Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.573
Filter
1.
Immunity ; 57(9): 2005-2007, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39260352

ABSTRACT

Aging leads to alterations that precipitate or aggravate several diseases that occur across our lifespan. In the CNS, aging affects the capacity to maintain and repair the myelin sheaths that protect axons and facilitate neuronal signaling. Tiwari et al. report aging-associated transcriptional responses in microglia after demyelination, which could be reversed by epigenetic remodeling after BCG vaccination.


Subject(s)
Aging , BCG Vaccine , Myelin Sheath , Remyelination , BCG Vaccine/immunology , Humans , Aging/immunology , Animals , Myelin Sheath/immunology , Myelin Sheath/metabolism , Microglia/immunology , Demyelinating Diseases/immunology , Epigenesis, Genetic , Mice , Vaccination
2.
Immunity ; 57(9): 2000-2002, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39260350

ABSTRACT

Our brain is not an immune-privileged island isolated from peripheries, but how non-neuronal brain cells interact with the peripheral system is not well understood. Wei et al. report that microglia in the hypothalamic paraventricular nucleus (PVN) with unique vasculature can detect ATP derived from hemodynamic disturbance. These microglia in the PVN regulate the response to hypertension via ATP-P2Y12-C/EBPß signaling.


Subject(s)
Blood Pressure , Brain , Microglia , Paraventricular Hypothalamic Nucleus , Microglia/immunology , Microglia/physiology , Microglia/metabolism , Animals , Humans , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/immunology , Paraventricular Hypothalamic Nucleus/physiology , Blood Pressure/physiology , Brain/immunology , Adenosine Triphosphate/metabolism , Signal Transduction , Hypertension/immunology , Hypertension/physiopathology , CCAAT-Enhancer-Binding Protein-beta/metabolism
3.
Virol J ; 21(1): 219, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285274

ABSTRACT

BACKGROUND: To investigate the mechanism of Golgi matrix protein 130(GM130) regulating the antiviral immune response of TLR3 after herpes simplex virus type 1(HSV-1) infection of microglia cells. We explored the regulatory effects of berberine on the immune response mediated by GM130 and TLR3. METHODS: An in vitro model of HSV-1 infection was established by infecting BV2 cells with HSV-1. RESULTS: Compared to the uninfected group, the Golgi apparatus (GA) fragmentation and GM130 decreased after HSV-1 infection; TLR3 increased at 6 h and began to decrease at 12 h after HSV-1 infection; the secretion of interferon-beta(IFN-ß), tumour necrosis factor alpha(TNF-α), and interleukin-6(IL-6) increased after infection. Knockdown of GM130 aggravated fragmentation of the GA and caused TLR3 to further decrease, and the virus titer also increased significantly. GM130 knockdown inhibits the increase in TLR3 and inflammatory factors induced by TLR3 agonists and increases the viral titer. Overexpression of GM130 alleviated fragmentation of the GA induced by HSV-1, partially restored the levels of TLR3, and reduced viral titers. GM130 overexpression reversed the reduction in TLR3 and inflammatory cytokine levels induced by TLR3 inhibitors. Therefore, the decrease in GM130 levels caused by HSV-1 infection leads to increased viral replication by inhibiting TLR3-mediated innate immunity. Berberine can protect the GA and reverse the downregulation of GM130, as well as the downregulation of TLR3 and its downstream factors after HSV-1 infection, reducing the virus titer. CONCLUSIONS: In microglia, one mechanism of HSV-1 immune escape is disruption of the GM130/TLR3 pathway. Berberine protects the GA and enhances TLR3-mediated antiviral immune responses.


