Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.971
Filter
1.
Oncol Rep ; 52(5)2024 11.
Article in English | MEDLINE | ID: mdl-39219268

ABSTRACT

Acute myeloid leukemia (AML) is a predominant form of leukemia. Central nervous system (CNS) involvement complicates its diagnosis due to limited diagnostic tools, as well as its treatment due to inadequate therapeutic methodologies and poor prognosis. Furthermore, its incidence rate is unclear. The mechanisms of AML cell mobilization from the bone marrow (BM) to the CNS are not fully elucidated, and the molecular factors contributing to CNS infiltration are insufficiently recognized. The present review aimed to enhance the understanding of CNS involvement of AML and its impact on CNS. The latest research on the pathways and mechanisms facilitating AML cells to escape the BM and infiltrate the CNS was reviewed. Additionally, novel therapeutic strategies targeting specific molecules and genes for treating CNS involvement in AML were examined.


Subject(s)
Central Nervous System Neoplasms , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/therapy , Central Nervous System/pathology , Central Nervous System/metabolism , Bone Marrow/pathology , Bone Marrow/metabolism
2.
Front Endocrinol (Lausanne) ; 15: 1385650, 2024.
Article in English | MEDLINE | ID: mdl-39224124

ABSTRACT

Introduction: Fanconi anemia (FA) is a genomic instability disorder associated with congenital abnormalities, including short stature and the presence of central nervous system anomalies, especially in the hypothalamic-pituitary area. Thus, differences in pituitary size could associate with the short stature observed in these patients. Our aim was to evaluate whether central nervous system abnormalities and pituitary gland volume correlate with height and hormone deficiencies in these patients. Methods: In this cross-sectional exploratory study 21 patients diagnosed with FA between 2017 and 2022 in a Spanish Reference Center were investigated. Magnetic resonance imaging (MRI) was performed and pituitary volume calculated and corelated with height and other endocrine parameters. Results: The percentage of abnormalities in our series was 81%, with a small pituitary (pituitary volume less than 1 SD) being the most frequent, followed by Chiari malformation type 1. The median value of pituitary volume was -1.03 SD (IQR: -1.56, -0.36). Short stature was found in 66.7% [CI95% 43-85.4]. Total volume (mm3) increases significantly with age and in pubertal stages. There were no differences between volume SD and pubertal stage, or the presence of endocrine deficiencies. No correlations were found between pituitary volume and the presence of short stature. The intraclass correlation index (ICC) average for volume was 0.85 [CI95% 0.61-0.94] indicating a good-to-excellent correlation of measurements. Discussion: Central nervous system anomalies are part of the FA phenotype, the most frequent after pituitary hypoplasia being posterior fossa abnormalities, which may have clinical repercussions in the patient. It is therefore necessary to identify those who could be candidates for neurosurgical intervention. The size of the pituitary gland is smaller in these patients, but this does not seem to be related to hormone deficiency and short stature or exposure to a low dose of total body irradiation.


Subject(s)
Fanconi Anemia , Magnetic Resonance Imaging , Pituitary Gland , Humans , Male , Female , Pituitary Gland/diagnostic imaging , Pituitary Gland/pathology , Pituitary Gland/abnormalities , Cross-Sectional Studies , Fanconi Anemia/pathology , Fanconi Anemia/complications , Child , Adolescent , Child, Preschool , Adult , Young Adult , Central Nervous System/abnormalities , Central Nervous System/pathology , Central Nervous System/diagnostic imaging , Organ Size
3.
J Clin Exp Hematop ; 64(3): 237-241, 2024.
Article in English | MEDLINE | ID: mdl-39343611

ABSTRACT

Since the identification of the TTMV::RARA fusion in pediatric cases resembling acute promyelocytic leukemia (APL) by Astolfi et al. in 2021, several similar cases have been reported worldwide. In this report, we present a case of relapsed APL in an adolescent patient, who exhibited the TTMV::RARA fusion gene. This patient exhibited extensive central nervous system involvement and experienced bone marrow necrosis during disease recurrence. Despite achieving complete remission after re-induction chemotherapy, the patient experienced a rapid second relapse, highlighting the extremely aggressive nature of this subtype. These clinical manifestations contribute to the growing recognition of this rare disease.


