1
|
Yuasa-Kawada J, Kinoshita-Kawada M, Hiramoto M, Yamagishi S, Mishima T, Yasunaga S, Tsuboi Y, Hattori N, Wu JY. Neuronal guidance signaling in neurodegenerative diseases: Key regulators that function at neuron-glia and neuroimmune interfaces. Neural Regen Res 2026; 21:612-635. [PMID: 39995079 DOI: 10.4103/nrr.nrr-d-24-01330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
The nervous system processes a vast amount of information, performing computations that underlie perception, cognition, and behavior. During development, neuronal guidance genes, which encode extracellular cues, their receptors, and downstream signal transducers, organize neural wiring to generate the complex architecture of the nervous system. It is now evident that many of these neuroguidance cues and their receptors are active during development and are also expressed in the adult nervous system. This suggests that neuronal guidance pathways are critical not only for neural wiring but also for ongoing function and maintenance of the mature nervous system. Supporting this view, these pathways continue to regulate synaptic connectivity, plasticity, and remodeling, and overall brain homeostasis throughout adulthood. Genetic and transcriptomic analyses have further revealed many neuronal guidance genes to be associated with a wide range of neurodegenerative and neuropsychiatric disorders. Although the precise mechanisms by which aberrant neuronal guidance signaling drives the pathogenesis of these diseases remain to be clarified, emerging evidence points to several common themes, including dysfunction in neurons, microglia, astrocytes, and endothelial cells, along with dysregulation of neuron-microglia-astrocyte, neuroimmune, and neurovascular interactions. In this review, we explore recent advances in understanding the molecular and cellular mechanisms by which aberrant neuronal guidance signaling contributes to disease pathogenesis through altered cell-cell interactions. For instance, recent studies have unveiled two distinct semaphorin-plexin signaling pathways that affect microglial activation and neuroinflammation. We discuss the challenges ahead, along with the therapeutic potentials of targeting neuronal guidance pathways for treating neurodegenerative diseases. Particular focus is placed on how neuronal guidance mechanisms control neuron-glia and neuroimmune interactions and modulate microglial function under physiological and pathological conditions. Specifically, we examine the crosstalk between neuronal guidance signaling and TREM2, a master regulator of microglial function, in the context of pathogenic protein aggregates. It is well-established that age is a major risk factor for neurodegeneration. Future research should address how aging and neuronal guidance signaling interact to influence an individual's susceptibility to various late-onset neurological diseases and how the progression of these diseases could be therapeutically blocked by targeting neuronal guidance pathways.
Collapse
Affiliation(s)
| | | | | | - Satoru Yamagishi
- Department of Optical Neuroanatomy, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takayasu Mishima
- Division of Neurology, Department of Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| | - Shin'ichiro Yasunaga
- Department of Biochemistry, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Jane Y Wu
- Department of Neurology, Center for Genetic Medicine, Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Di Stefano J, Di Marco F, Cicalini I, FitzGerald U, Pieragostino D, Verhoye M, Ponsaerts P, Van Breedam E. Generation, interrogation, and future applications of microglia-containing brain organoids. Neural Regen Res 2025; 20:3448-3460. [PMID: 39665813 PMCID: PMC11974650 DOI: 10.4103/nrr.nrr-d-24-00921] [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: 08/13/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Brain organoids encompass a large collection of in vitro stem cell-derived 3D culture systems that aim to recapitulate multiple aspects of in vivo brain development and function. First, this review provides a brief introduction to the current state-of-the-art for neuro-ectoderm brain organoid development, emphasizing their biggest advantages in comparison with classical two-dimensional cell cultures and animal models. However, despite their usefulness for developmental studies, a major limitation for most brain organoid models is the absence of contributing cell types from endodermal and mesodermal origin. As such, current research is highly investing towards the incorporation of a functional vasculature and the microglial immune component. In this review, we will specifically focus on the development of immune-competent brain organoids. By summarizing the different approaches applied to incorporate microglia, it is highlighted that immune-competent brain organoids are not only important for studying neuronal network formation, but also offer a clear future as a new tool to study inflammatory responses in vitro in 3D in a brain-like environment. Therefore, our main focus here is to provide a comprehensive overview of assays to measure microglial phenotype and function within brain organoids, with an outlook on how these findings could better understand neuronal network development or restoration, as well as the influence of physical stress on microglia-containing brain organoids. Finally, we would like to stress that even though the development of immune-competent brain organoids has largely evolved over the past decade, their full potential as a pre-clinical tool to study novel therapeutic approaches to halt or reduce inflammation-mediated neurodegeneration still needs to be explored and validated.
Collapse
Affiliation(s)
- Julia Di Stefano
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
| | - Federica Di Marco
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Ilaria Cicalini
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Una FitzGerald
- CÚRAM, Center for Research in Medical Devices, Biomedical Engineering, University of Galway, Ireland
- Galway Neuroscience Center, University of Galway, Ireland
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- μNEURO Research Center of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
3
|
Yang B, Li Z, Li P, Liu Y, Ding X, Feng E. Piezo1 in microglial cells: Implications for neuroinflammation and tumorigenesis. Channels (Austin) 2025; 19:2492161. [PMID: 40223276 PMCID: PMC12005408 DOI: 10.1080/19336950.2025.2492161] [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: 12/11/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025] Open
Abstract
Microglia, the central nervous system (CNS) resident immune cells, are pivotal in regulating neurodevelopment, maintaining neural homeostasis, and mediating neuroinflammatory responses. Recent research has highlighted the importance of mechanotransduction, the process by which cells convert mechanical stimuli into biochemical signals, in regulating microglial activity. Among the various mechanosensitive channels, Piezo1 has emerged as a key player in microglia, influencing their behavior under both physiological and pathological conditions. This review focuses on the expression and role of Piezo1 in microglial cells, particularly in the context of neuroinflammation and tumorigenesis. We explore how Piezo1 mediates microglial responses to mechanical changes within the CNS, such as alterations in tissue stiffness and fluid shear stress, which are common in conditions like multiple sclerosis, Alzheimer's disease, cerebral ischemia, and gliomas. The review also discusses the potential of targeting Piezo1 for therapeutic intervention, given its involvement in the modulation of microglial activity and its impact on disease progression. This review integrates findings from recent studies to provide a comprehensive overview of Piezo1's mechanistic pathways in microglial function. These insights illuminate new possibilities for developing targeted therapies addressing CNS disorders with neuroinflammation and pathological tissue mechanics.
Collapse
Affiliation(s)
- Bo Yang
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhenyu Li
- Department of Neonatology, Children’s Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Peiliang Li
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yuhan Liu
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xinghuan Ding
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Enshan Feng
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Matsuo K, Watanabe M, Inamine S, Matsushima T, Kyuragi S, Maeda Y, Katsuki R, Ohgidani M, Yamasaki R, Isobe N, Nakao T, Kato TA. The flow cytometric analysis of depression focusing on modern-type depression and hikikomori: Exploring the link between subtypes of depression and immunological imbalances. DIALOGUES IN CLINICAL NEUROSCIENCE 2025; 27:13-25. [PMID: 39831784 PMCID: PMC11748865 DOI: 10.1080/19585969.2025.2452842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
INTRODUCTION Depression includes different phenotypes. Modern-type depression (MTD) is a gateway disorder to pathological social withdrawal, known as hikikomori. Adverse childhood experiences (ACEs) are also important aetiologies of depression. Recently, immune imbalance has been proposed as a biological basis of depression. We hypothesised that peripheral immunological characteristics may be involved in subtyping of depression. METHODS 21 patients with major depressive disorder (MDD) and 24 healthy controls (HC) were recruited. Peripheral blood mononuclear cells (PBMCs) were examined for surface antigens by flow cytometry. Participants were administered psychological scales such as Patient Health Questionnaire (PHQ)-9, Modern-Type Depression Trait Scale (TACS-22), Hikikomori Questionnaire (HQ-25), Child Abuse and Trauma Scale (CATS). RESULTS MDD group showed significantly higher percentage of B cells than HC group (p = 0.032). MDD group presented a negative correlation between: PHQ-9 and CD8 T effector memory cells (r= -0.639, p = 0.002), TACS-22 and monocytes (r= -0.459, p = 0.036), HQ-25 and NK T cells (r= -0.638, p = 0.004), CATS and Intermediate monocytes (r= -0.594, p = 0.009). CONCLUSIONS MTD traits, hikikomori tendencies, and ACEs were correlated with specific characteristics of peripheral immune cells. Our results suggest that immune imbalance influences the diverse presentations of depression. Further validation is warranted by large-scale prospective studies.
Collapse
Affiliation(s)
- Keitaro Matsuo
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shogo Inamine
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshio Matsushima
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sota Kyuragi
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuhiro Maeda
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryoko Katsuki
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Ohgidani
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Hokkaido, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Nakao
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro A. Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
5
|
Ning W, Lv S, Wang Q, Xu Y. The pivotal role of microglia in injury and the prognosis of subarachnoid hemorrhage. Neural Regen Res 2025; 20:1829-1848. [PMID: 38993136 PMCID: PMC11691474 DOI: 10.4103/nrr.nrr-d-24-00241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/18/2024] [Accepted: 05/25/2024] [Indexed: 07/13/2024] Open
Abstract
Subarachnoid hemorrhage leads to a series of pathological changes, including vascular spasm, cellular apoptosis, blood-brain barrier damage, cerebral edema, and white matter injury. Microglia, which are the key immune cells in the central nervous system, maintain homeostasis in the neural environment, support neurons, mediate apoptosis, participate in immune regulation, and have neuroprotective effects. Increasing evidence has shown that microglia play a pivotal role in the pathogenesis of subarachnoid hemorrhage and affect the process of injury and the prognosis of subarachnoid hemorrhage. Moreover, microglia play certain neuroprotective roles in the recovery phase of subarachnoid hemorrhage. Several approaches aimed at modulating microglia function are believed to attenuate subarachnoid hemorrhage injury. This provides new targets and ideas for the treatment of subarachnoid hemorrhage. However, an in-depth and comprehensive summary of the role of microglia after subarachnoid hemorrhage is still lacking. This review describes the activation of microglia after subarachnoid hemorrhage and their roles in the pathological processes of vasospasm, neuroinflammation, neuronal apoptosis, blood-brain barrier disruption, cerebral edema, and cerebral white matter lesions. It also discusses the neuroprotective roles of microglia during recovery from subarachnoid hemorrhage and therapeutic advances aimed at modulating microglial function after subarachnoid hemorrhage. Currently, microglia in subarachnoid hemorrhage are targeted with TLR inhibitors, nuclear factor-κB and STAT3 pathway inhibitors, glycine/tyrosine kinases, NLRP3 signaling pathway inhibitors, Gasdermin D inhibitors, vincristine receptor α receptor agonists, ferroptosis inhibitors, genetic modification techniques, stem cell therapies, and traditional Chinese medicine. However, most of these are still being evaluated at the laboratory stage. More clinical studies and data on subarachnoid hemorrhage are required to improve the treatment of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Wenjing Ning
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Shi Lv
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Qian Wang
- Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong Province, China
| | - Yuzhen Xu
- Department of Rehabilitation, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| |
Collapse
|
6
|
Chen Y, Deng H, Zhang N. Autophagy-targeting modulation to promote peripheral nerve regeneration. Neural Regen Res 2025; 20:1864-1882. [PMID: 39254547 PMCID: PMC11691477 DOI: 10.4103/nrr.nrr-d-23-01948] [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: 11/27/2023] [Revised: 02/22/2024] [Accepted: 03/29/2024] [Indexed: 09/11/2024] Open
Abstract
Nerve regeneration following traumatic peripheral nerve injuries and neuropathies is a complex process modulated by diverse factors and intricate molecular mechanisms. Past studies have focused on factors that stimulate axonal outgrowth and myelin regeneration. However, recent studies have highlighted the pivotal role of autophagy in peripheral nerve regeneration, particularly in the context of traumatic injuries. Consequently, autophagy-targeting modulation has emerged as a promising therapeutic approach to enhancing peripheral nerve regeneration. Our current understanding suggests that activating autophagy facilitates the rapid clearance of damaged axons and myelin sheaths, thereby enhancing neuronal survival and mitigating injury-induced oxidative stress and inflammation. These actions collectively contribute to creating a favorable microenvironment for structural and functional nerve regeneration. A range of autophagy-inducing drugs and interventions have demonstrated beneficial effects in alleviating peripheral neuropathy and promoting nerve regeneration in preclinical models of traumatic peripheral nerve injuries. This review delves into the regulation of autophagy in cell types involved in peripheral nerve regeneration, summarizing the potential drugs and interventions that can be harnessed to promote this process. We hope that our review will offer novel insights and perspectives on the exploitation of autophagy pathways in the treatment of peripheral nerve injuries and neuropathies.
