1
|
Gopalakrishnan B, Galili U, Saenger M, Burket NJ, Koss W, Lokender MS, Wolfe KM, Husak SJ, Stark CJ, Solorio L, Cox A, Dunbar A, Shi R, Li J. α-Gal Nanoparticles in CNS Trauma: II. Immunomodulation Following Spinal Cord Injury (SCI) Improves Functional Outcomes. Tissue Eng Regen Med 2024; 21:437-453. [PMID: 38308742 PMCID: PMC10987462 DOI: 10.1007/s13770-023-00616-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: 07/06/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Previous investigations have shown that local application of nanoparticles presenting the carbohydrate moiety galactose-α-1,3-galactose (α-gal epitopes) enhance wound healing by activating the complement system and recruiting pro-healing macrophages to the injury site. Our companion in vitro paper suggest α-gal epitopes can similarly recruit and polarize human microglia toward a pro-healing phenotype. In this continuation study, we investigate the in vivo implications of α-gal nanoparticle administration directly to the injured spinal cord. METHODS α-Gal knock-out (KO) mice subjected to spinal cord crush were injected either with saline (control) or with α-gal nanoparticles immediately following injury. Animals were assessed longitudinally with neurobehavioral and histological endpoints. RESULTS Mice injected with α-gal nanoparticles showed increased recruitment of anti-inflammatory macrophages to the injection site in conjunction with increased production of anti-inflammatory markers and a reduction in apoptosis. Further, the treated group showed increased axonal infiltration into the lesion, a reduction in reactive astrocyte populations and increased angiogenesis. These results translated into improved sensorimotor metrics versus the control group. CONCLUSIONS Application of α-gal nanoparticles after spinal cord injury (SCI) induces a pro-healing inflammatory response resulting in neuroprotection, improved axonal ingrowth into the lesion and enhanced sensorimotor recovery. The data shows α-gal nanoparticles may be a promising avenue for further study in CNS trauma.
Collapse
Affiliation(s)
- Bhavani Gopalakrishnan
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Uri Galili
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Megan Saenger
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Noah J Burket
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Wendy Koss
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Manjari S Lokender
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Kaitlyn M Wolfe
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Samantha J Husak
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Collin J Stark
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
| | - Abigail Cox
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, 47907, USA
| | - August Dunbar
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
| | - Riyi Shi
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Jianming Li
- Center for Paralysis Research, Purdue University, West Lafayette, IN, 47907, USA.
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
2
|
Ma Q, Chen J, Kong X, Zeng Y, Chen Z, Liu H, Liu L, Lu S, Wang X. Interactions between CNS and immune cells in tuberculous meningitis. Front Immunol 2024; 15:1326859. [PMID: 38361935 PMCID: PMC10867975 DOI: 10.3389/fimmu.2024.1326859] [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: 10/24/2023] [Accepted: 01/10/2024] [Indexed: 02/17/2024] Open
Abstract
The central nervous system (CNS) harbors its own special immune system composed of microglia in the parenchyma, CNS-associated macrophages (CAMs), dendritic cells, monocytes, and the barrier systems within the brain. Recently, advances in the immune cells in the CNS provided new insights to understand the development of tuberculous meningitis (TBM), which is the predominant form of Mycobacterium tuberculosis (M.tb) infection in the CNS and accompanied with high mortality and disability. The development of the CNS requires the protection of immune cells, including macrophages and microglia, during embryogenesis to ensure the accurate development of the CNS and immune response following pathogenic invasion. In this review, we summarize the current understanding on the CNS immune cells during the initiation and development of the TBM. We also explore the interactions of immune cells with the CNS in TBM. In the future, the combination of modern techniques should be applied to explore the role of immune cells of CNS in TBM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shuihua Lu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Xiaomin Wang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Calder PC, Bach-Faig A, Bevacqua T, Caballero Lopez CG, Chen ZY, Connolly D, Koay WL, Meydani SN, Pinar AS, Ribas-Filho D, Pierre A. Vital role for primary healthcare providers: urgent need to educate the community about daily nutritional self-care to support immune function and maintain health. BMJ Nutr Prev Health 2023; 6:392-401. [PMID: 38618551 PMCID: PMC11009526 DOI: 10.1136/bmjnph-2023-000755] [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: 08/21/2023] [Accepted: 11/22/2023] [Indexed: 04/16/2024] Open
Abstract
The importance of self-care to improve health and social well-being is well recognised. Nevertheless, there remains a need to encourage people to better understand how their body works, and how to keep it healthy. Because of its important role, part of this understanding should be based on why the immune system must be supported. This highly complex system is essential for defending against pathogens, but also for maintaining health throughout the body by preserving homeostasis and integrity. Accordingly, the immune system requires active management for optimal functioning and to reduce the risk of chronic diseases. In addition to regular exercise, healthy sleeping patterns, cultivating mental resilience, adequate nutrition through healthy and diverse dietary habits is key to the daily support of immune function. Diet and the immune system are closely intertwined, and a poor diet will impair immunity and increase the risk of acute and chronic diseases. To help elucidate the roles of primary healthcare providers in supporting individuals to engage in self-care, an international group of experts reviewed the evidence for the roles of the immune system in maintaining health and for nutrition in daily immune support, and discussed implications for population health and clinical practice.
Collapse
Affiliation(s)
- Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Reseaech Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Anna Bach-Faig
- Faculty of Health Sciences, Open University of Catalonia, Barcelona, Spain
- Food and Nutrition Area, Barcelona Official College of Pharmacists, Barcelona, Spain
| | | | | | - Zheng-Yu Chen
- International Pharmaceutical Federation, Shanghai, China
| | | | | | - Simin N Meydani
- Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, USA
| | | | - Durval Ribas-Filho
- Padre Albino Foundation, Faculty of Medicine, Catanduva, São Paulo, Brazil
| | | |
Collapse
|
4
|
Shang J, Jiang C, Cai J, Chen Z, Jin S, Wang F, Liang W, Wang Y, Zhang S, Hu X. Knowledge Mapping of Macrophage in Spinal Cord Injury: A Bibliometric Analysis. World Neurosurg 2023; 180:e183-e197. [PMID: 37714458 DOI: 10.1016/j.wneu.2023.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND Spinal cord injury (SCI) is a devastating condition, often leading to significant disability and impairment. As crucial immune cells, macrophages play a critical role in the pathophysiology of SCI. Understanding the current state of knowledge and research trends related to macrophages in SCI is crucial for developing effective therapeutic interventions. METHODS Using search strategies, we retrieved relevant articles from the Web of Science Core Collection (WOSCC), resulting in a robust dataset for analysis. VOSviewer, Citespace, and PRISM were employed for analysis and visualization. Various bibliometric indicators, including publication trends, citation analysis, co-authorship networks, and keyword analysis, were utilized to assess the scholarly landscape of macrophage research in SCI. RESULTS Our findings revealed a steady increase in publications over the past 33 years, indicating a growing interest in this field. We identified Popovich Phillip G was the most influential author, Ohio State University was the most influential institution, and identification of 2 distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord was the most influential paper in this field. CONCLUSIONS This bibliometric analysis provides a comprehensive overview of the current knowledge landscape and research trends regarding macrophages in SCI. Neuroinflammation and macrophage polarization, transplation and molecular mechanism were emerging research areas and novel directions. Our study serves as a valuable resource for researchers in spinal cord injury research and therapeutic development.
Collapse
Affiliation(s)
- JinXiang Shang
- Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing City, China
| | - Chao Jiang
- Orthopedics, Shaoxing People's Hospital, Shaoxing City, China
| | - Junhui Cai
- Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing City, China
| | - ZhuoLin Chen
- Orthopedics, Shaoxing People's Hospital, Shaoxing City, China
| | - SongTao Jin
- Orthopedics, Shaoxing People's Hospital, Shaoxing City, China
| | - Fei Wang
- Orthopedics, Shaoxing People's Hospital, Shaoxing City, China
| | - WenQing Liang
- Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine, Zhoushan City, China
| | - YaQing Wang
- Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing City, China
| | - SongOu Zhang
- School of Medicine, Ningbo University, Ningbo City, China
| | - Xujun Hu
- Orthopedics, Shaoxing People's Hospital, Shaoxing City, China.
| |
Collapse
|
5
|
Salvador AFM, Dykstra T, Rustenhoven J, Gao W, Blackburn SM, Bhasiin K, Dong MQ, Guimarães RM, Gonuguntla S, Smirnov I, Kipnis J, Herz J. Age-dependent immune and lymphatic responses after spinal cord injury. Neuron 2023; 111:2155-2169.e9. [PMID: 37148871 PMCID: PMC10523880 DOI: 10.1016/j.neuron.2023.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 02/13/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
Spinal cord injury (SCI) causes lifelong debilitating conditions. Previous works demonstrated the essential role of the immune system in recovery after SCI. Here, we explored the temporal changes of the response after SCI in young and aged mice in order to characterize multiple immune populations within the mammalian spinal cord. We revealed substantial infiltration of myeloid cells to the spinal cord in young animals, accompanied by changes in the activation state of microglia. In contrast, both processes were blunted in aged mice. Interestingly, we discovered the formation of meningeal lymphatic structures above the lesion site, and their role has not been examined after contusive injury. Our transcriptomic data predicted lymphangiogenic signaling between myeloid cells in the spinal cord and lymphatic endothelial cells (LECs) in the meninges after SCI. Together, our findings delineate how aging affects the immune response following SCI and highlight the participation of the spinal cord meninges in supporting vascular repair.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Taitea Dykstra
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Justin Rustenhoven
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland 1023, New Zealand
| | - Wenqing Gao
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Susan M Blackburn
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kesshni Bhasiin
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Michael Q Dong
- Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Rafaela Mano Guimarães
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Sriharsha Gonuguntla
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Igor Smirnov
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| | - Jasmin Herz
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
6
|
Floriddia E. In conversation with Michal Schwartz. Nat Neurosci 2023; 26:715-718. [PMID: 37156871 DOI: 10.1038/s41593-023-01318-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
|
7
|
Castellani G, Croese T, Peralta Ramos JM, Schwartz M. Transforming the understanding of brain immunity. Science 2023; 380:eabo7649. [PMID: 37023203 DOI: 10.1126/science.abo7649] [Citation(s) in RCA: 86] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Contemporary studies have completely changed the view of brain immunity from envisioning the brain as isolated and inaccessible to peripheral immune cells to an organ in close physical and functional communication with the immune system for its maintenance, function, and repair. Circulating immune cells reside in special niches in the brain's borders, the choroid plexus, meninges, and perivascular spaces, from which they patrol and sense the brain in a remote manner. These niches, together with the meningeal lymphatic system and skull microchannels, provide multiple routes of interaction between the brain and the immune system, in addition to the blood vasculature. In this Review, we describe current ideas about brain immunity and their implications for brain aging, diseases, and immune-based therapeutic approaches.