Subject(s)
Bone Marrow , Leukemia, Promyelocytic, Acute , Necrosis , Humans , Leukemia, Promyelocytic, Acute/diagnosis , Leukemia, Promyelocytic, Acute/pathology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Adolescent , Bone Marrow/pathology , Oncogene Proteins, Fusion/genetics , Male , Recurrence , Retinoic Acid Receptor alpha/genetics , Central Nervous System/pathology
4.
Signal Transduct Target Ther ; 9(1): 254, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39327467

ABSTRACT

The downregulation of Cadm4 (Cell adhesion molecular 4) is a prominent feature in demyelination diseases, yet, the underlying molecular mechanism remains elusive. Here, we reveal that Cadm4 undergoes specific palmitoylation at cysteine-347 (C347), which is crucial for its stable localization on the plasma membrane (PM). Mutation of C347 to alanine (C347A), blocking palmitoylation, causes Cadm4 internalization from the PM and subsequent degradation. In vivo experiments introducing the C347A mutation (Cadm4-KI) lead to severe myelin abnormalities in the central nervous system (CNS), characterized by loss, demyelination, and hypermyelination. We further identify ZDHHC3 (Zinc finger DHHC-type palmitoyltransferase 3) as the enzyme responsible for catalyzing Cadm4 palmitoylation. Depletion of ZDHHC3 reduces Cadm4 palmitoylation and diminishes its PM localization. Remarkably, genetic deletion of ZDHHC3 results in decreased Cadm4 palmitoylation and defects in CNS myelination, phenocopying the Cadm4-KI mouse model. Consequently, altered Cadm4 palmitoylation impairs neuronal transmission and cognitive behaviors in both Cadm4-KI and ZDHHC3 knockout mice. Importantly, attenuated ZDHHC3-Cadm4 signaling significantly influences neuroinflammation in diverse demyelination diseases. Mechanistically, we demonstrate the predominant expression of Cadm4 in the oligodendrocyte lineage and its potential role in modulating cell differentiation via the WNT-ß-Catenin pathway. Together, our findings propose that dysregulated ZDHHC3-Cadm4 signaling contributes to myelin abnormalities, suggesting a common pathological mechanism underlying demyelination diseases associated with neuroinflammation.


Subject(s)
Acyltransferases , Central Nervous System , Lipoylation , Myelin Sheath , Lipoylation/genetics , Animals , Acyltransferases/genetics , Mice , Humans , Myelin Sheath/genetics , Myelin Sheath/metabolism , Myelin Sheath/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Demyelinating Diseases/metabolism , Mice, Knockout
5.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39273598

ABSTRACT

C-C Chemokine Receptor 7 (CCR7) mediates T-cell acute lymphoblastic leukemia (T-ALL) invasion of the central nervous system (CNS) mediated by chemotactic migration to C-C chemokine ligand 19 (CCL19). To determine if a CCL19 antagonist, CCL198-83, could inhibit CCR7-induced chemotaxis and signaling via CCL19 but not CCL21, we used transwell migration and Ca2+ mobilization signaling assays. We found that in response to CCL19, human T-ALL cells employ ß2 integrins to invade human brain microvascular endothelial cell monolayers. In vivo, using an inducible mouse model of T-ALL, we found that we were able to increase the survival of the mice treated with CCL198-83 when compared to non-treated controls. Overall, our results describe a targetable cell surface receptor, CCR7, which can be inhibited to prevent ß2-integrin-mediated T-ALL invasion of the CNS and potentially provides a platform for the pharmacological inhibition of T-ALL cell entry into the CNS.