Collapse
Affiliation(s)
- Yan Chen
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Reproductive Endocrinology and Reproductive Regulation, Sichuan University, Chengdu, Sichuan Province, China
| | - Hongxia Deng
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Reproductive Endocrinology and Reproductive Regulation, Sichuan University, Chengdu, Sichuan Province, China
| | - Nannan Zhang
- Key Laboratory of Birth Defects and Women and Children’s Diseases, Ministry of Education, Sichuan University, Chengdu, Sichuan Province, China
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
7
|
Wang M, Jin G, Duan T, Li R, Gao Y, Yu M, Xu Y. Microglial phagocytosis and regulatory mechanisms: Key players in the pathophysiology of depression. Neuropharmacology 2025; 271:110383. [PMID: 39993469 DOI: 10.1016/j.neuropharm.2025.110383] [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: 09/08/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/26/2025]
Abstract
Depression is a globally prevalent emotional disorder with a complex pathophysiology. Microglia are resident immune cells in the central nervous system, playing crucial roles in regulating inflammation, synaptic plasticity, immune phagocytosis, and other functions, thereby exerting significant impacts on neuropsychiatric disorders like depression. Increasing research indicates that abnormal phagocytic function of microglia in the brain is involved in depression, showing excessive or insufficient phagocytosis in different states. Here, we have provided a review of the signaling molecules involved in microglial phagocytosis in depression, including "eat me" signals such as phosphatidylserine (PS), complement, and "don't eat me" signals such as CD47, CD200 and related receptors. Furthermore, we discuss the regulatory effects of existing pharmaceuticals and dietary nutrients on microglial phagocytosis in depression, emphasizing the need for tailored modulation based on the varying phagocytic states of microglia. This review aims to facilitate a deeper understanding of the role of microglial phagocytosis in depression and provide a roadmap for potential therapeutic strategies for depression targeting microglial phagocytosis.
Collapse
Affiliation(s)
- Man Wang
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Guimin Jin
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Tingting Duan
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Run Li
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Yubin Gao
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China
| | - Ming Yu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China.
| | - Yuhao Xu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212001, China; Department of Neuroimaging Laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
8
|
Zhang X, Yan S, Zhang X, Huang D, Zhou J, Song X, Hao Y, Wang X, Yan J. TREML2 enhances sensitivity of acute myeloid leukemia cells to chemotherapy by inhibiting the NF-κB/CXCL10 pathway. BLOOD SCIENCE 2025; 7:e00223. [PMID: 40109578 PMCID: PMC11922426 DOI: 10.1097/bs9.0000000000000223] [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: 11/04/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
The triggering receptors expressed on myeloid cells (TREMs) family of cell surface receptors are mainly expressed by myeloid cells. The expression profile of TREM-like 2 (TREML2), a TREM family member, in patients with acute myeloid leukemia (AML) is unknown. In this study, we aimed to elucidate the role of TREML2 in the development of AML. We analyzed the TREML2 expression profile in patients with AML. TREML2 was expressed at lower levels in patients with AML than in healthy individuals. The partial remission (PR) + no remission (NR) group showed lower TREML2 expression levels and a poorer chemotherapy response than that observed in the complete remission group. Overall survival was significantly shorter in the group with low TREML2 expression levels than in the group with high TREML2 expression levels. TREML2 inhibited the proliferation of AML cells and enhanced the sensitivity of AML cells to doxorubicin. Mechanistically, TREML2 reduced C-X-C motif chemokine ligand 10 expression levels by inhibiting the nuclear factor kappa B pathway. Taken together, we demonstrate that TREML2 has diagnostic value as a potential indicator of AML and that upregulation of TREML2 may be a new strategy to overcome doxorubicin resistance for AML treatment.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Shuheng Yan
- University of California, Davis, College of Biological Science, 1 Shields Ave, Davis, CA 95616, United States
| | - Xuehong Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044, China
| | - Dan Huang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Jiayin Zhou
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Xiaoting Song
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Yuchao Hao
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Xijia Wang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, Dalian, China
| | - Jinsong Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Blood Stem Cell Transplantation Institute, Dalian Key Laboratory of Hematology, Diamond Bay Institute of Hematology, the Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
Lawrence AB, Brown SM, Bradford BM, Mabbott NA, Bombail V, Rutherford KMD. Non-neuronal brain biology and its relevance to animal welfare. Neurosci Biobehav Rev 2025; 173:106136. [PMID: 40185375 DOI: 10.1016/j.neubiorev.2025.106136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Non-neuronal cells constitute a significant portion of brain tissue and are seen as having key roles in brain homeostasis and responses to challenges. This review illustrates how non-neuronal biology can bring new perspectives to animal welfare through understanding mechanisms that determine welfare outcomes and highlighting interventions to improve welfare. Most obvious in this respect is the largely unrecognised relevance of neuroinflammation to animal welfare which is increasingly found to have roles in determining how animals respond to challenges. We start by introducing non-neuronal cells and review their involvement in affective states and cognition often seen as core psychological elements of animal welfare. We find that the evidence for a causal involvement of glia in cognition is currently more advanced than the corresponding evidence for affective states. We propose that translational research on affective disorders could usefully apply welfare science derived approaches for assessing affective states. Using evidence from translational research, we illustrate the involvement of non-neuronal cells and neuroinflammatory processes as mechanisms modulating resilience to welfare challenges including disease, pain, and social stress. We review research on impoverished environments and environmental enrichment which suggests that environmental conditions which improve animal welfare also improve resilience to challenges through balancing pro- and anti-inflammatory non-neuronal processes. We speculate that non-neuronal biology has relevance to animal welfare beyond neuro-inflammation including facilitating positive affective states. We acknowledge the relevance of neuronal biology to animal welfare whilst proposing that non-neuronal biology provides additional and relevant insights to improve animals' lives.
Collapse
Affiliation(s)
- Alistair B Lawrence
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK; Scotland's Rural College (SRUC), Edinburgh EH9 3JG, UK.
| | - Sarah M Brown
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Barry M Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | | | | |
Collapse
|
10
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial NOX2 as a therapeutic target in traumatic brain injury: Mechanisms, consequences, and potential for neuroprotection. Ageing Res Rev 2025; 108:102735. [PMID: 40122395 DOI: 10.1016/j.arr.2025.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability worldwide, with secondary injury mechanisms, including neuroinflammation and oxidative stress, driving much of its chronic pathology. While NADPH oxidase 2 (NOX2)-mediated reactive oxygen species (ROS) production is a recognized factor in TBI, the specific role of microglial NOX2 in perpetuating oxidative and inflammatory damage remains underexplored. Addressing this gap is critical, as current therapeutic approaches primarily target acute symptoms and fail to interrupt the persistent neuroinflammation that contributes to progressive neurodegeneration. Besides NOX, other ROS-generating enzymes, such as CYP1B1, COX2, and XO, also play crucial roles in triggering oxidative stress and neuroinflammatory conditions in TBI. However, this review highlights the pathophysiological role of microglial NOX2 in TBI, focusing on its activation following injury and its impact on ROS generation, neuroinflammatory signaling, and neuronal loss. These insights reveal NOX2 as a critical driver of secondary injury, linked to worsened outcomes, particularly in aged individuals where NOX2 activation is more pronounced. In addition, this review evaluates emerging therapeutic approaches targeting NOX2, such as GSK2795039 and other selective NOX2 inhibitors, which show potential in reducing ROS levels, limiting neuroinflammation, and preserving neurological functions. By highlighting the specific role of NOX2 in microglial ROS production and secondary neurodegeneration, this study advocates for NOX2 inhibition as a promising strategy to improve TBI outcomes by addressing the unmet need for therapies targeting long-term inflammation and neuroprotection. Our review highlights the potential of NOX2-targeted interventions to disrupt the cycle of oxidative stress and inflammation, ultimately offering a pathway to mitigate the chronic impact of TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
11
|
Saito Y, Fujiwara Y, Yamaguchi YL, Tanaka SS, Miura K, Hizukuri Y, Yamashiro K, Hayashi Y, Nakashima Y, Komohara Y. Rodent monocyte-derived macrophages do not express CD163: Comparative analysis using macrophages from living boreoeutherians. Dev Dyn 2025. [PMID: 40355384 DOI: 10.1002/dvdy.70036] [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: 08/02/2024] [Revised: 02/27/2025] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND CD163 is a scavenger receptor predominantly expressed on the surfaces of macrophages in various mammalian species and is a marker of anti-inflammatory (M2-like) macrophages. High density of CD163-positive tumor-associated macrophages (TAMs) is associated with worse prognosis in various patient tumors. Interestingly, studies on mice have shown that CD163-positive TAMs only infiltrate the margins of tumor tissues, not the center. Based on these observations, we hypothesized that circulating monocyte-derived macrophages (MDMs), which are the origin of most TAMs, do not express CD163 in mice. RESULTS We examined CD163 expression in MDMs, differentiated from healthy animals in vitro, and in normal, pathogenic, and tumorigenic macrophages infiltrating various tumors and organs across multiple species including primates, rodents, cetartiodactylans, and carnivores. We found that MDMs, including TAMs, do not express CD163 in mice. Our findings also suggest that murine CD163-positive macrophages likely originate from a specific subset of resident macrophages, namely fetal liver monocytes/macrophages, as indicated by fetal analysis. Furthermore, we revealed that the CD163-negative expression pattern in MDMs is a trait shared by the rodent clade. CONCLUSIONS Rodent MDMs do not express CD163, a phenotype not shared with MDMs of other mammals. Our findings caution against the extrapolation of rodent experimental results to other animal models.