Collapse
Affiliation(s)
- Giulia Castellani
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Tommaso Croese
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
8
|
Sterner RC, Sterner RM. Immune response following traumatic spinal cord injury: Pathophysiology and therapies. Front Immunol 2023; 13:1084101. [PMID: 36685598 PMCID: PMC9853461 DOI: 10.3389/fimmu.2022.1084101] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating condition that is often associated with significant loss of function and/or permanent disability. The pathophysiology of SCI is complex and occurs in two phases. First, the mechanical damage from the trauma causes immediate acute cell dysfunction and cell death. Then, secondary mechanisms of injury further propagate the cell dysfunction and cell death over the course of days, weeks, or even months. Among the secondary injury mechanisms, inflammation has been shown to be a key determinant of the secondary injury severity and significantly worsens cell death and functional outcomes. Thus, in addition to surgical management of SCI, selectively targeting the immune response following SCI could substantially decrease the progression of secondary injury and improve patient outcomes. In order to develop such therapies, a detailed molecular understanding of the timing of the immune response following SCI is necessary. Recently, several studies have mapped the cytokine/chemokine and cell proliferation patterns following SCI. In this review, we examine the immune response underlying the pathophysiology of SCI and assess both current and future therapies including pharmaceutical therapies, stem cell therapy, and the exciting potential of extracellular vesicle therapy.
Collapse
Affiliation(s)
- Robert C. Sterner
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Rosalie M. Sterner
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States,*Correspondence: Rosalie M. Sterner,
| |
Collapse
|
9
|
Zhao J, Wang X, He Y, Xu P, Lai L, Chung Y, Pan X. The Role of T Cells in Alzheimer's Disease Pathogenesis. Crit Rev Immunol 2023; 43:15-23. [PMID: 37943150 DOI: 10.1615/critrevimmunol.2023050145] [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: 11/10/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder associated with memory decline and cognitive impairment, which is related to hallmark protein aggregates, amyloid-β (Аβ) plaques and neurofibrillary tangles; the latter are accumulated with hyperphosphorylated Tau protein. Immune cells play an important role in AD pathogenesis. Although the role of T cells in AD remains controversial, studies have shown that T cell deficiency is associated with increased AD pathology. In contrast, transplantation of T cells reduces AD pathology. T cells can help B cells generate anti-Аβ antibody to neutralize the toxin of Аβ and hyperphosphorylated Tau. T cells also activate macrophages to phagocytose misfolded proteins including Аβ and Tau. Recent data have also shown that AD animals have a damaged thymic microenvironment, especially thymic epithelial cells (TECs), resulting in decreased T cell numbers, which contribute to AD pathology. Therefore, regulation of T cell regeneration, for example by rejuvenating the thymic microenvironment, has the potential to be used in the treatment of AD.
Collapse
Affiliation(s)
- Jin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, 510515, China; ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangdong, 510515, China
| | - Xiaofang Wang
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT 06030, USA
| | - Yusheng He
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA; University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA
| | - Younggie Chung
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT 06030, USA
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangdong, 510515, China
| |
Collapse
|
10
|
Peralta Ramos JM, Kviatcovsky D, Schwartz M. Targeting the immune system towards novel therapeutic avenues to fight brain aging and neurodegeneration. Eur J Neurosci 2022; 56:5413-5427. [PMID: 35075702 DOI: 10.1111/ejn.15609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
The incidence of age-related dementia is growing with increased longevity, yet there are currently no disease-modifying therapies for these devastating disorders. Studies over the last several years have led to an evolving awareness of the role of the immune system in supporting brain maintenance and repair, displaying a diverse repertoire of functions while orchestrating the crosstalk between the periphery and the brain. Here, we provide insights into the current understanding of therapeutic targets that could be adopted to modulate immune cell fate, either systemically or locally, to defeat brain aging and neurodegeneration.
Collapse
Affiliation(s)
| | - Denise Kviatcovsky
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
11
|
Kühl B, Beyerbach M, Baumgärtner W, Gerhauser I. Characterization of microglia/macrophage phenotypes in the spinal cord following intervertebral disc herniation. Front Vet Sci 2022; 9:942967. [PMID: 36262531 PMCID: PMC9574228 DOI: 10.3389/fvets.2022.942967] [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: 05/13/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Dogs frequently suffer from traumatic spinal cord injury (SCI). Most cases of SCI have a favorable prognosis but 40-50% of dogs with paraplegia and absence of nociception do not regain ambulatory abilities, eventually leading to euthanasia. Microglia and infiltrating macrophages play a crucial role in inflammatory process after SCI. However, little is known about microglia/macrophage phenotypes representing a potential target for future therapeutic strategies. In the present study, the microglia/macrophage phenotype was characterized by immunohistochemistry in the morphologically unaltered canine spinal cord (10 control dogs) and during acute and subacute SCI (1-4 and 5-10 days post injury, 9 and 8 dogs, respectively) using antibodies directed against IBA1, MAC387, MHC-II, lysozyme, EGR2, myeloperoxidase, CD18, CD204 and lectin from Griffonia simplicifolia (BS-1). The expression of these markers was also analyzed in the spleen as reference for the phenotype of histiocytic cells. Histological lesions were absent in controls. In acute SCI, 4 dogs showed mild to moderate hemorrhages, 2 dogs bilateral gray matter necrosis and 6 dogs mild multifocal axonal swellings and myelin sheath dilation. One dog with acute SCI did not show histological alterations except for few dilated myelin sheaths. In subacute SCI, variable numbers of gitter cells, axonal changes and dilated myelin sheaths were present in all dogs and large areas of tissue necrosis in 2 dogs. Neuronal chromatolysis was found in 3 dogs with acute and subacute SCI, respectively. In control dogs, microglia/macrophage constitutively expressed IBA1 and rarely other markers. In acute SCI, a similar marker expression was found except for an increase in MAC387-positive cells in the spinal cord white matter due to an infiltration of few blood-borne macrophages. In subacute SCI, increased numbers of microglia/macrophages expressed CD18, CD204 and MHC-II in the gray matter SCI indicating enhanced antigen recognition, processing and presentation as well as cell migration and phagocytosis during this stage. Interestingly, only CD204-positive cells were upregulated in the white matter, which might be related to gray-white matter heterogeneity of microglia as previously described in humans. The present findings contribute to the understanding of the immunological processes during SCI in a large animal model for human SCI.
Collapse
Affiliation(s)
- Bianca Kühl
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Martin Beyerbach
- Institute for Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany,*Correspondence: Wolfgang Baumgärtner
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
12
|
Nadine S, Correia CR, Mano JF. Engineering immunomodulatory hydrogels and cell-laden systems towards bone regeneration. BIOMATERIALS ADVANCES 2022; 140:213058. [PMID: 35933955 DOI: 10.1016/j.bioadv.2022.213058] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
The well-known synergetic interplay between the skeletal and immune systems has changed the design of advanced bone tissue engineering strategies. The immune system is essential during the bone lifetime, with macrophages playing multiple roles in bone healing and biomaterial integration. If in the past, the most valuable aspect of implants was to avoid immune responses of the host, nowadays, it is well-established how important are the crosstalks between immune cells and bone-engineered niches for an efficient regenerative process to occur. For that, it is essential to recapitulate the multiphenotypic cellular environment of bone tissue when designing new approaches. Indeed, the lack of osteoimmunomodulatory knowledge may be the explanation for the poor translation of biomaterials into clinical practice. Thus, smarter hydrogels incorporating immunomodulatory bioactive factors, stem cells, and immune cells are being proposed to develop a new generation of bone tissue engineering strategies. This review highlights the power of immune cells to upgrade the development of innovative engineered strategies, mainly focusing on orthopaedic and dental applications.
Collapse
Affiliation(s)
- Sara Nadine
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Clara R Correia
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João F Mano
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
13
|
Helmreich DL. Profiles of women in science: Michal Schwartz, Professor of Neuroimmunology, at the Weizmann Institute of Science, Rehovot, Israel. Eur J Neurosci 2022; 56:4271-4274. [PMID: 35801336 DOI: 10.1111/ejn.15765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Dana L Helmreich
- The Ernest J. Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
14
|
Schwartz M, Cahalon L. The vicious cycle governing the brain–immune system relationship in neurodegenerative diseases. Curr Opin Immunol 2022; 76:102182. [DOI: 10.1016/j.coi.2022.102182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/11/2022]
|
15
|
Cell-based and stem-cell-based treatments for spinal cord injury: evidence from clinical trials. Lancet Neurol 2022; 21:659-670. [DOI: 10.1016/s1474-4422(21)00464-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/01/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022]
|
16
|
Progression in translational research on spinal cord injury based on microenvironment imbalance. Bone Res 2022; 10:35. [PMID: 35396505 PMCID: PMC8993811 DOI: 10.1038/s41413-022-00199-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 11/14/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) leads to loss of motor and sensory function below the injury level and imposes a considerable burden on patients, families, and society. Repair of the injured spinal cord has been recognized as a global medical challenge for many years. Significant progress has been made in research on the pathological mechanism of spinal cord injury. In particular, with the development of gene regulation, cell sequencing, and cell tracing technologies, in-depth explorations of the SCI microenvironment have become more feasible. However, translational studies related to repair of the injured spinal cord have not yielded significant results. This review summarizes the latest research progress on two aspects of SCI pathology: intraneuronal microenvironment imbalance and regenerative microenvironment imbalance. We also review repair strategies for the injured spinal cord based on microenvironment imbalance, including medications, cell transplantation, exosomes, tissue engineering, cell reprogramming, and rehabilitation. The current state of translational research on SCI and future directions are also discussed. The development of a combined, precise, and multitemporal strategy for repairing the injured spinal cord is a potential future direction.