Subject(s)
CD18 Antigens , Chemokine CCL19 , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Receptors, CCR7 , Receptors, CCR7/metabolism , Receptors, CCR7/genetics , Animals , Humans , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Chemokine CCL19/metabolism , CD18 Antigens/metabolism , Central Nervous System/metabolism , Central Nervous System/pathology , Cell Line, Tumor , Chemotaxis/drug effects , Chemokine CCL21/metabolism , Cell Movement/drug effects , Neoplasm Invasiveness
6.
Bull Exp Biol Med ; 177(4): 528-533, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39266920

ABSTRACT

CNS diseases associated with compromised blood supply and/or vascular integrity are one of the leading causes of mortality and disability in adults worldwide and are also among 10 most common causes of death in children. Angiogenesis is an essential element of regeneration processes upon nervous tissue damage and can play a crucial role in neuroprotection. Here we review the features of cerebral vascular regeneration after ischemic stroke, including the complex interactions between endothelial cells and other brain cell types (neural stem cells, astrocytes, microglia, and oligodendrocytes). The mechanisms of reciprocal influence of angiogenesis and neurogenesis, the role of astrocytes in the formation of the blood-brain barrier, and roles of microglia and oligodendrocytes in vascular regeneration are discussed. Understanding the mechanisms of angiogenesis regulation in CNS is of critical importance for the development of new treatments of neurovascular pathologies.


Subject(s)
Astrocytes , Blood-Brain Barrier , Ischemic Stroke , Neovascularization, Physiologic , Neural Stem Cells , Neurogenesis , Humans , Ischemic Stroke/physiopathology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Neovascularization, Physiologic/physiology , Neurogenesis/physiology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/physiology , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglia/physiology , Microglia/pathology , Microglia/metabolism , Microglia/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Neuroglia/metabolism , Neuroglia/pathology , Brain Ischemia/physiopathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Central Nervous System/blood supply , Central Nervous System/metabolism , Central Nervous System/pathology , Brain/blood supply , Brain/pathology , Brain/metabolism , Brain/physiopathology , Angiogenesis
7.
Int J Mol Sci ; 25(17)2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39273659

ABSTRACT

GM1 gangliosidosis is a lysosomal storage disorder characterized by the accumulation of GM1 ganglioside, leading to severe neurodegeneration and early mortality. The disease primarily affects the central nervous system, causing progressive neurodegeneration, including widespread neuronal loss and gliosis. To gain a deeper understanding of the neuropathology associated with GM1 gangliosidosis, we employed single-nucleus RNA sequencing to analyze brain tissues from both GM1 gangliosidosis model mice and control mice. No significant changes in cell proportions were detected between the two groups of animals. Differential expression analysis revealed cell type-specific changes in gene expression in neuronal and glial cells. Functional analysis highlighted the neurodegenerative processes, oxidative phosphorylation, and neuroactive ligand-receptor interactions as the significantly affected pathways. The contribution of the impairment of neurotransmitter system disruption and neuronal circuitry disruption was more important than neuroinflammatory responses to GM1 pathology. In 16-week-old GM1 gangliosidosis mice, no microglial or astrocyte activation or increased expression of innate immunity genes was detected. This suggested that nerve degeneration did not induce the inflammatory response but rather promoted glial cell clearance. Our findings provide a crucial foundation for understanding the cellular and molecular mechanisms of GM1 gangliosidosis, potentially guiding future therapeutic strategies.


Subject(s)
Disease Models, Animal , Gangliosidosis, GM1 , Animals , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Gangliosidosis, GM1/pathology , Mice , Transcriptome , Neuroglia/metabolism , Neuroglia/pathology , Gene Expression Profiling , Neurons/metabolism , Neurons/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Brain/metabolism , Brain/pathology , G(M1) Ganglioside/metabolism , Single-Cell Analysis , Mice, Inbred C57BL
8.
J Virol ; 98(8): e0056024, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39087762