Collapse
Affiliation(s)
- Yoichi Saito
- Laboratory of Bioengineering, Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuka L Yamaguchi
- Laboratory of Molecular Embryology, Faculty of Health Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Satomi S Tanaka
- Laboratory of Molecular Embryology, Faculty of Health Science, Kumamoto Health Science University, Kumamoto, Japan
| | - Kyoko Miura
- Department of Aging and Longevity Research, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | | | | | - Yuta Nakashima
- Laboratory of Bioengineering, Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
- Institute of Industrial Nanomaterials, Kumamoto University, Kumamoto, Japan
- International Research Organization for Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
- Fusion Oriented Research for Disruptive Science and Technology, Japan Science and Technology Agency, Saitama, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
12
|
Naydovich LR, Orthmann-Murphy JL, Markowitz CE. Beyond relapses: How BTK inhibitors are shaping the future of progressive MS treatment. Neurotherapeutics 2025:e00602. [PMID: 40345950 DOI: 10.1016/j.neurot.2025.e00602] [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: 12/24/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Multiple sclerosis is a biologically and clinically heterogenous inflammatory demyelinating disease, driven by relapsing and progressive mechanisms, all individuals experiencing varying degrees of both. Existing highly effective therapies target peripheral inflammation and reduce relapse rates but have limited efficacy in progressive MS due to poor blood-brain barrier penetration and inability to address neurodegeneration. Bruton's tyrosine kinase (BTK) inhibitors represent an emerging therapeutic class offering a novel mechanism targeting BTK, which is expressed by both B cells and myeloid cells, including microglia within the CNS. Pre-clinical, Phase II, and Phase III clinical trials have demonstrated promising results in modulating progressive disease in both relapsing and non-relapsing MS patients. In contrast, the evidence regarding impact on relapse biology remains mixed and somewhat inconclusive. This review highlights gaps in current therapeutic strategies, examines the latest evidence for the efficacy and safety of BTK inhibitors in MS, and explores the future landscape of MS treatment.
Collapse
Affiliation(s)
- Laura R Naydovich
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| | | | - Clyde E Markowitz
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
13
|
Ho MFS, Farkas O, Faria AV, Plemel JR, Kerr BJ. A recent history of immune cell sex differences in the peripheral nervous system in persistent pain states. Brain Behav Immun 2025:S0889-1591(25)00181-3. [PMID: 40345628 DOI: 10.1016/j.bbi.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/23/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025] Open
Abstract
Pain is entwined with inflammation, and biological sex often influences mechanisms of the immune system. Due to possible differences in inflammatory mechanisms, women are predisposed to autoimmune diseases and chronic pain. Despite sex as a critical variable in clinical cases of autoimmune conditions and its pain comorbidities, fundamental investigations have long underrepresented female subjects in their studies. Fundamental research in the 2010 s, however, identified a binary sex specific mechanism for pain in rodents: male pain is microglia-driven while female pain is T cell-driven. Since then, studies have expanded in neuro-immunology to indicate that the sex differences and immune cells involved in these processes take on more elaborate roles when expanded to other causal modalities and anatomical levels of neuropathic and inflammatory pain. In this mini-review, we highlight updated roles for macrophages, T cells, and B cells in the peripheral nervous system during persistent pain conditions: neuropathic pain and inflammatory pain. We discuss sex similarities and sex differences in these cell types. By parsing out the sex specific roles of immune cells in persistent pain states we may be better positioned to find immune-based therapies that can effectively target chronic pain in sex-biased autoimmune conditions.
Collapse
Affiliation(s)
| | - Olivia Farkas
- Neuroscience and Mental Health Institute, University of Alberta, Canada
| | | | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Canada; Department of Medicine, Division of Neurology, University of Alberta, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Canada; Departments of Pharmacology and Physiology, University of Alberta, Canada; Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
14
|
Ferreira GAM, Pinto LAM. Neural Stem Cell-Derived Astrogliogenesis: The Hidden Player of the Adult Hippocampal Cytogenic Niche. Glia 2025. [PMID: 40326621 DOI: 10.1002/glia.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
The adult mammalian brain exhibits remarkable forms of neural plasticity, enabling it to adapt and reorganize in response to internal and external stimuli. These plastic mechanisms include cytogenesis, the capacity of producing new neuronal and glial cells in restricted brain regions through processes known as neuro- and gliogenesis, respectively. Although many advances have been made in understanding adult brain plastic processes associated with cell genesis, as well as its functional and behavioral implications, most of the evidence is focused on neuronal cells. Even though astrocytes play a critical role in maintaining a neurochemical and electrophysiological homeostasis in the brain and provide a pivotal support to neuronal activity, the molecular mechanisms underlying the formation and functional integration of newly formed astroglial cells are poorly understood. However, some studies have provided key insights into the molecular mechanisms driving the generation of adult neural stem cell (NSC)-derived astrocytes, focusing on the dentate gyrus of the hippocampal cytogenic niche. Recent work has demonstrated that intrinsic and extrinsic factors can modulate astrogliogenesis. In the context of neuropathogenesis, this mechanism may be compromised in the hippocampus, contributing to functional and behavioral impairments. Here, we review the mechanisms underlying NSC-derived hippocampal astrogliogenesis, examining current perspectives on how adult-born astrocytes develop in the adult brain, their functional relevance, and the intricate regulation of the astrogliogenic process.
Collapse
Affiliation(s)
- Gonçalo Alexandre Martins Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Luísa Alexandra Meireles Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Bn'ML-Behavioral and Molecular Lab, Braga, Portugal
| |
Collapse
|
15
|
Lacoste B, Prat A, Freitas-Andrade M, Gu C. The Blood-Brain Barrier: Composition, Properties, and Roles in Brain Health. Cold Spring Harb Perspect Biol 2025; 17:a041422. [PMID: 38951020 PMCID: PMC12047665 DOI: 10.1101/cshperspect.a041422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Blood vessels are critical to deliver oxygen and nutrients to tissues and organs throughout the body. The blood vessels that vascularize the central nervous system (CNS) possess unique properties, termed the blood-brain barrier (BBB), which allow these vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. This precise control of CNS homeostasis allows for proper neuronal function and protects the neural tissue from toxins and pathogens, and alterations of this barrier are important components of the pathogenesis and progression of various neurological diseases. The physiological barrier is coordinated by a series of physical, transport, and metabolic properties possessed by the brain endothelial cells (ECs) that form the walls of the blood vessels. These properties are regulated by interactions between different vascular, perivascular, immune, and neural cells. Understanding how these cell populations interact to regulate barrier properties is essential for understanding how the brain functions in both health and disease contexts.
Collapse
Affiliation(s)
- Baptiste Lacoste
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, Ontario K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Moises Freitas-Andrade
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, Ontario K1H 8M5, Canada
| | - Chenghua Gu
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
16
|
Wei S, Song X, Mou Y, Yang T, Wang Y, Wang H, Ren C, Song X. New insights into pathogenisis and therapies of P2X7R in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:108. [PMID: 40325043 PMCID: PMC12053563 DOI: 10.1038/s41531-025-00980-7] [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: 11/07/2024] [Accepted: 04/27/2025] [Indexed: 05/07/2025] Open
Abstract
Parkinson's disease (PD), a prevalent neurodegenerative disorder, is linked to genetics and environment, but its mechanisms remain unclear. Emerging evidence connects purinergic signaling-particularly ATP-sensitive P2X7 receptor (P2X7R)-to PD. P2X7R expression is elevated in PD patients, and its antagonist BBG mitigates 6-OHDA-induced dopaminergic neuron death. This review discusses P2X7R's structure, neural functions, PD-related mechanisms, and therapeutic potential as a targert.
Collapse
Affiliation(s)
- Shizhuang Wei
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Xiaoyu Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Yakui Mou
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Ting Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Yao Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Hanrui Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Chao Ren
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Department of Neurology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai Yuhuangding Hospital, Yantai, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Yantai, China.
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China.
| |
Collapse
|
17
|
Frosch M, Prinz M. Niche-specific therapeutic targeting of myeloid cells in the central nervous system. Immunity 2025:S1074-7613(25)00135-9. [PMID: 40324377 DOI: 10.1016/j.immuni.2025.03.016] [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: 01/15/2025] [Revised: 03/18/2025] [Accepted: 03/18/2025] [Indexed: 05/07/2025]
Abstract
The central nervous system (CNS) can be subdivided into distinct anatomical and functional compartments, including the parenchyma, perivascular space, leptomeninges, and dura mater, etc. Each compartment hosts distinct immune cell populations, such as monocytes and diverse macrophages, which play critical roles in local tissue homeostasis and regional disease pathogenesis. Advances in single-cell technologies have revealed complex immune cell compositions and functions in these anatomical regions. This review summarizes the latest approaches for modulating myeloid cell subsets in a compartment-specific manner, including cellular strategies such as stem cell therapy, ex vivo gene treatment, bone marrow transplantation, as well as non-cellular strategies like antibodies, small molecules, and viral gene delivery to augment CNS immune responses and improve disease outcomes. We also discuss the challenges and requirements of translating targeting strategies from mice to humans.
Collapse
Affiliation(s)
- Maximilian Frosch
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany; CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
18
|
Karimova AF, Khalitova AR, Suezov R, Markov N, Mukhamedshina Y, Rizvanov AA, Huber M, Simon HU, Brichkina A. Immunometabolism of tumor-associated macrophages: A therapeutic perspective. Eur J Cancer 2025; 220:115332. [PMID: 40048925 DOI: 10.1016/j.ejca.2025.115332] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 04/26/2025]
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in the tumor microenvironment (TME), actively contributing to the formation of an immunosuppressive niche that fosters tumor progression. Consequently, there has been a growing interest in targeting TAMs as a promising avenue for cancer therapy. Recent advances in the field of immunometabolism have shed light on the influence of metabolic adaptations on macrophage physiology in the context of cancer. Here, we discuss the key metabolic pathways that shape the phenotypic diversity of macrophages. We place special emphasis on how metabolic reprogramming impacts the activation status of TAMs and their functions within the TME. Additionally, we explore alterations in TAM metabolism and their effects on phagocytosis, production of cytokines/chemokines and interaction with cytotoxic T and NK immune cells. Moreover, we examine the application of nanomedical approaches to target TAMs and assess the clinical significance of modulating the metabolism of TAMs as a strategy to develop new anti-cancer therapies. Taken together, in this comprehensive review article focusing on TAMs, we provide invaluable insights for the development of effective immunotherapeutic strategies and the enhancement of clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Adelya F Karimova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Adelya R Khalitova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Roman Suezov
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany
| | - Nikita Markov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Yana Mukhamedshina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, Kazan, Russia
| | - Magdalena Huber
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany
| | - Hans-Uwe Simon
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia; Institute of Pharmacology, University of Bern, Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Anna Brichkina
- Institute of Systems Immunology, Center for Tumor and Immune Biology, Philipps University of Marburg, Marburg, Germany.
| |
Collapse
|
19
|
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
|
20
|
Song KW, Lim M, Monje M. Complex neural-immune interactions shape glioma immunotherapy. Immunity 2025:S1074-7613(25)00180-3. [PMID: 40324379 DOI: 10.1016/j.immuni.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025]
Abstract
Rich neural-immune interactions in the central nervous system (CNS) shape its function and create a unique immunological microenvironment for immunotherapy in CNS malignancies. Far from the now-debunked concept of CNS "immune privilege," it is now understood that unique immunological niches and constant immune surveillance of the brain contribute in multifaceted ways to brain health and robustly influence immunotherapy approaches for CNS cancers. Challenges include immune-suppressive and neurotoxicity-promoting crosstalk between brain, immune, and tumor cells. Developing effective immunotherapies for cancers of the nervous system will require a deeper understanding of these neural-immune-malignant cell interactions. Here, we review progress and challenges in immunotherapy for gliomas of the brain and spinal cord in light of these unique neural-immune interactions and highlight future work needed to optimize promising immunotherapies for gliomas.