Collapse
|
17
|
Patsalos A, Halasz L, Medina-Serpas MA, Berger WK, Daniel B, Tzerpos P, Kiss M, Nagy G, Fischer C, Simandi Z, Varga T, Nagy L. A growth factor-expressing macrophage subpopulation orchestrates regenerative inflammation via GDF-15. J Exp Med 2022; 219:e20210420. [PMID: 34846534 PMCID: PMC8635277 DOI: 10.1084/jem.20210420] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/03/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Muscle regeneration is the result of the concerted action of multiple cell types driven by the temporarily controlled phenotype switches of infiltrating monocyte-derived macrophages. Pro-inflammatory macrophages transition into a phenotype that drives tissue repair through the production of effectors such as growth factors. This orchestrated sequence of regenerative inflammatory events, which we termed regeneration-promoting program (RPP), is essential for proper repair. However, it is not well understood how specialized repair-macrophage identity develops in the RPP at the transcriptional level and how induced macrophage-derived factors coordinate tissue repair. Gene expression kinetics-based clustering of blood circulating Ly6Chigh, infiltrating inflammatory Ly6Chigh, and reparative Ly6Clow macrophages, isolated from injured muscle, identified the TGF-β superfamily member, GDF-15, as a component of the RPP. Myeloid GDF-15 is required for proper muscle regeneration following acute sterile injury, as revealed by gain- and loss-of-function studies. Mechanistically, GDF-15 acts both on proliferating myoblasts and on muscle-infiltrating myeloid cells. Epigenomic analyses of upstream regulators of Gdf15 expression identified that it is under the control of nuclear receptors RXR/PPARγ. Finally, immune single-cell RNA-seq profiling revealed that Gdf15 is coexpressed with other known muscle regeneration-associated growth factors, and their expression is limited to a unique subpopulation of repair-type macrophages (growth factor-expressing macrophages [GFEMs]).
Collapse
Affiliation(s)
- Andreas Patsalos
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Laszlo Halasz
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Miguel A. Medina-Serpas
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Wilhelm K. Berger
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Bence Daniel
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Petros Tzerpos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Máté Kiss
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gergely Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Zoltan Simandi
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL
| | - Tamas Varga
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Laszlo Nagy
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
18
|
Salvador AFM, Kipnis J. Immune response after central nervous system injury. Semin Immunol 2022; 59:101629. [PMID: 35753867 DOI: 10.1016/j.smim.2022.101629] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/07/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
Traumatic injuries of the central nervous system (CNS) affect millions of people worldwide, and they can lead to severely damaging consequences such as permanent disability and paralysis. Multiple factors can obstruct recovery after CNS injury. One of the most significant is the progressive neuronal death that follows the initial mechanical impact, leading to the loss of undamaged cells via a process termed secondary neurodegeneration. Efforts to define treatments that limit the spread of damage, while important, have been largely ineffectual owing to gaps in the mechanistic understanding that underlies the persisting neuronal cell death. Inflammation, with its influx of immune cells that occurs shortly after injury, has been associated with secondary neurodegeneration. However, the role of the immune system after CNS injury is far more complex. Studies have indicated that the immune response after CNS injury is detrimental, owing to immune cell-produced factors (e.g., pro-inflammatory cytokines, free radicals, neurotoxic glutamate) that worsen tissue damage. Our lab and others have also demonstrated the beneficial immune response that occurs after CNS injury, with the release of growth factors such as brain-derived growth factor (BDNF) and interleukin (IL-10) and the clearance of apoptotic and myelin debris by immune cells1-4. In this review, we first discuss the multifaceted roles of the immune system after CNS injury. We then speculate on how advancements in single-cell RNA technologies can dramatically change our understanding of the immune response, how the spinal cord meninges serve as an important site for hosting immunological processes critical for recovery, and how the origin of peripherally recruited immune cells impacts their function in the injured CNS.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jonathan Kipnis
- Department of Pathology & Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
19
|
Dervan A, Franchi A, Almeida-Gonzalez FR, Dowling JK, Kwakyi OB, McCoy CE, O’Brien FJ, Hibbitts A. Biomaterial and Therapeutic Approaches for the Manipulation of Macrophage Phenotype in Peripheral and Central Nerve Repair. Pharmaceutics 2021; 13:2161. [PMID: 34959446 PMCID: PMC8706646 DOI: 10.3390/pharmaceutics13122161] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/18/2022] Open
Abstract
Injury to the peripheral or central nervous systems often results in extensive loss of motor and sensory function that can greatly diminish quality of life. In both cases, macrophage infiltration into the injury site plays an integral role in the host tissue inflammatory response. In particular, the temporally related transition of macrophage phenotype between the M1/M2 inflammatory/repair states is critical for successful tissue repair. In recent years, biomaterial implants have emerged as a novel approach to bridge lesion sites and provide a growth-inductive environment for regenerating axons. This has more recently seen these two areas of research increasingly intersecting in the creation of 'immune-modulatory' biomaterials. These synthetic or naturally derived materials are fabricated to drive macrophages towards a pro-repair phenotype. This review considers the macrophage-mediated inflammatory events that occur following nervous tissue injury and outlines the latest developments in biomaterial-based strategies to influence macrophage phenotype and enhance repair.
Collapse
Affiliation(s)
- Adrian Dervan
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Antonio Franchi
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Francisco R. Almeida-Gonzalez
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Jennifer K. Dowling
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Ohemaa B. Kwakyi
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- School of Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Claire E. McCoy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (J.K.D.); (O.B.K.); (C.E.M.)
- FutureNeuro SFI Research Centre, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| | - Alan Hibbitts
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, D02 YN77 Dublin, Ireland; (A.D.); (A.F.); (F.R.A.-G.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity College Dublin, D02 R590 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, D02 YN77 Dublin, Ireland
| |
Collapse
|
20
|
Gao J, Khang M, Liao Z, Detloff M, Lee JS. Therapeutic targets and nanomaterial-based therapies for mitigation of secondary injury after spinal cord injury. Nanomedicine (Lond) 2021; 16:2013-2028. [PMID: 34402308 PMCID: PMC8411395 DOI: 10.2217/nnm-2021-0113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/29/2021] [Indexed: 12/31/2022] Open
Abstract
Spinal cord injury (SCI) and the resulting neurological trauma commonly result in complete or incomplete neurological dysfunction and there are few effective treatments for primary SCI. However, the following secondary SCI, including the changes of microvasculature, inflammatory response and oxidative stress around the injury site, may provide promising therapeutic targets. The advances of nanomaterials hold promise for delivering therapeutics to alleviate secondary SCI and promote functional recovery. In this review, we highlight recent achievements of nanomaterial-based therapy, specifically targeting blood-spinal cord barrier disruption, mitigation of the inflammatory response and lightening of oxidative stress after spinal cord injury.
Collapse
Affiliation(s)
- Jun Gao
- Department of Bioengineering, Drug Design, Development & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
| | - Minkyung Khang
- Department of Bioengineering, Drug Design, Development & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| | - Zhen Liao
- Department of Bioengineering, Drug Design, Development & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| | - Megan Detloff
- Department of Neurobiology & Anatomy, Drexel University, Philadelphia, PA 19129, USA
| | - Jeoung Soo Lee
- Department of Bioengineering, Drug Design, Development & Delivery (4D) Laboratory, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
21
|
Immune cell compartmentalization for brain surveillance and protection. Nat Immunol 2021; 22:1083-1092. [PMID: 34429552 DOI: 10.1038/s41590-021-00994-2] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023]
Abstract
For decades, it was commonly accepted that the brain is secluded from peripheral immune activity and is self-sufficient for its maintenance and repair. This simplistic perception was based on the presence of resident immune cells, the microglia, and barrier systems within the brain, and the assumption that the central nervous system (CNS) lacks lymphatic drainage. This view was revised with the discoveries that higher functions of the CNS, homeostasis and repair are supported by peripheral innate and adaptive immune cells. The findings of bone marrow-derived immune cells in specialized niches, and the renewed observation that a lymphatic drainage system exists within the brain, further contributed to this revised model. In this Review, we describe the immune niches within the brain, the contribution of professional immune cells to brain functions, the bidirectional relationships between the CNS and the immune system and the relevance of immune components to brain aging and neurodegenerative diseases.
Collapse
|
22
|
Patsalos A, Tzerpos P, Wei X, Nagy L. Myeloid cell diversification during regenerative inflammation: Lessons from skeletal muscle. Semin Cell Dev Biol 2021; 119:89-100. [PMID: 34016524 PMCID: PMC8530826 DOI: 10.1016/j.semcdb.2021.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/27/2021] [Accepted: 05/03/2021] [Indexed: 12/11/2022]
Abstract
Understanding the mechanisms of tissue and organ regeneration in adult animals and humans is of great interest from a basic biology as well as a medical, therapeutical point of view. It is increasingly clear that the relatively limited ability to regenerate tissues and organs in mammals as oppose to lower vertebrates is the consequence of evolutionary trade-offs and changes during development and aging. Thus, the coordinated interaction of the immune system, particularly the innate part of it, and the injured, degenerated parenchymal tissues such as skeletal muscle, liver, lung, or kidney shape physiological and also pathological processes. In this review, we provide an overview of how morphologically and functionally complete (ad integrum) regeneration is achieved using skeletal muscle as a model. We will review recent advances about the differentiation, activation, and subtype specification of circulating monocyte to resolution or repair-type macrophages during the process we term regenerative inflammation, resulting in complete restoration of skeletal muscle in murine models of toxin-induced injury.
