1
|
Zhao W, Zhang Z, Xie M, Ding F, Zheng X, Sun S, Du J. Exploring tumor-associated macrophages in glioblastoma: from diversity to therapy. NPJ Precis Oncol 2025; 9:126. [PMID: 40316746 PMCID: PMC12048723 DOI: 10.1038/s41698-025-00920-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Glioblastoma is the most aggressive and lethal cancer of the central nervous system, presenting substantial treatment challenges. The current standard treatment, which includes surgical resection followed by temozolomide and radiation, offers limited success. While immunotherapies, such as immune checkpoint inhibitors, have proven effective in other cancers, they have not demonstrated significant efficacy in GBM. Emerging research highlights the pivotal role of tumor-associated macrophages (TAMs) in supporting tumor growth, fostering treatment resistance, and shaping an immunosuppressive microenvironment. Preclinical studies show promising results for therapies targeting TAMs, suggesting potential in overcoming these barriers. TAMs consist of brain-resident microglia and bone marrow-derived macrophages, both exhibiting diverse phenotypes and functions within the tumor microenvironment. This review delves into the origin, heterogeneity, and functional roles of TAMs in GBM, underscoring their dual roles in tumor promotion and suppression. It also summarizes recent progress in TAM-targeted therapies, which may, in combination with other treatments like immunotherapy, pave the way for more effective and personalized strategies against this aggressive malignancy.
Collapse
Affiliation(s)
- Wenwen Zhao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhi Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mingyuan Xie
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Ding
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangrong Zheng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shicheng Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianyang Du
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
2
|
Shirahama S, Okunuki Y, Lee MY, Karg MM, Refaian N, Krasniqi D, Connor KM, Gregory-Ksander MS, Ksander BR. Preventing the antigen-presenting function of retinal microglia blocks autoimmune neuroinflammation by dendritic cell-primed CD4 + T cells. J Autoimmun 2025; 153:103417. [PMID: 40239533 DOI: 10.1016/j.jaut.2025.103417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/10/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Autoimmune uveitis is a major cause of blindness and experimental autoimmune uveitis (EAU) is mediated by interphotoreceptor retinoid-binding protein specific effector CD4+ T cells that infiltrate the retina. At least two MHC Class II (MHC II) antigen-presenting cell (APC) events are required for uveitis to develop. The first occurs in the secondary lymphoid organs when dendritic cells (DCs) activate and expand effector CD4+ T cells that enter the circulation and migrate systemically. The second APC event occurs when DC-primed effector CD4+ T cells infiltrate the retina and are restimulated by the relevant autoantigen. Importantly, if this second restimulation does not occur, then uveitis does not develop. However, it is still unclear which cell type(s) function as APCs within the retina. There are two candidate MHC II+ cell types-resident microglia and infiltrating DCs. We used the inducible Cre-lox approach to develop mouse strains in which MHC II was knocked out specifically on microglia using either the P2ry12 or Tmem119 gene to drive recombination. We also used Itgax (CD11c encoding gene) to drive recombination in DCs. Using this approach, we uncovered that the second APC event was mediated by MHC II+ microglia and not infiltrating MHC II+ DCs. Therefore, microglia are an important therapeutic target that can prevent and/or diminish uveitis even in the presence of circulating retinal autoantigen-specific effector CD4+ T cells.
Collapse
Affiliation(s)
- Shintaro Shirahama
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA.
| | - Yoko Okunuki
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - May Y Lee
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Margarete M Karg
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Nasrin Refaian
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Drenushe Krasniqi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kip M Connor
- Angiogenesis Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA.
| | - Meredith S Gregory-Ksander
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Bruce R Ksander
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
3
|
Theis T, Kumar S, Shah P, Patel M, Tadmori I, Ayala C, Tschang M, Young W, Schachner M. Depletion of Cell Adhesion Molecule L1 from Microglia and Macrophages Reduces Recovery After Spinal Cord Injury. Int J Mol Sci 2025; 26:3285. [PMID: 40244153 PMCID: PMC11989546 DOI: 10.3390/ijms26073285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
The young mammalian central nervous system regenerates after spinal cord injury and recovers locomotion, whereas adult mice only show limited recovery that depends on the injury severity, genetic background, and physical therapy. At the molecular level, key regulators that contribute to recovery are cell adhesion molecules, such as L1CAM (L1). At the cell surface, L1 functions as a homotypic receptor that signal-transduces crucial functions in neuronal migration and survival, neurite outgrowth, myelination, formation of synapses, and synaptic plasticity. In the adult central nervous system, L1 is expressed only by neurons. We now show that L1 is unexpectedly also expressed by 26% microglia, freshly isolated from a 7-day-old mouse brain. At postnatal day 21, only 3% of microglia are L1-positive. Using a mouse mutant in which L1 is deleted specifically in monocytes of 10- to 14-week-old mice, functional recovery was reduced up to 4 weeks after injury at lower thoracic spinal levels. Also, NF200-immunoreactive and 5-HT-immunoreactive fibers were found decreased below the injury site as compared to wild-type mice. In conclusion, microglial cells that express L1 stimulate neurite outgrowth in vitro, improve functional recovery after spinal cord injury in adult mice, and increase fiber densities caudal to the lesion site.
Collapse
Affiliation(s)
- Thomas Theis
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08844, USA;
| | - Pratiksha Shah
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Mukti Patel
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Iman Tadmori
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Carlos Ayala
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Monica Tschang
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Wise Young
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| |
Collapse
|
4
|
Ju XD, Zhang PH, Li Q, Bai QY, Hu B, Xu J, Lu C. Peripheral Blood Monocytes as Biomarkers of Neurodevelopmental Disorders: A Systematic Review and Meta-Analysis. Res Child Adolesc Psychopathol 2025; 53:583-595. [PMID: 40053221 DOI: 10.1007/s10802-025-01303-3] [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] [Accepted: 02/20/2025] [Indexed: 04/26/2025]
Abstract
Accumulating evidence implicates immune dysregulation and chronic inflammation in neurodevelopmental disorders (NDDs), often manifesting as abnormal alterations in peripheral blood immune cell levels. The mononuclear phagocyte system, including monocytes and microglia, has been increasingly recognized for its involvement in the pathogenesis of NDDs. However, due to inconsistent findings in the literature, whether monocytes can serve as a reliable biomarker for NDDs remains controversial. To address this issue, we conducted a systematic review and meta-analysis of studies examining monocyte counts in NDD individuals. A comprehensive search was conducted across PubMed, Web of Science, and Scopus databases. Variables extracted for analysis encompassed the author's name, year of study, sample size, patient's age, type of disease, mean, standard deviation of monocytes and sex ratio. A total of 2503 articles were found by searching the three databases. After removed duplicates and screening titles, abstracts, and full texts, 17 articles met the inclusion criteria, and 20 independent studies were included in the meta-analysis. The results indicated significantly increased monocyte counts in 5 type NDDs compared to Typical Development (TD) groups (g = 0.36, 95%CI [0.23, 0.49]). Subgroup analyses revealed no significant differences in monocyte counts across different NDD types, gender, or age. These findings suggest that aberrant alterations in monocyte counts are common in NDD cases, indicating their potential as biomarkers for these conditions. Future research should further investigate the role of monocyte in understanding the mechanisms, early detection, and clinical diagnosis of NDDs.
Collapse
Affiliation(s)
- Xing-Da Ju
- School of Psychology, Northeast Normal University, Changchun, China
- Jilin Provincial Key Laboratory of Cognitive Neuroscience and Brain Development, Changchun, China
- Autism Centre of Excellence, Northeast Normal University, Changchun, China
| | - Pai-Hao Zhang
- School of Psychology, Northeast Normal University, Changchun, China
| | - Qiang Li
- School of Psychology, Northeast Normal University, Changchun, China
| | - Qiu-Yu Bai
- Yancheng College of Mechatronic Technology, Yancheng, China
| | - Bo Hu
- School of Psychology, Northeast Normal University, Changchun, China
- School of Social and Behavioral Science, Nanjing University, Nanjing, China
| | - Jing Xu
- School of Life Sciences, Northeast Normal University, Changchun, China
| | - Chang Lu
- School of Psychology, Northeast Normal University, Changchun, China.
- Jilin Provincial Key Laboratory of Cognitive Neuroscience and Brain Development, Changchun, China.
| |
Collapse
|
5
|
Ismailov A, Spallone A, Belogurov A, Herbert A, Poptsova M. Molecular biology of the deadliest cancer - glioblastoma: what do we know? Front Immunol 2025; 16:1530305. [PMID: 40191211 PMCID: PMC11968700 DOI: 10.3389/fimmu.2025.1530305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Glioblastomas are the most prevalent primary brain tumors and are associated with a dramatically poor prognosis. Despite an intensive treatment approach, including maximal surgical tumor removal followed by radio- and chemotherapy, the median survival for glioblastoma patients has remained around 18 months for decades. Glioblastoma is distinguished by its highly complex mechanisms of immune evasion and pronounced heterogeneity. This variability is apparent both within the tumor itself, which can exhibit multiple phenotypes simultaneously, and in its surrounding microenvironment. Another key feature of glioblastoma is its "cold" microenvironment, characterized by robust immunosuppression. Recent advances in single-cell RNA sequencing have uncovered new promising insights, revealing previously unrecognized aspects of this tumor. In this review, we consolidate current knowledge on glioblastoma cells and its microenvironment, with an emphasis on their biological properties and unique patterns of molecular communication through signaling pathways. The evidence underscores the critical need for personalized poly-immunotherapy and other approaches to overcome the plasticity of glioblastoma stem cells. Analyzing the tumor microenvironment of individual patients using single-cell transcriptomics and implementing a customized immunotherapeutic strategy could potentially improve survival outcomes for those facing this formidable disease.
Collapse
Affiliation(s)
- Aly Ismailov
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
| | - Aldo Spallone
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
- Laboratory of Hormonal Regulation Proteins, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
| | - Alexey Belogurov
- Laboratory of Hormonal Regulation Proteins, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
- Scientific and Educational Institute of Fundamental Medicine named after V.I. Pokrovsky, Department of Biological Chemistry, Russian University of Medicine, Moscow, Russia
| | - Alan Herbert
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
- Discovery Department, InsideOutBio, Boston, MA, United States
| | - Maria Poptsova
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
| |
Collapse
|
6
|
Tuddenham JF, Taga M, Haage V, Marshe VS, Roostaei T, White C, Lee AJ, Fujita M, Khairallah A, Zhang Y, Green G, Hyman B, Frosch M, Hopp S, Beach TG, Serrano GE, Corboy J, Habib N, Klein HU, Soni RK, Teich AF, Hickman RA, Alcalay RN, Shneider N, Schneider J, Sims PA, Bennett DA, Olah M, Menon V, De Jager PL. A cross-disease resource of living human microglia identifies disease-enriched subsets and tool compounds recapitulating microglial states. Nat Neurosci 2024; 27:2521-2537. [PMID: 39406950 DOI: 10.1038/s41593-024-01764-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/13/2024] [Indexed: 12/06/2024]
Abstract
Human microglia play a pivotal role in neurological diseases, but we still have an incomplete understanding of microglial heterogeneity, which limits the development of targeted therapies directly modulating their state or function. Here, we use single-cell RNA sequencing to profile 215,680 live human microglia from 74 donors across diverse neurological diseases and CNS regions. We observe a central divide between oxidative and heterocyclic metabolism and identify microglial subsets associated with antigen presentation, motility and proliferation. Specific subsets are enriched in susceptibility genes for neurodegenerative diseases or the disease-associated microglial signature. We validate subtypes in situ with an RNAscope-immunofluorescence pipeline and high-dimensional MERFISH. We also leverage our dataset as a classification resource, finding that induced pluripotent stem cell model systems capture substantial in vivo heterogeneity. Finally, we identify and validate compounds that recapitulate certain subtypes in vitro, including camptothecin, which downregulates the signature of disease-enriched subtypes and upregulates a signature previously associated with Alzheimer's disease.
Collapse
Affiliation(s)
- John F Tuddenham
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Mariko Taga
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Verena Haage
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Victoria S Marshe
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Tina Roostaei
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles White
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Annie J Lee
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Masashi Fujita
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anthony Khairallah
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Gilad Green
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Bradley Hyman
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthew Frosch
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Sarah Hopp
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | | | | | - John Corboy
- Department of Neurology, University of Colorado, and Rocky Mountain Multiple Sclerosis Center at the University of Colorado, Aurora, CO, USA
| | - Naomi Habib
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hans-Ulrich Klein
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Rajesh Kumar Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Andrew F Teich
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Richard A Hickman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Movement Disorders Division, Neurological Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Neil Shneider
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Eleanor and Lou Gehrig ALS Center, Columbia University Medical Center, New York, NY, USA
| | - Julie Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Marta Olah
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Philip L De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
7
|
Solomou G, Young AMH, Bulstrode HJCJ. Microglia and macrophages in glioblastoma: landscapes and treatment directions. Mol Oncol 2024; 18:2906-2926. [PMID: 38712663 PMCID: PMC11619806 DOI: 10.1002/1878-0261.13657] [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: 11/28/2023] [Revised: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 05/08/2024] Open
Abstract
Glioblastoma is the most common primary malignant tumour of the central nervous system and remains uniformly and rapidly fatal. The tumour-associated macrophage (TAM) compartment comprises brain-resident microglia and bone marrow-derived macrophages (BMDMs) recruited from the periphery. Immune-suppressive and tumour-supportive TAM cell states predominate in glioblastoma, and immunotherapies, which have achieved striking success in other solid tumours have consistently failed to improve survival in this 'immune-cold' niche context. Hypoxic and necrotic regions in the tumour core are found to enrich, especially in anti-inflammatory and immune-suppressive TAM cell states. Microglia predominate at the invasive tumour margin and express pro-inflammatory and interferon TAM cell signatures. Depletion of TAMs, or repolarisation towards a pro-inflammatory state, are appealing therapeutic strategies and will depend on effective understanding and classification of TAM cell ontogeny and state based on new single-cell and spatial multi-omic in situ profiling. Here, we explore the application of these datasets to expand and refine TAM characterisation, to inform improved modelling approaches, and ultimately underpin the effective manipulation of function.