Collapse
Affiliation(s)
- Kun-Wei Song
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA, USA; Howard Hughes Medical Institute, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
21
|
Augusto-Oliveira M, Arrifano GDP, Leal-Nazaré CG, Chaves-Filho A, Santos-Sacramento L, Lopes-Araujo A, Tremblay MÈ, Crespo-Lopez ME. Morphological diversity of microglia: Implications for learning, environmental adaptation, ageing, sex differences and neuropathology. Neurosci Biobehav Rev 2025; 172:106091. [PMID: 40049541 DOI: 10.1016/j.neubiorev.2025.106091] [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: 11/06/2024] [Revised: 02/21/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Microglia are the brain resident macrophages that respond rapidly to any insult. These non-neuroectodermal cells are decorated with plenty of receptors allowing them to recognise and respond precisely to a multitude of stimuli. To do so, microglia undergo structural and functional changes aiming to actively keep the brain's homeostasis. However, some microglial responses, when sustained or exacerbated, can contribute to neuropathology and neurodegeneration. Many microglial molecular and cellular changes were identified that display a strong correlation with neuronal damage and neuroinflammation/disease status, as well as present key sex-related differences that modulate microglial outcomes. Nevertheless, the relationship between microglial structural and functional features is just beginning to be unravelled. Several reports show that microglia undergo soma and branch remodelling in response to environmental stimuli, ageing, neurodegenerative diseases, trauma, and systemic inflammation, suggesting a complex form and function link. Also, it is reasonable overall to suppose that microglia diminishing their process length and ramification also reduce their monitoring activity of synapses, which is critical for detecting any synaptic disturbance and performing synaptic remodelling. Elucidating the complex interactions between microglial morphological plasticity and its functional implications appears essential for the understanding of complex cognitive and behavioural processes in health and neuropathological conditions.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| | - Gabriela de Paula Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Caio Gustavo Leal-Nazaré
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Adriano Chaves-Filho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada
| | - Leticia Santos-Sacramento
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Amanda Lopes-Araujo
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER)
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada; Department of Molecular Medicine, Université Laval, Québec, Qubec, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, Québec, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada; Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, British Columbia, Canada; Women's Health Research Institute, British Columbia, Canada; College Member of the Royal Society of Canada, Canada.
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, PA, Brazil; Amazonian Institute on Mercury (Instituto Amazônico do Mercúrio - IAMER).
| |
Collapse
|
22
|
Malcangio M, Sideris-Lampretsas G. How microglia contribute to the induction and maintenance of neuropathic pain. Nat Rev Neurosci 2025; 26:263-275. [PMID: 40128335 DOI: 10.1038/s41583-025-00914-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2025] [Indexed: 03/26/2025]
Abstract
Neuropathic pain is a debilitating condition caused by damage to the nervous system that results in changes along the pain pathway that lead to persistence of the pain sensation. Unremitting pain conditions are associated with maladaptive plasticity, disruption of neuronal activity that favours excitation over inhibition, and engagement of immune cells. The substantial progress made over the last two decades in the neuroimmune interaction research area points to a mechanistic role of spinal cord microglia, which are resident immune cells of the CNS. Microglia respond to and modulate neuronal activity during establishment and persistence of neuropathic pain states, and microglia-neuron pathways provide targets that can be exploited to attenuate abnormal neuronal activity and provide pain relief.
Collapse
Affiliation(s)
- Marzia Malcangio
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London, UK.
| | | |
Collapse
|
23
|
Ratnapriya R, Grassman F, Chen R, Hewitt A, Du J, Saban DR, Klaver CCW, Ash J, Stambolian D, Tumminia SJ, Qian J, Husain D, Iyengar SK, den Hollander AI. Functional genomics in age-related macular degeneration: From genetic associations to understanding disease mechanisms. Exp Eye Res 2025; 254:110344. [PMID: 40089136 PMCID: PMC12048874 DOI: 10.1016/j.exer.2025.110344] [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: 11/11/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Genome-wide association studies have been remarkably successful in identifying genetic variants associated with age-related macular degeneration (AMD), demonstrating a strong genetic component largely driven by common variants. However, progress in translating these genetic findings into a deeper understanding of disease mechanisms and new therapies has been slow. Slow progress in this area can be attributed to limited knowledge of the functional impact of AMD-associated non-coding variants on gene function, the molecular mechanisms and cell types underlying disease. This review offers a comprehensive overview of functional genomics approaches to uncover the genetic, epigenetic, cellular and molecular mechanisms underlying AMD and outlines future directions for research.
Collapse
Affiliation(s)
- Rinki Ratnapriya
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, USA
| | - Felix Grassman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Institute for Clinical Research and Systems Medicine, Health and Medical University, Potsdam, Germany
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Alex Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, USA
| | - Daniel R Saban
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA; Department of Integrative Immunobiology, Duke University, Durham, NC, USA
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands; Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands; Institute of Molecular and Clinical Ophthalmology, University of Basel, Basel, Switzerland
| | - John Ash
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Jiang Qian
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Deeba Husain
- Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Sudha K Iyengar
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Anneke I den Hollander
- Genomics Research Center, AbbVie, North Chicago, IL, USA; Genomics Research Center, AbbVie, Cambridge, MA, USA.
| |
Collapse
|
24
|
Heneka MT, van der Flier WM, Jessen F, Hoozemanns J, Thal DR, Boche D, Brosseron F, Teunissen C, Zetterberg H, Jacobs AH, Edison P, Ramirez A, Cruchaga C, Lambert JC, Laza AR, Sanchez-Mut JV, Fischer A, Castro-Gomez S, Stein TD, Kleineidam L, Wagner M, Neher JJ, Cunningham C, Singhrao SK, Prinz M, Glass CK, Schlachetzki JCM, Butovsky O, Kleemann K, De Jaeger PL, Scheiblich H, Brown GC, Landreth G, Moutinho M, Grutzendler J, Gomez-Nicola D, McManus RM, Andreasson K, Ising C, Karabag D, Baker DJ, Liddelow SA, Verkhratsky A, Tansey M, Monsonego A, Aigner L, Dorothée G, Nave KA, Simons M, Constantin G, Rosenzweig N, Pascual A, Petzold GC, Kipnis J, Venegas C, Colonna M, Walter J, Tenner AJ, O'Banion MK, Steinert JR, Feinstein DL, Sastre M, Bhaskar K, Hong S, Schafer DP, Golde T, Ransohoff RM, Morgan D, Breitner J, Mancuso R, Riechers SP. Neuroinflammation in Alzheimer disease. Nat Rev Immunol 2025; 25:321-352. [PMID: 39653749 DOI: 10.1038/s41577-024-01104-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 02/20/2025]
Abstract
Increasing evidence points to a pivotal role of immune processes in the pathogenesis of Alzheimer disease, which is the most prevalent neurodegenerative and dementia-causing disease of our time. Multiple lines of information provided by experimental, epidemiological, neuropathological and genetic studies suggest a pathological role for innate and adaptive immune activation in this disease. Here, we review the cell types and pathological mechanisms involved in disease development as well as the influence of genetics and lifestyle factors. Given the decade-long preclinical stage of Alzheimer disease, these mechanisms and their interactions are driving forces behind the spread and progression of the disease. The identification of treatment opportunities will require a precise understanding of the cells and mechanisms involved as well as a clear definition of their temporal and topographical nature. We will also discuss new therapeutic strategies for targeting neuroinflammation, which are now entering the clinic and showing promise for patients.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg.
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Jeroen Hoozemanns
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Dietmar Rudolf Thal
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), Leuven, Belgium
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Charlotte Teunissen
- Department of Laboratory Medicine, VUMC Amsterdam, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Paul Edison
- Division of Neurology, Department of Brain Sciences, Imperial College London, London, UK
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Washington School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Agustin Ruiz Laza
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Jose Vicente Sanchez-Mut
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Alicante, Spain
| | - Andre Fischer
- Clinic for Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
- Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Disease (DZNE), Göttingen, Germany
| | - Sergio Castro-Gomez
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Michael Wagner
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jonas J Neher
- Biomedical Center Munich, Biochemistry, Medical Faculty, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - Sim K Singhrao
- Brain and Behaviour Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kilian Kleemann
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip L De Jaeger
- Center for Translational and 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
| | - Hannah Scheiblich
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Guy C Brown
- Deparment of Biochemistry, University of Cambridge, Cambridge, UK
| | - Gary Landreth
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Miguel Moutinho
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Ising
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Karabag
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Darren J Baker
- Department of Paediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York City, NY, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Malu Tansey
- College of Medicine, University of Florida, Gainsville, FL, USA
| | - Alon Monsonego
- Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Guillaume Dorothée
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Hôpital Saint-Antoine, Paris, France
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Neta Rosenzweig
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University of Bonn, Bonn, Germany
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto Biosanitario de Granada (ibs.Granada), Granada, Spain
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jochen Walter
- Center of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behaviour, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Joern R Steinert
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Douglas L Feinstein
- Department of NeuroAnesthesia, University of Illinois at Chicago, Chicago, IL, USA
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Kiran Bhaskar
- Department of Molecular Genetics & Microbiology and Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Todd Golde
- Department of Pharmacology and Chemical Biology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
| | | | - David Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John Breitner
- Department of Psychiatry, McGill University Faculty of Medicine, Montreal, Québec, Canada
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sean-Patrick Riechers
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
| |
Collapse
|
25
|
Du S, Kipnis J. Beyond the brain: microglia-like cells regulate peripheral neuronal soma size. Cell Mol Immunol 2025; 22:466-467. [PMID: 40200088 PMCID: PMC12041247 DOI: 10.1038/s41423-025-01276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Affiliation(s)
- Siling Du
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St Louis, St Louis, MO, USA.