Collapse
Affiliation(s)
- Andreas Patsalos
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Petros Tzerpos
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Xiaoyan Wei
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Laszlo Nagy
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA; Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
23
|
David S, López-Vales R. Bioactive Lipid Mediators in the Initiation and Resolution of Inflammation after Spinal Cord Injury. Neuroscience 2021; 466:273-297. [PMID: 33951502 DOI: 10.1016/j.neuroscience.2021.04.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
Neuroinflammation is a prominent feature of the response to CNS trauma. It is also an important hallmark of various neurodegenerative diseases in which inflammation contributes to the progression of pathology. Inflammation in the CNS can contribute to secondary damage and is therefore an excellent therapeutic target for a range of neurological conditions. Inflammation in the nervous system is complex and varies in its fine details in different conditions. It involves a wide variety of secreted factors such as chemokines and cytokines, cell adhesion molecules, and different cell types that include resident cell of the CNS, as well as immune cells recruited from the peripheral circulation. Added to this complexity is the fact that some aspects of inflammation are beneficial, while other aspects can induce secondary damage in the acute, subacute and chronic phases. Understanding these aspects of the inflammatory profile is essential for developing effective therapies. Bioactive lipids constitute a large group of molecules that modulate the initiation and the resolution of inflammation. Dysregulation of these bioactive lipid pathways can lead to excessive acute inflammation, and failure to resolve this by specialized pro-resolution lipid mediators can lead to the development of chronic inflammation. The focus of this review is to discuss the effects of bioactive lipids in spinal cord trauma and their potential for therapies.
Collapse
Affiliation(s)
- Samuel David
- Centre for Research in Neuroscience, BRaIN Program, The Research Institute of the McGill University Health Centre, 1650 Cedar Avenue, Montreal, Quebec H3G 1A4, Canada.
| | - Rubén López-Vales
- Departament de Biologia Cellular, Fisiologia i Inmunologia, Institut de Neurociències, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Universitat Autònoma de Barcelona, 08193 Bellaterra, Catalonia, Spain
| |
Collapse
|
24
|
Abstract
Tissue-resident macrophages are present in most tissues with developmental, self-renewal, or functional attributes that do not easily fit into a textbook picture of a plastic and multifunctional macrophage originating from hematopoietic stem cells; nor does it fit a pro- versus anti-inflammatory paradigm. This review presents and discusses current knowledge on the developmental biology of macrophages from an evolutionary perspective focused on the function of macrophages, which may aid in study of developmental, inflammatory, tumoral, and degenerative diseases. We also propose a framework to investigate the functions of macrophages in vivo and discuss how inherited germline and somatic mutations may contribute to the roles of macrophages in diseases.
Collapse
Affiliation(s)
- Nehemiah Cox
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Maria Pokrovskii
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| |
Collapse
|
25
|
Harnessing the Benefits of Neuroinflammation: Generation of Macrophages/Microglia with Prominent Remyelinating Properties. J Neurosci 2021; 41:3366-3385. [PMID: 33712513 DOI: 10.1523/jneurosci.1948-20.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 11/21/2022] Open
Abstract
Excessive inflammation within the CNS is injurious, but an immune response is also required for regeneration. Macrophages and microglia adopt different properties depending on their microenvironment, and exposure to IL4 and IL13 has been used to elicit repair. Unexpectedly, while LPS-exposed macrophages and microglia killed neural cells in culture, the addition of LPS to IL4/IL13-treated macrophages and microglia profoundly elevated IL10, repair metabolites, heparin binding epidermal growth factor trophic factor, antioxidants, and matrix-remodeling proteases. In C57BL/6 female mice, the generation of M(LPS/IL4/IL13) macrophages required TLR4 and MyD88 signaling, downstream activation of phosphatidylinositol-3 kinase/mTOR and MAP kinases, and convergence on phospho-CREB, STAT6, and NFE2. Following mouse spinal cord demyelination, local LPS/IL4/IL13 deposition markedly increased lesional phagocytic macrophages/microglia, lactate and heparin binding epidermal growth factor, matrix remodeling, oligodendrogenesis, and remyelination. Our data show that a prominent reparative state of macrophages/microglia is generated by the unexpected integration of pro- and anti-inflammatory activation cues. The results have translational potential, as the LPS/IL4/IL13 mixture could be locally applied to a focal CNS injury to enhance neural regeneration and recovery.SIGNIFICANCE STATEMENT The combination of LPS and regulatory IL4 and IL13 signaling in macrophages and microglia produces a previously unknown and particularly reparative phenotype devoid of pro-inflammatory neurotoxic features. The local administration of LPS/IL4/IL13 into spinal cord lesion elicits profound oligodendrogenesis and remyelination. The careful use of LPS and IL4/IL13 mixture could harness the known benefits of neuroinflammation to enable repair in neurologic insults.
Collapse
|
26
|
Schmidt E, Raposo P, Vavrek R, Fouad K. Inducing inflammation following subacute spinal cord injury in female rats: A double-edged sword to promote motor recovery. Brain Behav Immun 2021; 93:55-65. [PMID: 33358981 DOI: 10.1016/j.bbi.2020.12.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/20/2020] [Accepted: 12/16/2020] [Indexed: 12/20/2022] Open
Abstract
The inflammatory response following spinal cord injury is associated with increased tissue damage and impaired functional recovery. However, inflammation can also promote plasticity and the secretion of growth-promoting substances. Previously we have shown that inducing inflammation with a systemic injection of lipopolysaccharide in the chronic (8 weeks) stage of spinal cord injury enhances neuronal sprouting and the efficacy of rehabilitative training in rats. Here, we tested whether administration of lipopolysaccharide in female rats in the subacute (10 days) stage of spinal cord injury would have a similar effect. Since the lesioned environment is already in a pro-inflammatory state at this earlier time after injury, we hypothesized that triggering a second immune response may not be beneficial for recovery. Contrary to our hypothesis, we found that eliciting an inflammatory response 10 days after spinal cord injury enhanced the recovery of the ipsilesional forelimb in rehabilitative training. Compared to rats that received rehabilitative training without treatment, rats that received systemic lipopolysaccharide showed restored motor function without the use of compensatory strategies that translated beyond the trained task. Furthermore, lipopolysaccharide treatment paradoxically promoted the resolution of chronic neuroinflammation around the lesion site. Unfortunately, re-triggering a systemic immune response after spinal cord injury also resulted in a long-term increase in anxiety-like behaviour.
Collapse
Affiliation(s)
- Emma Schmidt
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Pamela Raposo
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Romana Vavrek
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada; Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada.
| |
Collapse
|
27
|
Sharma S, Tiarks G, Haight J, Bassuk AG. Neuropathophysiological Mechanisms and Treatment Strategies for Post-traumatic Epilepsy. Front Mol Neurosci 2021; 14:612073. [PMID: 33708071 PMCID: PMC7940684 DOI: 10.3389/fnmol.2021.612073] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death in young adults and a risk factor for acquired epilepsy. Severe TBI, after a period of time, causes numerous neuropsychiatric and neurodegenerative problems with varying comorbidities; and brain homeostasis may never be restored. As a consequence of disrupted equilibrium, neuropathological changes such as circuit remodeling, reorganization of neural networks, changes in structural and functional plasticity, predisposition to synchronized activity, and post-translational modification of synaptic proteins may begin to dominate the brain. These pathological changes, over the course of time, contribute to conditions like Alzheimer disease, dementia, anxiety disorders, and post-traumatic epilepsy (PTE). PTE is one of the most common, devastating complications of TBI; and of those affected by a severe TBI, more than 50% develop PTE. The etiopathology and mechanisms of PTE are either unknown or poorly understood, which makes treatment challenging. Although anti-epileptic drugs (AEDs) are used as preventive strategies to manage TBI, control acute seizures and prevent development of PTE, their efficacy in PTE remains controversial. In this review, we discuss novel mechanisms and risk factors underlying PTE. We also discuss dysfunctions of neurovascular unit, cell-specific neuroinflammatory mediators and immune response factors that are vital for epileptogenesis after TBI. Finally, we describe current and novel treatments and management strategies for preventing PTE.
Collapse
Affiliation(s)
- Shaunik Sharma
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Grant Tiarks
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Joseph Haight
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| | - Alexander G Bassuk
- Medical Laboratories, Department of Pediatrics, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
28
|
Fiani B, Arshad MA, Shaikh ES, Baig A, Farooqui M, Ayub MA, Zafar A, Quadri SA. Current updates on various treatment approaches in the early management of acute spinal cord injury. Rev Neurosci 2021; 32:513-530. [PMID: 33565738 DOI: 10.1515/revneuro-2020-0148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023]
Abstract
Spinal cord injury (SCI) is a debilitating condition which often leads to a severe disability and ultimately impact patient's physical, psychological, and social well-being. The management of acute SCI has evolved over the couple of decades due to improved understanding of injury mechanisms and increasing knowledge of disease. Currently, the early management of acute SCI patient includes pharmacological agents, surgical intervention and newly experimental neuroprotective strategies. However, many controversial areas are still surrounding in the current treatment strategies for acute SCI, including the optimal timing of surgical intervention, early versus delayed decompression outcome benefits, the use of methylprednisolone. Due to the lack of consensus, the optimal standard of care has been varied across treatment centres. The authors have shed a light on the current updates on early treatment approaches and neuroprotective strategies in the initial management of acute SCI in order to protect the early neurologic injury and reduce the future disability.