Subject(s)
Down-Regulation , Herpesvirus 1, Human , Immunity, Innate , Microglia , Toll-Like Receptor 3 , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Microglia/virology , Microglia/immunology , Microglia/drug effects , Animals , Mice , Cell Line , Immune Evasion , Berberine/pharmacology , Cytokines/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Herpes Simplex/immunology , Herpes Simplex/virology
4.
Cells ; 13(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39272998

ABSTRACT

There is an urgent need for effective disease-modifying therapeutic interventions for Alzheimer's disease (AD)-the most prevalent cause of dementia with a profound socioeconomic burden. Most clinical trials targeting the classical hallmarks of this disease-ß-amyloid plaques and neurofibrillary tangles-failed, showed discrete clinical effects, or were accompanied by concerning side effects. There has been an ongoing search for novel therapeutic targets. Neuroinflammation, now widely recognized as a hallmark of all neurodegenerative diseases, has been proven to be a major contributor to AD pathology. Here, we summarize the role of neuroinflammation in the pathogenesis and progression of AD and discuss potential targets such as microglia, TREM2, the complement system, inflammasomes, and cytosolic DNA sensors. We also present an overview of ongoing studies targeting specific innate immune system components, highlighting the progress in this field of drug research while bringing attention to the delicate nature of innate immune modulations in AD.


Subject(s)
Alzheimer Disease , Immunity, Innate , Alzheimer Disease/immunology , Humans , Animals , Microglia/immunology , Microglia/metabolism , Inflammasomes/metabolism , Inflammasomes/immunology , Molecular Targeted Therapy , Membrane Glycoproteins , Receptors, Immunologic
5.
Cell Death Dis ; 15(9): 676, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39278921

ABSTRACT

Tumor-associated macrophages/microglia (TAMs) are highly plastic and heterogeneous immune cells that can be immune-supportive or tumor-supportive depending of the microenvironment. TAMs are the most abundant immune cells in glioblastoma (GB), and play a key role in immunosuppression. Therefore, TAMs reprogramming toward immune-supportive cells is a promising strategy to overcome immunosuppression. By leveraging scRNAseq human GB databases, we identified that Inhibitor of Apoptosis Proteins (IAP) were expressed by TAMs. To investigate their role in TAMs-related immunosuppression, we antagonized IAP using the central nervous system permeant SMAC mimetic GDC-0152 (SMg). On explants and cultured immune cells isolated from human GB samples, SMg modified TAMs activity. We showed that SMg treatment promoted microglia pro-apoptotic and anti-tumoral function via caspase-3 pro-inflammatory cleavage and the inhibition of tumoroids growth. Then we designed a relevant immunogenic mouse GB model to decipher the spatio-temporal densities, distribution, phenotypes and function of TAMs with or without SMg treatment. We used 3D imaging techniques, a transgenic mouse with fluorescent TAM subsets and mass cytometry. We confirmed that SMg promoted microglia activation, antigen-presenting function and tumor infiltration. In addition, we observed a remodeling of blood vessels, a decrease in anti-inflammatory macrophages and an increased level of monocytes and their mo-DC progeny. This remodeling of the TAM landscape is associated with an increase in CD8 T cell density and activation. Altogether, these results demonstrated that SMg drives the immunosuppressive basal microglia toward an active phenotype with pro-apoptotic and anti-tumoral function and modifies the GB immune landscape. This identifies IAP as targets of choice for a potential mechanism-based therapeutic strategy and SMg as a promising molecule for this application.


Subject(s)
Glioblastoma , Microglia , Phenotype , Tumor Microenvironment , Glioblastoma/immunology , Glioblastoma/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Animals , Microglia/drug effects , Microglia/metabolism , Microglia/immunology , Humans , Mice , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Apoptosis Regulatory Proteins/metabolism , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Cell Line, Tumor , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Transgenic
6.
Acta Neuropathol ; 148(1): 31, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39196440

ABSTRACT

Alzheimer´s disease (AD) stands out as the most common chronic neurodegenerative disorder. AD is characterized by progressive cognitive decline and memory loss, with neurodegeneration as its primary pathological feature. The role of neuroinflammation in the disease course has become a focus of intense research. While microglia, the brain's resident macrophages, have been pivotal to study central immune inflammation, recent evidence underscores the contributions of other cellular entities to the neuroinflammatory process. In this article, we review the inflammatory role of microglia and astrocytes, focusing on their interactions with AD's core pathologies, amyloid beta deposition, and tau tangle formation. Additionally, we also discuss how different modes of regulated cell death in AD may impact the chronic neuroinflammatory environment. This review aims to highlight the evolving landscape of neuroinflammatory research in AD and underscores the importance of considering multiple cellular contributors when developing new therapeutic strategies.