- Department of Pathology and Immunology, School of Medicine, Washington University in St Louis, St Louis, MO, USA.
| |
Collapse
|
26
|
Zhao Y, Wang T, Liu J, Wang Z, Lu Y. Emerging brain organoids: 3D models to decipher, identify and revolutionize brain. Bioact Mater 2025; 47:378-402. [PMID: 40026825 PMCID: PMC11869974 DOI: 10.1016/j.bioactmat.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Brain organoids are an emerging in vitro 3D brain model that is integrated from pluripotent stem cells. This model mimics the human brain's developmental process and disease-related phenotypes to a certain extent while advancing the development of human brain-based biological intelligence. However, many limitations of brain organoid culture (e.g., lacking a functional vascular system, etc.) prevent in vitro-cultured organoids from truly replicating the human brain in terms of cell type and structure. To improve brain organoids' scalability, efficiency, and stability, this paper discusses important contributions of material biology and microprocessing technology in solving the related limitations of brain organoids and applying the latest imaging technology to make real-time imaging of brain organoids possible. In addition, the related applications of brain organoids, especially the development of organoid intelligence combined with artificial intelligence, are analyzed, which will help accelerate the rational design and guidance of brain organoids.
Collapse
Affiliation(s)
- Yuli Zhao
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Ting Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Jiajun Liu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
- Tianjin Industrial Microbiology Key Laboratory, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ze Wang
- College of Life Sciences, Shenyang Normal University, Shenyang, 110034, Liaoning, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
27
|
Strackeljan L, Baidoe-Ansah D, Mirzapourdelavar H, Jia S, Kaushik R, Cangalaya C, Dityatev A. Partial microglial depletion through inhibition of colony-stimulating factor 1 receptor improves synaptic plasticity and cognitive performance in aged mice. Exp Neurol 2025; 387:115186. [PMID: 39956381 DOI: 10.1016/j.expneurol.2025.115186] [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: 11/13/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Microglia depletion, followed by repopulation, improves cognitive functions in the aged mouse brain. However, even temporal ablation of microglia puts the brain at a high risk of infection. Hence, in the present work, we studied if the partial reduction of microglia with PLX3397 (pexidartinib), an inhibitor of the colony-stimulating factor 1 receptor (CSF1R), could bring similar benefits as reported for microglia ablation. Aged (two-years-old) mice were treated with PLX3397 for a total of 6 weeks, which reduced microglia numbers in the hippocampus and retrosplenial cortex (RSC) to the levels seen in young mice and resulted in layer-specific ablation in the expression of microglial complement protein C1q mediating synaptic remodeling. This treatment boosted long-term potentiation in the CA1 region and improved performance in the hippocampus-dependent novel object location recognition task. Although PLX3397 treatment did not alter the number or total intensity of Wisteria floribunda agglutinin-positive perineuronal nets (PNNs) in the CA1 region of the hippocampus, it changed the fine structure of PNNs. It also elevated the expression of perisynaptic proteoglycan brevican, presynaptic vGluT1 at excitatory synapses, and vGAT in inhibitory ones in the CA1 stratum radiatum. Thus, targeting the CSF1R may provide a safe and efficient strategy to boost synaptic and cognitive functions in the aged brain.
Collapse
Affiliation(s)
- Luisa Strackeljan
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - David Baidoe-Ansah
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Shaobo Jia
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Rahul Kaushik
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Carla Cangalaya
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany; Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany.
| |
Collapse
|
28
|
Shakiba M, Tuveson DA. Macrophages and fibroblasts as regulators of the immune response in pancreatic cancer. Nat Immunol 2025; 26:678-691. [PMID: 40263612 DOI: 10.1038/s41590-025-02134-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the few cancers that has yet to benefit from immunotherapies. This is primarily a result of its characteristic 'cold' tumor microenvironment composed of cancer-associated fibroblasts (CAFs), a dense network of extracellular matrix and several immune cell types, the most abundant of which are the tumor-associated macrophages (TAMs). Advances in single-cell and spatial technologies have elucidated the vast functional heterogeneity of CAFs and TAMs, their symbiotic relationship and their cooperative role in the tumor microenvironment. In this Review, we provide an overview of the heterogeneity of CAFs and TAMs, how they establish an immunosuppressive microenvironment and their collaboration in the remodeling of the extracellular matrix. Finally, we examine why the impact of immunotherapy in PDAC has been limited and how a detailed molecular and spatial understanding of the combined role of CAFs and TAMs is paramount to the design of effective therapies.
Collapse
Affiliation(s)
- Mojdeh Shakiba
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
- Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
29
|
Tseng CEJ, Guma E, McDougle CJ, Hooker JM, Zürcher NR. Regional skull translocator protein elevation in autistic adults detected by PET-MRI. Brain Behav Immun 2025; 126:70-79. [PMID: 39904469 DOI: 10.1016/j.bbi.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/06/2025] Open
Abstract
Immune processes have been implicated in the pathophysiology of autism spectrum disorder (ASD). Brain borders, such as the skull, have recently been highlighted as sites where neuro-immune interactions occur with key consequences for brain immunity. Translocator protein (TSPO), a mitochondrial protein involved in immune functions, was measured in the skull using [11C]PBR28 positron emission tomography-magnetic resonance imaging (PET-MRI) in 38 autistic adults (26 males, 12 females) and 29 age-and sex-matched healthy controls (19 males, 10 females). [11C]PBR28 uptake relative to a pseudo-reference region assessed using standardized uptake value ratio (SUVR) revealed elevated TSPO in autistic adults in frontal and temporal skull. We did not observe an association between [11C]PBR28 uptake in total or regional skull areas and autism symptom severity. C-reactive protein levels were positively associated with [11C]PBR28 uptake in the total skull across participants. Lastly, [11C]PBR28 uptake in the total skull was stable across a 4-month period. This work indicates regional TSPO elevations in the skull in autistic adults, which may suggest immune involvement.
Collapse
Affiliation(s)
- Chieh-En Jane Tseng
- Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging Charlestown MA USA; Harvard Medical School Boston MA USA
| | - Elisa Guma
- Harvard Medical School Boston MA USA; Lurie Center for Autism, Massachusetts General Hospital Lexington MA USA
| | - Christopher J McDougle
- Harvard Medical School Boston MA USA; Lurie Center for Autism, Massachusetts General Hospital Lexington MA USA
| | - Jacob M Hooker
- Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging Charlestown MA USA; Harvard Medical School Boston MA USA; Lurie Center for Autism, Massachusetts General Hospital Lexington MA USA
| | - Nicole R Zürcher
- Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging Charlestown MA USA; Harvard Medical School Boston MA USA; Lurie Center for Autism, Massachusetts General Hospital Lexington MA USA.
| |
Collapse
|
30
|
Byatt TC, Razaghi E, Tüzüner S, Simões FC. Immune-mediated cardiac development and regeneration. Semin Cell Dev Biol 2025; 171:103613. [PMID: 40315634 DOI: 10.1016/j.semcdb.2025.103613] [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: 12/29/2024] [Revised: 03/18/2025] [Accepted: 04/16/2025] [Indexed: 05/04/2025]
Abstract
The complex interplay between the immune and cardiovascular systems during development, homeostasis and regeneration represents a rapidly evolving field in cardiac biology. Single cell technologies, spatial mapping and computational analysis have revolutionised our understanding of the diversity and functional specialisation of immune cells within the heart. From the earliest stages of cardiogenesis, where primitive macrophages guide heart tube formation, to the complex choreography of inflammation and its resolution during regeneration, immune cells emerge as central orchestrators of cardiac fate. Translating these fundamental insights into clinical applications represents a major challenge and opportunity for the field. In this Review, we decode the immunological blueprint of heart development and regeneration to transform cardiovascular disease treatment and unlock the regenerative capacity of the human heart.
Collapse
Affiliation(s)
- Timothy C Byatt
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Ehsan Razaghi
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Selin Tüzüner
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Filipa C Simões
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
31
|
Gunasegaran B, Krishnamurthy S, Chow SS, Villanueva MD, Guller A, Ahn SB, Heng B. Comparative Analysis of HMC3 and C20 Microglial Cell Lines Reveals Differential Myeloid Characteristics and Responses to Immune Stimuli. Immunology 2025; 175:84-102. [PMID: 39961658 PMCID: PMC11982601 DOI: 10.1111/imm.13900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 04/11/2025] Open
Abstract
Microglia are the primary resident immune cells of the central nervous system (CNS) that respond to injury and infections. Being critical to CNS homeostasis, microglia also have been shown to contribute to neurodegenerative diseases and brain cancer. Hence, microglia are regarded as a potential therapeutic target in CNS diseases, resulting in an increased demand for reliable in vitro models. Two human microglia cell lines (HMC3 and C20) are being used in multiple in vitro studies, however, the knowledge of their biological and immunological characteristics remains limited. Our aim was to identify and compare the biological changes in these immortalised immune cells under normal physiological and immunologically challenged conditions. Using high-resolution quantitative mass spectrometry, we have examined in-depth proteomic profiles of non-stimulated and LPS or IFN-γ challenged HMC3 and C20 cells. Our findings reveal that HMC3 cells responded to both treatments through upregulation of immune, metabolic, and antiviral pathways, while C20 cells showed a response associated with mitochondrial and immune activities. Additionally, the secretome analysis demonstrated that both cell lines release IL-6 in response to LPS, while IFN-γ treatment resulted in altered kynurenine pathway activity, highlighting distinct immune and metabolic adaptations.
Collapse
Affiliation(s)
- Bavani Gunasegaran
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Shivani Krishnamurthy
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Sharron S. Chow
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Millijoy D. Villanueva
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
- Computational Neurosurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Anna Guller
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
- Computational Neurosurgery (CNS) Lab, Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Seong Beom Ahn
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Benjamin Heng
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| |
Collapse
|
32
|
Balla J, Rathore APS, St. John AL. Maternal IgE Influence on Fetal and Infant Health. Immunol Rev 2025; 331:e70029. [PMID: 40281548 PMCID: PMC12032057 DOI: 10.1111/imr.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Immunoglobulin E (IgE) is the most recently discovered and evolved mammalian antibody type, best known for interacting with mast cells (MCs) as immune effectors. IgE-mediated antigen sensing by MC provides protection from parasites, venomous animals, bacteria, and other insults to barrier tissues exposed to the environment. IgE and MCs act as inflammation amplifiers and immune response adjuvants. Thus, IgE production and memory formation are greatly constrained and require specific licensing. Failure of regulation gives rise to allergic disease, one of the top causes of chronic illness. Increasing evidence suggests allergy development often starts early in life, including prenatally, with maternal influence being central in shaping the offspring's immune system. Although IgE often exists before birth, an endogenous source of IgE-producing B cells has not been identified. This review discusses the mechanisms of maternal IgE transfer into the offspring, its interactions with offspring MCs and antigen-presenting cells, and the consequences for allergic inflammation and allergen sensitization development. We discuss the multifaceted effects of pre-existing IgG, IgE, and their glycosylation on maternal IgE transfer and functionality in the progeny. Understanding the IgE-mediated mechanisms predisposing for early life allergy development may allow their targeting with existing therapeutics and guide the development of new ones.