Collapse
Affiliation(s)
- Brian Fiani
- Department of Neurosurgery, Desert Regional Medical Center, 1150 N. Indian Canyon Drive, Palm Springs, CA92262, USA
| | - Mohammad Arsal Arshad
- Department of Neurosurgery, Desert Regional Medical Center, 1150 N. Indian Canyon Drive, Palm Springs, CA92262, USA
| | - Emad Salman Shaikh
- Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
| | - Aqsa Baig
- Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Mudassir Farooqui
- Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Muhammed Abubakar Ayub
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Atif Zafar
- Department of Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Syed A Quadri
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
29
|
Macrophage migration inhibitory factor as a therapeutic target after traumatic spinal cord injury: a systematic review. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2021; 30:1474-1494. [PMID: 33486594 DOI: 10.1007/s00586-021-06718-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/25/2020] [Accepted: 01/01/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Macrophages play an important role in mediating damage after Spinal cord injury (SCI) by secreting macrophage migration inhibitory factor (MMIF) as a secondary injury mediator. We aimed to systematically review the role of MMIF as a therapeutic target after traumatic SCI. METHODS Our systematic review has been performed according to the PRISMA 2009 Checklist. A systematic search in the scientific databases was carried out for studies published before 20 February 2019 from major databases. Two researchers independently screened titles. The risk of bias of eligible articles was assessed, and data were extracted. Finally, we systematically analyzed and interpreted related data. RESULTS 785 papers were selected for the title and abstract screening. 12 papers were included for data extraction. Eight animal studies were of high quality and the remaining two were of medium quality. One of the two human studies was of poor quality and the other was of fair quality. MMIF as a pro-inflammatory mediator can cause increased susceptibility to glutamate-related neurotoxicity, increased nitrite production, increased ERK activation, and increased COX2/PGE2 signaling pathway activation and subsequent stimulation of CCL5-related chemotaxis. Two human studies and six animal studies demonstrated that MMIF level increases after SCI. MMIF inhibition might be a potential therapeutic target in SCI by multiple different mechanisms (6/12 studies). CONCLUSION Most animal studies demonstrate significant neurologic improvement after administration of MMIF inhibitors, but these inhibitors have not been studied in humans yet. Further clinical trials are need to further understand MMIF inhibitor utility in acute or chronic SCI. LEVEL OF EVIDENCE I Diagnostic: individual cross-sectional studies with the consistently applied reference standard and blinding.
Collapse
|
30
|
Mesquida-Veny F, Del Río JA, Hervera A. Macrophagic and microglial complexity after neuronal injury. Prog Neurobiol 2020; 200:101970. [PMID: 33358752 DOI: 10.1016/j.pneurobio.2020.101970] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/12/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022]
Abstract
Central nervous system (CNS) injuries do not heal properly in contrast to normal tissue repair, in which functional recovery typically occurs. The reason for this dichotomy in wound repair is explained in part by macrophage and microglial malfunction, affecting both the extrinsic and intrinsic barriers to appropriate axonal regeneration. In normal healing tissue, macrophages promote the repair of injured tissue by regulating transitions through different phases of the healing response. In contrast, inflammation dominates the outcome of CNS injury, often leading to secondary damage. Therefore, an understanding of the molecular mechanisms underlying this dichotomy is critical to advance in neuronal repair therapies. Recent studies highlight the plasticity and complexity of macrophages and microglia beyond the classical view of the M1/M2 polarization paradigm. This plasticity represents an in vivo continuous spectrum of phenotypes with overlapping functions and markers. Moreover, macrophage and microglial plasticity affect many events essential for neuronal regeneration after injury, such as myelin and cell debris clearance, inflammation, release of cytokines, and trophic factors, affecting both intrinsic neuronal properties and extracellular matrix deposition. Until recently, this complexity was overlooked in the translation of therapies modulating these responses for the treatment of neuronal injuries. However, recent studies have shed important light on the underlying molecular mechanisms of this complexity and its transitions and effects on regenerative events. Here we review the complexity of macrophages and microglia after neuronal injury and their roles in regeneration, as well as the underlying molecular mechanisms, and we discuss current challenges and future opportunities for treatment.
Collapse
Affiliation(s)
- Francina Mesquida-Veny
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain; Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - José Antonio Del Río
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain; Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Arnau Hervera
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Spain; Institute of Neuroscience, University of Barcelona, 08028 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain.
| |
Collapse
|
31
|
Xiao Q, Guo T, Li J, Li L, Chen K, Zhou L, Wu W, So KF, Ramakrishna S, Liu B, Rong L, Chen G, Xing X, He L. Macrophage polarization induced by sustained release of 7,8-DHF from aligned PLLA fibers potentially for neural stem cell neurogenesis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111415. [PMID: 33255017 DOI: 10.1016/j.msec.2020.111415] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 01/06/2023]
Abstract
Neural stem cells (NSCs)-based regenerative medicine provides unprecedented therapeutic potential in neural insults. However, NSC-based neurogenesis is strongly influenced by the inflammatory environment after injury, which is mainly modulated by macrophages' secretion effects. In this study, we adopted poly L-lactic acid (PLLA) aligned fibers to guide macrophages elongating along the fiber directions and polarizing phenotypically toward anti-inflammatory M2 type. 7,8-DHF was loaded within the fibers with a sustained and controlled release pattern to promote the polarization of the macrophages and secretion of various anti-inflammatory factors. NSCs showed enhanced neuronal differentiation in the presence of the conditioned medium (CM) from M2 macrophages cultured on the 7,8-DHF-loaded PLLA aligned fibers. Moreover, M2-CM promoted neurogenesis by enhancing neurite outgrowth of NSC-derived neurons. In summary, we provided a novel therapeutic strategy for NSC neurogenesis by manipulating macrophage classification into anti-inflammatory M2 phenotypes with the 7,8-DHF-loaded PLLA aligned fibers, existing potential applications in treating neural injuries.
Collapse
Affiliation(s)
- Qiao Xiao
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Ting Guo
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Jun Li
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Liming Li
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Kaixin Chen
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Libing Zhou
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Wutian Wu
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Department of Biotechnology, Key Laboratory of Virology of Guangzhou, College of Life Science and Technology, Jinan University, Guangzhou 510630, China
| | - Kwok-Fai So
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Seeram Ramakrishna
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Bin Liu
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Limin Rong
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Guoqiang Chen
- The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Xiwen Xing
- Department of Biotechnology, Key Laboratory of Virology of Guangzhou, College of Life Science and Technology, Jinan University, Guangzhou 510630, China.
| | - Liumin He
- MOE Joint International Research Laboratory of CNS Regeneration, Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China.
| |
Collapse
|
32
|
Zhao J, Su M, Lin Y, Liu H, He Z, Lai L. Administration of Amyloid Precursor Protein Gene Deleted Mouse ESC-Derived Thymic Epithelial Progenitors Attenuates Alzheimer's Pathology. Front Immunol 2020; 11:1781. [PMID: 32849642 PMCID: PMC7431620 DOI: 10.3389/fimmu.2020.01781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/03/2020] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of dementia in older adults. Although amyloid-beta (Aβ) plaque deposition and chronic neuroinflammation in the central nervous system (CNS) contribute to AD pathology, neither Aβ plaque removal nor anti-inflammatory therapy has shown much clinical success, suggesting that the combinational therapies for the disease-causative factors may be needed for amelioration. Recent data also suggest that systemic immunity in AD should be boosted, rather than suppressed, to drive an immune-dependent cascade needed for Aβ clearance and brain repair. Thymic epithelial cells (TECs) not only play a critical role in supporting T cell development but also mediate the deletion of autoreactive T cells by expressing autoantigens. We have reported that embryonic stem cells (ESCs) can be selectively induced to differentiate into thymic epithelial progenitors (TEPs) in vitro that further develop into TECs in vivo to support T cell development. We show here that transplantation of mouse ESC (mESC)-TEPs into AD mice reduced cerebral Aβ plaque load and improved cognitive performance, in correlation with an increased number of T cells, enhanced choroid plexus (CP) gateway activity, and increased number of macrophages in the brain. Furthermore, transplantation of the amyloid precursor protein (APP) gene deleted mESC-TEPs (APP-/-) results in more effective reduction of AD pathology as compared to wild-type (APP+/+) mESC-TEPs. This is associated with the generation of Aβ-specific T cells, which leads to an increase of anti-Aβ antibody (Ab)-producing B cells in the spleen and enhanced levels of anti-Aβ antibodies in the serum, as well as an increase of Aβ phagocytosing macrophages in the CNS. Our results suggest that transplantation of APP-/- human ESC- or induced pluripotent stem cell (iPSC)-derived TEPs may provide a new tool to mitigate AD in patients.
Collapse
Affiliation(s)
- Jin Zhao
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Tissue Engineering and Stem Cell Research Center, Department of Immunology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States
| | - Yujun Lin
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States
| | - Haiyan Liu
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States
| | - Zhixu He
- Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, China.,Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States.,University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
33
|
Schwartz M, Peralta Ramos JM, Ben-Yehuda H. A 20-Year Journey from Axonal Injury to Neurodegenerative Diseases and the Prospect of Immunotherapy for Combating Alzheimer's Disease. THE JOURNAL OF IMMUNOLOGY 2020; 204:243-250. [PMID: 31907265 DOI: 10.4049/jimmunol.1900844] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022]
Abstract
The understanding of the dialogue between the brain and the immune system has undergone dramatic changes over the last two decades, with immense impact on the perception of neurodegenerative diseases, mental dysfunction, and many other brain pathologic conditions. Accumulated results have suggested that optimal function of the brain is dependent on support from the immune system, provided that this immune response is tightly controlled. Moreover, in contrast to the previous prevailing dogma, it is now widely accepted that circulating immune cells are needed for coping with brain pathologies and that their optimal effect is dependent on their type, location, and activity. In this perspective, we describe our own scientific journey, reviewing the milestones in attaining this understanding of the brain-immune axis integrated with numerous related studies by others. We then explain their significance in demonstrating the possibility of harnessing the immune system in a well-controlled manner for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Michal Schwartz
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142; and .,Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | | - Hila Ben-Yehuda
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
34
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
35
|
Lau YCC, Ding JA, Simental A, Mirzoyan H, Lee W, Diamante G, Cely I, Tran M, Morselli M, Dang J, Kaczor-Urbanowicz KE, Sayre J, Stiles L, Yang X, Pellegrini M, Fiala M. Omega-3 fatty acids increase OXPHOS energy for immune therapy of Alzheimer disease patients. FASEB J 2020; 34:9982-9994. [PMID: 32614485 DOI: 10.1096/fj.202000669rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/03/2020] [Accepted: 05/05/2020] [Indexed: 11/11/2022]
Abstract
Sporadic late-onset Alzheimer disease (LOAD) preceded by mild cognitive impairment (MCI) is the most common type of dementia. Long-term studies of immunity to pathogenic amyloid-β (Aβ) in LOAD are lacking. Innate immunity of LOAD patients is malfunctioning in phagocytosis and degradation of Aβ and LOAD patients' macrophage transcriptome and metabolome are deregulated. We previously showed omega-3 fatty acid (ω-3)-mediated repair of unfolded protein response and here we show much broader transcriptomic effects. ω-3 treatment in vitro and ω-3 supplementation by the drink Smartfish (SMF) in vivo increased the transcripts of the genes and pathways of immunity, glycolysis, tricarboxylic acid cycle, OX-PHOS, nicotinamide dinucleotide (NAD+ ) synthesis, and reversed the defects in Aβ phagocytosis. In both peripheral blood mononuclear cells (PBMC) and macrophages, ω-3 increased ATP-linked oxygen consumption rate (OCR) and ω-3 with carnitine was superior to ω-3. ω-3 treatment in vitro and supplementation by the ω-3 drink SMF in vivo rescued macrophage phagocytosis when glycolysis or glycosylation were blocked. ω-3 provide flexible energy for immune clearance of the brain throughout the diurnal cycle, even in hypo- or hyper-glycemia. In certain LOAD patients, ω-3 may delay progression to dementia.