ABSTRACT

Powassan virus (POWV) is an emergent tick-borne flavivirus that causes fatal encephalitis in the elderly and long-term neurologic sequelae in survivors. How age contributes to severe POWV encephalitis remains an enigma, and no animal models have assessed age-dependent POWV neuropathology. Inoculating C57BL/6 mice with a POWV strain (LI9) currently circulating in Ixodes ticks resulted in age-dependent POWV lethality 10-20 dpi. POWV infection of 50-week-old mice was 82% fatal with lethality sequentially reduced by age to 7.1% in 10-week-old mice. POWV LI9 was neuroinvasive in mice of all ages, causing acute spongiform CNS pathology and reactive gliosis 5-15 dpi that persisted in survivors 30 dpi. High CNS viral loads were found in all mice 10 dpi. However, by 15 dpi, viral loads decreased by 2-4 logs in 10- to 40-week-old mice, while remaining at high levels in 50-week-old mice. Age-dependent differences in CNS viral loads 15 dpi occurred concomitantly with striking changes in CNS cytokine responses. In the CNS of 50-week-old mice, POWV induced Th1-type cytokines (IFNγ, IL-2, IL-12, IL-4, TNFα, IL-6), suggesting a neurodegenerative pro-inflammatory M1 microglial program. By contrast, in 10-week-old mice, POWV-induced Th2-type cytokines (IL-10, TGFß, IL-4) were consistent with a neuroprotective M2 microglial phenotype. These findings correlate age-dependent CNS cytokine responses and viral loads with POWV lethality and suggest potential neuroinflammatory therapeutic targets. Our results establish the age-dependent lethality of POWV in a murine model that mirrors human POWV severity and long-term CNS pathology in the elderly. IMPORTANCE: Powassan virus is an emerging tick-borne flavivirus causing lethal encephalitis in aged individuals. We reveal an age-dependent POWV murine model that mirrors human POWV encephalitis and long-term CNS damage in the elderly. We found that POWV is neuroinvasive and directs reactive gliosis in all age mice, but at acute stages selectively induces pro-inflammatory Th1 cytokine responses in 50-week-old mice and neuroprotective Th2 cytokine responses in 10-week-old mice. Our findings associate CNS viral loads and divergent cytokine responses with age-dependent POWV lethality and survival outcomes. Responses of young mice suggest potential therapeutic targets and approaches for preventing severe POWV encephalitis that may be broadly applicable to other neurodegenerative diseases. Our age-dependent murine POWV model permits analysis of vaccines that prevent POWV lethality, and therapeutics that resolve severe POWV encephalitis.


Subject(s)
Cytokines , Disease Models, Animal , Encephalitis Viruses, Tick-Borne , Encephalitis, Tick-Borne , Mice, Inbred C57BL , Neuroglia , Viral Load , Animals , Mice , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/virology , Encephalitis, Tick-Borne/mortality , Encephalitis, Tick-Borne/pathology , Cytokines/metabolism , Cytokines/immunology , Neuroglia/virology , Neuroglia/immunology , Neuroglia/pathology , Female , Age Factors , Ixodes/virology , Ixodes/immunology , Central Nervous System/virology , Central Nervous System/immunology , Central Nervous System/pathology , Brain/virology , Brain/pathology , Brain/immunology
9.
Int J Mol Sci ; 25(16)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39201536

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two neuroprotective and anti-inflammatory molecules of the central nervous system (CNS). Both bind to three G protein-coupled receptors, namely PAC1, VPAC1 and VPAC2, to elicit their beneficial effects in various CNS diseases, including multiple sclerosis (MS). In this study, we assessed the expression and distribution of PACAP/VIP receptors in the normal-appearing white matter (NAWM) of MS donors with a clinical history of either relapsing-remitting MS (RRMS), primary MS (PPMS), secondary progressive MS (SPMS) or in aged-matched non-MS controls. Gene expression studies revealed MS-subtype specific changes in PACAP and VIP and in the receptors' levels in the NAWM, which were partly corroborated by immunohistochemical analyses. Most PAC1 immunoreactivity was restricted to myelin-producing cells, whereas VPAC1 reactivity was diffused within the neuropil and in axonal bundles, and VPAC2 in small vessel walls. Within and around lesioned areas, glial cells were the predominant populations showing reactivity for the different PACAP/VIP receptors, with distinctive patterns across MS subtypes. Together, these data identify the differential expression patterns of PACAP/VIP receptors among the different MS clinical entities. These results may offer opportunities for the development of personalized therapeutic approaches to treating MS and/or other demyelinating disorders.