Collapse
Affiliation(s)
- Georgios Solomou
- Wellcome MRC Cambridge Stem Cell InstituteUniversity of CambridgeUK
- Department of NeurosurgeryAddenbrooke's HospitalCambridgeUK
| | - Adam M. H. Young
- Wellcome MRC Cambridge Stem Cell InstituteUniversity of CambridgeUK
- Department of NeurosurgeryAddenbrooke's HospitalCambridgeUK
| | - Harry J. C. J. Bulstrode
- Wellcome MRC Cambridge Stem Cell InstituteUniversity of CambridgeUK
- Department of NeurosurgeryAddenbrooke's HospitalCambridgeUK
| |
Collapse
|
8
|
Sousa NS, Bica M, Brás MF, Sousa AC, Antunes IB, Encarnação IA, Costa TM, Martins IB, Barbosa-Morais NL, Sousa-Victor P, Neves J. The immune landscape of murine skeletal muscle regeneration and aging. Cell Rep 2024; 43:114975. [PMID: 39541212 DOI: 10.1016/j.celrep.2024.114975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 09/16/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Age-related alterations in the immune system are starting to emerge as key contributors to impairments found in aged organs. A decline in regenerative capacity is a hallmark of tissue aging; however, the contribution of immune aging to regenerative failure is just starting to be explored. Here, we apply a strategy combining single-cell RNA sequencing with flow cytometry, histological analysis, and functional assays to perform a complete analysis of the immune environment of the aged regenerating skeletal muscle on a time course following injury with single-cell resolution. Our results reveal an unanticipated complexity and functional heterogeneity in immune populations within the skeletal muscle that have been regarded as homogeneous. Furthermore, we uncover a profound remodeling of both myeloid and lymphoid compartments in aging. These discoveries challenge established notions on immune regulation of skeletal muscle regeneration, providing a set of potential targets to improve skeletal muscle health and regenerative capacity in aging.
Collapse
Affiliation(s)
- Neuza S Sousa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Marta Bica
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Margarida F Brás
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Ana C Sousa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Inês B Antunes
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Isabel A Encarnação
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal
| | - Tiago M Costa
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Inês B Martins
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | | | - Pedro Sousa-Victor
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal.
| | - Joana Neves
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisbon, Portugal.
| |
Collapse
|
9
|
Gern OL, Pavlou A, Mulenge F, Busker LM, Ghita L, Aringo A, Costa B, Spanier J, Waltl I, Stangel M, Kalinke U. MAVS signaling shapes microglia responses to neurotropic virus infection. J Neuroinflammation 2024; 21:264. [PMID: 39425188 PMCID: PMC11490141 DOI: 10.1186/s12974-024-03258-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
Viral encephalitis is characterized by a series of immunological reactions that can control virus infection in the brain, but dysregulated responses may cause excessive inflammation and brain damage. Microglia are brain-resident myeloid cells that are specialized in surveilling the local CNS environment and in case of viral brain infection they contribute to the control of the infection and to restriction of viral dissemination. Here, we report that after exposure to neurotropic vesicular stomatitis virus (VSV), murine in vitro microglia cultures showed rapid upregulation of a broad range of pro-inflammatory and antiviral genes, which were stably expressed over the entire 8 h infection period. Additionally, a set of immunomodulatory genes was upregulated between 6 and 8 h post infection. In microglia cultures, the induction of several immune response pathways including cytokine responses was dependent on mitochondrial antiviral-signaling protein (MAVS). Consequently, in Mavs-deficient microglia the control of virus propagation failed as indicated by augmented virus titers and the accumulation of viral transcripts. Thus, in the analyzed in vitro system, MAVS signaling is critically required to achieve full microglia activation and to mediate profound antiviral effects. In Mavs-deficient mice, intranasal VSV instillation caused higher disease severity than in WT mice and virus dissemination was noticed beyond the olfactory bulb. Virus spread to inner regions of the olfactory bulb, i.e., the granular cell layer, correlated with the recruitment of highly inflammatory non-microglia myeloid cells into the olfactory bulb in Mavs-/- mice. Furthermore, increased cytokine levels were detected in the nasal cavity, the olfactory bulb and in other brain regions. Thus, microglial MAVS signaling is critically needed for virus sensing, full microglia activation, and for orchestration of protective immunity in the virus-infected CNS.
Collapse
Affiliation(s)
- Olivia Luise Gern
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Andreas Pavlou
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Felix Mulenge
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Lena Mareike Busker
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Luca Ghita
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
- Genentech, South San Francisco, CA, USA
| | - Angela Aringo
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Bibiana Costa
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany
| | - Martin Stangel
- Department of Neurology, Hannover Medical School, 30625, Hannover, Germany
- Translational Medicine Neuroscience, Biomedical Research, Novartis Pharma AG, Basel, 4056, Switzerland
- Center of Systems Neuroscience, Hannover, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, Centre for Experimental and Clinical Infection Research, TWINCORE, Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, 30625, Hannover, Germany.
- Cluster of Excellence-Resolving Infection Susceptibility (RESIST, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| |
Collapse
|
10
|
Pan J, Fores-Martos J, Delpirou Nouh C, Jensen TD, Vallejo K, Cayrol R, Ahmadian S, Ashley EA, Greicius MD, Cobos I. Deciphering glial contributions to CSF1R-related disorder via single-nuclear transcriptomic profiling: a case study. Acta Neuropathol Commun 2024; 12:139. [PMID: 39217398 PMCID: PMC11365264 DOI: 10.1186/s40478-024-01853-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
CSF1R-related disorder (CSF1R-RD) is a neurodegenerative condition that predominantly affects white matter due to genetic alterations in the CSF1R gene, which is expressed by microglia. We studied an elderly man with a hereditary, progressive dementing disorder of unclear etiology. Standard genetic testing for leukodystrophy and other neurodegenerative conditions was negative. Brain autopsy revealed classic features of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), including confluent white matter degeneration with axonal spheroids and pigmented glial cells in the affected white matter, consistent with CSF1R-RD. Subsequent long-read sequencing identified a novel deletion in CSF1R that was not detectable with short-read exome sequencing. To gain insight into potential mechanisms underlying white matter degeneration in CSF1R-RD, we studied multiple brain regions exhibiting varying degrees of white matter pathology. We found decreased CSF1R transcript and protein across brain regions, including intact white matter. Single nuclear RNA sequencing (snRNAseq) identified two disease-associated microglial cell states: lipid-laden microglia (expressing GPNMB, ATG7, LGALS1, LGALS3) and inflammatory microglia (expressing IL2RA, ATP2C1, FCGBP, VSIR, SESN3), along with a small population of CD44+ peripheral monocyte-derived macrophages exhibiting migratory and phagocytic signatures. GPNMB+ lipid-laden microglia with ameboid morphology represented the end-stage disease microglia state. Disease-associated oligodendrocytes exhibited cell stress signatures and dysregulated apoptosis-related genes. Disease-associated oligodendrocyte precursor cells (OPCs) displayed a failure in their differentiation into mature myelin-forming oligodendrocytes, as evidenced by upregulated LRP1, PDGFRA, SOX5, NFIA, and downregulated NKX2-2, NKX6.2, SOX4, SOX8, TCF7L2, YY1, ZNF488. Overall, our findings highlight microglia-oligodendroglia crosstalk in demyelination, with CSF1R dysfunction promoting phagocytic and inflammatory microglia states, an arrest in OPC differentiation, and oligodendrocyte depletion.
Collapse
Affiliation(s)
- Jie Pan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jaume Fores-Martos
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Claire Delpirou Nouh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tanner D Jensen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristen Vallejo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Romain Cayrol
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Saman Ahmadian
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Euan A Ashley
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Inma Cobos
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
11
|
Fang Y, Dou A, Shen Y, Li T, Liu H, Cui Y, Xie K. Association of triglyceride-glucose index and delirium in patients with sepsis: a retrospective study. Lipids Health Dis 2024; 23:227. [PMID: 39054513 PMCID: PMC11271053 DOI: 10.1186/s12944-024-02213-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE It is well known that glucose and lipid metabolism disorders and insulin resistance are common in sepsis, which affect the occurrence and prognosis of multiple organ dysfunction in septic patients. Previous study reported the predictive value of triglyceride-glucose index (TyG), a clinical indicator for insulin resistance, in postoperative delirium patients. However, it remains unclear whether the TyG index is a novel predictive biomarker for sepsis-associated delirium. The aim of this study is to explore the relationship between TyG index and the risk of delirium in patients with sepsis. METHODS Adult septic patients were identified from the MIMIC-IV database and divided into four groups based on the mean value of TyG. The primary outcome was the incidence of delirium. The association between TyG and the risk of developing delirium was evaluated by restricted cubic spline (RCS), multivariate logistic regression and subgroup analysis. Propensity Score Matching (PSM) method was used to balance the baseline data. RESULTS A total of 3,331 septic patients were included in the analysis, and further divided into four groups: Q1 (TyG ≤ 8.67), Q2 (8.67 < TyG ≤ 9.08), Q3 (9.08 < TyG ≤ 9.61), and Q4 (TyG > 9.61). The RCS curves demonstrated a non-linear positive relationship between TyG index and the risk of developing delirium, and an optimal cut-of value 9.09 was recommended. After balancing the baseline information by PSM, patients in the TyG > 9.09 group had a significant higher incidence of delirium compared with those in the TyG ≤ 9.09 group. In logistic regression analysis, TyG > 9.09 was significantly associated with lower risk of developing delirium in both original cohort (OR 1.54-1.78, all P < 0.001) and the PSM cohort (OR 1.41-1.48, all P < 0.001). No association was found between the TyG index and mortality (all P > 0.05). In subgroup analysis, our findings were consistent (all OR > 1 in all subgroups). CONCLUSION Our study demonstrated an independent association between TyG index and increased risk of delirium in septic patients, indicating that TyG index can serve as a biomarker for delirium in sepsis.
Collapse
Affiliation(s)
- Yipeng Fang
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Aizhen Dou
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Yuehao Shen
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Tianyu Li
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Haiying Liu
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China
| | - Yan Cui
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China.
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22, Qixiangtai Road, Heping District, Tianjin, 300070, China.
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, 154th Anshan Road, Tianjin, 300052, China.
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
12
|
Gardner RS, Kyle M, Hughes K, Zhao LR. Single-Cell RNA Sequencing Reveals Immunomodulatory Effects of Stem Cell Factor and Granulocyte Colony-Stimulating Factor Treatment in the Brains of Aged APP/PS1 Mice. Biomolecules 2024; 14:827. [PMID: 39062541 PMCID: PMC11275138 DOI: 10.3390/biom14070827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) leads to progressive neurodegeneration and dementia. AD primarily affects older adults with neuropathological changes including amyloid-beta (Aβ) deposition, neuroinflammation, and neurodegeneration. We have previously demonstrated that systemic treatment with combined stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) (SCF+G-CSF) reduces the Aβ load, increases Aβ uptake by activated microglia and macrophages, reduces neuroinflammation, and restores dendrites and synapses in the brains of aged APPswe/PS1dE9 (APP/PS1) mice. However, the mechanisms underlying SCF+G-CSF-enhanced brain repair in aged APP/PS1 mice remain unclear. This study used a transcriptomic approach to identify the potential mechanisms by which SCF+G-CSF treatment modulates microglia and peripheral myeloid cells to mitigate AD pathology in the aged brain. After injections of SCF+G-CSF for 5 consecutive days, single-cell RNA sequencing was performed on CD11b+ cells isolated from the brains of 28-month-old APP/PS1 mice. The vast majority of cell clusters aligned with transcriptional profiles of microglia in various activation states. However, SCF+G-CSF treatment dramatically increased a cell population showing upregulation of marker genes related to peripheral myeloid cells. Flow cytometry data also revealed an SCF+G-CSF-induced increase of cerebral CD45high/CD11b+ active phagocytes. SCF+G-CSF treatment robustly increased the transcription of genes implicated in immune cell activation, including gene sets that regulate inflammatory processes and cell migration. The expression of S100a8 and S100a9 was robustly enhanced following SCF+G-CSF treatment in all CD11b+ cell clusters. Moreover, the topmost genes differentially expressed with SCF+G-CSF treatment were largely upregulated in S100a8/9-positive cells, suggesting a well-conserved transcriptional profile related to SCF+G-CSF treatment in resident and peripherally derived CD11b+ immune cells. This S100a8/9-associated transcriptional profile contained notable genes related to pro-inflammatory and anti-inflammatory responses, neuroprotection, and Aβ plaque inhibition or clearance. Altogether, this study reveals the immunomodulatory effects of SCF+G-CSF treatment in the aged brain with AD pathology, which will guide future studies to further uncover the therapeutic mechanisms.
Collapse
Affiliation(s)
| | | | | | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, 750 E. Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
13
|
Gardner R, Kyle M, Hughes K, Zhao LR. Single cell RNA sequencing reveals immunomodulatory effects of stem cell factor and granulocyte colony-stimulating factor treatment in the brains of aged APP/PS1 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593359. [PMID: 38766064 PMCID: PMC11100789 DOI: 10.1101/2024.05.09.593359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Alzheimers disease leads to progressive neurodegeneration and dementia. Alzheimers disease primarily affects older adults with neuropathological changes including amyloid beta deposition, neuroinflammation, and neurodegeneration. We have previously demonstrated that systemic treatment with combined stem cell factor, SCF, and granulocyte colony stimulating factor, GCSF, reduces amyloid beta load, increases amyloid beta uptake by activated microglia and macrophages, reduces neuroinflammation, and restores dendrites and synapses in the brains of aged APP-PS1 mice. However, the mechanisms underlying SCF-GCSF-enhanced brain repair in aged APP-PS1 mice remain unclear. This study used a transcriptomic approach to identify potential mechanisms by which SCF-GCSF treatment modulates microglia and peripheral myeloid cells to mitigate Alzheimers disease pathology in the aged brain. After injections of SCF-GCSF for 5 consecutive days, single cell RNA sequencing was performed on CD11b positive cells isolated from the brains of 28-month-old APP-PS1 mice. The vast majority of cell clusters aligned with transcriptional profiles of microglia in various activation states. However, SCF-GCSF treatment dramatically increased a cell population showing upregulation of marker genes related to peripheral myeloid cells. Flow cytometry data also revealed an SCF-GCSF-induced increase of cerebral CD45high-CD11b positive active phagocytes. SCF-GCSF treatment robustly increased the transcription of genes implicated in immune cell activation, including gene sets that regulate inflammatory processes and cell migration. Expression of S100a8 and S100a9 were robustly enhanced following SCF-GCSF treatment in all CD11b positive cell clusters. Moreover, the topmost genes differentially expressed with SCF-GCSF treatment were largely upregulated in S100a8-S100a9 positive cells, suggesting a well-conserved transcriptional profile related to SCF-GCSF treatment in resident and peripherally derived CD11b positive immune cells. This S100a8-S100a9-associated transcriptional profile contained notable genes related to proinflammatory and antiinflammatory responses, neuroprotection, and amyloid beta plaque inhibition or clearance. Altogether, this study reveals immunomodulatory effects of SCF-GCSF treatment in the aged brain with Alzheimers disease pathology, which will guide future studies to further uncover the therapeutic mechanisms.