Subject(s)
Alzheimer Disease , Microglia , Neuroinflammatory Diseases , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/immunology , Humans , Microglia/pathology , Microglia/metabolism , Microglia/immunology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/immunology , Animals , Inflammation/pathology , Astrocytes/pathology , Astrocytes/metabolism , Astrocytes/immunology , Brain/pathology , Brain/metabolism , Amyloid beta-Peptides/metabolism
7.
J Neuroimmunol ; 395: 578433, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39168018

ABSTRACT

This review offers a comprehensive examination of the role of microglia in the pathogenesis of autoimmune uveitis, an inflammatory eye disease with significant potential for vision impairment. Central to our discussion is the dual nature of microglial cells, which act as both protectors and potential perpetrators in the immune surveillance of the retina. We explore the mechanisms of microglial activation, highlighting the key signaling pathways involved, such as NF-κB, JAK/STAT, MAPK, and PI3K/Akt. The review also delves into the genetic and environmental factors influencing microglial behavior, underscoring their complex interaction in disease manifestation. Advanced imaging techniques and emerging biomarkers for microglial activation, pivotal in diagnosing and monitoring the disease, are critically assessed. Additionally, we discuss current and novel therapeutic strategies targeting microglial activity, emphasizing the shift towards more precise and personalized interventions. This article aims to provide a nuanced understanding of microglial dynamics in autoimmune uveitis, offering insights into potential avenues for effective treatment and management.


Subject(s)
Autoimmune Diseases , Microglia , Uveitis , Humans , Uveitis/immunology , Microglia/immunology , Microglia/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Animals , Signal Transduction/immunology
8.
Brain Behav Immun ; 122: 444-455, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39191349

ABSTRACT

Alzheimer's disease (AD) is linked to toxic Aß plaques in the brain and activation of innate responses. Recent findings however suggest that the disease may also depend on the adaptive immunity, as B cells exacerbate and CD8+ T cells limit AD-like pathology in mouse models of amyloidosis. Here, by artificially blocking or augmenting CD8+ T cells in the brain of 5xFAD mice, we provide evidence that AD-like pathology is promoted by pathogenic, proinflammatory cytokines and exhaustion markers expressing CXCR6+ CD39+CD73+/- CD8+ TRM-like cells. The CD8+ T cells appear to act by targeting disease associated microglia (DAM), as we find them in tight complexes with microglia around Aß plaques in the brain of mice and humans with AD. We also report that these CD8+ T cells are induced by B cells in the periphery, further underscoring the pathogenic importance of the adaptive immunity in AD. We propose that CD8+ T cells and B cells should be considered as therapeutic targets for control of AD, as their ablation at the onset of AD is sufficient to decrease CD8+ T cells in the brain and block the amyloidosis-linked neurodegeneration.


Subject(s)
Alzheimer Disease , Amyloidosis , Brain , CD8-Positive T-Lymphocytes , Disease Models, Animal , Microglia , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Amyloidosis/immunology , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Brain/immunology , Brain/pathology , Brain/metabolism , Microglia/immunology , Microglia/metabolism , Mice, Transgenic , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Humans , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/immunology , Adaptive Immunity/immunology , Cytokines/metabolism , Female , Mice, Inbred C57BL , Male
9.
Adv Neurobiol ; 37: 263-286, 2024.
Article in English | MEDLINE | ID: mdl-39207697