Collapse
Affiliation(s)
- Yik Chai Charles Lau
- Division of Oral Biology and Oral Medicine, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA.,Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Jessica Aliyah Ding
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aracely Simental
- Division of Oral Biology and Oral Medicine, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA.,Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Hayk Mirzoyan
- Division of Oral Biology and Oral Medicine, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA.,Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - William Lee
- Division of Oral Biology and Oral Medicine, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA.,Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ingrid Cely
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michelle Tran
- Division of Oral Biology and Oral Medicine, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA.,Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Marco Morselli
- Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Johnny Dang
- Division of Oral Biology and Oral Medicine, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA.,Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Division of Oral Biology and Oral Medicine, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA.,Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - James Sayre
- UCLA School of Public Health, University of California, Los Angeles, Los Angeles, CA, USA
| | - Linsey Stiles
- Mitochondrial and Metabolism Core, UCLA School of Medicine, Los Angeles, CA, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA.,Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| | - Milan Fiala
- Division of Oral Biology and Oral Medicine, UCLA School of Dentistry and Medicine, Los Angeles, CA, USA.,Department of Molecular, Cell, and Developmental Biology, UCLA School of Life Sciences, Los Angeles, CA, USA
| |
Collapse
|
36
|
Yip PK, Bowes AL, Hall JCE, Burguillos MA, Ip THR, Baskerville T, Liu ZH, Mohamed MAEK, Getachew F, Lindsay AD, Najeeb SUR, Popovich PG, Priestley JV, Michael-Titus AT. Docosahexaenoic acid reduces microglia phagocytic activity via miR-124 and induces neuroprotection in rodent models of spinal cord contusion injury. Hum Mol Genet 2020; 28:2427-2448. [PMID: 30972415 DOI: 10.1093/hmg/ddz073] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/31/2019] [Accepted: 04/01/2019] [Indexed: 12/20/2022] Open
Abstract
Microglia are activated after spinal cord injury (SCI), but their phagocytic mechanisms and link to neuroprotection remain incompletely characterized. Docosahexaenoic acid (DHA) has been shown to have significant neuroprotective effects after hemisection and compression SCI and can directly affect microglia in these injury models. In rodent contusion SCI, we demonstrate that DHA (500 nmol/kg) administered acutely post-injury confers neuroprotection and enhances locomotor recovery, and also exerts a complex modulation of the microglial response to injury. In rodents, at 7 days after SCI, the level of phagocytosed myelin within Iba1-positive or P2Y12-positive cells was significantly lower after DHA treatment, and this occurred in parallel with an increase in intracellular miR-124 expression. Furthermore, intraspinal administration of a miR-124 inhibitor significantly reduced the DHA-induced decrease in myelin phagocytosis in mice at 7 days post-SCI. In rat spinal primary microglia cultures, DHA reduced the phagocytic response to myelin, which was associated with an increase in miR-124, but not miR-155. A similar response was observed in a microglia cell line (BV2) treated with DHA, and the effect was blocked by a miR-124 inhibitor. Furthermore, the phagocytic response of BV2 cells to stressed neurones was also reduced in the presence of DHA. In peripheral monocyte-derived macrophages, the expression of the M1, but not the M0 or M2 phenotype, was reduced by DHA, but the phagocytic activation was not altered. These findings show that DHA induces neuroprotection in contusion injury. Furthermore, the improved outcome is via a miR-124-dependent reduction in the phagocytic response of microglia.
Collapse
Affiliation(s)
- Ping K Yip
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Amy L Bowes
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jodie C E Hall
- Centre for Brain and Spinal Cord Repair, Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - Miguel A Burguillos
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Instituto de Biomedicina de Sevilla (IBiS)/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla and, Sevilla, Spain
| | - T H Richard Ip
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tracey Baskerville
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Zhuo-Hao Liu
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Chang Gung Medical College and University, Chang Gung Memorial Hospital, Department of Neurosurgery, 5 Fu-Shin Street, Linkou, Taiwan
| | - Moumin A E K Mohamed
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Fanuelle Getachew
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anna D Lindsay
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Saif-Ur-Rehman Najeeb
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Phillip G Popovich
- Centre for Brain and Spinal Cord Repair, Department of Neuroscience, Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - John V Priestley
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Adina T Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
37
|
Yagura K, Ohtaki H, Tsumuraya T, Sato A, Miyamoto K, Kawada N, Suzuki K, Nakamura M, Kanzaki K, Dohi K, Izumizaki M, Hiraizumi Y, Honda K. The enhancement of CCL2 and CCL5 by human bone marrow-derived mesenchymal stem/stromal cells might contribute to inflammatory suppression and axonal extension after spinal cord injury. PLoS One 2020; 15:e0230080. [PMID: 32155215 PMCID: PMC7064230 DOI: 10.1371/journal.pone.0230080] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/20/2020] [Indexed: 01/04/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem/stromal cells (hMSCs) have shown potential in facilitating recovery from spinal cord injury (SCI) through communicating with microglia/macrophages (MG/MΦ). We here focused on chemokines as a candidate for the communication. Selected MG/MΦ-related chemokines were determined gene expression after SCI and further focused CCL2/CCR2 and CCL5/CCR5 to estimate role of the chemokines by hMSCs. Male C57/BL6 mice were subjected to spinal cord transection. Gene expression was assayed in the spinal cords following SCI for selected MG/MΦ-related chemokines and their receptors. hMSCs (5×105 cells) were then transplanted into parenchyma of the spinal cord, and the expressions of the Ccl2/Ccr2 and Ccl5/Ccr5 axes, inflammation, MG/MΦ-polarization, and axonal regeneration were evaluated to measure the influence of the hMSCs. Finally, mouse CCL5 was injected into the spinal cords. Acute increases in gene expression after SCI were observed for most chemokines, including Ccl2; chronic increases were observed for Ccl5. CCL2+-cells merged with NeuN+-neurons. CCR2+ immunoreactivity was principally observed in Ly-6G+/iNOS+-granulocytes on postoperative day (pod) 1, and CCL5+ and CCR5+ immunoreactivity overlapped with NeuN+-neurons and F4/80+-MG/MΦ on pod 14. The hMSC transplantation enhanced Ccl2 and Ccl5 and improved locomotor activity. The hMSC implantation did not alter the number of Ly-6G+/CCR2+ but decreased Il1, Elane, and Mpo on pod 3. Conversely, hMSC transplantation increased expression of Zc3h12a (encodes MCP-1-induced protein) on pod 14. Moreover, hMSC increased the Aif1, and two alternatively activated macrophage (AAM)-related genes, Arg1 and Chil3 (Ym1), as well as axonal regenerative markers, Dpysl2 and Gap43. Gene expression indicative of AAM polarization and axonal regeneration were partially recovered by CCL5 injection. These results suggest that hMSC implantation increases Ccl2 and Ccl5, improves locomotor activity, enhances MG/MΦ polarization to AAM, and increases the gene expression of axonal regenerative markers. These functions of hMSCs might be partially mediated by the CCL2/CCR2 and CCL5/CCR5 axes.
Collapse
Affiliation(s)
- Kazumichi Yagura
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, Fujigaoka, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Hirokazu Ohtaki
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
- * E-mail:
| | - Tomomi Tsumuraya
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, Fujigaoka, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Atsushi Sato
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, Fujigaoka, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Kazuyuki Miyamoto
- Department of Emergency & Clinical Care Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Naoto Kawada
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Keisuke Suzuki
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
- Department of Emergency & Clinical Care Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Motoyasu Nakamura
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
- Department of Emergency & Clinical Care Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Koji Kanzaki
- Department of Orthopedic Surgery, Showa University Fujigaoka Hospital, Fujigaoka, Aoba-ku, Yokohama, Kanagawa, Japan
| | - Kenji Dohi
- Department of Emergency & Clinical Care Medicine, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Yutaka Hiraizumi
- Department of Orthopedic Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| | - Kazuho Honda
- Department of Anatomy, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
38
|
Macrophage Transplantation Fails to Improve Repair of Critical-Sized Calvarial Defects. J Craniofac Surg 2020; 30:2640-2645. [PMID: 31609958 DOI: 10.1097/scs.0000000000005797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Over 500,000 bone grafting procedures are performed every year in the United States for neoplastic and traumatic lesions of the craniofacial skeleton, costing $585 million in medical care. Current bone grafting procedures are limited, and full-thickness critical-sized defects (CSDs) of the adult human skull thus pose a substantial reconstructive challenge for the craniofacial surgeon. Cell-based strategies have been shown to safely and efficaciously accelerate the rate of bone formation in CSDs in animals. The authors recently demonstrated that supraphysiological transplantation of macrophages seeded in pullalan-collagen composite hydrogels significantly accelerated wound healing in wild type and diabetic mice, an effect mediated in part by enhancing angiogenesis. In this study, the authors investigated the bone healing effects of macrophage transplantation into CSDs of mice. METHODS CD1 athymic nude mice (60 days of age) were anesthetized, and unilateral full-thickness critical-sized (4 mm in diameter) cranial defects were created in the right parietal bone, avoiding cranial sutures. Macrophages were isolated from FVB-L2G mice and seeded onto hydroxyapatite-poly (lactic-co-glycolic acid) (HA-PLGA) scaffolds (1.0 × 10 cells per CSD). Scaffolds were incubated for 24 hours before they were placed into the CSDs. Macrophage survival was assessed using three-dimensional in vivo imaging system (3D IVIS)/micro-CT. Micro-CT at 0, 2, 4, 6, and 8 weeks was performed to evaluate gross bone formation, which was quantified using Adobe Photoshop. Microscopic evidence of bone regeneration was assessed at 8 weeks by histology. Bone formation and macrophage survival were compared at each time point using independent samples t tests. RESULTS Transplantation of macrophages at supraphysiological concentration had no effect on the formation of bones in CSDs as assessed by either micro-CT data at any time point analyzed (all P > 0.05). These results were corroborated by histology. 3D IVIS/micro-CT demonstrated survival of macrophages through 8 weeks. CONCLUSION Supraphysiologic delivery of macrophages to CSDs of mice had no effect on bone formation despite survival of transplanted macrophages through to 8 weeks posttransplantation. Further research into the physiological effects of macrophages on bone regeneration is needed to assess whether recapitulation of these conditions in macrophage-based therapy can promote the healing of large cranial defects.