Collapse
|
14
|
Felício R, Franco A, Corrêa-Moreira D, Martinez de Carvalho F, Guimarães A, Vargas FR. Humoral immune transcriptome signature in myelomeningocele patients. J Reprod Immunol 2024; 163:104224. [PMID: 38479055 DOI: 10.1016/j.jri.2024.104224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Myelomeningocele (MMC) results from incomplete closure of the neural tube, and has a complex multifactorial etiology, including an inflammatory microenvironment. OBJECTIVE We evaluated the contribution of humoral immune response for development of inflammatory milieu. METHODS Using public repository Gene Expression Omnibus (GEO), we retrieve dataset transcriptome from the amniotic fluid of ten fetuses with myelomeningocele and ten healthy control fetuses to found differential gene expression associated with disturbances and inflammatory signatures in MMC. The identified DEGs were submitted to enrichment, network, and matrix correlation analyses. RESULTS Our initial analysis revealed 90 DEGs in MMC, mainly associated with signaling pathways of inflammation, including the immune modules, humoral immune response and IFN-type I signatures. Protein-protein analysis (PPI) revealed an association with three protein networks; positive regulation of B cell proliferation constituted the largest network. Matrix correlation analyses showed that MMC alters the co-expression of genes related to inflammatory processes that promote microenvironment inflammation. CONCLUSION These results revealed an altered humoral immune response in MMC patients, contributing to an inflammatory profile and providing opportunities for identifying potential biomarkers in myelomeningocele disease.
Collapse
Affiliation(s)
- Rfm Felício
- Birth Defects Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rede de Atenção à Saúde nas Anomalias Congênitas do Sistema Nervoso Central, Instituo Oswaldo Cruz, Fundação Oswaldo Cruz, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Almm Franco
- Birth Defects Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Programa de Pós-Graduação em Ciências Biológicas (Genética), Universidade Federal do Rio de Janeiro, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - D Corrêa-Moreira
- Rede de Atenção à Saúde nas Anomalias Congênitas do Sistema Nervoso Central, Instituo Oswaldo Cruz, Fundação Oswaldo Cruz, Brazil; Laboratory of Taxonomy, Biochesmistry and Bioprospecting of Fungi, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - F Martinez de Carvalho
- Birth Defects Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rede de Atenção à Saúde nas Anomalias Congênitas do Sistema Nervoso Central, Instituo Oswaldo Cruz, Fundação Oswaldo Cruz, Brazil; Programa de Pós-Graduação em Ciências Biológicas (Genética), Universidade Federal do Rio de Janeiro, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Acr Guimarães
- Laboratory of Taxonomy, Biochesmistry and Bioprospecting of Fungi, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - F R Vargas
- Birth Defects Epidemiology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Rede de Atenção à Saúde nas Anomalias Congênitas do Sistema Nervoso Central, Instituo Oswaldo Cruz, Fundação Oswaldo Cruz, Brazil; Programa de Pós-Graduação em Ciências Biológicas (Genética), Universidade Federal do Rio de Janeiro, Brazil; Laboratory for Applied Genomics and Bioinnovations, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Departamento de Genética e Biologia Molecular, Universidade Federal do Estado do Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Pallarés-Moratalla C, Bergers G. The ins and outs of microglial cells in brain health and disease. Front Immunol 2024; 15:1305087. [PMID: 38665919 PMCID: PMC11043497 DOI: 10.3389/fimmu.2024.1305087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Microglia are the brain's resident macrophages that play pivotal roles in immune surveillance and maintaining homeostasis of the Central Nervous System (CNS). Microglia are functionally implicated in various cerebrovascular diseases, including stroke, aneurysm, and tumorigenesis as they regulate neuroinflammatory responses and tissue repair processes. Here, we review the manifold functions of microglia in the brain under physiological and pathological conditions, primarily focusing on the implication of microglia in glioma propagation and progression. We further review the current status of therapies targeting microglial cells, including their re-education, depletion, and re-population approaches as therapeutic options to improve patient outcomes for various neurological and neuroinflammatory disorders, including cancer.
Collapse
|
16
|
Potru PS, Vidovic N, Wiemann S, Russ T, Trautmann M, Spittau B. A Custom Panel for Profiling Microglia Gene Expression. Cells 2024; 13:630. [PMID: 38607069 PMCID: PMC11012202 DOI: 10.3390/cells13070630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
Despite being immune cells of the central nervous system (CNS), microglia contribute to CNS development, maturation, and homeostasis, and microglia dysfunction has been implicated in several neurological disorders. Recent advancements in single-cell studies have uncovered unique microglia-specific gene expression. However, there is a need for a simple yet elegant multiplexed approach to quantifying microglia gene expression. To address this, we have designed a NanoString nCounter technology-based murine microglia-specific custom codeset comprising 178 genes. We analyzed RNA extracted from ex vivo adult mouse microglia, primary mouse microglia, the BV2 microglia cell line, and mouse bone marrow monocytes using our custom panel. Our findings reveal a pattern where homeostatic genes exhibit heightened expression in adult microglia, followed by primary cells, and are absent in BV2 cells, while reactive markers are elevated in primary microglia and BV2 cells. Analysis of publicly available data sets for the genes present in the panel revealed that the panel could reliably reflect the changes in microglia gene expression in response to various factors. These findings highlight that the microglia panel used offers a swift and cost-effective means to assess microglial cells and can be used to study them in varying contexts, ranging from normal homeostasis to disease models.
Collapse
Affiliation(s)
- Phani Sankar Potru
- Bielefeld University, Medical School OWL, Anatomy and Cell Biology, 33615 Bielefeld, Germany; (P.S.P.); (N.V.); (S.W.); (T.R.)
| | - Natascha Vidovic
- Bielefeld University, Medical School OWL, Anatomy and Cell Biology, 33615 Bielefeld, Germany; (P.S.P.); (N.V.); (S.W.); (T.R.)
| | - Susanne Wiemann
- Bielefeld University, Medical School OWL, Anatomy and Cell Biology, 33615 Bielefeld, Germany; (P.S.P.); (N.V.); (S.W.); (T.R.)
| | - Tamara Russ
- Bielefeld University, Medical School OWL, Anatomy and Cell Biology, 33615 Bielefeld, Germany; (P.S.P.); (N.V.); (S.W.); (T.R.)
| | - Marcel Trautmann
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, 48149 Münster, Germany
| | - Björn Spittau
- Bielefeld University, Medical School OWL, Anatomy and Cell Biology, 33615 Bielefeld, Germany; (P.S.P.); (N.V.); (S.W.); (T.R.)
| |
Collapse
|
17
|
Parsons BD, Medina-Luna D, Scur M, Pinelli M, Gamage GS, Chilvers RA, Hamon Y, Ahmed IHI, Savary S, Makrigiannis AP, Braverman NE, Rodriguez-Alcazar JF, Latz E, Karakach TK, Di Cara F. Peroxisome deficiency underlies failures in hepatic immune cell development and antigen presentation in a severe Zellweger disease model. Cell Rep 2024; 43:113744. [PMID: 38329874 DOI: 10.1016/j.celrep.2024.113744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Peroxisome biogenesis disorders (PBDs) represent a group of metabolic conditions that cause severe developmental defects. Peroxisomes are essential metabolic organelles, present in virtually every eukaryotic cell and mediating key processes in immunometabolism. To date, the full spectrum of PBDs remains to be identified, and the impact PBDs have on immune function is unexplored. This study presents a characterization of the hepatic immune compartment of a neonatal PBD mouse model at single-cell resolution to establish the importance and function of peroxisomes in developmental hematopoiesis. We report that hematopoietic defects are a feature in a severe PBD murine model. Finally, we identify a role for peroxisomes in the regulation of the major histocompatibility class II expression and antigen presentation to CD4+ T cells in dendritic cells. This study adds to our understanding of the mechanisms of PBDs and expands our knowledge of the role of peroxisomes in immunometabolism.
Collapse
Affiliation(s)
- Brendon D Parsons
- University of Alberta, Department of Laboratory Medicine and Pathology, Edmonton, AB T6G 1C9, Canada
| | - Daniel Medina-Luna
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Michal Scur
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Marinella Pinelli
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Gayani S Gamage
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Rebecca A Chilvers
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Yannick Hamon
- Aix Marseille University, CNRS, INSERM au Centre d'Immunologie de Marseille Luminy, 13288 Marseille, France
| | - Ibrahim H I Ahmed
- Dalhousie University, Department of Pharmacology, Halifax, NS B3H 4R2, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Stéphane Savary
- University of Bourgogne, Laboratoire Bio-PeroxIL EA7270, Dijon, France
| | - Andrew P Makrigiannis
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Nancy E Braverman
- Research Institute of the McGill University Children's Hospital, Montreal, QC H4A 3J1, Canada
| | | | - Eicke Latz
- University of Bonn, Institute of Innate Immunity, Medical Faculty, 53127 Bonn, Germany
| | - Tobias K Karakach
- Dalhousie University, Department of Pharmacology, Halifax, NS B3H 4R2, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| | - Francesca Di Cara
- University of Alberta, Department of Laboratory Medicine and Pathology, Edmonton, AB T6G 1C9, Canada; Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada.
| |
Collapse
|
18
|
Zhao Q, Lai K. Role of immune inflammation regulated by macrophage in the pathogenesis of age-related macular degeneration. Exp Eye Res 2024; 239:109770. [PMID: 38145794 DOI: 10.1016/j.exer.2023.109770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
Age-related macular degeneration (AMD) can lead to irreversible impairment of visual function, and the number of patients with AMD has been increasing globally. The immunoinflammatory theory is an important pathogenic mechanism of AMD, with macrophages serving as the primary inflammatory infiltrating cells in AMD lesions. Its powerful immunoinflammatory regulatory function has attracted considerable attention. Herein, we provide an overview of the involvement of macrophage-regulated immunoinflammation in different stages of AMD. Additionally, we summarize novel therapeutic approaches for AMD, focusing on targeting macrophages, such as macrophage/microglia modulators, reduction of macrophage aggregation in the subretinal space, modulation of macrophage effector function, macrophage phenotypic alterations, and novel biomimetic nanocomposites development based on macrophage-associated functional properties. We aimed to provide a basis and reference for the further exploration of AMD pathogenesis, developmental influences, and new therapeutic approaches.
Collapse
Affiliation(s)
- Qin Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, No.7 Jinsui Road, Guangzhou, 510060, China
| | - Kunbei Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, No.7 Jinsui Road, Guangzhou, 510060, China.
| |
Collapse
|
19
|
Priori EC, Ratto D, De Luca F, Sandionigi A, Savino E, Giammello F, Romeo M, Brandalise F, Roda E, Rossi P. Hericium erinaceus Extract Exerts Beneficial Effects on Gut-Neuroinflammaging-Cognitive Axis in Elderly Mice. BIOLOGY 2023; 13:18. [PMID: 38248449 PMCID: PMC10813749 DOI: 10.3390/biology13010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/23/2024]
Abstract
Ageing is a biological phenomenon that determines the impairment of cognitive performances, in particular, affecting memory. Inflammation and cellular senescence are known to be involved in the pathogenesis of cognitive decline. The gut microbiota-brain axis could exert a critical role in influencing brain homeostasis during ageing, modulating neuroinflammation, and possibly leading to inflammaging. Due to their anti-ageing properties, medicinal mushrooms can be utilised as a resource for developing pharmaceuticals and functional foods. Specifically, Hericium erinaceus (He), thanks to its bioactive metabolites, exerts numerous healthy beneficial effects, such as reinforcing the immune system, counteracting ageing, and improving cognitive performance. Our previous works demonstrated the capabilities of two months of He1 standardised extract oral supplementation in preventing cognitive decline in elderly frail mice. Herein, we showed that this treatment did not change the overall gut microbiome composition but significantly modified the relative abundance of genera specifically involved in cognition and inflammation. Parallelly, a significant decrease in crucial markers of inflammation and cellular senescence, i.e., CD45, GFAP, IL6, p62, and γH2AX, was demonstrated in the dentate gyrus and Cornus Ammonis hippocampal areas through immunohistochemical experiments. In summary, we suggested beneficial and anti-inflammatory properties of He1 in mouse hippocampus through the gut microbiome-brain axis modulation.
Collapse
Affiliation(s)
- Erica Cecilia Priori
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | - Daniela Ratto
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | - Fabrizio De Luca
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | - Anna Sandionigi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy;
- Quantia Consulting S.r.l., Via Petrarca 20, 22066 Mariano Comense, Italy
| | - Elena Savino
- Department of Earth and Environmental Science, University of Pavia, 27100 Pavia, Italy;
| | - Francesca Giammello
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | - Marcello Romeo
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| | | | - Elisa Roda
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy; (E.C.P.); (D.R.); (F.D.L.); (F.G.); (M.R.)
| |
Collapse
|
20
|
du Chatinier A, Velilla IQ, Meel MH, Hoving EW, Hulleman E, Metselaar DS. Microglia in pediatric brain tumors: The missing link to successful immunotherapy. Cell Rep Med 2023; 4:101246. [PMID: 37924816 PMCID: PMC10694606 DOI: 10.1016/j.xcrm.2023.101246] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/10/2023] [Accepted: 09/26/2023] [Indexed: 11/06/2023]
Abstract
Brain tumors are the leading cause of cancer-related mortality in children. Despite the development of immunotherapeutic strategies for adult brain tumors, progress in pediatric neuro-oncology has been hindered by the complex and poorly understood nature of the brain's immune system during early development, a phase that is critical for the onset of many pediatric brain tumors. A defining characteristic of these tumors is the abundance of microglia, the resident immune cells of the central nervous system. In this review, we explore the concept of microglial diversity across brain regions and throughout development and discuss how their maturation stage may contribute to tumor growth in children. We also summarize the current knowledge on the roles of microglia in common pediatric brain tumor entities and provide examples of myeloid-based immunotherapeutic strategies. Our review underscores the importance of microglial plasticity in pediatric brain tumors and its significance for developing effective immunotherapeutic strategies.