Subject(s)
Multiple Sclerosis , Pituitary Adenylate Cyclase-Activating Polypeptide , Vasoactive Intestinal Peptide , White Matter , Humans , White Matter/metabolism , White Matter/pathology , Male , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Middle Aged , Female , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Adult , Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II/genetics , Aged , Receptors, Vasoactive Intestinal Polypeptide, Type I/metabolism , Receptors, Vasoactive Intestinal Polypeptide, Type I/genetics , Autopsy , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Central Nervous System/metabolism , Central Nervous System/pathology , Multiple Sclerosis, Relapsing-Remitting/metabolism , Multiple Sclerosis, Relapsing-Remitting/pathology
10.
Int J Mol Sci ; 25(15)2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39125815

ABSTRACT

Neurological symptoms associated with COVID-19, acute and long term, suggest SARS-CoV-2 affects both the peripheral and central nervous systems (PNS/CNS). Although studies have shown olfactory and hematogenous invasion into the CNS, coinciding with neuroinflammation, little attention has been paid to susceptibility of the PNS to infection or to its contribution to CNS invasion. Here we show that sensory and autonomic neurons in the PNS are susceptible to productive infection with SARS-CoV-2 and outline physiological and molecular mechanisms mediating neuroinvasion. Our infection of K18-hACE2 mice, wild-type mice, and golden Syrian hamsters, as well as primary peripheral sensory and autonomic neuronal cultures, show viral RNA, proteins, and infectious virus in PNS neurons, satellite glial cells, and functionally connected CNS tissues. Additionally, we demonstrate, in vitro, that neuropilin-1 facilitates SARS-CoV-2 neuronal entry. SARS-CoV-2 rapidly invades the PNS prior to viremia, establishes a productive infection in peripheral neurons, and results in sensory symptoms often reported by COVID-19 patients.


Subject(s)
COVID-19 , Neuropilin-1 , SARS-CoV-2 , Animals , SARS-CoV-2/physiology , SARS-CoV-2/pathogenicity , COVID-19/virology , COVID-19/pathology , COVID-19/metabolism , Mice , Neuropilin-1/metabolism , Neuropilin-1/genetics , Viremia/virology , Central Nervous System/virology , Central Nervous System/pathology , Central Nervous System/metabolism , Sensory Receptor Cells/virology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology , Mesocricetus , Humans , Angiotensin-Converting Enzyme 2/metabolism , Mice, Inbred C57BL , Virus Internalization , Male
11.
Commun Biol ; 7(1): 811, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965360

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a demyelinating disease affecting the central nervous system (CNS) in animals that parallels several clinical and molecular traits of multiple sclerosis in humans. Herpes simplex virus type 1 (HSV-1) infection mainly causes cold sores and eye diseases, yet eventually, it can also reach the CNS, leading to acute encephalitis. Notably, a significant proportion of healthy individuals are likely to have asymptomatic HSV-1 brain infection with chronic brain inflammation due to persistent latent infection in neurons. Because cellular senescence is suggested as a potential factor contributing to the development of various neurodegenerative disorders, including multiple sclerosis, and viral infections may induce a premature senescence state in the CNS, potentially increasing susceptibility to such disorders, here we examine the presence of senescence-related markers in the brains and spinal cords of mice with asymptomatic HSV-1 brain infection, EAE, and both conditions. Across all scenarios, we find a significant increases of senescence biomarkers in the CNS with some differences depending on the analyzed group. Notably, some senescence biomarkers are exclusively observed in mice with the combined conditions. These results indicate that asymptomatic HSV-1 brain infection and EAE associate with a significant expression of senescence biomarkers in the CNS.