ABSTRACT

Microglia are best known as the resident phagocytes of the central nervous system (CNS). As a resident brain immune cell population, microglia play key roles during the initiation, propagation, and resolution of inflammation. The discovery of resident adaptive immune cells in the CNS has unveiled a relationship between microglia and adaptive immune cells for CNS immune-surveillance during health and disease. The interaction of microglia with elements of the peripheral immune system and other CNS resident cells mediates a fine balance between neuroprotection and tissue damage. In this chapter, we highlight the innate immune properties of microglia, with a focus on how pattern recognition receptors, inflammatory signaling cascades, phagocytosis, and the interaction between microglia and adaptive immune cells regulate events that initiate an inflammatory or neuroprotective response within the CNS that modulates immune-mediated disease exacerbation or resolution.


Subject(s)
Immunity, Innate , Microglia , Phagocytosis , Receptors, Pattern Recognition , Humans , Microglia/immunology , Microglia/metabolism , Animals , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Inflammation/immunology , Signal Transduction , Central Nervous System/immunology , Central Nervous System/metabolism , Adaptive Immunity/immunology
10.
PLoS Comput Biol ; 20(8): e1012356, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39186798

ABSTRACT

Alzheimer's disease (AD) involves aggregation of amyloid ß and tau, neuron loss, cognitive decline, and neuroinflammatory responses. Both resident microglia and peripheral immune cells have been associated with the immune component of AD. However, the relative contribution of resident and peripheral immune cell types to AD predisposition has not been thoroughly explored due to their similarity in gene expression and function. To study the effects of AD-associated variants on cis-regulatory elements, we train convolutional neural network (CNN) regression models that link genome sequence to cell type-specific levels of open chromatin, a proxy for regulatory element activity. We then use in silico mutagenesis of regulatory sequences to predict the relative impact of candidate variants across these cell types. We develop and apply criteria for evaluating our models and refine our models using massively parallel reporter assay (MPRA) data. Our models identify multiple AD-associated variants with a greater predicted impact in peripheral cells relative to microglia or neurons. Our results support their use as models to study the effects of AD-associated variants and even suggest that peripheral immune cells themselves may mediate a component of AD predisposition. We make our library of CNN models and predictions available as a resource for the community to study immune and neurological disorders.


Subject(s)
Alzheimer Disease , Genetic Predisposition to Disease , Neural Networks, Computer , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Humans , Genetic Predisposition to Disease/genetics , Microglia/immunology , Computational Biology/methods , Neurons
11.
Adv Neurobiol ; 37: 53-63, 2024.
Article in English | MEDLINE | ID: mdl-39207686

ABSTRACT

Since the classical studies of Pío del Río-Hortega, microglia research has come a long way. In particular, recent advances in bulk and single-cell (sc) transcriptomics have yielded many fascinating new insights into these intriguing immune cells at the interface with the central nervous system (CNS), both in small animal models and human samples. In parallel, tools developed by advanced mouse genetics have revealed the unique ontogeny of microglia and their striking dynamic interactions with other cells in the brain parenchyma. In this chapter, we will discuss various applications of the Cre/loxP-based approach that have enabled the study of microglia in their physiological context of the mouse brain. We will highlight selected key findings that have shaped our current understanding of these cells and discuss the technical intricacies of the Cre/loxP approach and some remaining challenges.


Subject(s)
Brain , Microglia , Animals , Mice , Brain/cytology , Brain/immunology , Brain/metabolism , Integrases/metabolism , Microglia/immunology , Microglia/metabolism , Mutagenesis/immunology , Single-Cell Gene Expression Analysis
12.
Adv Neurobiol ; 37: 39-51, 2024.
Article in English | MEDLINE | ID: mdl-39207685