Collapse
Affiliation(s)
- Aimée du Chatinier
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Irene Querol Velilla
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Michaël Hananja Meel
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Eelco Wieger Hoving
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Esther Hulleman
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands
| | - Dennis Serge Metselaar
- Department of Neuro-oncology, Princess Máxima Center for Paediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, the Netherlands.
| |
Collapse
|
21
|
Kellogg CM, Pham K, Machalinski AH, Porter HL, Blankenship HE, Tooley KB, Stout MB, Rice HC, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Microglial MHC-I induction with aging and Alzheimer's is conserved in mouse models and humans. GeroScience 2023; 45:3019-3043. [PMID: 37393197 PMCID: PMC10643718 DOI: 10.1007/s11357-023-00859-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023] Open
Abstract
Major histocompatibility complex I (MHC-I) CNS cellular localization and function is still being determined after previously being thought to be absent from the brain. MHC-I expression has been reported to increase with brain aging in mouse, rat, and human whole tissue analyses, but the cellular localization was undetermined. Neuronal MHC-I is proposed to regulate developmental synapse elimination and tau pathology in Alzheimer's disease (AD). Here, we report that across newly generated and publicly available ribosomal profiling, cell sorting, and single-cell data, microglia are the primary source of classical and non-classical MHC-I in mice and humans. Translating ribosome affinity purification-qPCR analysis of 3-6- and 18-22-month-old (m.o.) mice revealed significant age-related microglial induction of MHC-I pathway genes B2m, H2-D1, H2-K1, H2-M3, H2-Q6, and Tap1 but not in astrocytes and neurons. Across a timecourse (12-23 m.o.), microglial MHC-I gradually increased until 21 m.o. and then accelerated. MHC-I protein was enriched in microglia and increased with aging. Microglial expression, and absence in astrocytes and neurons, of MHC-I-binding leukocyte immunoglobulin-like (Lilrs) and paired immunoglobin-like type 2 (Pilrs) receptor families could enable cell -autonomous MHC-I signaling and increased with aging in mice and humans. Increased microglial MHC-I, Lilrs, and Pilrs were observed in multiple AD mouse models and human AD data across methods and studies. MHC-I expression correlated with p16INK4A, suggesting an association with cellular senescence. Conserved induction of MHC-I, Lilrs, and Pilrs with aging and AD opens the possibility of cell-autonomous MHC-I signaling to regulate microglial reactivation with aging and neurodegeneration.
Collapse
Affiliation(s)
- Collyn M Kellogg
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kevin Pham
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Adeline H Machalinski
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Hunter L Porter
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Harris E Blankenship
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla B Tooley
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Heather C Rice
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Amanda L Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Sarah R Ocañas
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA.
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
22
|
Hamel R, Peruzzotti-Jametti L, Ridley K, Testa V, Yu B, Rowitch D, Marioni JC, Pluchino S. Time-resolved single-cell RNAseq profiling identifies a novel Fabp5+ subpopulation of inflammatory myeloid cells with delayed cytotoxic profile in chronic spinal cord injury. Heliyon 2023; 9:e18339. [PMID: 37636454 PMCID: PMC10450865 DOI: 10.1016/j.heliyon.2023.e18339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 08/29/2023] Open
Abstract
Traumatic spinal cord injuries (SCI) are a group of highly debilitating pathologies affecting thousands annually, and adversely affecting quality of life. Currently, no fully restorative therapies exist, and SCI still results in significant personal, societal and financial burdens. Inflammation plays a major role in the evolution of SCI, with myeloid cells, including bone marrow derived macrophages (BMDMs) and microglia (MG) being primary drivers of both early secondary pathogenesis and delayed wound healing events. The precise role of myeloid cell subsets is unclear as upon crossing the blood-spinal cord barrier, infiltrating bone marrow derived macrophages (BMDMs) may take on the morphology of resident microglia, and upregulate canonical microglia markers, thus making the two populations difficult to distinguish. Here, we used time-resolved scRNAseq and transgenic fate-mapping to chart the transcriptional profiles of tissue-resident and -infiltrating myeloid cells in a mouse model of thoracic contusion SCI. Our work identifies a novel subpopulation of foam cell-like inflammatory myeloid cells with increased expression of Fatty Acid Binding Protein 5 (Fabp5) and comprise both tissue-resident and -infiltrating cells. Fabp5+ inflammatory myeloid cells display a delayed cytotoxic profile that is predominant at the lesion epicentre and extends into the chronic phase of SCI.
Collapse
Affiliation(s)
- Regan Hamel
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | | | - Veronica Testa
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Bryan Yu
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - David Rowitch
- Cambridge Stem Cell Institute, University of Cambridge, UK
| | - John C. Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
Claeys W, Van Hoecke L, Lernout H, De Nolf C, Van Imschoot G, Van Wonterghem E, Verhaege D, Castelein J, Geerts A, Van Steenkiste C, Vandenbroucke RE. Experimental hepatic encephalopathy causes early but sustained glial transcriptional changes. J Neuroinflammation 2023; 20:130. [PMID: 37248507 DOI: 10.1186/s12974-023-02814-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 05/21/2023] [Indexed: 05/31/2023] Open
Abstract
Hepatic encephalopathy (HE) is a common complication of liver cirrhosis, associated with high morbidity and mortality, for which no brain-targeted therapies exist at present. The interplay between hyperammonemia and inflammation is thought to drive HE development. As such, astrocytes, the most important ammonia-metabolizing cells in the brain, and microglia, the main immunomodulatory cells in the brain, have been heavily implicated in HE development. As insight into cellular perturbations driving brain pathology remains largely elusive, we aimed to investigate cell-type specific transcriptomic changes in the HE brain. In the recently established mouse bile duct ligation (BDL) model of HE, we performed RNA-Seq of sorted astrocytes and microglia at 14 and 28 days after induction. This revealed a marked transcriptional response in both cell types which was most pronounced in microglia. In both cell types, pathways related to inflammation and hypoxia, mechanisms commonly implicated in HE, were enriched. Additionally, astrocytes exhibited increased corticoid receptor and oxidative stress signaling, whereas microglial transcriptome changes were linked to immune cell attraction. Accordingly, both monocytes and neutrophils accumulated in the BDL mouse brain. Time-dependent changes were limited in both cell types, suggesting early establishment of a pathological phenotype. While HE is often considered a unique form of encephalopathy, astrocytic and microglial transcriptomes showed significant overlap with previously established gene expression signatures in other neuroinflammatory diseases like septic encephalopathy and stroke, suggesting common pathophysiological mechanisms. Our dataset identifies key molecular mechanisms involved in preclinical HE and provides a valuable resource for development of novel glial-directed therapeutic strategies.
Collapse
Affiliation(s)
- Wouter Claeys
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Ghent University, 9000, Ghent, Belgium
- Liver Research Center Ghent, Ghent University Hospital, Ghent University, 9000, Ghent, Belgium
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Lien Van Hoecke
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Hannah Lernout
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- IBD Research Unit, Department of Internal Medicine and Paediatrics, Ghent University, 9000, Ghent, Belgium
| | - Clint De Nolf
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, 9000, Ghent, Belgium
| | - Griet Van Imschoot
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Elien Van Wonterghem
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Daan Verhaege
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Jonas Castelein
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium
| | - Anja Geerts
- Hepatology Research Unit, Department of Internal Medicine and Paediatrics, Ghent University, 9000, Ghent, Belgium
- Liver Research Center Ghent, Ghent University Hospital, Ghent University, 9000, Ghent, Belgium
- Department of Gastroenterology and Hepatology, Ghent University Hospital, Ghent, Belgium
| | - Christophe Van Steenkiste
- Department of Gastroenterology and Hepatology, Antwerp University, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- Barriers in Inflammation, VIB Center for Inflammation Research, VIB, Technologiepark-Zwijnaarde 71, 9052, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9000, Ghent, Belgium.
| |
Collapse
|
24
|
Spitzer D, Khel MI, Pütz T, Zinke J, Jia X, Sommer K, Filipski K, Thorsen F, Freiman TM, Günther S, Plate KH, Harter PN, Liebner S, Reiss Y, Di Tacchio M, Guérit S, Devraj K. A flow cytometry-based protocol for syngenic isolation of neurovascular unit cells from mouse and human tissues. Nat Protoc 2023; 18:1510-1542. [PMID: 36859615 DOI: 10.1038/s41596-023-00805-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/16/2022] [Indexed: 03/03/2023]
Abstract
The neurovascular unit (NVU), composed of endothelial cells, pericytes, juxtaposed astrocytes and microglia together with neurons, is essential for proper central nervous system functioning. The NVU critically regulates blood-brain barrier (BBB) function, which is impaired in several neurological diseases and is therefore a key therapeutic target. To understand the extent and cellular source of BBB dysfunction, simultaneous isolation and analysis of NVU cells is needed. Here, we describe a protocol for the EPAM-ia method, which is based on flow cytometry for simultaneous isolation and analysis of endothelial cells, pericytes, astrocytes and microglia. This method is based on differential processing of NVU cell types using enzymes, mechanical homogenization and filtration specific for each cell type followed by combining them for immunostaining and fluorescence-activated cell sorting. The gating strategy encompasses cell-type-specific and exclusion markers for contaminating cells to isolate the major NVU cell types. This protocol takes ~6 h for two sets of one or two animals. The isolation part requires experience in animal handling, fresh tissue processing and immunolabeling for flow cytometry. Sorted NVU cells can be used for downstream applications including transcriptomics, proteomics and cell culture. Multiple cell-type analyses using UpSet can then be applied to obtain robust targets from single or multiple NVU cell types in neurological diseases associated with BBB dysfunction. The EPAM-ia method is also amenable to isolation of several other cell types, including cancer cells and immune cells. This protocol is applicable to healthy and pathological tissue from mouse and human sources and to several cell types compared with similar protocols.
Collapse
Affiliation(s)
- Daniel Spitzer
- Department of Neurology, Goethe University, Frankfurt, Germany.,Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Maryam I Khel
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Tim Pütz
- Department of Neurology, Goethe University, Frankfurt, Germany.,Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Jenny Zinke
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Xiaoxiong Jia
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Kathleen Sommer
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Katharina Filipski
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Frits Thorsen
- The Molecular Imaging Center, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Thomas M Freiman
- Department of Neurosurgery, University Medical Center Rostock, Rostock, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Karl H Plate
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Patrick N Harter
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Liebner
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany
| | - Yvonne Reiss
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK) Partner site Frankfurt/Mainz, Frankfurt, Germany.,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Sylvaine Guérit
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany
| | - Kavi Devraj
- Edinger Institute (Institute of Neurology), Goethe University, Frankfurt, Germany. .,Center for Personalized Translational Epilepsy Research (CePTER), Frankfurt, Germany.
| |
Collapse
|
25
|
Evidence for Extensive Duplication and Subfunctionalization of FCRL6 in Armadillo ( Dasypus novemcinctus). Int J Mol Sci 2023; 24:ijms24054531. [PMID: 36901962 PMCID: PMC10003336 DOI: 10.3390/ijms24054531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
The control of infections by the vertebrate adaptive immune system requires careful modulation to optimize defense and minimize harm to the host. The Fc receptor-like (FCRL) genes encode immunoregulatory molecules homologous to the receptors for the Fc portion of immunoglobulin (FCR). To date, nine different genes (FCRL1-6, FCRLA, FCRLB and FCRLS) have been identified in mammalian organisms. FCRL6 is located at a separate chromosomal position from the FCRL1-5 locus, has conserved synteny in mammals and is situated between the SLAMF8 and DUSP23 genes. Here, we show that this three gene block underwent repeated duplication in Dasypus novemcinctus (nine-banded armadillo) resulting in six FCRL6 copies, of which five appear functional. Among 21 mammalian genomes analyzed, this expansion was unique to D. novemcinctus. Ig-like domains that derive from the five clustered FCRL6 functional gene copies show high structural conservation and sequence identity. However, the presence of multiple non-synonymous amino acid changes that would diversify individual receptor function has led to the hypothesis that FCRL6 endured subfunctionalization during evolution in D. novemcinctus. Interestingly, D. novemcinctus is noteworthy for its natural resistance to the Mycobacterium leprae pathogen that causes leprosy. Because FCRL6 is chiefly expressed by cytotoxic T and NK cells, which are important in cellular defense responses against M. leprae, we speculate that FCRL6 subfunctionalization could be relevant for the adaptation of D. novemcinctus to leprosy. These findings highlight the species-specific diversification of FCRL family members and the genetic complexity underlying evolving multigene families critical for modulating adaptive immune protection.
Collapse
|
26
|
Sharpe MA, Baskin DS, Johnson RD, Baskin AM. Acquisition of Immune Privilege in GBM Tumors: Role of Prostaglandins and Bile Salts. Int J Mol Sci 2023; 24:3198. [PMID: 36834607 PMCID: PMC9958596 DOI: 10.3390/ijms24043198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Based on the postulate that glioblastoma (GBM) tumors generate anti-inflammatory prostaglandins and bile salts to gain immune privilege, we analyzed 712 tumors in-silico from three GBM transcriptome databases for prostaglandin and bile synthesis/signaling enzyme-transcript markers. A pan-database correlation analysis was performed to identify cell-specific signal generation and downstream effects. The tumors were stratified by their ability to generate prostaglandins, their competency in bile salt synthesis, and the presence of bile acid receptors nuclear receptor subfamily 1, group H, member 4 (NR1H4) and G protein-coupled bile acid receptor 1 (GPBAR1). The survival analysis indicates that tumors capable of prostaglandin and/or bile salt synthesis are linked to poor outcomes. Tumor prostaglandin D2 and F2 syntheses are derived from infiltrating microglia, whereas prostaglandin E2 synthesis is derived from neutrophils. GBMs drive the microglial synthesis of PGD2/F2 by releasing/activating complement system component C3a. GBM expression of sperm-associated heat-shock proteins appears to stimulate neutrophilic PGE2 synthesis. The tumors that generate bile and express high levels of bile receptor NR1H4 have a fetal liver phenotype and a RORC-Treg infiltration signature. The bile-generating tumors that express high levels of GPBAR1 are infiltrated with immunosuppressive microglia/macrophage/myeloid-derived suppressor cells. These findings provide insight into how GBMs generate immune privilege and may explain the failure of checkpoint inhibitor therapy and provide novel targets for treatment.