Collapse
Affiliation(s)
- Jozef Balla
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
| | - Abhay P. S. Rathore
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Ashley L. St. John
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- SingHealth Duke‐NUS Global Health InstituteSingaporeSingapore
| |
Collapse
|
33
|
Bastos J, O'Brien C, Vara-Pérez M, Mampay M, van Olst L, Barry-Carroll L, Kancheva D, Leduc S, Lievens AL, Ali L, Vlasov V, Meysman L, Shakeri H, Roelandt R, Van Hove H, De Vlaminck K, Scheyltjens I, Yaqoob F, Lombroso SI, Breugelmans M, Faron G, Gomez-Nicola D, Gate D, Bennett FC, Movahedi K. Monocytes can efficiently replace all brain macrophages and fetal liver monocytes can generate bona fide SALL1 + microglia. Immunity 2025:S1074-7613(25)00169-4. [PMID: 40311613 DOI: 10.1016/j.immuni.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025]
Abstract
Microglia and border-associated macrophages (BAMs) are critical for brain health, and their dysfunction is associated to disease. Replacing brain macrophages holds substantial therapeutic promise but remains challenging. Here, we demonstrate that monocytes can efficiently replace all brain macrophages. Monocytes readily replaced embryonal BAMs upon their depletion and engrafted as monocyte-derived microglia (Mo-Microglia) upon more sustained niche availability. Mo-Microglia expanded comparably to their embryonic counterparts and showed similar longevity. However, monocytes were unable to replicate the distinct identity of embryonically derived BAMs and microglia. Using xenotransplantation, we found that human monocytes exhibited similar behavior, enabling identification of putative Mo-Microglia in Alzheimer's disease individuals. In mice and humans, monocyte ontogeny shaped their identity as brain macrophages. Importantly, mouse fetal liver monocytes exhibited a distinct epigenetic landscape and could develop a bona fide microglial identity. Our results illuminate brain macrophage development and highlight monocytes as an abundant progenitor source for brain macrophage replacement therapies.
Collapse
Affiliation(s)
- Jonathan Bastos
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carleigh O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mónica Vara-Pérez
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Myrthe Mampay
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lynn van Olst
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Liam Barry-Carroll
- School of Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK; Nutrineuro, UMR 1286 INRAE, Bordeaux University, Bordeaux INP, Bordeaux, France
| | - Daliya Kancheva
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sophia Leduc
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ayla Line Lievens
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Leen Ali
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Vladislav Vlasov
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laura Meysman
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hadis Shakeri
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ria Roelandt
- VIB Single Cell Core, VIB, Ghent/Leuven, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Hannah Van Hove
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karen De Vlaminck
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Scheyltjens
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sonia I Lombroso
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria Breugelmans
- Department of Obstetrics and Prenatal Medicine, UZ Brussel, VUB, Brussels, Belgium
| | - Gilles Faron
- Department of Obstetrics and Prenatal Medicine, UZ Brussel, VUB, Brussels, Belgium
| | - Diego Gomez-Nicola
- School of Biological Sciences, Southampton General Hospital, University of Southampton, Southampton, UK
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kiavash Movahedi
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
34
|
Lopez-Atalaya JP, Bhojwani-Cabrera AM. Type I interferon signalling and interferon-responsive microglia in health and disease. FEBS J 2025. [PMID: 40299722 DOI: 10.1111/febs.70126] [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: 09/19/2024] [Revised: 03/31/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025]
Abstract
Recent evidence suggests that type I interferon (IFN-I) signalling extends beyond its canonical roles in antiviral defence and immunomodulation. Over the past decade, dysregulated IFN-I signalling has been linked to genetic disorders and neurodegenerative diseases, where it may contribute to neurological impairments. Microglia have emerged as key mediators of IFN-I responses in the central nervous system. A distinct transcriptional state responsive to interferons has recently been identified in microglia. The activation of the IFN-I pathway in these cells is now recognised as pivotal in both development and neurodegeneration. This review is divided into two main sections: the first examines the broader role of IFN-I signalling in the central nervous system, particularly its contribution to neurological dysfunction; the second focuses on the specific state of interferon-responsive microglia, exploring its mechanisms and relevance in neurodegenerative conditions. Finally, we discuss how these areas intersect and their implications for both healthy and diseased states.
Collapse
Affiliation(s)
- Jose P Lopez-Atalaya
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Alicante, Spain
| | - Aysha M Bhojwani-Cabrera
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, Alicante, Spain
| |
Collapse
|
35
|
Zhan X, Wang S, Bèchet N, Gouras G, Wen G. Perivascular macrophages in the central nervous system: insights into their roles in health and disease. Cell Death Dis 2025; 16:350. [PMID: 40295513 PMCID: PMC12037809 DOI: 10.1038/s41419-025-07592-2] [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: 11/05/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
Perivascular macrophages (PVMs) are a specialized subset of macrophages situated near blood vessels in the brain. Their strategic positioning around these vessels enables them to perform key functions in immune surveillance and response to inflammation and injury. These cells are crucial for modulating the immune response within the brain, contributing to normal central nervous system (CNS) processes. In pathological conditions, the role of PVMs becomes more complex. Depending on the specific disease or injury, they may contribute to inflammation, blood-brain barrier (BBB) dysfunction, and the clearance of abnormal materials. PVMs are implicated in degenerative diseases, cerebrovascular impairment, and microhemorrhages associated with amyloid-β immunotherapy. Despite their important roles in the CNS, research on PVMs remains limited, and the mechanisms underlying their involvement in both physiological and pathological processes within the brain are not yet fully elucidated. Therefore, this review will focus on the current advancements in PVM research, including their origin, classification, roles in neuroinflammation and neuroprotection, and their potential roles as therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoni Zhan
- School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China
- Neural Plasticity and Repair Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Shuying Wang
- School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Nicholas Bèchet
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Department of Clinical Science, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Gunnar Gouras
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Gehua Wen
- School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China.
- Experimental Dementia Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
36
|
Van Hove H, Glück C, Mildenberger W, Petrova E, Maheshwari U, Häne P, Kreiner V, Bijnen M, Mussak C, Utz SG, Droux J, Ingelfinger F, Ashworth C, Stifter SA, Roussel E, Lelios I, Vermeer M, Huang SF, Zhou Q, Chen Z, Calvet C, Bourgeois S, Schaffenrath J, Razansky D, Juang JX, Asano K, Pelczar P, Mundt S, Weber B, Wegener S, Tugues S, Stockmann C, Becher B, Keller A, El Amki M, Greter M. Interleukin-34-dependent perivascular macrophages promote vascular function in the brain. Immunity 2025:S1074-7613(25)00166-9. [PMID: 40315842 DOI: 10.1016/j.immuni.2025.04.003] [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: 01/21/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 05/04/2025]
Abstract
The development of most macrophages depends on the colony-stimulating factor 1 (CSF-1) receptor, which has two ligands: CSF-1 and interleukin-34 (IL-34). While IL-34 is required for the homeostasis of microglia, the parenchymal macrophages in the central nervous system (CNS), it is unclear whether brain border-associated macrophages (BAMs) also depend on this cytokine. Here, we demonstrated that the embryonic development of murine BAMs in the choroid plexus, leptomeninges, and perivascular spaces required CSF-1, while IL-34 was critical for their maintenance in adulthood. In the brain, Il34 was expressed by mural cells and perivascular fibroblasts, and its transgenic deletion in these cells interrupted BAM maintenance. Il34 deficiency coincided with transcriptional changes in vascular cells, leading to increased flow velocity and vasomotion in pial and penetrating arterioles. Similarly, Mrc1CreCsf1rfl/fl mice lacking CD206+ perivascular BAMs exhibited increased hemodynamics in arterial networks. These findings reveal a crosstalk between vascular cells and CNS macrophages regulating cerebrovascular function.
Collapse
Affiliation(s)
- Hannah Van Hove
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Chaim Glück
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Wiebke Mildenberger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Ekaterina Petrova
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Upasana Maheshwari
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Philipp Häne
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Victor Kreiner
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Mitchell Bijnen
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Caroline Mussak
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sebastian G Utz
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jeanne Droux
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Florian Ingelfinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Christian Ashworth
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sebastian A Stifter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Elsa Roussel
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Iva Lelios
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Marijne Vermeer
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sheng-Fu Huang
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Quanyu Zhou
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Zhenyue Chen
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Charlotte Calvet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Johanna Schaffenrath
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland; Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Jean X Juang
- Department of Biochemistry and Structural Biology, University of Texas Science Center, San Antonio, TX 78229, USA
| | - Kenichi Asano
- Laboratory of Immune regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Bruno Weber
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Sonia Tugues
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Mohamad El Amki
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
37
|
Hattori Y. Microglial colonization routes and their impacts on cellular diversity. Neurosci Res 2025:S0168-0102(25)00078-1. [PMID: 40288616 DOI: 10.1016/j.neures.2025.04.004] [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: 03/24/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Microglia are the resident immune cells of the central nervous system. Unlike other glial cells-such as astrocytes and oligodendrocytes-which originate from neural stem cells alongside neurons, microglia derive from erythromyeloid progenitors that emerge in the yolk sac during early embryonic development. Once they reach the brain, microglia expand their population through proliferation during development. A growing body of research has revealed that microglia play diverse roles throughout life, both in physiological and pathological contexts. With recent advancements in single-cell transcriptomics, it has become increasingly evident that microglia exhibit substantial heterogeneity in their gene expression patterns. While various functions and subtypes of microglia are being uncovered, the mechanisms underlying their diversity remain largely unknown. Two key hypotheses may explain how microglial diversity arises. One possibility is that their diversity is influenced by the different colonization routes they take before settling in the brain. Alternatively, microglia may acquire distinct properties in response to their local environment. This review explores both possibilities, with a particular focus on the first hypothesis, drawing on recent findings that highlight the multiple routes microglia utilize to colonize the brain. It discusses how these processes contribute to the establishment of microglial diversity during brain development.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan.
| |
Collapse
|
38
|
Bhattacharya S, Deka J, Avallone T, Todd L. The neuroimmune interface in retinal regeneration. Prog Retin Eye Res 2025; 106:101361. [PMID: 40287050 DOI: 10.1016/j.preteyeres.2025.101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/12/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Retinal neurodegeneration leads to irreversible blindness due to the mammalian nervous system's inability to regenerate lost neurons. Efforts to regenerate retina involve two main strategies: stimulating endogenous cells to reprogram into neurons or transplanting stem-cell derived neurons into the degenerated retina. However, both approaches must overcome a major barrier in getting new neurons to grow back down the optic nerve and connect to appropriate visual targets in environments that differ significantly from developmental conditions. While immune privilege has historically been associated with the central nervous system, an emerging literature highlights the active role of immune cells in shaping neurodegeneration and regeneration. This review explores the neuroimmune interface in retinal repair, dissecting how immune interactions influence glial reprogramming, transplantation outcomes, and axonal regeneration. By integrating insights from regenerative species with mammalian models, we highlight novel immunomodulatory strategies to optimize retinal regeneration.
