrated bioinformatics analysis. Front Immunol 2023; 14:1175873. [PMID: 37588593 PMCID: PMC10425804 DOI: 10.3389/fimmu.2023.1175873] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/06/2023] [Indexed: 08/18/2023] Open
Abstract
Background Inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD), is a chronic, progressive, and recurrent intestinal condition that poses a significant global health burden. The high prevalence of neuropsychiatric comorbidities in IBD necessitates the development of targeted management strategies. Methods Leveraging genetic data from genome-wide association studies and Immunochip genotype analyses of nearly 150,000 individuals, we conducted a two-sample Mendelian randomization study to elucidate the driving force of IBD, UC, and CD on cortical reshaping. Genetic variants mediating the causality were collected to disclose the biological pathways linking intestinal inflammation to brain dysfunction. Results Here, 115, 69, and 98 instrumental variables genetically predicted IBD, UC, and CD. We found that CD significantly decreased the surface area of the temporal pole gyrus (β = -0.946 mm2, P = 0.005, false discovery rate-P = 0.085). Additionally, we identified suggestive variations in cortical surface area and thickness induced by exposure across eight functional gyri. The top 10 variant-matched genes were STAT3, FOS, NFKB1, JAK2, STAT4, TYK2, SMAD3, IL12B, MYC, and CCL2, which are interconnected in the interaction network and play a role in inflammatory and immune processes. Conclusion We explore the causality between intestinal inflammation and altered cortical morphology. It is likely that neuroinflammation-induced damage, impaired neurological function, and persistent nociceptive input lead to morphological changes in the cerebral cortex, which may trigger neuropsychiatric disorders.
Collapse
Affiliation(s)
- Shubei He
- Department of Gastroenterology, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Institute of Digestive Diseases of the People's Liberation Army, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Cholestatic Liver Diseases Center, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| | - Ying Peng
- Department of Gastroenterology, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Institute of Digestive Diseases of the People's Liberation Army, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Cholestatic Liver Diseases Center, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiaofang Chen
- Department of Gastroenterology, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Institute of Digestive Diseases of the People's Liberation Army, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Cholestatic Liver Diseases Center, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
- Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing, China
| | - Ying Ou
- Department of Psychiatry, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
46
|
Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 2023; 8:267. [PMID: 37433768 PMCID: PMC10336149 DOI: 10.1038/s41392-023-01486-5] [Citation(s) in RCA: 456] [Impact Index Per Article: 228.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 07/13/2023] Open
Abstract
Studies in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Amyotrophic lateral sclerosis, Huntington's disease, and so on, have suggested that inflammation is not only a result of neurodegeneration but also a crucial player in this process. Protein aggregates which are very common pathological phenomenon in neurodegeneration can induce neuroinflammation which further aggravates protein aggregation and neurodegeneration. Actually, inflammation even happens earlier than protein aggregation. Neuroinflammation induced by genetic variations in CNS cells or by peripheral immune cells may induce protein deposition in some susceptible population. Numerous signaling pathways and a range of CNS cells have been suggested to be involved in the pathogenesis of neurodegeneration, although they are still far from being completely understood. Due to the limited success of traditional treatment methods, blocking or enhancing inflammatory signaling pathways involved in neurodegeneration are considered to be promising strategies for the therapy of neurodegenerative diseases, and many of them have got exciting results in animal models or clinical trials. Some of them, although very few, have been approved by FDA for clinical usage. Here we comprehensively review the factors affecting neuroinflammation and the major inflammatory signaling pathways involved in the pathogenicity of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic lateral sclerosis. We also summarize the current strategies, both in animal models and in the clinic, for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Weifeng Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China
| | - Dan Xiao
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, P.R. China
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China.