Collapse
Affiliation(s)
- Martyn A. Sharpe
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
| | - David S. Baskin
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
- Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ryan D. Johnson
- Kenneth R. Peak Brain and Pituitary Tumor Treatment Center, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
| | - Alexandra M. Baskin
- Department of Natural Science, Marine Science, Hawaii Pacific University, Honolulu, HI 96801, USA
| |
Collapse
|
27
|
Ahmed MH, Hernández-Verdin I, Quissac E, Lemaire N, Guerin C, Guyonnet L, Zahr N, Mouton L, Santin M, Petiet A, Schmitt C, Bouchoux G, Canney M, Sanson M, Verreault M, Carpentier A, Idbaih A. Low-Intensity Pulsed Ultrasound-Mediated Blood-Brain Barrier Opening Increases Anti-Programmed Death-Ligand 1 Delivery and Efficacy in Gl261 Mouse Model. Pharmaceutics 2023; 15:pharmaceutics15020455. [PMID: 36839777 PMCID: PMC9967384 DOI: 10.3390/pharmaceutics15020455] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Therapeutic antibodies targeting immune checkpoints have shown limited efficacy in clinical trials in glioblastoma (GBM) patients. Ultrasound-mediated blood-brain barrier opening (UMBO) using low-intensity pulsed ultrasound improved drug delivery to the brain. We explored the safety and the efficacy of UMBO plus immune checkpoint inhibitors in preclinical models of GBM. A blood-brain barrier (BBB) opening was performed using a 1 MHz preclinical ultrasound system in combination with 10 µL/g microbubbles. Brain penetration of immune checkpoint inhibitors was determined, and immune cell populations were evaluated using flow cytometry. The impact of repeated treatments on survival was determined. In syngeneic GL261-bearing immunocompetent mice, we showed that UMBO safely and repeatedly opened the BBB. BBB opening was confirmed visually and microscopically using Evans blue dye and magnetic resonance imaging. UMBO plus anti-PDL-1 was associated with a significant improvement of overall survival compared to anti-PD-L1 alone. Using mass spectroscopy, we showed that the penetration of therapeutic antibodies can be increased when delivered intravenously compared to non-sonicated brains. Furthermore, we observed an enhancement of activated microglia percentage when combined with anti-PD-L1. Here, we report that the combination of UMBO and anti-PD-L1 dramatically increases GL261-bearing mice's survival compared to their counterparts treated with anti-PD-L1 alone. Our study highlights the BBB as a limitation to overcome in order to increase the efficacy of anti-PD-L1 in GBM and supports clinical trials combining UMBO and in GBM patients.
Collapse
Affiliation(s)
- Mohammed H. Ahmed
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
- School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9NH, UK
- Correspondence: (M.H.A.); (A.I.); Tel.: +44-(0)-20-7836-5454 (M.H.A.); +33-01-42-16-03-85 (A.I.); Fax: +33-01-42-16-04-18 (A.I.)
| | - Isaias Hernández-Verdin
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Emie Quissac
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Nolwenn Lemaire
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Coralie Guerin
- Cytometry Department, Institute Curie, F-75006 Paris, France
| | - Lea Guyonnet
- Cytometry Department, Institute Curie, F-75006 Paris, France
| | - Noël Zahr
- Pharmacokinetics and Therapeutic Drug Monitoring Unit, Inserm, CIC-1901, UMR ICAN 1166, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Laura Mouton
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Mathieu Santin
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Alexandra Petiet
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Charlotte Schmitt
- CarThera, Institut du Cerveau et de la Moelle Épinière (ICM), F-75013 Paris, France
| | - Guillaume Bouchoux
- CarThera, Institut du Cerveau et de la Moelle Épinière (ICM), F-75013 Paris, France
| | - Michael Canney
- CarThera, Institut du Cerveau et de la Moelle Épinière (ICM), F-75013 Paris, France
| | - Marc Sanson
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, DMU Neurosciences, Service de Neurologie 2-Mazarin, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Maïté Verreault
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Alexandre Carpentier
- CarThera, Institut du Cerveau et de la Moelle Épinière (ICM), F-75013 Paris, France
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, AP-HP, DMU Neurosciences, Service de Neurologie 2-Mazarin, Hôpital de la Pitié Salpêtrière, Sorbonne Université, F-75013 Paris, France
| | - Ahmed Idbaih
- CarThera, Institut du Cerveau et de la Moelle Épinière (ICM), F-75013 Paris, France
- Correspondence: (M.H.A.); (A.I.); Tel.: +44-(0)-20-7836-5454 (M.H.A.); +33-01-42-16-03-85 (A.I.); Fax: +33-01-42-16-04-18 (A.I.)
| |
Collapse
|
28
|
Sun H, Gao Q, Zhu G, Han C, Yan H, Wang T. Identification of influential observations in high-dimensional survival data through robust penalized Cox regression based on trimming. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:5352-5378. [PMID: 36896549 DOI: 10.3934/mbe.2023248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Penalized Cox regression can efficiently be used for the determination of biomarkers in high-dimensional genomic data related to disease prognosis. However, results of Penalized Cox regression is influenced by the heterogeneity of the samples who have different dependent structure between survival time and covariates from most individuals. These observations are called influential observations or outliers. A robust penalized Cox model (Reweighted Elastic Net-type maximum trimmed partial likelihood estimator, Rwt MTPL-EN) is proposed to improve the prediction accuracy and identify influential observations. A new algorithm AR-Cstep to solve Rwt MTPL-EN model is also proposed. This method has been validated by simulation study and application to glioma microarray expression data. When there were no outliers, the results of Rwt MTPL-EN were close to the Elastic Net (EN). When outliers existed, the results of EN were impacted by outliers. And whenever the censored rate was large or low, the robust Rwt MTPL-EN performed better than EN. and could resist the outliers in both predictors and response. In terms of outliers detection accuracy, Rwt MTPL-EN was much higher than EN. The outliers who "lived too long" made EN perform worse, but were accurately detected by Rwt MTPL-EN. Through the analysis of glioma gene expression data, most of the outliers identified by EN were those "failed too early", but most of them were not obvious outliers according to risk estimated from omics data or clinical variables. Most of the outliers identified by Rwt MTPL-EN were those who "lived too long", and most of them were obvious outliers according to risk estimated from omics data or clinical variables. Rwt MTPL-EN can be adopted to detect influential observations in high-dimensional survival data.
Collapse
Affiliation(s)
- Hongwei Sun
- Department of Health Statistics, School of Public Health and Management, Binzhou Medical University, Yantai City, Shandong 264003, China
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan City, Shanxi 030001, China
| | - Qian Gao
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan City, Shanxi 030001, China
| | - Guiming Zhu
- Department of Health Statistics, School of Public Health and Management, Binzhou Medical University, Yantai City, Shandong 264003, China
| | - Chunlei Han
- Department of Health Statistics, School of Public Health and Management, Binzhou Medical University, Yantai City, Shandong 264003, China
| | - Haosen Yan
- Department of Health Statistics, School of Public Health and Management, Binzhou Medical University, Yantai City, Shandong 264003, China
| | - Tong Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan City, Shanxi 030001, China
| |
Collapse
|
29
|
Khan F, Pang L, Dunterman M, Lesniak MS, Heimberger AB, Chen P. Macrophages and microglia in glioblastoma: heterogeneity, plasticity, and therapy. J Clin Invest 2023; 133:163446. [PMID: 36594466 PMCID: PMC9797335 DOI: 10.1172/jci163446] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive tumor in the central nervous system and contains a highly immunosuppressive tumor microenvironment (TME). Tumor-associated macrophages and microglia (TAMs) are a dominant population of immune cells in the GBM TME that contribute to most GBM hallmarks, including immunosuppression. The understanding of TAMs in GBM has been limited by the lack of powerful tools to characterize them. However, recent progress on single-cell technologies offers an opportunity to precisely characterize TAMs at the single-cell level and identify new TAM subpopulations with specific tumor-modulatory functions in GBM. In this Review, we discuss TAM heterogeneity and plasticity in the TME and summarize current TAM-targeted therapeutic potential in GBM. We anticipate that the use of single-cell technologies followed by functional studies will accelerate the development of novel and effective TAM-targeted therapeutics for GBM patients.
Collapse
|
30
|
Kantor AB, Akassoglou K, Stavenhagen JB. Fibrin-Targeting Immunotherapy for Dementia. J Prev Alzheimers Dis 2023; 10:647-660. [PMID: 37874085 PMCID: PMC11227370 DOI: 10.14283/jpad.2023.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Blood-brain barrier (BBB) disruption is an early event in the development of Alzheimer's disease. It precedes extracellular deposition of amyloid-β in senile plaques and blood vessel walls, the intracellular accumulation of neurofibrillary tangles containing phosphorylated tau protein, microglial activation, and neuronal cell death. BBB disruption allows the coagulation protein fibrinogen to leak from the blood into the brain, where it is converted by thrombin cleavage into fibrin and deposits in the parenchyma and CNS vessels. Fibrinogen cleavage by thrombin exposes a cryptic epitope termed P2 which can bind CD11b and CD11c on microglia, macrophages and dendritic cells and trigger an inflammatory response toxic to neurons. Indeed, genetic and pharmacological evidence demonstrates a causal role for fibrin in innate immune cell activation and the development of neurodegenerative diseases. The P2 inflammatory epitope is spatially and compositionally distinct from the coagulation epitope on fibrin. Mouse monoclonal antibody 5B8, which targets the P2 epitope without interfering with the clotting process, has been shown to reduce neurodegeneration and neuroinflammation in animal models of Alzheimer's disease and multiple sclerosis. The selectivity and efficacy of this anti-human fibrin-P2 antibody in animal models supports the development of a monoclonal antibody drug targeting fibrin P2 for the treatment of neurodegenerative diseases. THN391 is a humanized, affinity-matured antibody which has a 100-fold greater affinity for fibrin P2 and improved development properties compared to the parental 5B8 antibody. It is currently in a Phase 1 clinical trial.
Collapse
Affiliation(s)
- A B Kantor
- Jeffrey Stavenhagen, PhD, Therini Bio, Inc, Sacramento, CA, USA,
| | | | | |
Collapse
|
31
|
Olivares-González L, Velasco S, Gallego I, Esteban-Medina M, Puras G, Loucera C, Martínez-Romero A, Peña-Chilet M, Pedraz JL, Rodrigo R. An SPM-Enriched Marine Oil Supplement Shifted Microglia Polarization toward M2, Ameliorating Retinal Degeneration in rd10 Mice. Antioxidants (Basel) 2022; 12:antiox12010098. [PMID: 36670960 PMCID: PMC9855087 DOI: 10.3390/antiox12010098] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 01/04/2023] Open
Abstract
Retinitis pigmentosa (RP) is the most common inherited retinal dystrophy causing progressive vision loss. It is accompanied by chronic and sustained inflammation, including M1 microglia activation. This study evaluated the effect of an essential fatty acid (EFA) supplement containing specialized pro-resolving mediators (SPMs), on retinal degeneration and microglia activation in rd10 mice, a model of RP, as well as on LPS-stimulated BV2 cells. The EFA supplement was orally administered to mice from postnatal day (P)9 to P18. At P18, the electrical activity of the retina was examined by electroretinography (ERG) and innate behavior in response to light were measured. Retinal degeneration was studied via histology including the TUNEL assay and microglia immunolabeling. Microglia polarization (M1/M2) was assessed by flow cytometry, qPCR, ELISA and histology. Redox status was analyzed by measuring antioxidant enzymes and markers of oxidative damage. Interestingly, the EFA supplement ameliorated retinal dysfunction and degeneration by improving ERG recording and sensitivity to light, and reducing photoreceptor cell loss. The EFA supplement reduced inflammation and microglia activation attenuating M1 markers as well as inducing a shift to the M2 phenotype in rd10 mouse retinas and LPS-stimulated BV2 cells. It also reduced oxidative stress markers of lipid peroxidation and carbonylation. These findings could open up new therapeutic opportunities based on resolving inflammation with oral supplementation with SPMs such as the EFA supplement.