Subject(s)
Brain , Cellular Senescence , Herpes Simplex , Herpesvirus 1, Human , Multiple Sclerosis , Animals , Mice , Brain/virology , Brain/pathology , Brain/metabolism , Multiple Sclerosis/virology , Multiple Sclerosis/pathology , Multiple Sclerosis/metabolism , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/pathogenicity , Herpes Simplex/virology , Herpes Simplex/pathology , Female , Mice, Inbred C57BL , Encephalomyelitis, Autoimmune, Experimental/virology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Phenotype , Central Nervous System/virology , Central Nervous System/metabolism , Central Nervous System/pathology , Spinal Cord/virology , Spinal Cord/metabolism , Spinal Cord/pathology , Biomarkers/metabolism , Encephalitis, Herpes Simplex/virology , Encephalitis, Herpes Simplex/pathology , Encephalitis, Herpes Simplex/metabolism
12.
Cell Mol Immunol ; 21(9): 943-958, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38997413

ABSTRACT

Many newly emerging and re-emerging viruses have neuroinvasive potential, underscoring viral encephalitis as a global research priority. Upon entry of the virus into the CNS, severe neurological life-threatening conditions may manifest that are associated with high morbidity and mortality. The currently available therapeutic arsenal against viral encephalitis is rather limited, emphasizing the need to better understand the conditions of local antiviral immunity within the infected CNS. In this review, we discuss new insights into the pathophysiology of viral encephalitis, with a focus on myeloid cells and CD8+ T cells, which critically contribute to protection against viral CNS infection. By illuminating the prerequisites of myeloid and T cell activation, discussing new discoveries regarding their transcriptional signatures, and dissecting the mechanisms of their recruitment to sites of viral replication within the CNS, we aim to further delineate the complexity of antiviral responses within the infected CNS. Moreover, we summarize the current knowledge in the field of virus infection and neurodegeneration and discuss the potential links of some neurotropic viruses with certain pathological hallmarks observed in neurodegeneration.


Subject(s)
Central Nervous System , Humans , Animals , Central Nervous System/immunology , Central Nervous System/virology , Central Nervous System/pathology , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , CD8-Positive T-Lymphocytes/immunology , Virus Replication , Antiviral Agents/therapeutic use
14.
J Integr Neurosci ; 23(6): 119, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38940087

ABSTRACT

OBJECTIVES: The majority of neuromyelitis optica spectrum disorders (NMOSD) patients are seropositive for aquaporin-4 (AQP4)-specific antibodies [also named neuromyelitis optica immunoglobulin G antibodies (NMO-IgG)]. Although NMO-IgG can induce pathological changes in the central nervous system (CNS), the immunological changes in the CNS and peripheral tissue remain largely unknown. We investigated whether NMO-IgG binds to tissue expressing AQP4 and induces immunological changes in the peripheral tissue and CNS. METHODS: C57BL/6 female mice were assigned into an NMOSD or control group. Pathological and immunological changes in peripheral tissue and CNS were measured by immunostaining and flow cytometry, respectively. Motor impairment was measured by open-field test. RESULTS: We found that NMO-IgG did bind to astrocyte- and AQP4-expressing peripheral tissue, but induced glial fibrillary acidic protein and AQP4 loss only in the CNS. NMO-IgG induced the activation of microglia and modulated microglia polarization toward the classical (M1) phenotype, but did not affect innate or adaptive immune cells in the peripheral immune system, such as macrophages, neutrophils, Th17/Th1, or IL-10-producing B cells. In addition, NMOSD mice showed significantly less total distance traveled and higher immobility time in the open field. CONCLUSIONS: We found that injection of human NMO-IgG led to astrocytopathic lesions with microglial activation in the CNS. However, there were no significant pathological or immunological changes in the peripheral tissues.