| |
Collapse
|
47
|
Inoue Y, Shue F, Bu G, Kanekiyo T. Pathophysiology and probable etiology of cerebral small vessel disease in vascular dementia and Alzheimer's disease. Mol Neurodegener 2023; 18:46. [PMID: 37434208 PMCID: PMC10334598 DOI: 10.1186/s13024-023-00640-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is commonly caused by vascular injuries in cerebral large and small vessels and is a key driver of age-related cognitive decline. Severe VCID includes post-stroke dementia, subcortical ischemic vascular dementia, multi-infarct dementia, and mixed dementia. While VCID is acknowledged as the second most common form of dementia after Alzheimer's disease (AD) accounting for 20% of dementia cases, VCID and AD frequently coexist. In VCID, cerebral small vessel disease (cSVD) often affects arterioles, capillaries, and venules, where arteriolosclerosis and cerebral amyloid angiopathy (CAA) are major pathologies. White matter hyperintensities, recent small subcortical infarcts, lacunes of presumed vascular origin, enlarged perivascular space, microbleeds, and brain atrophy are neuroimaging hallmarks of cSVD. The current primary approach to cSVD treatment is to control vascular risk factors such as hypertension, dyslipidemia, diabetes, and smoking. However, causal therapeutic strategies have not been established partly due to the heterogeneous pathogenesis of cSVD. In this review, we summarize the pathophysiology of cSVD and discuss the probable etiological pathways by focusing on hypoperfusion/hypoxia, blood-brain barriers (BBB) dysregulation, brain fluid drainage disturbances, and vascular inflammation to define potential diagnostic and therapeutic targets for cSVD.
Collapse
Affiliation(s)
- Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Guojun Bu
- SciNeuro Pharmaceuticals, Rockville, MD 20850 USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| |
Collapse
|
48
|
Huseby CJ, Delvaux E, Brokaw DL, Coleman PD. Blood RNA transcripts reveal similar and differential alterations in fundamental cellular processes in Alzheimer's disease and other neurodegenerative diseases. Alzheimers Dement 2023; 19:2618-2632. [PMID: 36541444 PMCID: PMC11633037 DOI: 10.1002/alz.12880] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Dysfunctional processes in Alzheimer's disease and other neurodegenerative diseases lead to neural degeneration in the central and peripheral nervous system. Research demonstrates that neurodegeneration of any kind is a systemic disease that may even begin outside of the region vulnerable to the disease. Neurodegenerative diseases are defined by the vulnerabilities and pathology occurring in the regions affected. METHOD A random forest machine learning analysis on whole blood transcriptomes from six neurodegenerative diseases generated unbiased disease-classifying RNA transcripts subsequently subjected to pathway analysis. RESULTS We report that transcripts of the blood transcriptome selected for each of the neurodegenerative diseases represent fundamental biological cell processes including transcription regulation, degranulation, immune response, protein synthesis, apoptosis, cytoskeletal components, ubiquitylation/proteasome, and mitochondrial complexes that are also affected in the brain and reveal common themes across six neurodegenerative diseases. CONCLUSION Neurodegenerative diseases share common dysfunctions in fundamental cellular processes. Identifying regional vulnerabilities will reveal unique disease mechanisms. HIGHLIGHTS Transcriptomics offer information about dysfunctional processes. Comparing multiple diseases will expose unique malfunctions within diseases. Blood RNA can be used ante mortem to track expression changes in neurodegenerative diseases. Protocol standardization will make public datasets compatible.
Collapse
Affiliation(s)
- Carol J. Huseby
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Elaine Delvaux
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Danielle L. Brokaw
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Paul D. Coleman
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
49
|
Benarroch E. What Are the Roles of Pericytes in the Neurovascular Unit and Its Disorders? Neurology 2023; 100:970-977. [PMID: 37188542 PMCID: PMC10186232 DOI: 10.1212/wnl.0000000000207379] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/15/2023] [Indexed: 05/17/2023] Open
|
50
|
Yao B, Delaidelli A, Vogel H, Sorensen PH. Pediatric Brain Tumours: Lessons from the Immune Microenvironment. Curr Oncol 2023; 30:5024-5046. [PMID: 37232837 PMCID: PMC10217418 DOI: 10.3390/curroncol30050379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/01/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
In spite of recent advances in tumour molecular subtyping, pediatric brain tumours (PBTs) remain the leading cause of cancer-related deaths in children. While some PBTs are treatable with favourable outcomes, recurrent and metastatic disease for certain types of PBTs remains challenging and is often fatal. Tumour immunotherapy has emerged as a hopeful avenue for the treatment of childhood tumours, and recent immunotherapy efforts have been directed towards PBTs. This strategy has the potential to combat otherwise incurable PBTs, while minimizing off-target effects and long-term sequelae. As the infiltration and activation states of immune cells, including tumour-infiltrating lymphocytes and tumour-associated macrophages, are key to shaping responses towards immunotherapy, this review explores the immune landscape of the developing brain and discusses the tumour immune microenvironments of common PBTs, with hopes of conferring insights that may inform future treatment design.
Collapse
Affiliation(s)
- Betty Yao
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
| | - Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hannes Vogel
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Poul H. Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; (B.Y.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|