Collapse
Affiliation(s)
- Sucheta Bhattacharya
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Jugasmita Deka
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Thomas Avallone
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Levi Todd
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
39
|
Vara-Pérez M, Movahedi K. Border-associated macrophages as gatekeepers of brain homeostasis and immunity. Immunity 2025:S1074-7613(25)00168-2. [PMID: 40324381 DOI: 10.1016/j.immuni.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 05/07/2025]
Abstract
The brain's border tissues serve as essential hubs for neuroimmune regulation and the trafficking of biomaterials to and from the brain. These complex tissues-including the meninges, perivascular spaces, choroid plexus, and circumventricular organs-balance the brain's need for immune privilege with immune surveillance and blood-brain communication. Macrophages are integral components of these tissues, taking up key strategic positions within the brain's circulatory system. These border-associated macrophages, or "BAMs," are therefore emerging as pivotal for brain homeostasis and disease. BAMs perform trophic functions that help to support border homeostasis but also act as immune sentinels essential for border defense. In this review, we integrate recent findings on BAM origins, cell states, and functions, aiming to provide global insights and perspectives on the complex relationship between these macrophages and their border niche.
Collapse
Affiliation(s)
- Mónica Vara-Pérez
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kiavash Movahedi
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
40
|
Kim JS, Trzebanski S, Shin SH, Schori L, Frumer Friedman GR, Ilani NC, Kadam A, Vicario R, Aust O, Bugaeva P, Piatek S, Ismajli LK, Hoffmann CJ, Scheller M, Boura-Halfon S, Kaushansky N, Golani O, Solomon A, Liu Z, Amann L, Böhm-Sturm P, Koch SP, Wenger N, Ginhoux F, Prinz M, Avraham R, Harms C, Geissmann F, Müller-Tidow C, Uderhardt S, Milenkovic I, Shlush L, Jung S. Clonal hematopoiesis-associated motoric deficits caused by monocyte-derived microglia accumulating in aging mice. Cell Rep 2025; 44:115609. [PMID: 40279248 DOI: 10.1016/j.celrep.2025.115609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/27/2025] Open
Abstract
Microglia are parenchymal brain macrophages that are established during embryogenesis and form a self-containing cellular compartment that resists seeding with cells derived from adult definitive hematopoiesis. We report that monocyte-derived macrophages (MoMΦs) accumulate in the brain of aging mice with distinct topologies, including the nigrostriatum and medulla but not the frontal cortex. Parenchymal MoMΦs adopt bona fide microglia morphology and expression profiles. Due to their hematopoietic stem cell (HSC) derivation, monocyte-derived microglia (MoMg) are unlike yolk-sac-derived cells, targets of clonal hematopoiesis (CH). Indeed, using a chimeric transfer model, we show that the hematopoietic expression of DNMT3AR882H, a prominent human CH variant, renders MoMg pathogenic and promotes motor deficits resembling atypical Parkinsonian disorders. Collectively, we establish that MoMg progressively seed the brain of healthy aging mice, accumulate in selected areas, and, when carrying a somatic mutation associated with CH, can cause brain pathology.
Collapse
Affiliation(s)
- Jung-Seok Kim
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sébastien Trzebanski
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sun-Hye Shin
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lior Schori
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Gal Ronit Frumer Friedman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Noa Chapal Ilani
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Aditee Kadam
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rocio Vicario
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Oliver Aust
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Exploratory Research Unit, Optical Imaging Centre Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Polina Bugaeva
- Department of Neurology with Experimental Neurology, Center for Stroke Research Berlin and Einstein Center for Neuroscience Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sylwia Piatek
- Department of Neurology with Experimental Neurology, Center for Stroke Research Berlin and Einstein Center for Neuroscience Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Laura Kate Ismajli
- Department of Neurology with Experimental Neurology, Center for Stroke Research Berlin and Einstein Center for Neuroscience Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Christian Johannes Hoffmann
- Department of Neurology with Experimental Neurology, Center for Stroke Research Berlin and Einstein Center for Neuroscience Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Marina Scheller
- Department of Medicine, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Sigalit Boura-Halfon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nathali Kaushansky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ofra Golani
- MICC Cell Observatory, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Aryeh Solomon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lukas Amann
- Institute of Neuropathology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Böhm-Sturm
- Department of Neurology with Experimental Neurology, Center for Stroke Research Berlin and Einstein Center for Neuroscience Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany; Charité Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence and Charité Core Facility, 7T Experimental MRIs, Berlin, Germany
| | - Stefan Paul Koch
- Department of Neurology with Experimental Neurology, Center for Stroke Research Berlin and Einstein Center for Neuroscience Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany; Charité Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence and Charité Core Facility, 7T Experimental MRIs, Berlin, Germany
| | - Nikolaus Wenger
- Department of Neurology with Experimental Neurology, Center for Stroke Research Berlin and Einstein Center for Neuroscience Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Singapore Immunology Network, Agency for Science, Technology & Research, Singapore, Singapore; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Marco Prinz
- Institute of Neuropathology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany; Signalling Research Centre's BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Roi Avraham
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Christoph Harms
- Department of Neurology with Experimental Neurology, Center for Stroke Research Berlin and Einstein Center for Neuroscience Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carsten Müller-Tidow
- Department of Medicine, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Uderhardt
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany; Exploratory Research Unit, Optical Imaging Centre Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ivan Milenkovic
- Department of Neurology, Medical University Vienna, Wien, Austria
| | - Liran Shlush
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Steffen Jung
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
41
|
Thorp EB, Ananthakrishnan A, Lantz CW. Decoding immune cell interactions during cardiac allograft vasculopathy: insights derived from bioinformatic strategies. Front Cardiovasc Med 2025; 12:1568528. [PMID: 40342971 PMCID: PMC12058854 DOI: 10.3389/fcvm.2025.1568528] [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: 01/30/2025] [Accepted: 04/04/2025] [Indexed: 05/11/2025] Open
Abstract
Chronic allograft vasculopathy (CAV) is a major cause of late graft failure in heart transplant recipients, characterized by progressive intimal thickening and diffuse narrowing of the coronary arteries. Unlike atherosclerosis, CAV exhibits a distinct cellular composition and lesion distribution, yet its pathogenesis remains incompletely understood. A major challenge in CAV research has been the limited application of advanced "-omics" technologies, which have revolutionized the study of other vascular diseases. Recent advancements in single-cell and spatial transcriptomics, proteomics, and metabolomics have begun to uncover the complex immune-endothelial-stromal interactions driving CAV progression. Notably, single-cell RNA sequencing has identified previously unrecognized immune cell populations and signaling pathways implicated in endothelial injury and vascular remodeling after heart transplantation. Despite these breakthroughs, studies applying these technologies to CAV remain sparse, limiting the translation of these insights into clinical practice. This review aims to bridge this gap by summarizing recent findings from single-cell and multi-omic approaches, highlighting key discoveries, and discussing their implications for understanding CAV pathogenesis.
Collapse
Affiliation(s)
- Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Aparnaa Ananthakrishnan
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Connor W. Lantz
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
42
|
Medeiros-Furquim T, Miedema A, Schilder E, Brouwer N, Holtman IR, Kooistra SM, Eggen BJL. Microglia endotoxin tolerance is retained after enforced repopulation. Brain Behav Immun 2025; 128:512-528. [PMID: 40274001 DOI: 10.1016/j.bbi.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Microglia are crucial for CNS homeostasis and are involved in a wide range of neurodegenerative and neuroinflammatory diseases. Systemic inflammation and infections can contribute to neurodegeneration later in life by affecting microglia. Like other innate immune cells, microglia can develop innate immune memory (IIM) in response to an inflammatory challenge, altering their response to subsequent stimuli. IIM can ameliorate or worsen CNS pathology, but it is unclear if IIM can be reversed to restore microglia functions. Here, we investigated whether microglia depletion-repopulation by inhibition of the colony-stimulating factor 1 receptor with BLZ945 reversed LPS-induced microglia endotoxin tolerance in mice. Repopulated microglia displayed a reduced expression of homeostatic genes and genes related to mitochondrial respiration and TCA cycle metabolism and an increased expression of immune effector and activation genes. Nonetheless, the blunted inflammatory gene response after LPS-preconditioning was retained after a depletion-repopulation cycle. Our study highlights the persistence of endotoxin tolerance in microglia after a depletion-repopulation cycle, which might impact the potential effectiveness of strategies targeted at microglia depletion for clinical applications.
Collapse
Affiliation(s)
- Tiago Medeiros-Furquim
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anneke Miedema
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Edwin Schilder
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nieske Brouwer
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Inge R Holtman
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Susanne M Kooistra
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Bart J L Eggen
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
43
|
Qiu W, Zheng Z, Wang J, Cai Y, Zou J, Huang Z, Yang P, Ye W, Jin M, Zhang D, Little PJ, Zhou Q, Liu Z. Targeting mitochondrial DNA-STING-NF-κB Axis-mediated microglia activation by cryptotanshinone alleviates ischemic retinopathy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156779. [PMID: 40279967 DOI: 10.1016/j.phymed.2025.156779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/27/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Ischemic retinopathy, a leading cause of vision impairment, involves oxidative stress and dysregulated inflammation, with microglia playing a key role. Cryptotanshinone (CTS), a bioactive compound from Salvia miltiorrhiza, exhibits anti-inflammatory and antioxidant properties and thus has the potential for development as a therapeutic agent. However, the actual mechanism of action of CTS in ischemic retinopathy is not known. Overactivation of the STING pathway in microglia is critical in ischemic retinopathy pathogenesis and a potential target of CTS. PURPOSE This study aimed to explore whether CTS alleviates ischemic retinopathy by modulating microglial STING signaling. METHODS Oxygen-induced retinopathy (OIR) mice and hypoxia-induced microglial cells were used. CTS efficacy in ischemic retinopathy was evaluated at multiple stages using fluorescein fundus angiography, electroretinogram, H&E staining, and Western blotting of relevant proteins. Network pharmacology and RNA sequencing identified STING as a key target. Furthermore, surface plasmon resonance (SPR), molecular docking, and site-directed mutagenesis were systematically employed to elucidate the precise binding interface between CTS and the STING protein. STING activation and knockout models were employed to further investigate the mechanisms of action of CTS. RESULTS CTS treatment reduced microglial activation and pathological retinal angiogenesis, and protected both retinal function and structure in OIR mice. Network pharmacology, RNA sequencing, and experimental validation demonstrated a significant link between the protective effect of CTS and the inhibition of STING signaling. Mechanistically, CTS suppressed cytosolic mtDNA release, blocked STING translocation from the ER to the Golgi, and enhanced lysosomal STING degradation. These CTS-mediated effects were abolished by STING activation and absent in Sting-deficient OIR mice. Notably, CTS combined with anti-VEGF therapy showed synergistic efficacy in suppressing pathological retinal neovascularization. CONCLUSION CTS, a natural inhibitor of STING, alleviated ischemic retinopathy by inhibiting the mtDNA-STING-NF-κB signaling pathway via multifaceted mechanisms in microglia.