ABSTRACT

Microglial cells are unique tissue-resident macrophages located in the parenchyma of the central nervous system (CNS). A recent comparative transcriptional study on microglia across more than 20 species from leach across chicken and many more up to humans revealed multiple conserved features. The results indicate the imperative role of microglia over the last 500 million years (Geirsdottir et al. Cell 181:746, 2020). Improved understanding of microglial evolution provides essential insights into conserved and divergent microglial pathways and will have implications for future development of microglia-based therapies to treat CNS disorders. Not only therapeutic approaches may be rethought, but also the understanding of sex specificity of the immune system within the CNS needs to be renewed. Besides revealing the highly detailed characteristics of microglia, the former paradigm of microglia being the only CNS-resident immune cells was outdated by the identification of CNS-associated macrophages (CAMs) as CNS interface residents, who, most likely, accompanied microglia in evolution over the past million years.


Subject(s)
Biological Evolution , Macrophages , Microglia , Animals , Humans , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/cytology , Macrophages/metabolism , Macrophages/immunology , Microglia/immunology , Microglia/metabolism
13.
Adv Neurobiol ; 37: 15-37, 2024.
Article in English | MEDLINE | ID: mdl-39207684

ABSTRACT

The term 'microglia' was first introduced into the scientific literature a century ago. The various eras of microglial research have been defined not only by the number of reports subsequently generated but, more critically, also by the concepts that have shaped our present-day views and understanding of microglia. Key methods, technologies, and models, as well as seminal discoveries made possible through their deployment have enabled breakthroughs, and now pave the way for lines of investigation that could not have been anticipated even a decade ago. Advances in our understanding of the microglial origin, forms, and functions have relied fundamentally on parallel developments in immunology. As the 'neuro-immune' cells of the brain, microglia are now under the spotlight in various disciplines. This chapter surveys the gradual processes and precipitous events that helped form ideas concerning the developmental origin of microglia and their roles in health and disease. It first covers the dawning phase during which the early pioneers of microglial research discovered cellular entities and already assigned functions to them. Following a recess period, the 1960s brought about a renaissance of active interest, with the development of tools and models-and fundamental notions on microglial contributions to central nervous system (CNS) pathologies. These seminal efforts laid the foundation for the awakening of a sweeping research era beginning in the 1980s and spurred on by a blast of immunological discoveries. Finally, this chapter stresses the advancements in molecular, genetic, and imaging approaches to the study of microglia with the turn of the millennium, enabling insights into virtually all facets of microglial physiology. Moving forward, it is clear that the future holds substantial promise for further discoveries. The next epoch in the history of microglial research has just begun.


Subject(s)
Microglia , Animals , Humans , Central Nervous System/cytology , Central Nervous System/immunology , Central Nervous System/metabolism , History, 20th Century , History, 21st Century , Microglia/immunology , Microglia/metabolism
14.
Adv Neurobiol ; 37: 569-578, 2024.
Article in English | MEDLINE | ID: mdl-39207713

ABSTRACT

Microglia are macrophages residing in the central nervous system, where they perform immune surveillance, synaptic remodeling, neurogenesis, and monitor signals arising from brain injuries or potential pathogens.Commonly, rodent models are used for studying microglia because of the available transgenic mouse lines in which specific genetic manipulations are successfully accomplished. However, human and rodents microglia showed significant differences, which are reflected in different morphological and functional properties. These differences are in genetic and transcriptomic, but also in the expression of signaling molecules and age-associated changes.Several strategies are available to study human microglia, as using surgical brain resections from epileptic and tumoral tissues and from post mortem brain samples. In addition, the generation of human-induced pluripotent stem cells (hPSCs) and the possibility to differentiate them in microglia-like cells provide unique opportunities to compare microglia functions between rodents' and human brain.The use of human ex vivo and in vitro brain models allows the study of human microglia, mimicking in vivo conditions. This will be useful for a better understanding of the real live behavior and functions of microglia in the human brain. This chapter aims to highlight significant similarities and differences between human and rodent microglia in order to re-evaluate mouse models of different human brain disorders, proposing the use of in vitro and ex vivo human brain models.Studies on living human microglia in the brain may help to define divergences from animal models and to improve clinical interventions to treat brain pathologies, using alternatives targets.