Collapse
Affiliation(s)
- Lorena Olivares-González
- Group of Pathophysiology and Therapies for Vision Disorders, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain
| | - Sheyla Velasco
- Group of Pathophysiology and Therapies for Vision Disorders, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain
| | - Idoia Gallego
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Health Institute Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Marina Esteban-Medina
- Computational Medicine Platform, Andalusian Public Foundation Progress and Health-FPS Hospital Virgen del Rocío, 41013 Seville, Spain
- Systems and Computational Medicine Group, Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | - Gustavo Puras
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Health Institute Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Carlos Loucera
- Computational Medicine Platform, Andalusian Public Foundation Progress and Health-FPS Hospital Virgen del Rocío, 41013 Seville, Spain
- Systems and Computational Medicine Group, Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
| | | | - María Peña-Chilet
- Computational Medicine Platform, Andalusian Public Foundation Progress and Health-FPS Hospital Virgen del Rocío, 41013 Seville, Spain
- Systems and Computational Medicine Group, Institute of Biomedicine of Seville, IBiS, University Hospital Virgen del Rocío/CSIC/University of Seville, 41013 Seville, Spain
- Bioinformatics in Rare Diseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, 41013 Seville, Spain
| | - José Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), 01006 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Health Institute Carlos III, 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
| | - Regina Rodrigo
- Group of Pathophysiology and Therapies for Vision Disorders, Príncipe Felipe Research Center (CIPF), 46012 Valencia, Spain
- Biomedical Research Networking Center in Rare Diseases (CIBERER), Health Institute Carlos III, 28029 Madrid, Spain
- Department of Physiology, University of Valencia (UV), 46100 Burjassot, Spain
- Department of Anatomy and Physiology, Catholic University of Valencia San Vicente Mártir, 46001 Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics UV-IIS La Fe, 46026 Valencia, Spain
- Correspondence: ; Tel.: +34-96-328-96-80
| |
Collapse
|
32
|
Neuronal CaMKK2 promotes immunosuppression and checkpoint blockade resistance in glioblastoma. Nat Commun 2022; 13:6483. [PMID: 36309495 PMCID: PMC9617949 DOI: 10.1038/s41467-022-34175-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 10/14/2022] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma (GBM) is notorious for its immunosuppressive tumor microenvironment (TME) and is refractory to immune checkpoint blockade (ICB). Here, we identify calmodulin-dependent kinase kinase 2 (CaMKK2) as a driver of ICB resistance. CaMKK2 is highly expressed in pro-tumor cells and is associated with worsened survival in patients with GBM. Host CaMKK2, specifically, reduces survival and promotes ICB resistance. Multimodal profiling of the TME reveals that CaMKK2 is associated with several ICB resistance-associated immune phenotypes. CaMKK2 promotes exhaustion in CD8+ T cells and reduces the expansion of effector CD4+ T cells, additionally limiting their tumor penetrance. CaMKK2 also maintains myeloid cells in a disease-associated microglia-like phenotype. Lastly, neuronal CaMKK2 is required for maintaining the ICB resistance-associated myeloid phenotype, is deleterious to survival, and promotes ICB resistance. Our findings reveal CaMKK2 as a contributor to ICB resistance and identify neurons as a driver of immunotherapeutic resistance in GBM.
Collapse
|
33
|
Çetin G, Studencka-Turski M, Venz S, Schormann E, Junker H, Hammer E, Völker U, Ebstein F, Krüger E. Immunoproteasomes control activation of innate immune signaling and microglial function. Front Immunol 2022; 13:982786. [PMID: 36275769 PMCID: PMC9584546 DOI: 10.3389/fimmu.2022.982786] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and play a major role in the regulation of brain homeostasis. To maintain their cellular protein homeostasis, microglia express standard proteasomes and immunoproteasomes (IP), a proteasome isoform that preserves protein homeostasis also in non-immune cells under challenging conditions. The impact of IP on microglia function in innate immunity of the CNS is however not well described. Here, we establish that IP impairment leads to proteotoxic stress and triggers the unfolded and integrated stress responses in mouse and human microglia models. Using proteomic analysis, we demonstrate that IP deficiency in microglia results in profound alterations of the ubiquitin-modified proteome among which proteins involved in the regulation of stress and immune responses. In line with this, molecular analysis revealed chronic activation of NF-κB signaling in IP-deficient microglia without further stimulus. In addition, we show that IP impairment alters microglial function based on markers for phagocytosis and motility. At the molecular level IP impairment activates interferon signaling promoted by the activation of the cytosolic stress response protein kinase R. The presented data highlight the importance of IP function for the proteostatic potential as well as for precision proteolysis to control stress and immune signaling in microglia function.
Collapse
Affiliation(s)
- Gonca Çetin
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Maja Studencka-Turski
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Simone Venz
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Eileen Schormann
- Institute of Biochemistry, Charité – University Medicine Berlin, Berlin, Germany
| | - Heike Junker
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Hammer
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Frédéric Ebstein
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
| | - Elke Krüger
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald, Germany
- *Correspondence: Elke Krüger,
| |
Collapse
|
34
|
Plasschaert RN, DeAndrade MP, Hull F, Nguyen Q, Peterson T, Yan A, Loperfido M, Baricordi C, Barbarossa L, Yoon JK, Dogan Y, Unnisa Z, Schindler JW, van Til NP, Biasco L, Mason C. High-throughput analysis of hematopoietic stem cell engraftment after intravenous and intracerebroventricular dosing. Mol Ther 2022; 30:3209-3225. [PMID: 35614857 PMCID: PMC9552809 DOI: 10.1016/j.ymthe.2022.05.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 04/15/2022] [Accepted: 05/21/2022] [Indexed: 11/27/2022] Open
Abstract
Hematopoietic stem/progenitor cell gene therapy (HSPC-GT) has shown clear neurological benefit in rare diseases, which is achieved through the engraftment of genetically modified microglia-like cells (MLCs) in the brain. Still, the engraftment dynamics and the nature of engineered MLCs, as well as their potential use in common neurogenerative diseases, have remained largely unexplored. Here, we comprehensively characterized how different routes of administration affect the biodistribution of genetically engineered MLCs and other HSPC derivatives in mice. We generated a high-resolution single-cell transcriptional map of MLCs and discovered that they could clearly be distinguished from macrophages as well as from resident microglia by the expression of a specific gene signature that is reflective of their HSPC ontogeny and irrespective of their long-term engraftment history. Lastly, using murine models of Parkinson's disease and frontotemporal dementia, we demonstrated that MLCs can deliver therapeutically relevant levels of transgenic protein to the brain, thereby opening avenues for the clinical translation of HSPC-GT to the treatment of major neurological diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Aimin Yan
- AVROBIO, Inc, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | - Niek P van Til
- AVROBIO, Inc, Cambridge, MA 02139, USA; Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, VU University, and Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Luca Biasco
- AVROBIO, Inc, Cambridge, MA 02139, USA; Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Chris Mason
- AVROBIO, Inc, Cambridge, MA 02139, USA; Advanced Centre for Biochemical Engineering, University College London, London, UK.
| |
Collapse
|
35
|
Pang L, Khan F, Heimberger AB, Chen P. Mechanism and therapeutic potential of tumor-immune symbiosis in glioblastoma. Trends Cancer 2022; 8:839-854. [PMID: 35624002 PMCID: PMC9492629 DOI: 10.1016/j.trecan.2022.04.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/20/2022]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal form of brain tumor in human adults. Myeloid-lineage cells, including macrophages, microglia, myeloid-derived suppressor cells (MDSCs), and neutrophils, are the most frequent types of cell in the GBM tumor microenvironment (TME) that contribute to tumor progression. Emerging experimental evidence indicates that symbiotic interactions between cancer cells and myeloid cells are critical for tumor growth and immunotherapy resistance in GBM. In this review, we discuss the molecular mechanisms whereby cancer cells shape a myeloid cell-mediated immunosuppressive TME and, reciprocally, how such myeloid cells affect tumor progression and immunotherapy efficiency in GBM. Moreover, we highlight tumor-T cell symbiosis and summarize immunotherapeutic strategies intercepting this co-dependency in GBM.
Collapse
Affiliation(s)
- Lizhi Pang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Fatima Khan
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Amy B Heimberger
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peiwen Chen
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
36
|
Lei W, Ren Z, Su J, Zheng X, Gao L, Xu Y, Deng J, Xiao C, Sheng S, Cheng Y, Ma T, Liu Y, Wang P, Luo OJ, Chen G, Wang Z. Immunological risk factors for sepsis-associated delirium and mortality in ICU patients. Front Immunol 2022; 13:940779. [PMID: 36203605 PMCID: PMC9531264 DOI: 10.3389/fimmu.2022.940779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
Background A major challenge in intervention of critical patients, especially sepsis-associated delirium (SAD) intervention, is the lack of predictive risk factors. As sepsis and SAD are heavily entangled with inflammatory and immunological processes, to identify the risk factors of SAD and mortality in the intensive care unit (ICU) and determine the underlying molecular mechanisms, the peripheral immune profiles of patients in the ICU were characterized. Methods This study contains a cohort of 52 critical patients who were admitted to the ICU of the First Affiliated Hospital of Jinan University. Comorbidity, including sepsis and SAD, of this cohort was diagnosed and recorded. Furthermore, peripheral blood samples were collected on days 1, 3, and 5 of admission for peripheral immune profiling with blood routine examination, flow cytometry, ELISA, RNA-seq, and qPCR. Results The patients with SAD had higher mortality during ICU admission and within 28 days of discharge. Compared with survivors, nonsurvivors had higher neutrophilic granulocyte percentage, higher CRP concentration, lower monocyte count, lower monocyte percentage, lower C3 complement level, higher CD14loCD16+ monocytes percentage, and higher levels of IL-6 and TNFα. The CD14hiCD16- monocyte percentage manifested favorable prediction values for the occurrence of SAD. Differentially expressed genes between the nonsurvival and survival groups were mainly associated with immune response and metabolism process. The longitudinal expression pattern of SLC2A1 and STIMATE were different between nonsurvivors and survivors, which were validated by qPCR. Conclusions Nonsurvival critical patients have a distinct immune profile when compared with survival patients. CD14hiCD16- monocyte prevalence and expression levels of SLC2A1 and STIMATE may be predictors of SAD and 28-day mortality in ICU patients.
Collapse
Affiliation(s)
- Wen Lei
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Zhiyao Ren
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- National Health Commission (NHC) Key Laboratory of Male Reproduction and Genetics, Guangzhou, China
- Department of Central Laboratory, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
| | - Jun Su
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Sonograph, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xinglong Zheng
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Critical Care Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Yudai Xu
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Jieping Deng
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Chanchan Xiao
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Shuai Sheng
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Yu Cheng
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Tianshun Ma
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Yu Liu
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Pengcheng Wang
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
| | - Oscar Junhong Luo
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Zhigang Wang, ; Guobing Chen, ; Oscar Junhong Luo,
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
- Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Sonograph, The First Affiliated Hospital, Jinan University, Guangzhou, China
- *Correspondence: Zhigang Wang, ; Guobing Chen, ; Oscar Junhong Luo,
| | - Zhigang Wang
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Jinan University, Guangzhou, China
- Department of Critical Care Medicine, The First Affiliated Hospital, Jinan University, Guangzhou, China
- *Correspondence: Zhigang Wang, ; Guobing Chen, ; Oscar Junhong Luo,
| |
Collapse
|
37
|
Zhang Z, Wang X, Lin Y, Pan D. A multifaceted evaluation of microgliosis and differential cellular dysregulation of mammalian target of rapamycin signaling in neuronopathic Gaucher disease. Front Mol Neurosci 2022; 15:944883. [PMID: 36204141 PMCID: PMC9530712 DOI: 10.3389/fnmol.2022.944883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/03/2022] [Indexed: 11/18/2022] Open
Abstract
Neuronopathic Gaucher disease (nGD) is an inherited neurodegenerative disease caused by mutations in GBA1 gene and is associated with premature death. Neuroinflammation plays a critical role in disease pathogenesis which is characterized by microgliosis, reactive astrocytosis, and neuron loss, although molecular mechanisms leading to neuroinflammation are not well-understood. In this report, we developed a convenient tool to quantify microglia proliferation and activation independently and uncovered abnormal proliferation of microglia (∼2-fold) in an adult genetic nGD model. The nGD-associated pattern of inflammatory mediators pertinent to microglia phenotypes was determined, showing a unique signature favoring pro-inflammatory chemokines and cytokines. Moreover, highly polarized (up or down) dysregulations of mTORC1 signaling with varying lysosome dysfunctions (numbers and volume) were observed among three major cell types of nGD brain. Specifically, hyperactive mTORC1 signaling was detected in all disease-associated microglia (Iba1high) with concurrent increase in lysosome function. Conversely, the reduction of neurons presenting high mTORC1 activity was implicated (including Purkinje-like cells) which was accompanied by inconsistent changes of lysosome function in nGD mice. Undetectable levels of mTORC1 activity and low Lamp1 puncta were noticed in astrocytes of both diseased and normal mice, suggesting a minor involvement of mTORC1 pathway and lysosome function in disease-associated astrocytes. These findings highlight the differences and complexity of molecular mechanisms that are involved within various cell types of the brain. The quantifiable parameters established and nGD-associated pattern of neuroinflammatory mediators identified would facilitate the efficacy evaluation on microgliosis and further discovery of novel therapeutic target(s) in treating neuronopathic Gaucher disease.
Collapse
Affiliation(s)
- Zhenting Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Xiaohong Wang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Yi Lin
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Dao Pan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, United States
- *Correspondence: Dao Pan,
| |
Collapse
|
38
|
Lee SL, Lee MHH, Wu KJ, Chiang CW, Chang YX, Fang JD, Tung HH, Kuo LW, Wang Y. Post-ischemic protection of hepatocyte growth factor requires the type II transmembrane serine protease matriptase-A reciprocal regulation of the two for neuroprotection in stroke brain. FASEB J 2022; 36:e22494. [PMID: 35976173 DOI: 10.1096/fj.202200414r] [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: 03/24/2022] [Revised: 07/04/2022] [Accepted: 08/01/2022] [Indexed: 11/11/2022]
Abstract
In a rat middle cerebral artery occlusion (MACo) model of ischemic stroke, intracerebroventricular administration of human recombinant hepatocyte growth factor (HGF) mitigated motor impairment and cortical infarction. Recombinant HGF reduced MCAo-induced TNFα and IL1β expression, and alleviated perilesional reactivation of microglia and astrocyte. All of the aforementioned beneficial effects of HGF were antagonized by an inhibitor to the type II transmembrane serine protease matriptase (MTP). MCAo upregulated MTP mRNA and protein in the lesioned cortex. MTP protein, not the mRNA, was increased further by recombinant HGF but reduced when MTP inhibitor (MTPi) was added to the treatment. Changes of the endogenous active HGF by MCAo, HGF or MTPi paralleled with the changes of MTP protein under the same conditions whilst neither HGF mRNA nor the total endogenous HGF protein were altered. These data showed that the therapeutic effects of HGF in stroke brain is attributed to its proteolytic activation and that MTP is a main protease of the event. MCAo enhanced MTP mRNA and thus protein expression; the initial use of the recombinant active HGF stabilized MCAo-induced MTP protein and subsequent activation of endogenous latent HGF which in turn stabilized further MTP protein. A reciprocal regulation between MTP and HGF appears to be present where MTP promotes HGF activation and the active HGF prevents MTP protein turnover. This study, for the first time, shows that MTP can participate in neural protection in stroke brain through activation of HGF. The cycles of HGF-MTP regulation achieved preservation of the neurological activity.