Collapse
Affiliation(s)
- Wanlu Qiu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China; Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou 510006, China
| | - Zhihua Zheng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China; The Affiliated Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Traditional Chinese Medicine, School of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jiaojiao Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China; Key Laboratory of Big Data Mining and Precision Drug Design of Guangdong Medical University, Key Laboratory of Computer-Aided Drug Design of Dongguan City, Key Laboratory for Research and Development of Natural Drugs of Guangdong Province, School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Youran Cai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China; Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou 510006, China
| | - Jiami Zou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Ziqing Huang
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou 510006, China
| | - Pinglian Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Weile Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Mei Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia; Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Qing Zhou
- Department of Ophthalmology, the First Affiliated Hospital, Jinan University, Guangzhou 510006, China.
| | - Zhiping Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
44
|
Wu Z, Wang Y, Chen WW, Sun H, Chen X, Li X, Wang Z, Liang W, Wang SY, Luan X, Li Y, Huang S, Liang Y, Zhang J, Chen ZF, Wang G, Gao Y, Liu Y, Wang J, Liu Z, Shi P, Liu C, Lv L, Hou A, Wu C, Yao C, Hong Z, Dai J, Lu Z, Pan F, Chen X, Kettenmann H, Amit I, Speakman JR, Chen Y, Ginhoux F, Cui R, Huang T, Li H. Peripheral nervous system microglia-like cells regulate neuronal soma size throughout evolution. Cell 2025; 188:2159-2174.e15. [PMID: 40199320 DOI: 10.1016/j.cell.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/18/2024] [Accepted: 02/12/2025] [Indexed: 04/10/2025]
Abstract
Microglia, essential in the central nervous system (CNS), were historically considered absent from the peripheral nervous system (PNS). Here, we show a PNS-resident macrophage population that shares transcriptomic and epigenetic profiles as well as an ontogenetic trajectory with CNS microglia. This population (termed PNS microglia-like cells) enwraps the neuronal soma inside the satellite glial cell envelope, preferentially associates with larger neurons during PNS development, and is required for neuronal functions by regulating soma enlargement and axon growth. A phylogenetic survey of 24 vertebrates revealed an early origin of PNS microglia-like cells, whose presence is correlated with neuronal soma size (and body size) rather than evolutionary distance. Consistent with their requirement for soma enlargement, PNS microglia-like cells are maintained in vertebrates with large peripheral neuronal soma but absent when neurons evolve to have smaller soma. Our study thus reveals a PNS counterpart of CNS microglia that regulates neuronal soma size during both evolution and ontogeny.
Collapse
Affiliation(s)
- Zhisheng Wu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Department of Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Chemistry and Chemical Engineering, Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Yiheng Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Wei-Wei Chen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hua Sun
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; School of Life Sciences, Henan University, Henan, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women's and Children's Hospital, Jinan University, Shenzhen, China
| | - Xiaobo Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; College of Biological Sciences, China Agricultural University, Beijing, China
| | - Zeshuai Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Weizheng Liang
- Hebei Provincial Key Laboratory of Systems Biology and Gene Regulation, Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Shuang-Yin Wang
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Xuemei Luan
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yijiang Li
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Shangjin Huang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuteng Liang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jiaqi Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhou-Feng Chen
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, and Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| | - Guanlin Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China; Shanghai Qi Zhi Institute, Shanghai, China
| | - Yun Gao
- State Key Laboratory of Genetic Resources and Evolution, and Southwest Research Centre of Porcine Molecular Breeding and Translational Medicine in China, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yanan Liu
- State Key Laboratory of Genetic Resources and Evolution, and Southwest Research Centre of Porcine Molecular Breeding and Translational Medicine in China, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jun Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Peng Shi
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Cirong Liu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Longbao Lv
- National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Anli Hou
- Shenzhen Guangming District People's Hospital, Shenzhen, China
| | - Chenglin Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen Yao
- The First Affiliated Hospital of Shenzhen University/Shenzhen Second People's Hospital, Shenzhen, China
| | - Zexuan Hong
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ji Dai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhonghua Lu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fan Pan
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | | | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Yun Chen
- Department of Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China; School of Chemistry and Chemical Engineering, Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China.
| | - Florent Ginhoux
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Rongfeng Cui
- School of Ecology & State Key Laboratory of Biocontrol, Sun Yat-sen University, Shenzhen, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, China
| | - Tianwen Huang
- CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hanjie Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Shenzhen University of Advanced Technology, Shenzhen, China.
| |
Collapse
|
45
|
Godeanu S, Cătălin B. The Complementary Role of Morphology in Understanding Microglial Functional Heterogeneity. Int J Mol Sci 2025; 26:3811. [PMID: 40332469 PMCID: PMC12027755 DOI: 10.3390/ijms26083811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
A search of the PubMed database for publications on microglia reveals an intriguing shift in scientific interest over time. Dividing microglia into categories such as "resting" and "activated" or M1 versus M2 is nowadays obsolete, with the current research focusing on unraveling microglial heterogeneity. The onset of transcriptomics, especially single-cell RNA sequencing (scRNA-seq), has profoundly reshaped our understanding of microglia heterogeneity. Conversely, microglia morphology analysis can offer important insights regarding their activation state or involvement in tissue responses. This review explores microglial heterogeneity under homeostatic conditions, developmental stages, and disease states, with a focus on integrating transcriptomic data with morphological analysis. Beyond the core gene expression profile, regional differences are observed with cerebellar microglia exhibiting a uniquely immune-vigilant profile. During development, microglia express homeostatic genes before birth, yet the bushy appearance is a characteristic that appears later on. In neurodegeneration, microglia alternate between proinflammatory and neuroprotective roles, influenced by regional factors and disease onset. Understanding these structural adaptations may help identify specific microglial subpopulations for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sânziana Godeanu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Bogdan Cătălin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Building 48, University of Saarland, 66421 Homburg, Germany
| |
Collapse
|
46
|
Hewitt K, Huang XF. The Role of Microglial Exosomes in Clozapine Treatment: Effect on Cognition in Schizophrenia. J Neuroimmune Pharmacol 2025; 20:42. [PMID: 40238023 PMCID: PMC12003456 DOI: 10.1007/s11481-024-10160-8] [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: 06/05/2024] [Accepted: 11/22/2024] [Indexed: 04/18/2025]
Abstract
Schizophrenia is a complex neuropsychiatric disorder characterized by a spectrum of symptoms including cognitive impairments and psychotic episodes. Clozapine, an atypical antipsychotic drug, is a widely recognised treatment option for patients with drug-resistant schizophrenia, due to it having the highest efficacy out of all the antipsychotic drugs. Despite its efficacy, clozapine's impact on cognition and brain structure in schizophrenia patients remains a subject of ongoing research and debate, with accumulating evidence indicating negative impacts on cognitive performance and changes in brain volume. Changes in the immune system are linked to variations in cognitive functioning in schizophrenia. Previous research has indicated that microglia, the primary innate immune cells of the brain, have been associated with decreased cognitive performance when dysfunctional. Evidence suggests that brain structure may mediate the observed relationship between microglia and cognition. Microglial exosomes, integral to neuroinflammation and cellular communication, could provide insight into the neurobiological mechanisms underpinning the effects of clozapine treatment. This review focuses on the proposition that alterations in microglial exosome composition, particularly miRNAs, are involved in mediating clozapine's diverse effects on cognition by influencing brain macrostructure. This review aims to highlight new directions for future research that could lead to more effective and targeted therapeutic approaches in the management of schizophrenia.
Collapse
Affiliation(s)
- Kyle Hewitt
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, 2522, Australia
| | - Xu-Feng Huang
- School of Medical, Indigenous and Health Sciences, University of Wollongong, Wollongong, 2522, Australia.
| |
Collapse
|
47
|
Wu SR, Nowakowski TJ. Exploring human brain development and disease using assembloids. Neuron 2025; 113:1133-1150. [PMID: 40107269 PMCID: PMC12022838 DOI: 10.1016/j.neuron.2025.02.010] [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: 05/08/2024] [Revised: 01/10/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
How the human brain develops and what goes awry in neurological disorders represent two long-lasting questions in neuroscience. Owing to the limited access to primary human brain tissue, insights into these questions have been largely gained through animal models. However, there are fundamental differences between developing mouse and human brain, and neural organoids derived from human pluripotent stem cells (hPSCs) have recently emerged as a robust experimental system that mimics self-organizing and multicellular features of early human brain development. Controlled integration of multiple organoids into assembloids has begun to unravel principles of cell-cell interactions. Moreover, patient-derived or genetically engineered hPSCs provide opportunities to investigate phenotypic correlates of neurodevelopmental disorders and to develop therapeutic hypotheses. Here, we outline the advances in technologies that facilitate studies by using assembloids and summarize their applications in brain development and disease modeling. Lastly, we discuss the major roadblocks of the current system and potential solutions.
Collapse
Affiliation(s)
- Sih-Rong Wu
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
48
|
Schwartz M. Why resident microglial-like cells were missed in the peripheral nervous system. Cell Res 2025:10.1038/s41422-025-01119-2. [PMID: 40229554 DOI: 10.1038/s41422-025-01119-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
|
49
|
Brioschi S, Han CZ, Colonna M. Drivers and shapers of macrophages specification in the developing brain. Curr Opin Immunol 2025; 94:102558. [PMID: 40239283 DOI: 10.1016/j.coi.2025.102558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
The brain harbors two major macrophage populations: microglia reside within the brain parenchyma, while border-associated macrophages (BAMs) are situated at central nervous system (CNS) interfaces. BAMs can be further classified into distinct subsets based on their localization: perivascular macrophages surround blood vessels, meningeal macrophages reside in the leptomeninges, dura macrophages in the dura mater, and choroid plexus macrophages are confined to the choroid plexus. The environmental factors and molecular mechanisms driving the specification of these macrophage populations are still being elucidated. Deciphering the communication pathways between CNS macrophages and their tissue niches during development, homeostasis, and pathologic conditions offers significant potential for treating a wide range of brain disorders, from neurodevelopmental and neuroinflammatory diseases to neurovascular and neurodegenerative conditions. With this short review, we will address the current understanding and knowledge gaps in the field, as well as the future directions for the upcoming years.
Collapse
Affiliation(s)
- Simone Brioschi
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA
| | - Claudia Z Han
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA.
| |
Collapse
|
50
|
Randolph LN, Castiglioni C, Tavian M, Sturgeon CM, Ditadi A. Bloodhounds chasing the origin of blood cells. Trends Cell Biol 2025:S0962-8924(25)00067-4. [PMID: 40221343 DOI: 10.1016/j.tcb.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
The generation of blood cells during embryonic development involves a process resembling lineage reprogramming, where specialized cells within the vasculature become blood forming, or hemogenic. These hemogenic cells undergo rapid transcriptional and morphological changes as they appear to switch from an endothelial to blood identity. What controls this process and the exact nature of the hemogenic cells remains debated, with evidence supporting several hypotheses. In this opinion, we synthesize current knowledge and propose a model reconciling conflicting observations, integrating evolutionary and mechanistic insights into blood cell emergence.
Collapse
Affiliation(s)
- Lauren N Randolph
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Castiglioni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Manuela Tavian
- University of Strasbourg, INSERM UMR-S1109, FMTS, Strasbourg, France
| | - Christopher M Sturgeon
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Ditadi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|