Subject(s)
Microglia , Animals , Humans , Mice , Brain/cytology , Brain/immunology , Brain/metabolism , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Microglia/immunology , Microglia/metabolism
15.
J Neuroinflammation ; 21(1): 202, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154174

ABSTRACT

Growing evidence has implicated systemic infection as a significant risk factor for the development and advancement of Alzheimer's disease (AD). With the emergence of SARS-CoV-2 (COVID-19) and the resultant pandemic, many individuals from the same aging population vulnerable to AD suffered a severe systemic infection with potentially unidentified long-term consequences for survivors. To study the impact of COVID-19 survival on the brain's intrinsic immune system in a population also suffering from AD, we profiled post-mortem brain tissue from patients in the UF Neuromedicine Human Brain and Tissue Bank with a diagnosis of AD who survived a COVID-19 infection (COVID-AD) and contrasted our findings with AD patients who did not experience a COVID-19 infection, including a group of brain donors who passed away before arrival of SARS-CoV-2 in the United States. We assessed disease-relevant protein pathology and microglial and astrocytic markers by quantitative immunohistochemistry and supplemented these data with whole tissue gene expression analysis performed on the NanoString nCounter® platform. COVID-AD patients showed slightly elevated Aß burden in the entorhinal, fusiform, and inferior temporal cortices compared to non-COVID-AD patients, while tau pathology burden did not differ between groups. Analysis of microglia revealed a significant loss of microglial homeostasis as well as exacerbated microgliosis in COVID-AD patients compared to non-COVID-AD patients in a brain region-dependent manner. Furthermore, COVID-AD patients showed reduced cortical astrocyte numbers, independent of functional subtype. Transcriptomic analysis supported these histological findings and, in addition, identified a dysregulation of oligodendrocyte and myelination pathways in the hippocampus of COVID-AD patients. In summary, our data demonstrate a profound impact of COVID-19 infection on neuroimmune and glial pathways in AD patients persisting for months post-infection, highlighting the importance of peripheral to central neuroimmune crosstalk in neurodegenerative diseases.


Subject(s)
Alzheimer Disease , COVID-19 , Homeostasis , Humans , COVID-19/immunology , COVID-19/complications , COVID-19/pathology , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Male , Female , Aged , Aged, 80 and over , Homeostasis/physiology , Brain/pathology , Brain/immunology , Brain/metabolism , Neuroimmunomodulation/physiology , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Middle Aged , SARS-CoV-2 , Astrocytes/metabolism , Astrocytes/immunology , Astrocytes/pathology
16.
J Agric Food Chem ; 72(32): 17964-17976, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39096281

ABSTRACT

Spinal cord injury (SCI) is one of the most serious health problems, with no effective therapy. Recent studies indicate that Fisetin, a natural polyphenolic flavonoid, exhibits multiple functions, such as life-prolonging, antioxidant, antitumor, and neuroprotection. However, the restorative effects of Fisetin on SCI and the underlying mechanism are still unclear. In the present study, we found that Fisetin reduced LPS-induced apoptosis and oxidative damage in PC12 cells and reversed LPS-induced M1 polarization in BV2 cells. Additionally, Fisetin safely and effectively promoted the motor function recovery of SCI mice by attenuating neurological damage and promoting neurogenesis at the lesion. Moreover, Fisetin administration inhibited glial scar formation, modulated microglia/macrophage polarization, and reduced neuroinflammation. Network pharmacology, RNA-seq, and molecular biology revealed that Fisetin inhibited the activation of the JAK2/STAT3 signaling pathway. Notably, Colivelin TFA, an activator of JAK2/STAT3 signaling, attenuated Fis-mediated neuroinflammation inhibition and therapeutic effects on SCI mice. Collectively, Fisetin promotes functional recovery after SCI by inhibiting microglia/macrophage M1 polarization and the JAK2/STAT3 signaling pathway. Thus, Fisetin may be a promising therapeutic drug for the treatment of SCI.