Collapse
Affiliation(s)
- Sheau-Ling Lee
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,Biotechnology Center, National Chung Hsing University, Taichung, Taiwan, R.O.C
| | - Michelle Hui-Hsin Lee
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Chia-Wen Chiang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Yun-Xuan Chang
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Jung-Da Fang
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Hsiu-Hui Tung
- Institute of Cellular and Systems Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Li-Wei Kuo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| |
Collapse
|
39
|
Tang F, Pan Z, Wang Y, Lan T, Wang M, Li F, Quan W, Liu Z, Wang Z, Li Z. Advances in the Immunotherapeutic Potential of Isocitrate Dehydrogenase Mutations in Glioma. Neurosci Bull 2022; 38:1069-1084. [PMID: 35670952 PMCID: PMC9468211 DOI: 10.1007/s12264-022-00866-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/16/2022] [Indexed: 11/26/2022] Open
Abstract
Isocitrate dehydrogenase (IDH) is an essential metabolic enzyme in the tricarboxylic acid cycle (TAC). The high mutation frequency of the IDH gene plays a complicated role in gliomas. In addition to affecting gliomas directly, mutations in IDH can also alter their immune microenvironment and can change immune-cell function in direct and indirect ways. IDH mutations mediate immune-cell infiltration and function by modulating immune-checkpoint gene expression and chemokine secretion. In addition, IDH mutation-derived D2-hydroxyglutarate can be absorbed by surrounding immune cells, also affecting their functioning. In this review, we summarize current knowledge about the effects of IDH mutations as well as other gene mutations on the immune microenvironment of gliomas. We also describe recent preclinical and clinical data related to IDH-mutant inhibitors for the treatment of gliomas. Finally, we discuss different types of immunotherapy and the immunotherapeutic potential of IDH mutations in gliomas.
Collapse
Affiliation(s)
- Feng Tang
- Brain Glioma Center and Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Zhiyong Pan
- Brain Glioma Center and Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Yi Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Tian Lan
- Brain Glioma Center and Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Mengyue Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China
| | - Fengping Li
- Brain Glioma Center and Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Wei Quan
- Brain Glioma Center and Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Zhenyuan Liu
- Brain Glioma Center and Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China
| | - Zefen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China.
| | - Zhiqiang Li
- Brain Glioma Center and Department of Neurosurgery, Wuhan University Zhongnan Hospital, Wuhan, 430071, China.
| |
Collapse
|
40
|
Go J, Park HY, Lee DW, Maeng SY, Lee IB, Seo YJ, An JP, Oh WK, Lee CH, Kim KS. Humulus japonicus attenuates LPS-and scopolamine-induced cognitive impairment in mice. Lab Anim Res 2022; 38:21. [PMID: 35854340 PMCID: PMC9297604 DOI: 10.1186/s42826-022-00134-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/13/2022] [Indexed: 11/30/2022] Open
Abstract
Background Neuroinflammation plays an important role in cognitive decline and memory impairment in neurodegenerative disorders. Previously, we demonstrated that Humulus japonicus (HJ) has anti-inflammatory effects in rodent models of Alzheimer’s disease and Parkinson’s disease. The present study aimed to examine the protective potential of HJ extracts against lipopolysaccharide (LPS)-induced cognitive impairment and scopolamine-induced amnesia in mouse models. Cognitive improvement of mice was investigated by novel object recognition test. For analyzing effects on neuroinflammation, immunohistochemistry and quantitative real-time polymerase chain reaction (qRT-PCR) assays were performed. Results We found that the oral administration of HJ significantly improved cognitive dysfunction induced by LPS in a novel object recognition test. The LPS-induced activation of microglia was notably decreased by HJ treatment in the cortex and hippocampus. HJ administration with LPS also significantly increased the mRNA expression of interleukin (IL)-10 and decreased the mRNA expression of IL-12 in the parietal cortex of mice. The increased expression of LPS-induced complement C1q B chain (C1bq) and triggering receptor expressed on myeloid cells 2 (Trem2) genes was significantly suppressed by HJ treatment. In addition, HJ administration significantly improved novel object recognition in a scopolamine-induced amnesia mouse model. Conclusions These findings revealed that HJ has a beneficial effect on cognitive impairment and neuroinflammation induced by systemic inflammation and on amnesia induced by scopolamine in mice.
Collapse
Affiliation(s)
- Jun Go
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Gwahak-ro 125, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hye-Yeon Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Gwahak-ro 125, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Da Woon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Gwahak-ro 125, Yuseong-gu, Daejeon, 34141, Republic of Korea.,Department of Biomaterials Science, College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, 50463, Republic of Korea
| | - So-Young Maeng
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Gwahak-ro 125, Yuseong-gu, Daejeon, 34141, Republic of Korea.,College of Biosciences and Biotechnology, Chung-Nam National University, Daejeon, 34134, Republic of Korea
| | - In-Bok Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Gwahak-ro 125, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Yun Jeong Seo
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Gwahak-ro 125, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jin-Pyo An
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences College of Pharmacy, Seoul National University (SNU), Seoul, 08826, Republic of Korea
| | - Won Keun Oh
- Korea Bioactive Natural Material Bank, Research Institute of Pharmaceutical Sciences College of Pharmacy, Seoul National University (SNU), Seoul, 08826, Republic of Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Gwahak-ro 125, Yuseong-gu, Daejeon, 34141, Republic of Korea. .,Department of Functional Genomics, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Kyoung-Shim Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Gwahak-ro 125, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
41
|
Aubin RG, Troisi EC, Montelongo J, Alghalith AN, Nasrallah MP, Santi M, Camara PG. Pro-inflammatory cytokines mediate the epithelial-to-mesenchymal-like transition of pediatric posterior fossa ependymoma. Nat Commun 2022; 13:3936. [PMID: 35803925 PMCID: PMC9270322 DOI: 10.1038/s41467-022-31683-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022] Open
Abstract
Pediatric ependymoma is a devastating brain cancer marked by its relapsing pattern and lack of effective chemotherapies. This shortage of treatments is due to limited knowledge about ependymoma tumorigenic mechanisms. By means of single-nucleus chromatin accessibility and gene expression profiling of posterior fossa primary tumors and distal metastases, we reveal key transcription factors and enhancers associated with the differentiation of ependymoma tumor cells into tumor-derived cell lineages and their transition into a mesenchymal-like state. We identify NFκB, AP-1, and MYC as mediators of this transition, and show that the gene expression profiles of tumor cells and infiltrating microglia are consistent with abundant pro-inflammatory signaling between these populations. In line with these results, both TGF-β1 and TNF-α induce the expression of mesenchymal genes on a patient-derived cell model, and TGF-β1 leads to an invasive phenotype. Altogether, these data suggest that tumor gliosis induced by inflammatory cytokines and oxidative stress underlies the mesenchymal phenotype of posterior fossa ependymoma.
Collapse
Affiliation(s)
- Rachael G Aubin
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emma C Troisi
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Javier Montelongo
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Adam N Alghalith
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Maclean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mariarita Santi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Pablo G Camara
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
42
|
Wang X, Xu Y, Sun Q, Zhou X, Ma W, Wu J, Zhuang J, Sun C. New insights from the single-cell level: Tumor associated macrophages heterogeneity and personalized therapy. Biomed Pharmacother 2022; 153:113343. [PMID: 35785706 DOI: 10.1016/j.biopha.2022.113343] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/02/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are important immune cells in the tumor microenvironment, and their invasion in tumors is closely related to poor prognosis. Although TAMs are recognized as therapeutic targets, their heterogeneity makes studying tumor mechanism and developing drugs targeting TAMs difficult. The study of TAMs heterogeneity can be used to analyze the mechanism of tumor progression and drug resistance, and may provide possible treatment strategies for cancer patients. Single-cell RNA sequencing (scRNA-seq) can reveal the RNA expression profile for each TAM to distinguish heterogeneity, thereby providing a more efficient detection method and more accurate information for TAM-related studies. In this review, by summarizing the research progress in macrophage heterogeneity and other aspects of scRNA-seq over the past five years, we introduced the development of scRNA-seq technology and its application status in solid tumors, analyzed the advantages and selections of scRNA-seq in TAMs, and summarized the detailed specific research fields. To explore the mechanism of tumor progression and drug intervention from single cell level will provide new perspective for personalized treatment strategies targeting macrophages.
Collapse
Affiliation(s)
- Xiaomin Wang
- Special Medicine Department, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yiwei Xu
- Institute of Integrated Medicine, School of Medicine, Qingdao University, Qingdao, China
| | - Qi Sun
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xintong Zhou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - JiBiao Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China; College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China; Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China.
| |
Collapse
|
43
|
Yin W, Ping YF, Li F, Lv SQ, Zhang XN, Li XG, Guo Y, Liu Q, Li TR, Yang LQ, Yang KD, Liu YQ, Luo CH, Luo T, Wang WY, Mao M, Luo M, He ZC, Cao MF, Chen C, Miao JY, Zeng H, Wang C, Zhou L, Yang Y, Yang X, Wang QH, Feng H, Shi Y, Bian XW. A map of the spatial distribution and tumour-associated macrophage states in glioblastoma and grade-4 IDH-mutant astrocytoma. J Pathol 2022; 258:121-135. [PMID: 35723032 DOI: 10.1002/path.5984] [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: 03/22/2022] [Revised: 05/08/2022] [Accepted: 06/16/2022] [Indexed: 11/10/2022]
Abstract
Tumour-associated macrophages (TAMs) abundantly infiltrate high-grade gliomas and orchestrate immune response, but their diversity in isocitrate dehydrogenase (IDH)-differential grade-4 gliomas remains largely unknown. This study aimed to dissect the transcriptional states, spatial distribution and clinicopathological significance of distinct monocyte-derived TAM (Mo-TAM) and microglia-derived TAM (Mg-TAM) clusters across glioblastoma-IDH-wildtype and astrocytoma-IDH-mutant-grade 4 (Astro-IDH-mut-G4). Single-cell RNA sequencing was performed on four cases of human glioblastoma and three cases of Astro-IDH-mut-G4. Cell clustering, single-cell regulatory network inference and gene set enrichment analysis were performed to characterize the functional states of myeloid clusters. Spatial distribution of TAM subsets was determined in human glioma tissues using multiplex immunostaining. The prognostic value of different TAM-cluster specific geneset was evaluated in the TCGA glioma cohort. Profiling and unbiased clustering of 24,227 myeloid cells from glioblastoma and Astro-IDH-mut-G4 identified 9 myeloid cell clusters including monocyte, six Mo/Mg-TAM subsets, dendritic cell, and proliferative myeloid cluster. Different Mo/Mg-TAM clusters manifest functional and transcriptional diversity controlled by specific regulons. Multiplex immunostaining of subset-specific markers identified spatial enrichment of distinct TAM clusters at peri-vascular/necrotic areas in tumour parenchyma or at tumour-brain interface. Glioblastoma harboured a substantially higher number of monocytes and Mo-TAM-inflammatory cluster, whereas Astro-IDH-mut-G4 was with higher proportion of TAM subset mediating antigen presentation. Glioblastomas with higher proportion of monocytes exhibited a mesenchymal signature, increased angiogenesis and worse patient outcome. Our findings provide insight into myeloid cell diversity and its clinical relevance in IDH-differential grade-4 gliomas, and may serve as a resource for immunotherapy development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Wen Yin
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Fei Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Xiao-Ning Zhang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Xue-Gang Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Ying Guo
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Qing Liu
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Tian-Ran Li
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Liu-Qing Yang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Kai-Di Yang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Yu-Qi Liu
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Chun-Hua Luo
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Tao Luo
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Wen-Ying Wang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Min Mao
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Min Luo
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Zhi-Cheng He
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Mian-Fu Cao
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Cong Chen
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Jing-Ya Miao
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Hui Zeng
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Chao Wang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Lei Zhou
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Ying Yang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Xi Yang
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Qiang-Hu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, PR China
| | - Yu Shi
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University) and Key Laboratory of Tumour Immunopathology, Ministry of Education of China, Chongqing, PR China
| |
Collapse
|
44
|
Liao G, Dai N, Xiong T, Wang L, Diao X, Xu Z, Ni Y, Chen D, Jiang A, Lin H, Dai S, Bai J. Single-cell transcriptomics provides insights into the origin and microenvironment of human oesophageal high-grade intraepithelial neoplasia. Clin Transl Med 2022; 12:e874. [PMID: 35608199 PMCID: PMC9128161 DOI: 10.1002/ctm2.874] [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: 01/16/2022] [Revised: 04/01/2022] [Accepted: 04/26/2022] [Indexed: 11/10/2022] Open
Abstract
Background High‐grade intraepithelial neoplasia (HIN) is the precursor of oesophageal squamous cell carcinoma. The molecular and functional properties of HIN are determined by intrinsic origin cells and the extrinsic microenvironment. Yet, these factors are poorly understood. Methods We performed single‐cell RNA sequencing of cells from HINs and adjacent tissues from the human oesophagus. We analysed the heterogeneity of basal layer cells and confirmed it using immunostaining. Aneuploid cells in HIN were studied using primary cell culture combined with karyotype analysis. We reconstructed the lineage relationship between tumour and normal populations based on transcriptome similarity. Integration analysis was applied to our epithelial data and published invasive cancer data, and results were confirmed by immunostaining and 3D organoid functional experiments. We also analysed the tumour microenvironment of HIN. Results The basal layer contained two cell populations: KRT15highSTMN1low and KRT15highSTMN1high cells, which were located mainly in the interpapillary and papillary zones, respectively. The KRT15highSTMN1low population more closely resembled stem cells and transcriptome similarity revealed that HIN probably originated from these slow‐cycling KRT15highSTMN1low cells. 3D Organoid experiments and RNA‐sequencing showed that basal‐cell features and the differentiation ability of the normal epithelium were largely retained in HIN, but may change dramatically in tumour invasion stage. Moreover, the tumour microenvironment of HIN was characterised by both inflammation and immunosuppression. Conclusions Our study provides a comprehensive single‐cell transcriptome landscape of human oesophageal HIN. Our findings on the origin cells and unique microenvironment of HIN will allow for the development of strategies to block tumour progression and even prevent cancer initiation.