Subject(s)
Aquaporin 4 , Immunoglobulin G , Mice, Inbred C57BL , Neuromyelitis Optica , Animals , Neuromyelitis Optica/immunology , Neuromyelitis Optica/pathology , Aquaporin 4/immunology , Female , Humans , Mice , Disease Models, Animal , Microglia/metabolism , Microglia/immunology , Microglia/drug effects , Autoantibodies/immunology , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/immunology , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology
15.
Sci Transl Med ; 16(751): eadj9672, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38865481

ABSTRACT

Cancer treatment with anti-PD-1 immunotherapy can cause central nervous system immune-related adverse events (CNS-irAEs). The role of microglia in anti-PD-1 immunotherapy-induced CNS-irAEs is unclear. We found that anti-PD-1 treatment of mice caused morphological signs of activation and major histocompatibility complex (MHC) class II up-regulation on microglia. Functionally, anti-PD-1 treatment induced neurocognitive deficits in mice, independent of T cells, B cells, and natural killer cells. Instead, we found that microglia mediated these CNS-irAEs. Single-cell RNA sequencing revealed major transcriptional changes in microglia upon anti-PD-1 treatment. The anti-PD-1 effects were mediated by anti-PD-1 antibodies interacting directly with microglia and were not secondary to peripheral T cell activation. Using a proteomics approach, we identified spleen tyrosine kinase (Syk) as a potential target in activated microglia upon anti-PD-1 treatment. Syk inhibition reduced microglia activation and improved neurocognitive function without impairing anti-melanoma effects. Moreover, we analyzed CNS tissue from a patient cohort that had received anti-PD-1 treatment. Imaging mass cytometry revealed that anti-PD-1 treatment of patients was associated with increased surface marker expression indicative of microglia activation. In summary, we identified a disease-promoting role for microglia in CNS-irAEs driven by Syk and provide an inhibitor-based approach to interfere with this complication after anti-PD-1 immunotherapy.


Subject(s)
Central Nervous System , Immunotherapy , Microglia , Programmed Cell Death 1 Receptor , Animals , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Immunotherapy/adverse effects , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Humans , Central Nervous System/pathology , Central Nervous System/drug effects , Mice, Inbred C57BL , Syk Kinase/metabolism , Mice
16.
Toxicol Pathol ; 52(2-3): 114-122, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38828567

ABSTRACT

Gliosis, including microgliosis and astrocytosis, can be challenging to interpret in nonclinical studies. Incidences of glial foci in brains and spinal cords of control rats and nonhuman primates (NHPs) were reviewed in the historical control databases from two contract research organizations, including one specializing in neuropathology. In the brain, minimal to mild (grades 1-2) microgliosis was the most common diagnosis, especially in NHPs, although occasional moderate or marked microgliosis (grades 3 and 4) was encountered in both species. Microgliosis was more common in the cerebral cortex, cerebellum, and medulla oblongata in both species and was frequent in the white matter (brain), thalamus, and basal nuclei of NHPs. Gliosis ("not otherwise specified") of minimal severity was diagnosed in similar brain sub-sites for both species and was more common in NHPs compared with rats. Astrocytosis was most prominent in the cerebellum (molecular layer) of NHPs but was otherwise uncommon. In the spinal cord, microgliosis was most common in the lateral white matter tracts in rats and NHPs, and in the dorsal white matter tracts in NHPs. These data indicate that low-grade spontaneous glial responses occur with some frequency in control animals of two common nonclinical species.


Subject(s)
Gliosis , Spinal Cord , Animals , Gliosis/pathology , Rats , Spinal Cord/pathology , Male , Brain/pathology , Female , Central Nervous System/pathology , Macaca fascicularis
17.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167301, 2024 10.
Article in English | MEDLINE | ID: mdl-38878832