Collapse
|
39
|
Wang Y, Wei S, Song H, Zhang X, Wang W, Du N, Song T, Liang H, Chen X, Wang Y. Macrophage migration inhibitory factor derived from spinal cord is involved in activation of macrophages following gecko tail amputation. FASEB J 2019; 33:14798-14810. [PMID: 31689136 DOI: 10.1096/fj.201801966rrr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Macrophages and their initiation of acute inflammation have been defined to be functionally important in tissue repair and regeneration. In injury-induced production of macrophage migration inhibitory factor (MIF), which has been described as a pleiotropic protein that participates in multiple cellular and biologic processes, it is unknown whether it is involved in the regulation of macrophage events during the epimorphic regeneration. In the model of gecko tail amputation, the protein levels of gecko MIF (gMIF) have been determined to be significantly increased in the nerve cells of the spinal cord in association with the recruitment of macrophages to the lesion site. gMIF has been shown to interact with the CD74 receptor to promote the migration of macrophages through activation of Ras homolog gene family member A and to trigger inflammatory responses through MAPK signaling pathways. The determination of microsphere phagocytosis also indicated that gMIF could enhance macrophage phagocytosis. gMIF-mediated recruitment and activation of macrophages have been found to be necessary for gecko tail regeneration, as evidenced by the depletion of macrophages using clodronate liposomes. The results present a novel function of MIF during the epimorphic regeneration, which is beneficial for insights into its pleiotropic property.-Wang, Y., Wei, S., Song, H., Zhang, X., Wang, W., Du, N., Song, T., Liang, H., Chen, X., Wang, Y. Macrophage migration inhibitory factor derived from spinal cord is involved in activation of macrophages following gecko tail amputation.
Collapse
Affiliation(s)
- Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Sumei Wei
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Honghua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xuejie Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Wenjuan Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Nan Du
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Tiancheng Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Hao Liang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaojun Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
40
|
Macrophages with regulatory functions, a possible new therapeutic perspective in autoimmune diseases. Autoimmun Rev 2019; 18:102369. [PMID: 31404701 DOI: 10.1016/j.autrev.2019.102369] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 04/29/2019] [Indexed: 12/14/2022]
Abstract
Macrophages are pivotal cells involved in chronic inflammatory and autoimmune diseases. In fact, during these diseases, activated macrophages may play a critical role, promoting the inflammation as well as mediating the damage resolution. This dichotomy is referred to two end-stage phenotypes of macrophages, conventionally known as M1 and M2, playing a pro-inflammatory and anti-inflammatory role, respectively. The M1 macrophages are the mainly subset involved during inflammatory processes, producing pro-inflammatory mediators. Conversely, the M2 macrophages are proposed to contribute to the resolution phase of inflammation, when cells with pro-resolving property are recruited and activated. In fact, this subset of macrophages may activate regulatory T lymphocytes, which play a critical role in the maintenance of peripheral tolerance and preventing the occurrence of autoimmune diseases. On these bases, the polarization toward the M2 phenotype could play a therapeutic role for autoimmune diseases. In this Review we discussed the characteristic of M1 and M2 macrophages, focusing on the immunoregulatory role of M2 cells and their potential ability to control the inflammation and to promote the immunological tolerance.
Collapse
|
41
|
Song YH, Agrawal NK, Griffin JM, Schmidt CE. Recent advances in nanotherapeutic strategies for spinal cord injury repair. Adv Drug Deliv Rev 2019; 148:38-59. [PMID: 30582938 PMCID: PMC6959132 DOI: 10.1016/j.addr.2018.12.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/12/2018] [Accepted: 12/17/2018] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a devastating and complicated condition with no cure available. The initial mechanical trauma is followed by a secondary injury characterized by inflammatory cell infiltration and inhibitory glial scar formation. Due to the limitations posed by the blood-spinal cord barrier, systemic delivery of therapeutics is challenging. Recent development of various nanoscale strategies provides exciting and promising new means of treating SCI by crossing the blood-spinal cord barrier and delivering therapeutics. As such, we discuss different nanomaterial fabrication methods and provide an overview of recent studies where nanomaterials were developed to modulate inflammatory signals, target inhibitory factors in the lesion, and promote axonal regeneration after SCI. We also review emerging areas of research such as optogenetics, immunotherapy and CRISPR-mediated genome editing where nanomaterials can provide synergistic effects in developing novel SCI therapy regimens, as well as current efforts and barriers to clinical translation of nanomaterials.
Collapse
Affiliation(s)
- Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Nikunj K Agrawal
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jonathan M Griffin
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
42
|
Liu W, Kämpfe Nordström C, Danckwardt-Lillieström N, Rask-Andersen H. Human Inner Ear Immune Activity: A Super-Resolution Immunohistochemistry Study. Front Neurol 2019; 10:728. [PMID: 31354608 PMCID: PMC6635812 DOI: 10.3389/fneur.2019.00728] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Like the brain, the human inner ear was long thought to be devoid of immune activity. Only the endolymphatic sac (ES) was known to be endowed with white blood cells that could process antigens and serve as an immunologic defense organ for the entire inner ear. Unexpectedly, the cochlear and vestibular organs, including the eighth cranial nerve, were recently shown to contain macrophages whose functions and implication in ear disease are somewhat undefined. Here, we review recent inner ear findings in man and extend the analyses to the vestibular nerve using super-resolution structured illumination microscopy (SR-SIM). Materials and Methods: Human ESs and cochleae were collected during surgery to treat patients with vestibular schwannoma and life-threatening petro-clival meningioma compressing the brainstem. The ESs and cochleae were placed in fixative, decalcified, and rapidly frozen and cryostat sectioned. Antibodies against ionized calcium-binding adaptor molecule 1-expressing cells (IBA1 cells), laminin β2 and type IV collagen TUJ1, cytokine fractalkine (CX3CL1), toll-like receptor 4 (TLR4), CD68, CD11b, CD4, CD8, the major histocompatibility complex type II (MHCII), and the microglial marker TEME119 were used. Results: IBA1-positive cells were present in the ESs, the cochlea, central and peripheral axons of the cochlear nerve, and the vestibular nerve trunk. IBA1 cells were found in the cochlear lateral wall, spiral limbus, and spiral ganglion. Notable variants of IBA1 cells adhered to neurons with “synapse-like” specializations and cytoplasmic projections. Slender IBA1 cells occasionally protracted into the basal lamina of the Schwann cells and had intimate contact with surrounding axons. Discussion: The human eighth nerve may be under the control of a well-developed macrophage cell system. A small number of CD4+ and CD8+ cells were found in the ES and occasionally in the cochlea, mostly located in the peripheral region of Rosenthal's canal. A neuro-immunologic axis may exist in the human inner ear that could play a role in the protection of the auditory nerve. The implication of the macrophage system during disease, surgical interventions, and cell-based transplantation should be further explored.
Collapse
Affiliation(s)
- Wei Liu
- Section of Otolaryngology, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Charlotta Kämpfe Nordström
- Section of Otolaryngology, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | | | - Helge Rask-Andersen
- Section of Otolaryngology, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
43
|
Ballout N, Rochelle T, Brot S, Bonnet ML, Francheteau M, Prestoz L, Zibara K, Gaillard A. Characterization of Inflammation in Delayed Cortical Transplantation. Front Mol Neurosci 2019; 12:160. [PMID: 31293384 PMCID: PMC6603085 DOI: 10.3389/fnmol.2019.00160] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 06/07/2019] [Indexed: 01/30/2023] Open
Abstract
We previously reported that embryonic motor cortical neurons transplanted 1-week after lesion in the adult mouse motor cortex significantly enhances graft vascularization, survival, and proliferation of grafted cells, the density of projections developed by grafted neurons and improves functional repair and recovery. The purpose of the present study is to understand the extent to which post-traumatic inflammation following cortical lesion could influence the survival of grafted neurons and the development of their projections to target brain regions and conversely how transplanted cells can modulate host inflammation. For this, embryonic motor cortical tissue was grafted either immediately or with a 1-week delay into the lesioned motor cortex of adult mice. Immunohistochemistry (IHC) analysis was performed to determine the density and cell morphology of resident and peripheral infiltrating immune cells. Then, in situ hybridization (ISH) was performed to analyze the distribution and temporal mRNA expression pattern of pro-inflammatory or anti-inflammatory cytokines following cortical lesion. In parallel, we analyzed the protein expression of both M1- and M2-associated markers to study the M1/M2 balance switch. We have shown that 1-week after the lesion, the number of astrocytes, microglia, oligodendrocytes, and CD45+ cells were significantly increased along with characteristics of M2 microglia phenotype. Interestingly, the majority of microglia co-expressed transforming growth factor-β1 (TGF-β1), an anti-inflammatory cytokine, supporting the hypothesis that microglial activation is also neuroprotective. Our results suggest that the modulation of post-traumatic inflammation 1-week after cortical lesion might be implicated in the improvement of graft vascularization, survival, and density of projections developed by grafted neurons.