Collapse
Affiliation(s)
- Guobin Liao
- Department of Gastroenterology, the Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Nan Dai
- Cancer Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Tiantian Xiong
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, China
| | - Liang Wang
- Department of Gastroenterology, the Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Xinwei Diao
- Pathology, the Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Zhizhen Xu
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, China
| | - Yuanli Ni
- Chongqing University Cancer Hospital, Chongqing, China
| | - Dingrong Chen
- Department of Gastroenterology, the Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Airui Jiang
- Department of Gastroenterology, the Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Hui Lin
- Department of Gastroenterology, the Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Shuangshuang Dai
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, China
| | - Jianying Bai
- Department of Gastroenterology, the Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
45
|
Liu C, Sun Q, Xu J, Shen W, Li H, Yang L. The Role of Bone Morphogenetic Protein 4 in Microglial Polarization in the Process of Neuropathic Pain. J Inflamm Res 2022; 15:2803-2817. [PMID: 35535051 PMCID: PMC9078433 DOI: 10.2147/jir.s356531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/27/2022] [Indexed: 12/11/2022] Open
Abstract
Background Neuropathic pain (NP) is known to be highly correlated with microglial polarization, of which the regulatory mechanism remains to be elucidated. Here, the aim of this study is to further investigate the relationship between bone morphogenetic protein 4 (BMP4) and microglial polarization in the process of NP. Methods Firstly, normal adult rats received intrathecal BMP4 administration to assess BMP4ʹs effect on microglial polarization. Secondly, a BMP4 antagonist – Noggin – was applied to a rat NP model achieved by L5 spinal nerve ligation (SNL) to investigate whether antagonizing BMP4 signaling could alleviate allodynia by reversing the imbalance of the M1/M2 polarization ratio. In both experiments, Von-Frey filaments were used to test the changes in the paw withdrawal threshold (PWT), and Western blotting, immuno-fluorescence, PCR and flow cytometry were further performed to investigate microglial activity and the expression patterns of M1 and M2 markers, respectively. Results Firstly, BMP4 administration induced a significant PWT decrease and microglial activation in normal rats; Western blotting, PCR and flow cytometry further revealed that M1 markers including CD16, MHCII, and TNF-α showed a marked elevation after BMP4 application; while M2 markers, such as Arg-1, CD204 and IL-4, peaked at an early stage (P1 or P4) and then fell to the Sham level on P7, leading to a persistent imbalance of the M1/M2 ratio throughout the 1st week. Secondly, Noggin treatment significantly relieved allodynia and microglial activation in SNL rats. Moreover, Noggin persistently downregulated the M1 marker levels and simultaneously induced a late-stage elevation of M2 markers expressions, thereby reversing the imbalance of the M1/M2 polarization ratio. Conclusion Our results indicate that BMP4 has the ability to induce microglial polarization. Antagonizing BMP4 signaling can relieve pain behavior via mitigating microglial activation and reversing the imbalance of the M1/M2 polarization ratio in the process of NP.
Collapse
Affiliation(s)
- Changqing Liu
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
- Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, People’s Republic of China
| | - Qi Sun
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
- Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, People’s Republic of China
| | - Junmei Xu
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
- Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, People’s Republic of China
| | - Weiyun Shen
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
- Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, People’s Republic of China
| | - Hui Li
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
- Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, People’s Republic of China
- Correspondence: Hui Li, Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China, Fax +86 85295970, Email
| | - Lin Yang
- Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
- Hunan Province Center for Clinical Anesthesia and Anesthesiology, Research Institute of Central South University, Changsha, People’s Republic of China
- Lin Yang, Department of Anesthesiology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China, Fax +86 85295970, Email
| |
Collapse
|
46
|
Hohsfield LA, Tsourmas KI, Ghorbanian Y, Syage AR, Kim SJ, Cheng Y, Furman S, Inlay MA, Lane TE, Green KN. MAC2 is a long-lasting marker of peripheral cell infiltrates into the mouse CNS after bone marrow transplantation and coronavirus infection. Glia 2022; 70:875-891. [PMID: 35025109 PMCID: PMC8930563 DOI: 10.1002/glia.24144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 01/09/2023]
Abstract
Microglia are the primary resident myeloid cells of the brain responsible for maintaining homeostasis and protecting the central nervous system (CNS) from damage and infection. Monocytes and monocyte-derived macrophages arising from the periphery have also been implicated in CNS pathologies, however, distinguishing between different myeloid cell populations in the CNS has been difficult. Here, we set out to develop a reliable histological marker that can assess distinct myeloid cell heterogeneity and functional contributions, particularly in the context of disease and/or neuroinflammation. scRNAseq from brains of mice infected with the neurotropic JHM strain of the mouse hepatitis virus (JHMV), a mouse coronavirus, revealed that Lgals3 is highly upregulated in monocyte and macrophage populations, but not in microglia. Subsequent immunostaining for galectin-3 (encoded by Lgals3), also referred to as MAC2, highlighted the high expression levels of MAC2 protein in infiltrating myeloid cells in JHMV-infected and bone marrow (BM) chimeric mice, in stark contrast to microglia, which expressed little to no staining in these models. Expression of MAC2 was found even 6-10 months following BM-derived cell infiltration into the CNS. We also demonstrate that MAC2 is not a specific label for plaque-associated microglia in the 5xFAD mouse model, but only appears in a distinct subset of these cells in the presence of JHMV infection or during aging. Our data suggest that MAC2 can serve as a reliable and long-lasting histological marker for monocyte/macrophages in the brain, identifying an accessible approach to distinguishing resident microglia from infiltrating cells in the CNS under certain conditions.
Collapse
Affiliation(s)
- Lindsay A. Hohsfield
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Kate Inman Tsourmas
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Yasamine Ghorbanian
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Amber R. Syage
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Sung Jin Kim
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Yuting Cheng
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Susana Furman
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Matthew A. Inlay
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Thomas E. Lane
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Kim N. Green
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| |
Collapse
|
47
|
Herr SA, Gardeen SS, Low PS, Shi R. Targeted delivery of acrolein scavenger hydralazine in spinal cord injury using folate-linker-drug conjugation. Free Radic Biol Med 2022; 184:66-73. [PMID: 35398493 DOI: 10.1016/j.freeradbiomed.2022.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/30/2022] [Accepted: 04/03/2022] [Indexed: 11/26/2022]
Abstract
Oxidative stress has been shown to play a critical pathogenic role in functional loss after spinal cord injury (SCI). As a direct result of oxidative stress, lipid peroxidation-derived aldehydes have emerged as key culprits that sustain secondary injury and contribute significantly to pathological outcomes. Acrolein, a neurotoxin, has been shown to be elevated in SCI and can result in post-SCI neurological deficits. Reducing acrolein has therefore emerged as a novel and effective therapeutic strategy in SCI. Previous studies have revealed that hydralazine, an FDA approved blood pressure lowering medication, when administered after SCI shows strong acrolein scavenging capabilities and significantly improves cellular and behavioral outcomes. However, while effective at scavenging acrolein, hydralazine's blood pressure lowering activity can have a detrimental impact on neurotrauma patients. Here, our goal was to preserve the acrolein scavenging capability while mitigating the effect of hydralazine on blood pressure. We accomplished this using a folate-targeted delivery system to deploy hydralazine to the folate receptor positive inflammatory site of the cord injury. Using a model of rat SCI, we found that this system is effective for targeting the injury site, and that folate targeted hydralazine can scavenge acrolein without significantly impacting blood pressure.
Collapse
Affiliation(s)
- Seth A Herr
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University. Lynn Hall, 625 N Harrison St. West Lafayette, IN, 47907, USA.
| | - Spencer S Gardeen
- Department of Chemistry, College of Science, Purdue University. Drug Discovery Building, 720 Clinic Dr. West Lafayette, IN, 47907, USA.
| | - Philip S Low
- Department of Chemistry, College of Science, Purdue University. Drug Discovery Building, 720 Clinic Dr. West Lafayette, IN, 47907, USA.
| | - Riyi Shi
- Center for Paralysis Research & Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University. Lynn Hall, 625 N Harrison St. West Lafayette, IN, 47907, USA.
| |
Collapse
|
48
|
Hou Z, Wang L, Su D, Cai W, Zhu Y, Liu D, Huang S, Xu J, Pan Z, Tao J. Global MicroRNAs Expression Profile Analysis Reveals Possible Regulatory Mechanisms of Brain Injury Induced by Toxoplasma gondii Infection. Front Neurosci 2022; 16:827570. [PMID: 35360170 PMCID: PMC8961362 DOI: 10.3389/fnins.2022.827570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/16/2022] [Indexed: 11/13/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is an obligate intracellular parasitic protozoan that can cause toxoplasmosis in humans and other endotherms. T. gondii can manipulate the host gene expression profile by interfering with miRNA expression, which is closely associated with the molecular mechanisms of T. gondii-induced brain injury. However, it is unclear how T. gondii manipulates the gene expression of central nervous system (CNS) cells through modulation of miRNA expression in vivo during acute and chronic infection. Therefore, high-throughput sequencing was used to investigate expression profiles of brain miRNAs at 10, 25, and 50 days post-infection (DPI) in pigs infected with the Chinese I genotype T. gondii strain in this study. Compared with the control group 87, 68, and 135 differentially expressed miRNAs (DEMs) were identified in the infected porcine brains at 10, 25, and 50 DPI, respectively. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that a large number significantly enriched GO terms and KEGG pathways were found, and were mostly associated with stimulus or immune response, signal transduction, cell death or apoptosis, metabolic processes, immune system or diseases, and cancers. miRNA–gene network analysis revealed that the crucial connecting nodes, including DEMs and their target genes, might have key roles in the interactions between porcine brain and T. gondii. These results suggest that the regulatory strategies of T. gondii are involved in the modulation of a variety of host cell signaling pathways and cellular processes, containing unfolded protein response (UPR), oxidative stress (OS), autophagy, apoptosis, tumorigenesis, and inflammatory responses, by interfering with the global miRNA expression profile of CNS cells, allowing parasites to persist in the host CNS cells and contribute to pathological damage of porcine brain. To our knowledge, this is the first report on miRNA expression profile in porcine brains during acute and chronic T. gondii infection in vivo. Our results provide new insights into the mechanisms underlying T. gondii-induced brain injury during different infection stages and novel targets for developing therapeutic agents against T. gondii.
Collapse
Affiliation(s)
- Zhaofeng Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Lele Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Dingzeyang Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Weimin Cai
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Yu Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Dandan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Siyang Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Jinjun Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Zhiming Pan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
| | - Jianping Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou, China
- *Correspondence: Jianping Tao,
| |
Collapse
|
49
|
Ocañas SR, Pham KD, Blankenship HE, Machalinski AH, Chucair-Elliott AJ, Freeman WM. Minimizing the Ex Vivo Confounds of Cell-Isolation Techniques on Transcriptomic and Translatomic Profiles of Purified Microglia. eNeuro 2022; 9:ENEURO.0348-21.2022. [PMID: 35228310 PMCID: PMC8970438 DOI: 10.1523/eneuro.0348-21.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/10/2021] [Accepted: 01/25/2022] [Indexed: 01/21/2023] Open
Abstract
Modern molecular and biochemical neuroscience studies require analysis of specific cellular populations derived from brain tissue samples to disambiguate cell type-specific events. This is particularly true in the analysis of minority glial populations in the brain, such as microglia, which may be obscured in whole tissue analyses. Microglia have central functions in development, aging, and neurodegeneration and are a current focus of neuroscience research. A long-standing concern for glial biologists using in vivo models is whether cell isolation from CNS tissue could introduce ex vivo artifacts in microglia, which respond quickly to changes in the environment. Mouse microglia were purified by magnetic-activated cell sorting (MACS), as well as cytometer-based and cartridge-based fluorescence-activated cell sorting (FACS) approaches to compare and contrast performance. The Cx3cr1-NuTRAP mouse model was used to provide an endogenous fluorescent microglial marker and a microglial-specific translatome profile as a baseline comparison lacking cell isolation artifacts. All sorting methods performed similarly for microglial purity with main differences being in cell yield and time of isolation. Ex vivo activation signatures occurred principally during the initial tissue dissociation and cell preparation and not the cell sorting. The cell preparation-induced activational phenotype could be minimized by inclusion of transcriptional and translational inhibitors or non-enzymatic dissociation conducted entirely at low temperatures. These data demonstrate that a variety of microglial isolation approaches can be used, depending on experimental needs, and that inhibitor cocktails are effective at reducing cell preparation artifacts.
Collapse
Affiliation(s)
- Sarah R Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Kevin D Pham
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Harris E Blankenship
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Adeline H Machalinski
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Ana J Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Willard M Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK 73104
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| |
Collapse
|
50
|
Markwell SM, Ross JL, Olson CL, Brat DJ. Necrotic reshaping of the glioma microenvironment drives disease progression. Acta Neuropathol 2022; 143:291-310. [PMID: 35039931 DOI: 10.1007/s00401-021-02401-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma is the most common primary brain tumor and has a dismal prognosis. The development of central necrosis represents a tipping point in the evolution of these tumors that foreshadows aggressive expansion, swiftly leading to mortality. The onset of necrosis, severe hypoxia and associated radial glioma expansion correlates with dramatic tumor microenvironment (TME) alterations that accelerate tumor growth. In the past, most have concluded that hypoxia and necrosis must arise due to "cancer outgrowing its blood supply" when rapid tumor growth outpaces metabolic supply, leading to diffusion-limited hypoxia. However, growing evidence suggests that microscopic intravascular thrombosis driven by the neoplastic overexpression of pro-coagulants attenuates glioma blood supply (perfusion-limited hypoxia), leading to TME restructuring that includes breakdown of the blood-brain barrier, immunosuppressive immune cell accumulation, microvascular hyperproliferation, glioma stem cell enrichment and tumor cell migration outward. Cumulatively, these adaptations result in rapid tumor expansion, resistance to therapeutic interventions and clinical progression. To inform future translational investigations, the complex interplay among environmental cues and myriad cell types that contribute to this aggressive phenotype requires better understanding. This review focuses on contributions from intratumoral thrombosis, the effects of hypoxia and necrosis, the adaptive and innate immune responses, and the current state of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Steven M Markwell
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA
| | - James L Ross
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Cheryl L Olson
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA
| | - Daniel J Brat
- Department of Pathology, Northwestern Medicine Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave. Ward 3-140, Chicago, IL, USA.
| |
Collapse
|