ABSTRACT

A critical role for mitochondrial dysfunction has been shown in the pathogenesis of fibromyalgia. It is a chronic pain syndrome characterized by neuroinflammation and impaired oxidative balance in the central nervous system. Boswellia serrata (BS), a natural polyphenol, is a well-known able to influence the mitochondrial metabolism. The objective of this study was to evaluate the mitochondrial dysfunction and biogenesis in fibromyalgia and their modulation by BS. To induce the model reserpine (1 mg/Kg) was subcutaneously administered for three consecutive days and BS (100 mg/Kg) was given orally for twenty-one days. BS reduced pain like behaviors in reserpine-injected rats and the astrocytes activation in the dorsal horn of the spinal cord and prefrontal cortex that are recognized as key regions associated with the neuropathic pain. Vulnerability to neuroinflammation and impaired neuronal plasticity have been described as consequences of mitochondrial dysfunction. BS administration increased PGC-1α expression in the nucleus of spinal cord and brain tissues, promoting the expression of regulatory genes for mitochondrial biogenesis (NRF-1, Tfam and UCP2) and cellular antioxidant defence mechanisms (catalase, SOD2 and Prdx 3). According with these data BS reduced lipid peroxidation and the GSSG/GSH ratio and increased SOD activity in the same tissues. Our results also showed that BS administration mitigates cytochrome-c leakage by promoting mitochondrial function and supported the movement of PGC-1α protein into the nucleus restoring the quality control of mitochondria. Additionally, BS reduced Drp1 and Fis1, preventing both mitochondrial fission and cell death, and increased the expression of Mfn2 protein, facilitating mitochondrial fusion. Overall, our results showed important mitochondrial dysfunction in central nervous system in fibromyalgia syndrome and the role of BS in restoring mitochondrial dynamics.


Subject(s)
Fibromyalgia , Mitochondria , Fibromyalgia/metabolism , Fibromyalgia/pathology , Animals , Mitochondria/metabolism , Mitochondria/pathology , Rats , Male , Central Nervous System/metabolism , Central Nervous System/pathology , Organelle Biogenesis , Spinal Cord/metabolism , Spinal Cord/pathology , Oxidative Stress/drug effects , Rats, Wistar , Plant Extracts/pharmacology , Disease Models, Animal
18.
Cell Rep Med ; 5(7): 101622, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38917802

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) has been associated with different forms of immune compromise. This study analyzes the chemokine signals and attracted immune cells in cerebrospinal fluid (CSF) during PML to define immune cell subpopulations relevant for the PML immune response. In addition to chemokines that indicate a general state of inflammation, like CCL5 and CXCL10, the CSF of PML patients specifically contains CCL2 and CCL4. Single-cell transcriptomics of CSF cells suggests an enrichment of distinct CD4+ and CD8+ T cells expressing chemokine receptors CCR2, CCR5, and CXCR3, in addition to ITGA4 and the genetic PML risk genes STXBP2 and LY9. This suggests that specific immune cell subpopulations migrate into the central nervous system to mitigate PML, and their absence might coincide with PML development. Monitoring them might hold clues for PML risk, and boosting their recruitment or function before therapeutic immune reconstitution might improve its risk-benefit ratio.


Subject(s)
Cell Movement , Central Nervous System , Chemokines , Leukoencephalopathy, Progressive Multifocal , Humans , Leukoencephalopathy, Progressive Multifocal/pathology , Leukoencephalopathy, Progressive Multifocal/immunology , Chemokines/metabolism , Chemokines/genetics , Cell Movement/genetics , Central Nervous System/pathology , Central Nervous System/metabolism , Central Nervous System/immunology , CD8-Positive T-Lymphocytes/immunology , Male , Female , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Middle Aged , Aged
20.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892158

ABSTRACT

Neuroinflammatory conditions in the central nervous system (CNS) are implicated in the pathogenesis of several neuroimmune disorders such as acquired demyelinating syndromes, autoimmune encephalopathies, acute or chronic bacterial and viral CNS infections as well as multiple sclerosis (MS) [...].


Subject(s)
Neuroinflammatory Diseases , Humans , Neuroinflammatory Diseases/immunology , Animals , Multiple Sclerosis/therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/drug therapy , Central Nervous System/metabolism , Central Nervous System/pathology , Central Nervous System/immunology , Inflammation
SELECTION OF CITATIONS
SEARCH DETAIL