Collapse
Affiliation(s)
- Nissrine Ballout
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM U1084, Poitiers, France.,Laboratory of Stem Cells, PRASE, DSST, Department of Biology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Tristan Rochelle
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM U1084, Poitiers, France
| | - Sebastien Brot
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM U1084, Poitiers, France
| | - Marie-Laure Bonnet
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM U1084, Poitiers, France.,CHU Poitiers, Poitiers, France
| | - Maureen Francheteau
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM U1084, Poitiers, France
| | - Laetitia Prestoz
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM U1084, Poitiers, France
| | - Kazem Zibara
- Laboratory of Stem Cells, PRASE, DSST, Department of Biology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Afsaneh Gaillard
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM U1084, Poitiers, France
| |
Collapse
|
44
|
Kertser A, Baruch K, Deczkowska A, Weiner A, Croese T, Kenigsbuch M, Cooper I, Tsoory M, Ben-Hamo S, Amit I, Schwartz M. Corticosteroid signaling at the brain-immune interface impedes coping with severe psychological stress. SCIENCE ADVANCES 2019; 5:eaav4111. [PMID: 31149632 PMCID: PMC6541460 DOI: 10.1126/sciadv.aav4111] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 04/22/2019] [Indexed: 05/22/2023]
Abstract
The immune system supports brain plasticity and homeostasis, yet it is prone to changes following psychological stress. Thus, it remains unclear whether and how stress-induced immune alterations contribute to the development of mental pathologies. Here, we show that following severe stress in mice, leukocyte trafficking through the choroid plexus (CP), a compartment that mediates physiological immune-brain communication, is impaired. Blocking glucocorticoid receptor signaling, either systemically or locally through its genetic knockdown at the CP, facilitated the recruitment of Gata3- and Foxp3-expressing T cells to the brain and attenuated post-traumatic behavioral deficits. These findings functionally link post-traumatic stress behavior with elevated stress-related corticosteroid signaling at the brain-immune interface and suggest a novel therapeutic target to attenuate the consequences of severe psychological stress.
Collapse
Affiliation(s)
- A. Kertser
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - K. Baruch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - A. Deczkowska
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - A. Weiner
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - T. Croese
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - M. Kenigsbuch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - I. Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
| | - M. Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - S. Ben-Hamo
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - I. Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - M. Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- Corresponding author.
| |
Collapse
|
45
|
Milich LM, Ryan CB, Lee JK. The origin, fate, and contribution of macrophages to spinal cord injury pathology. Acta Neuropathol 2019; 137:785-797. [PMID: 30929040 PMCID: PMC6510275 DOI: 10.1007/s00401-019-01992-3] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
Virtually all phases of spinal cord injury pathogenesis, including inflammation, cell proliferation and differentiation, as well as tissue remodeling, are mediated in part by infiltrating monocyte-derived macrophages. It is now clear that these infiltrating macrophages have distinct functions from resident microglia and are capable of mediating both harmful and beneficial effects after injury. These divergent effects have been largely attributed to environmental cues, such as specific cytokines, that influence the macrophage polarization state. In this review, we also consider the possibility that different macrophage origins, including the spleen, bone marrow, and local self-renewal, may also affect macrophage fate, and ultimately their function that contribute to the complex pathobiology of spinal cord injury.
Collapse
Affiliation(s)
- Lindsay M Milich
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Christine B Ryan
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
46
|
Brennan FH, Popovich PG. Emerging targets for reprograming the immune response to promote repair and recovery of function after spinal cord injury. Curr Opin Neurol 2019; 31:334-344. [PMID: 29465433 DOI: 10.1097/wco.0000000000000550] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW In adult mammals, a traumatic spinal cord injury (SCI) elicits a chronic unregulated neuroinflammatory response accompanied by seemingly paradoxical suppression of systemic immunity. These SCI-induced changes in immune function contribute to poor neurological outcomes and enhanced morbidity or mortality. Nonspecific anti-inflammatory or proinflammatory therapies are ineffective and can even worsen outcomes. Therefore, recent experimental SCI research has advanced the understanding of how neuroimmune cross-talk contributes to spinal cord and systemic pathology. RECENT FINDINGS It is now appreciated that the immune response caused by injury to the brain or spinal cord encompasses heterogeneous elements that can drive events on the spectrum between exacerbating pathology and promoting tissue repair, within the spinal cord and throughout the body. Recent novel discoveries regarding the role and regulation of soluble factors, monocytes/macrophages, microRNAs, lymphocytes and systemic immune function are highlighted in this review. SUMMARY A more nuanced understanding of how the immune system responds and reacts to nervous system injury will present an array of novel therapeutic opportunities for clinical SCI and other forms of neurotrauma.
Collapse
Affiliation(s)
- Faith H Brennan
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | |
Collapse
|
47
|
Abstract
Emerging results support the concept that Alzheimer disease (AD) and age-related dementia are affected by the ability of the immune system to contain the brain's pathology. Accordingly, well-controlled boosting, rather than suppression of systemic immunity, has been suggested as a new approach to modify disease pathology without directly targeting any of the brain's disease hallmarks. Here, we provide a short review of the mechanisms orchestrating the cross-talk between the brain and the immune system. We then discuss how immune checkpoint blockade directed against the PD-1/PD-L1 pathways could be developed as an immunotherapeutic approach to combat this disease using a regimen that will address the needs to combat AD.
Collapse
|
48
|
Pappalardo F, Rajput AM, Motta S. Computational modeling of brain pathologies: the case of multiple sclerosis. Brief Bioinform 2019; 19:318-324. [PMID: 28011755 DOI: 10.1093/bib/bbw123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Indexed: 01/09/2023] Open
Abstract
The central nervous system is the most complex network of the human body. The existence and functionality of a large number of molecular species in human brain are still ambiguous and mostly unknown, thus posing a challenge to Science and Medicine. Neurological diseases inherit the same level of complexity, making effective treatments difficult to be found. Multiple sclerosis (MS) is a major neurological disease that causes severe inabilities and also a significant social burden on health care system: between 2 and 2.5 million people are affected by it, and the cost associated with it is significantly higher as compared with other neurological diseases because of the chronic nature of the disease and to the partial efficacy of current therapies. Despite difficulties in understanding and treating MS, many computational models have been developed to help neurologists. In the present work, we briefly review the main characteristics of MS and present a selection criteria of modeling approaches.
Collapse
Affiliation(s)
| | | | - Santo Motta
- Istitute for Applied Calculus (IAC) "M. Picone", National Research Council of Italy (CNR), Italy
| |
Collapse
|
49
|
Rosenzweig N, Dvir-Szternfeld R, Tsitsou-Kampeli A, Keren-Shaul H, Ben-Yehuda H, Weill-Raynal P, Cahalon L, Kertser A, Baruch K, Amit I, Weiner A, Schwartz M. PD-1/PD-L1 checkpoint blockade harnesses monocyte-derived macrophages to combat cognitive impairment in a tauopathy mouse model. Nat Commun 2019; 10:465. [PMID: 30692527 PMCID: PMC6349941 DOI: 10.1038/s41467-019-08352-5] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 01/02/2019] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous disorder with multiple etiologies. Harnessing the immune system by blocking the programmed cell death receptor (PD)-1 pathway in an amyloid beta mouse model was shown to evoke a sequence of immune responses that lead to disease modification. Here, blocking PD-L1, a PD-1 ligand, was found to have similar efficacy to that of PD-1 blocking in disease modification, in both animal models of AD and of tauopathy. Targeting PD-L1 in a tau-driven disease model resulted in increased immunomodulatory monocyte-derived macrophages within the brain parenchyma. Single cell RNA-seq revealed that the homing macrophages expressed unique scavenger molecules including macrophage scavenger receptor 1 (MSR1), which was shown here to be required for the effect of PD-L1 blockade in disease modification. Overall, our results demonstrate that immune checkpoint blockade targeting the PD-1/PD-L1 pathway leads to modification of common factors that go awry in AD and dementia, and thus can potentially provide an immunotherapy to help combat these diseases.
Collapse
Affiliation(s)
- Neta Rosenzweig
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Raz Dvir-Szternfeld
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | | | - Hadas Keren-Shaul
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Hila Ben-Yehuda
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Pierre Weill-Raynal
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Liora Cahalon
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Alex Kertser
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Kuti Baruch
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Assaf Weiner
- Department of Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
50
|
Chi ZL, Adini A, Birsner AE, Bazinet L, Akula JD, D'Amato RJ. PR1P ameliorates neurodegeneration through activation of VEGF signaling pathway and remodeling of the extracellular environment. Neuropharmacology 2018; 148:96-106. [PMID: 30594697 DOI: 10.1016/j.neuropharm.2018.12.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 11/26/2018] [Accepted: 12/26/2018] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases affect millions of people worldwide. Optic neuropathies are the most commonly occurring neurodegenerative diseases, characterized by progressive retinal ganglion cell (RGC) degeneration. We recently reported that Prominin-1, a protein found on the surface of stem cells, interacts with VEGF and enhances its activity. VEGF is known to have various protective roles in the nervous system. Subsequently, we have developed a 12-mer peptide derived from Prominin-1, named PR1P, and investigated its effects on neuronal survival of damaged RGCs in a rat model of optic nerve crush (ONC). PR1P prevented RGC apoptosis resulting in improvement of retinal function in the rat ONC model. PR1P treatment significantly increased phosphorylation of ERK and AKT and expression its downstream proteins c-fos and Egr-1 in the retina. Additionally, PR1P beneficially increased the MMP-9/TIMP-1 ratio and promoted glial activation in the retina of ONC rats. Thus, PR1P displayed neuroprotective effects through enhanced VEGF-driven neuronal survival and reconstruction of the extracellular environment in ONC model. Our data indicate that PR1P may be a promising new clinical candidate for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zai-Long Chi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, The Eye Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.
| | - Avner Adini
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Amy E Birsner
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Lauren Bazinet
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
| | - James D Akula
- Department of Ophthalmology, Boston Children's Hospital, Boston, 02115, MA, USA
| | - Robert J D'Amato
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA; Department of Ophthalmology, Harvard Medical School, Boston, 02115, MA, USA.
| |
Collapse
|