Subject(s)
Flavonols , Janus Kinase 2 , Macrophages , Microglia , STAT3 Transcription Factor , Signal Transduction , Spinal Cord Injuries , Animals , Humans , Male , Mice , Rats , Cell Polarity/drug effects , Flavonoids/pharmacology , Flavonoids/administration & dosage , Flavonols/pharmacology , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Microglia/immunology , PC12 Cells , Recovery of Function/drug effects , Signal Transduction/drug effects , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/immunology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics
17.
J Alzheimers Dis ; 100(s1): S363-S385, 2024.
Article in English | MEDLINE | ID: mdl-39177607

ABSTRACT

More than a century after the first description of Alzheimer's disease (AD), the road to a cure for this complex and heterogeneous neurodegenerative disorder has been paved by countless descriptive hypotheses and successive clinical trial failures. Auspiciously, the era of genome-wide association studies revolutionized the classical "neurocentric" view of AD by providing clues that brain-resident immune cells (i.e., microglia and astrocytes) are also key players in the pathological and clinical trajectory of this neurodegenerative disorder. Considering that the intercommunication among neurons, astrocytes, and microglia is fundamental for the functional organization of the brain, it is evident that the disruption of the proper functioning of this "triad" could contribute to the neuroinflammatory and neurodegenerative events that occur in the AD brain. Importantly, recent scientific progress in the burgeoning field of immunometabolism, a crossroad between metabolism and immune response, shed light on the importance of metabolic reprogramming of brain-resident immune cells in AD pathology. In this sense, the present review is aimed to summarize and discuss the current knowledge on the metabolic patterns of brain-resident immune cells during the AD continuum, putting a special focus on glucose, amino acids, and lipid metabolism. Changing the "old" picture of AD pathological basis by integrating the role of brain-resident immune cells it is imperative to establish new and feasible therapeutic interventions able to curb neuroinflammatory and neurodegenerative processes, and consequently cognitive deterioration.


Subject(s)
Alzheimer Disease , Brain , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Humans , Brain/metabolism , Brain/immunology , Brain/pathology , Animals , Microglia/metabolism , Microglia/immunology , Astrocytes/metabolism , Astrocytes/immunology , Lipid Metabolism/physiology
18.
Medicine (Baltimore) ; 103(31): e38691, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093774

ABSTRACT

Renal clear cell carcinoma (RCC) is a type of malignant tumor, which, in addition to surgical resection, radiotherapy, and chemotherapy, has been widely treated through immunotherapy recently. However, the influence of the tumor microenvironment and the infiltrating immune cells within it on immunotherapy remains unclear. It is imperative to study the interactions between various immune cells of RCC. The scRNA-seq dataset from GEO's database was used to analyze the immune cells present in tumor tissue and peripheral blood samples. Through quality control, clustering, and identification, the types and proportions of infiltrating immune cells were determined. The cellular differences were determined, and gene expression levels of the differentially present cells were investigated. A protein-protein interaction network analysis was performed using string. KEGG and GO analyses were performed to investigate abnormal activities. The microglia marker CD68 and CD1C+ B dendritic cells marker CD11C were detected using multiplex immunofluorescence staining. Many depleted CD8+ T cells (exhausted CD8+ T cells) appeared in tumor tissues as well as microglia. CD1C+ B dendritic cells did not infiltrate tumor tissues. HSPA1A was correlated with DNAJB1 in microglia. Compared with Paracancer tissues, microglia increased while CD1C+ B dendritic cells decreased in pathological stages I and I-II in cancerous tissues. An altered tumor microenvironment caused by increases in microglia in RCC in the early stage resulted in an inability of CD1C+ B dendritic cells to infiltrate, resulting in CD8+ T cells being unable to receive the antigens presented by them, and in turn being depleted in large quantities.


Subject(s)
Antigens, CD1 , CD8-Positive T-Lymphocytes , Carcinoma, Renal Cell , Dendritic Cells , Kidney Neoplasms , Microglia , Tumor Microenvironment , Humans , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Tumor Microenvironment/immunology , Microglia/immunology , Microglia/metabolism , Antigens, CD1/metabolism , Male , Neoplasm Staging , Female , Glycoproteins
19.
Adv Neurobiol ; 37: 287-302, 2024.
Article in English | MEDLINE | ID: mdl-39207698

ABSTRACT

Microglia are specialized immune cells that reside in the central nervous system (CNS) and play a crucial role in maintaining the homeostasis of the brain microenvironment. While traditionally regarded as a part of the innate immune system, recent research has highlighted their role in adaptive immunity. The CNS is no longer considered an immune-privileged organ, and increasing evidence suggests bidirectional communication between the immune system and the CNS. Microglia are sensitive to systemic immune signals and can respond to systemic inflammation by producing various inflammatory cytokines and chemokines. This response is mediated by activating pattern recognition receptors (PRRs), which recognize pathogen- and danger-associated molecular patterns in the systemic circulation. The microglial response to systemic inflammation has been implicated in several neurological conditions, including depression, anxiety, and cognitive impairment. Understanding the complex interplay between microglia and systemic immunity is crucial for developing therapeutic interventions to modulate immune responses in the CNS.


Subject(s)
Immunity, Innate , Microglia , Microglia/immunology , Microglia/metabolism , Humans , Animals , Immunity, Innate/immunology , Inflammation/immunology , Central Nervous System/immunology , Central Nervous System/metabolism , Cytokines/immunology , Cytokines/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Adaptive Immunity/immunology , Brain/immunology
20.
Proc Natl Acad Sci U S A ; 121(35): e2406421121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39159381

ABSTRACT

Viral infection is frequently assayed by ongoing expression of viral genes. These assays fail to identify cells that have been exposed to the virus but limit or inhibit viral replication. To address this limitation, we used a dual-labeling vesicular stomatitis virus (DL-VSV), which has a deletion of the viral glycoprotein gene, to allow evaluation of primary infection outcomes. This virus encodes Cre, which can stably mark any cell with even a minimal level of viral gene expression. Additionally, the virus encodes GFP, which distinguishes cells with higher levels of viral gene expression, typically due to genome replication. Stereotactic injections of DL-VSV into the murine brain showed that different cell types had very different responses to the virus. Almost all neurons hosted high levels of viral gene expression, while glial cells varied in their responses. Astrocytes (Sox9+) were predominantly productively infected, while oligodendrocytes (Sox10+) were largely abortively infected. Microglial cells (Iba1+) were primarily uninfected. Furthermore, we monitored the early innate immune response to viral infection and identified unique patterns of interferon (IFN) induction. Shortly after infection, microglia were the main producers of IFNb, whereas later, oligodendrocytes were the main producers. IFNb+ cells were primarily abortively infected regardless of cell type. Last, we investigated whether IFN signaling had any impact on the outcome of primary infection and did not observe significant changes, suggesting that intrinsic factors are likely responsible for determining the outcome of primary infection.


Subject(s)
Astrocytes , Animals , Mice , Astrocytes/virology , Astrocytes/metabolism , Virus Replication , Microglia/virology , Microglia/metabolism , Microglia/immunology , Neurons/virology , Neurons/metabolism , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Vesiculovirus/physiology , Vesiculovirus/immunology , Vesiculovirus/genetics , Oligodendroglia/virology , Oligodendroglia/metabolism , Vesicular Stomatitis/virology , Vesicular Stomatitis/immunology , Immunity, Innate , Mice, Inbred C57BL , Brain/virology , Brain/metabolism , Brain/immunology , Neuroglia/virology , Neuroglia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL