Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Nat Immunol ; 24(3): 545-557, 2023 03.
Article in English | MEDLINE | ID: mdl-36658241

ABSTRACT

The TREM2-DAP12 receptor complex sustains microglia functions. Heterozygous hypofunctional TREM2 variants impair microglia, accelerating late-onset Alzheimer's disease. Homozygous inactivating variants of TREM2 or TYROBP-encoding DAP12 cause Nasu-Hakola disease (NHD), an early-onset dementia characterized by cerebral atrophy, myelin loss and gliosis. Mechanisms underpinning NHD are unknown. Here, single-nucleus RNA-sequencing analysis of brain specimens from DAP12-deficient NHD individuals revealed a unique microglia signature indicating heightened RUNX1, STAT3 and transforming growth factor-ß signaling pathways that mediate repair responses to injuries. This profile correlated with a wound healing signature in astrocytes and impaired myelination in oligodendrocytes, while pericyte profiles indicated vascular abnormalities. Conversely, single-nuclei signatures in mice lacking DAP12 signaling reflected very mild microglial defects that did not recapitulate NHD. We envision that DAP12 signaling in microglia attenuates wound healing pathways that, if left unchecked, interfere with microglial physiological functions, causing pathology in human. The identification of a dysregulated NHD microglia signature sparks potential therapeutic strategies aimed at resetting microglia signaling pathways.


Subject(s)
Dementia , Subacute Sclerosing Panencephalitis , Animals , Humans , Mice , Brain/metabolism , Dementia/metabolism , Dementia/pathology , Membrane Glycoproteins/metabolism , Microglia/metabolism , Receptors, Immunologic/metabolism , Subacute Sclerosing Panencephalitis/metabolism , Subacute Sclerosing Panencephalitis/pathology
4.
Sci Rep ; 10(1): 7152, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32346002

ABSTRACT

Microglia, the innate immune cells of the central nervous system (CNS) survey their surroundings with their cytoplasmic processes, phagocytose debris and rapidly respond to injury. These functions are affected by the presence of beta-Amyloid (Aß) deposits, hallmark lesions of Alzheimer's disease (AD). We recently demonstrated that exchanging functionally altered endogenous microglia with peripheral myeloid cells did not change Aß-burden in a mouse model mimicking aspects of AD at baseline, and only mildly reduced Aß plaques upon stimulation. To better characterize these different myeloid cell populations, we used long-term in vivo 2-photon microscopy to compare morphology and basic functional parameters of brain populating peripherally-derived myeloid cells and endogenous microglia. While peripherally-derived myeloid cells exhibited increased process movement in the non-diseased brain, the Aß rich environment in an AD-like mouse model, which induced an alteration of surveillance functions in endogenous microglia, also restricted functional characteristics and response to CNS injury of newly recruited peripherally-derived myeloid cells. Our data demonstrate that the Aß rich brain environment alters the functional characteristics of endogenous microglia as well as newly recruited peripheral myeloid cells, which has implications for the role of myeloid cells in disease and the utilization of these cells in Alzheimer's disease therapy.


Subject(s)
Amyloid beta-Peptides/metabolism , Central Nervous System/metabolism , Myeloid Cells/metabolism , Animals , Humans , Mice
5.
Nat Med ; 26(1): 131-142, 2020 01.
Article in English | MEDLINE | ID: mdl-31932797

ABSTRACT

Glia have been implicated in Alzheimer's disease (AD) pathogenesis. Variants of the microglia receptor triggering receptor expressed on myeloid cells 2 (TREM2) increase AD risk, and activation of disease-associated microglia (DAM) is dependent on TREM2 in mouse models of AD. We surveyed gene-expression changes associated with AD pathology and TREM2 in 5XFAD mice and in human AD by single-nucleus RNA sequencing. We confirmed the presence of Trem2-dependent DAM and identified a previously undiscovered Serpina3n+C4b+ reactive oligodendrocyte population in mice. Interestingly, remarkably different glial phenotypes were evident in human AD. Microglia signature was reminiscent of IRF8-driven reactive microglia in peripheral-nerve injury. Oligodendrocyte signatures suggested impaired axonal myelination and metabolic adaptation to neuronal degeneration. Astrocyte profiles indicated weakened metabolic coordination with neurons. Notably, the reactive phenotype of microglia was less evident in TREM2-R47H and TREM2-R62H carriers than in non-carriers, demonstrating a TREM2 requirement in both mouse and human AD, despite the marked species-specific differences.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Transcriptome/genetics , Aged , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Axons/pathology , Brain/metabolism , Brain/pathology , Female , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Middle Aged , Nerve Degeneration/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Transcription, Genetic
6.
Acta Neuropathol ; 138(4): 613-630, 2019 10.
Article in English | MEDLINE | ID: mdl-31350575

ABSTRACT

Identification of multiple immune-related genetic risk factors for sporadic AD (sAD) have put the immune system center stage in mechanisms underlying this disorder. Comprehensive analysis of microglia in different stages of AD in human brains revealed microglia activation to follow the progression of AD neuropathological changes and requiring the co-occurrence of beta-Amyloid (Aß) and tau pathology. Carriers of AD-associated risk variants in TREM2 (Triggering receptor expressed on myeloid cells 2) showed a reduction of plaque-associated microglia and a substantial increase in dystrophic neurites and overall pathological tau compared with age and disease stage matched AD patients without TREM2 risk variants. These findings were substantiated by digital spatial profiling of the plaque microenvironment and targeted gene expression profiling on the NanoString nCounter system, which revealed striking brain region dependent differences in immune response patterns within individual cases. The demonstration of profound brain region and risk-variant specific differences in immune activation in human AD brains impacts the applicability of immune-therapeutic approaches for sAD and related neurodegenerative diseases.


Subject(s)
Alzheimer Disease/genetics , Brain/pathology , Membrane Glycoproteins/genetics , Microglia/pathology , Plaque, Amyloid/pathology , Receptors, Immunologic/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/immunology , Disease Progression , Humans , Male , Microglia/immunology , Neurites/immunology , Neurites/pathology , Plaque, Amyloid/immunology , tau Proteins/metabolism
7.
J Clin Invest ; 128(7): 2774-2786, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29634489

ABSTRACT

Activation of non-neuronal microglia is thought to play a causal role in spinal processing of neuropathic pain. To specifically investigate microglia-mediated effects in a model of neuropathic pain and overcome the methodological limitations of previous approaches exploring microglia function upon nerve injury, we selectively ablated resident microglia by intracerebroventricular ganciclovir infusion into male CD11b-HSVTK-transgenic mice, which was followed by a rapid, complete, and persistent (23 weeks) repopulation of the CNS by peripheral myeloid cells. In repopulated mice that underwent sciatic nerve injury, we observed a normal response to mechanical stimuli, but an absence of thermal hypersensitivity ipsilateral to the injured nerve. Furthermore, we found that neuronal expression of calcitonin gene-related peptide (CGRP), which is a marker of neurons essential for heat responses, was diminished in the dorsal horn of the spinal cord in repopulated mice. These findings identify distinct mechanisms for heat and mechanical hypersensitivity and highlight a crucial contribution of CNS myeloid cells in the facilitation of noxious heat.


Subject(s)
Central Nervous System/pathology , Central Nervous System/physiopathology , Hyperalgesia/pathology , Hyperalgesia/physiopathology , Myeloid Cells/pathology , Myeloid Cells/physiology , Animals , Calcitonin Gene-Related Peptide/physiology , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Microglia/physiology , Neuralgia/pathology , Neuralgia/physiopathology , Peptide Fragments/physiology , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/physiopathology , Peripheral Nerves/pathology , Peripheral Nerves/physiopathology , Spinal Cord Dorsal Horn/pathology , Spinal Cord Dorsal Horn/physiopathology
8.
Acta Ophthalmol ; 96(7): e789-e796, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29536657

ABSTRACT

PURPOSE: To assess the influence of retinal macrophages and microglia on the formation of choroidal neovascularization (CNV). Therefore, we used a transgenic mouse (CD11b-HSVTK) in which the application of ganciclovir (GCV) results in a depletion of CD11b+ cells. METHODS: We first investigated if a local depletion of CD11b+ macrophages and microglia in the retina is feasible. In a second step, the influence of CD11b+ cell depletion on CNV formation was analysed. One eye of each CD11b-HSVTK mouse was injected with GCV, and the fellow eye received sodium chloride solution (NaCl). Cell counting was performed at day 3 and 7 (one injection) or at day 14 and 21 (two injections). Choroidal neovascularization (CNV) was induced by argon laser and analysed at day 14. RESULTS: The most effective CD11b+ cell depletion was achieved 7 days after a single injection and 14 days after two injections of GCV. After two injections of GCV, we found a significant reduction of CD11b+ cells in central (52 ± 23.9 cells/mm2 ) and peripheral retina (53 ± 20.6 cells/mm2 ); compared to eyes received NaCl (216 ± 49.0 and 210 ± 50.5 cells/mm2 , p < 0.001, respectively). Regarding CNV areas, no statistical significance was found between the groups. CONCLUSION: The CD11b-HSVTK mouse is a feasible model for a local depletion of CD11b+ cells in the retina. Nevertheless, only a partial depletion of CD11b+ cells could be achieved compared to baseline data without any intravitreal injections. Our results did not reveal a significant reduction in CNV areas. In the light of previous knowledge, the potential influence of systemic immune cells on CNV formation might be more relevant than expected.


Subject(s)
CD11b Antigen/physiology , Choroidal Neovascularization/physiopathology , Disease Models, Animal , Macrophages/immunology , Microglia/immunology , Animals , Antiviral Agents/pharmacology , Calcium-Binding Proteins/physiology , Cell Count , Choroidal Neovascularization/metabolism , Female , Ganciclovir/pharmacology , Intravitreal Injections , Laser Coagulation , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/physiology , Microglia/pathology , Polymerase Chain Reaction , Simplexvirus/enzymology , Thymidine Kinase/genetics
9.
Nat Neurosci ; 21(3): 329-340, 2018 03.
Article in English | MEDLINE | ID: mdl-29463850

ABSTRACT

Though motor neurons selectively degenerate in amyotrophic lateral sclerosis, other cell types are likely involved in this disease. We recently generated rNLS8 mice in which human TDP-43 (hTDP-43) pathology could be reversibly induced in neurons and expected that microglia would contribute to neurodegeneration. However, only subtle microglial changes were detected during disease in the spinal cord, despite progressive motor neuron loss; microglia still reacted to inflammatory triggers in these mice. Notably, after hTDP-43 expression was suppressed, microglia dramatically proliferated and changed their morphology and gene expression profiles. These abundant, reactive microglia selectively cleared neuronal hTDP-43. Finally, when microgliosis was blocked during the early recovery phase using PLX3397, a CSF1R and c-kit inhibitor, rNLS8 mice failed to regain full motor function, revealing an important neuroprotective role for microglia. Therefore, reactive microglia exert neuroprotective functions in this amyotrophic lateral sclerosis model, and definition of the underlying mechanism could point toward novel therapeutic strategies.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/pathology , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/pathology , Aminopyridines/pharmacology , Animals , Gene Expression Profiling , Gliosis/pathology , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Mutation/genetics , Myeloid Cells/pathology , Pyrroles/pharmacology , Recovery of Function , Spinal Cord/pathology , Superoxide Dismutase-1/genetics
10.
Acta Neuropathol ; 132(3): 361-75, 2016 09.
Article in English | MEDLINE | ID: mdl-27393312

ABSTRACT

Diets high in fat (HFD) are known to cause an immune response in the periphery as well as the central nervous system. In peripheral adipose tissue, this immune response is primarily mediated by macrophages that are recruited to the tissue. Similarly, reactivity of microglia, the innate immune cells of the brain, has been shown to occur in the hypothalamus of mice fed a high-fat diet. To characterize the nature of the microglial response to diets high in fat in a temporal fashion, we studied the phenotypic spectrum of hypothalamic microglia of mice fed high-fat diet for 3 days and 8 weeks by assessing their tissue reaction and inflammatory signature. While we observed a significant increase in Iba1+ myeloid cells and a reaction of GFAP+ astrocytes in the hypothalamus after 8 weeks of HFD feeding, we found the hypothalamic myeloid cell reaction to be limited to endogenous microglia and not mediated by infiltrating myeloid cells. Moreover, obese humans were found to present with signs of hypothalamic gliosis and exacerbated microglia dystrophy, suggesting a targeted microglia response to diet in humans as well. Notably, the glial reaction occurring in the mouse hypothalamus was not accompanied by an increase in pro-inflammatory cytokines, but rather by an anti-inflammatory reaction. Gene expression analyses of isolated microglia not only confirmed this observation, but also revealed a downregulation of microglia genes important for sensing signals in the microenvironment. Finally, we demonstrate that long-term exposure of microglia to HFD in vivo does not impair the cell's ability to respond to additional stimuli, like lipopolysaccharide. Taken together, our findings support the notion that microglia react to diets high in fat in a region-specific manner in rodents as well as in humans; however, this response changes over time as it is not exclusively pro-inflammatory nor does exposure to HFD prime microglia in the hypothalamus.


Subject(s)
Astrocytes/metabolism , Central Nervous System/metabolism , Diet, High-Fat , Microglia/metabolism , Obesity/complications , Animal Feed , Animals , Gliosis/metabolism , Male , Mice, Inbred C57BL , Neurons/metabolism
11.
J Exp Med ; 212(11): 1811-8, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26458768

ABSTRACT

Although central nervous system-resident microglia are believed to be ineffective at phagocytosing and clearing amyloid-ß (Aß), a major pathological hallmark of Alzheimer's disease (AD), it has been suggested that peripheral myeloid cells constitute a heterogeneous cell population with greater Aß-clearing capabilities. Here, we demonstrate that the conditional ablation of resident microglia in CD11b-HSVTK (TK) mice is followed by a rapid repopulation of the brain by peripherally derived myeloid cells. We used this system to directly assess the ability of peripheral macrophages to reduce Aß plaque pathology and therefore depleted and replaced the pool of resident microglia with peripherally derived myeloid cells in Aß-carrying APPPS1 mice crossed to TK mice (APPPS1;TK). Despite a nearly complete exchange of resident microglia with peripheral myeloid cells, there was no significant change in Aß burden or APP processing in APPPS1;TK mice. Importantly, however, newly recruited peripheral myeloid cells failed to cluster around Aß deposits. Even additional anti-Aß antibody treatment aimed at engaging myeloid cells with amyloid plaques neither directed peripherally derived myeloid cells to amyloid plaques nor altered Aß burden. These data demonstrate that mere recruitment of peripheral myeloid cells to the brain is insufficient in substantially clearing Aß burden and suggest that specific additional triggers appear to be required to exploit the full potential of myeloid cell-based therapies for AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Myeloid Cells/physiology , Alzheimer Disease/metabolism , Animals , Cell Movement , Disease Models, Animal , Ganciclovir/pharmacology , Male , Mice , Microglia/physiology , Vaccination
12.
Acta Neuropathol ; 126(4): 461-77, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24224195

ABSTRACT

The identification of microglia-associated, neurological disease-causing mutations in patients, combined with studies in mouse models has highlighted microglia, the brain's intrinsic myeloid cells, as key modulators of pathogenesis and disease progression in neurodegenerative diseases. In Alzheimer's disease (AD) in particular, the activation and accumulation of microglial cells around b-Amyloid (Ab) plaques has long been described and is believed to result in chronic neuroinflammation­a term that, despite being commonly used, lacks a precise definition. This seemingly directed response of microglia to amyloid deposits conflicts with the fact that the increasing buildup of Ab plaques is not inhibited by these cells during disease progression. While recent evidence suggests that microglia lose their intrinsic beneficial function during the course of AD and may even acquire a ''toxic'' phenotype over time, Ab may also simply not be an appropriate trigger to induce phagocytosis and degradation by microglia in vivo. As recent experimental evidence has indicated the importance of the microglia in AD pathogenesis, future efforts aimed at tackling this disease via utilization or modulation of microglia or factors therefrom appear to be an exciting and challenging research front.


Subject(s)
Alzheimer Disease/pathology , Microglia/physiology , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Vaccines/therapeutic use , Amyloid beta-Peptides/immunology , Animals , Cellular Senescence/physiology , Humans , Macrophage Activation/physiology , Microglia/immunology , Microglia/metabolism , Myeloid Cells/physiology , Phenotype
13.
PLoS One ; 8(4): e60921, 2013.
Article in English | MEDLINE | ID: mdl-23577177

ABSTRACT

Microglial cells closely interact with senile plaques in Alzheimer's disease and acquire the morphological appearance of an activated phenotype. The significance of this microglial phenotype and the impact of microglia for disease progression have remained controversial. To uncover and characterize putative changes in the functionality of microglia during Alzheimer's disease, we directly assessed microglial behavior in two mouse models of Alzheimer's disease. Using in vivo two-photon microscopy and acute brain slice preparations, we found that important microglial functions - directed process motility and phagocytic activity - were strongly impaired in mice with Alzheimer's disease-like pathology compared to age-matched non-transgenic animals. Notably, impairment of microglial function temporally and spatially correlated with Aß plaque deposition, and phagocytic capacity of microglia could be restored by interventionally decreasing amyloid burden by Aß vaccination. These data suggest that major microglial functions progressively decline in Alzheimer's disease with the appearance of Aß plaques, and that this functional impairment is reversible by lowering Aß burden, e.g. by means of Aß vaccination.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Microglia/pathology , Plaque, Amyloid/pathology , Protein Multimerization , Amyloid beta-Peptides/genetics , Animals , Cell Movement , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Mutation , Phagocytosis , Protein Structure, Secondary , Spatio-Temporal Analysis
14.
Nat Med ; 18(12): 1812-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23178247

ABSTRACT

The pathology of Alzheimer's disease has an inflammatory component that is characterized by upregulation of proinflammatory cytokines, particularly in response to amyloid-ß (Aß). Using the APPPS1 Alzheimer's disease mouse model, we found increased production of the common interleukin-12 (IL-12) and IL-23 subunit p40 by microglia. Genetic ablation of the IL-12/IL-23 signaling molecules p40, p35 or p19, in which deficiency of p40 or its receptor complex had the strongest effect, resulted in decreased cerebral amyloid load. Although deletion of IL-12/IL-23 signaling from the radiation-resistant glial compartment of the brain was most efficient in mitigating cerebral amyloidosis, peripheral administration of a neutralizing p40-specific antibody likewise resulted in a reduction of cerebral amyloid load in APPPS1 mice. Furthermore, intracerebroventricular delivery of antibodies to p40 significantly reduced the concentration of soluble Aß species and reversed cognitive deficits in aged APPPS1 mice. The concentration of p40 was also increased in the cerebrospinal fluid of subjects with Alzheimer's disease, which suggests that inhibition of the IL-12/IL-23 pathway may attenuate Alzheimer's disease pathology and cognitive deficits.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Cognition/drug effects , Interleukin-12 Subunit p40/metabolism , Interleukin-12/metabolism , Signal Transduction/drug effects , Analysis of Variance , Animals , Antibodies/administration & dosage , Antibodies/pharmacology , Blotting, Western , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Injections, Intraperitoneal , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-12 Subunit p40/cerebrospinal fluid , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Mice , Mice, Transgenic , Microglia/metabolism , Real-Time Polymerase Chain Reaction
15.
Brain ; 135(Pt 6): 1964-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22492561

ABSTRACT

Ambivalent effects of interleukin-6 on the pathogenesis of ischaemic stroke have been reported. However, to date, the long-term actions of interleukin-6 after stroke have not been investigated. Here, we subjected interleukin-6 knockout (IL-6(-/-)) and wild-type control mice to mild brain ischaemia by 30-min filamentous middle cerebral artery occlusion/reperfusion. While ischaemic tissue damage was comparable at early time points, IL-6(-/-) mice showed significantly increased chronic lesion volumes as well as worse long-term functional outcome. In particular, IL-6(-/-) mice displayed an impaired angiogenic response to brain ischaemia with reduced numbers of newly generated endothelial cells and decreased density of perfused microvessels along with lower absolute regional cerebral blood flow and reduced vessel responsivity in ischaemic striatum at 4 weeks. Similarly, the early genomic activation of angiogenesis-related gene networks was strongly reduced and the ischaemia-induced signal transducer and activator of transcription 3 activation observed in wild-type mice was almost absent in IL-6(-/-) mice. In addition, systemic neoangiogenesis was impaired in IL-6(-/-) mice. Transplantation of interleukin-6 competent bone marrow into IL-6(-/-) mice (IL-6(chi)) did not rescue interleukin-6 messenger RNA expression or the early transcriptional activation of angiogenesis after stroke. Accordingly, chronic stroke outcome in IL-6(chi) mice recapitulated the major effects of interleukin-6 deficiency on post-stroke regeneration with significantly enhanced lesion volumes and reduced vessel densities. Additional in vitro experiments yielded complementary evidence, which showed that after stroke resident brain cells serve as the major source of interleukin-6 in a self-amplifying network. Treatment of primary cortical neurons, mixed glial cultures or immortalized brain endothelia with interleukin 6-induced robust interleukin-6 messenger RNA transcription in each case, whereas oxygen-glucose deprivation did not. However, oxygen-glucose deprivation of organotypic brain slices resulted in strong upregulation of interleukin-6 messenger RNA along with increased transcription of key angiogenesis-associated genes. In conclusion, interleukin-6 produced locally by resident brain cells promotes post-stroke angiogenesis and thereby affords long-term histological and functional protection.


Subject(s)
Infarction, Middle Cerebral Artery/complications , Interleukin-6/metabolism , Neovascularization, Pathologic/etiology , Analysis of Variance , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Bone Marrow Transplantation/methods , Brain/pathology , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Embryo, Mammalian , Endothelial Cells/pathology , Endothelin-1/genetics , Endothelin-1/metabolism , Enzyme-Linked Immunosorbent Assay , Gait Disorders, Neurologic/etiology , Gene Expression Profiling , Gene Expression Regulation/genetics , Glucose/deficiency , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypoxia/complications , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/surgery , Interleukin-6/genetics , Mice , Mice, Knockout/genetics , Microfilament Proteins/metabolism , Neovascularization, Pathologic/metabolism , Neuroglia/physiology , Neurons/drug effects , Oligonucleotide Array Sequence Analysis , Perfusion Imaging , Receptor, trkB/genetics , Receptor, trkB/metabolism , Rotarod Performance Test , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Tetrazolium Salts , Thiazoles
16.
Int J Cancer ; 130(10): 2291-9, 2012 May 15.
Article in English | MEDLINE | ID: mdl-21671470

ABSTRACT

Estrogen signaling plays an important role in breast carcinogenesis. An increased understanding of estrogen gene targets and their effects will allow for more directed and effective therapies for individuals with breast cancer, particularly those with estrogen receptor positive tumors resistant to tamoxifen therapy. Here, we identify YPEL3 as a growth suppressive protein downregulated by estrogen in estrogen receptor positive breast cancer cell lines. Estrogen repression of YPEL3 expression was found to be independent of p53 but dependent on estrogen receptor alpha expression. Importantly, YPEL3 expression, which is induced by the removal of estrogen or treatment with tamoxifen triggers cellular senescence in MCF-7 cells while loss of YPEL3 increases the growth rate of MCF-7 cells. Taken together these findings suggest that YPEL3 may represent a potential target for directed hormonal therapy for estrogen receptor positive breast cancer patients.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cellular Senescence/drug effects , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Tumor Suppressor Proteins/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Estrogens , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms, Hormone-Dependent , Tumor Suppressor Proteins/metabolism
17.
Cell Cycle ; 9(16): 3376-82, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20724842

ABSTRACT

Cellular senescence is an irreversible state of terminal growth arrest that requires functional p53. Acting to block tumor formation, induction of senescence has also been demonstrated to contribute to tumor clearance via the immune system following p53 reactivation. The Hdm2-antagonist, Nutlin-3a, has been shown to reactivate p53 and induce a quiescent state in various cancer cell lines, similar to the G(1) arrest observed upon RNAi targeting of Hdm2 in MCF7 breast cancer. In the present study we show that HdmX, a negative regulator of p53, impacts the senescence pathway. Specifically, overexpression of HdmX blocks Ras mediated senescence in primary human fibroblasts. The interaction of HdmX with p53 and the re-localization of HdmX to the nucleus through Hdm2 association appear to be required for this activity. Furthermore, inhibiting HdmX in prostate adenocarcinoma cells expressing wild-type p53, mutant Ras and high levels of HdmX induced cellular senescence as measured by an increase in irreversible b-galactosidase staining. Together these results suggest that HdmX overexpression may contribute to tumor formation by blocking senescence and that targeting HdmX may represent an attractive anti-cancer therapeutic approach.


Subject(s)
Cellular Senescence , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins , Cell Line , Fibroblasts/metabolism , Humans , Mutation , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , Tumor Suppressor Protein p53/metabolism
18.
Cancer Res ; 70(9): 3566-75, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20388804

ABSTRACT

Cellular senescence, the limited ability of cultured normal cells to divide, can result from cellular damage triggered through oncogene activation (premature senescence) or the loss of telomeres following successive rounds of DNA replication (replicative senescence). Although both processes require a functional p53 signaling pathway, relevant downstream p53 targets have been difficult to identify. Discovery of senescence activators is important because induction of tumor cell senescence may represent a therapeutic approach for the treatment of cancer. In microarray studies in which p53 was reactivated in MCF7 cells, we discovered that Yippee-like-3 (YPEL3), a member of a recently discovered family of putative zinc finger motif coding genes consisting of YPEL1-5, is a p53-regulated gene. YPEL3 expression induced by DNA damage leads to p53 recruitment to a cis-acting DNA response element located near the human YPEL3 promoter. Physiologic induction of YPEL3 results in a substantial decrease in cell viability associated with an increase in cellular senescence. Through the use of RNAi and H-ras induction of cellular senescence, we show that YPEL3 activates cellular senescence downstream of p53. Consistent with its growth suppressive activity, YPEL3 gene expression is repressed in ovarian tumor samples. One mechanism of YPEL3 downregulation in ovarian tumor cell lines seems to be hypermethylation of a CpG island upstream of the YPEL3 promoter. We believe these findings point to YPEL3 being a novel tumor suppressor, which upon induction triggers a permanent growth arrest in human tumor and normal cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Cellular Senescence/genetics , CpG Islands , DNA Methylation , HCT116 Cells , Hep G2 Cells , Humans , Neoplasms/metabolism , Neoplasms/pathology , Tumor Suppressor Proteins/biosynthesis
19.
Front Biosci ; 13: 3423-38, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508444

ABSTRACT

We have long promulgated the idea that microglial cells serve an entirely beneficial role in the central nervous system (CNS), not only as immunological sentinels to fend off potentially dangerous infections, but also as constitutively neuroprotective glia that help sustain neuronal function in the normal and especially in the injured CNS when microglia become activated. In recent years, we have reported on the presence of degenerating microglial cells, which are prominent in the brains of aged humans and humans with neurodegenerative diseases, and this has led us to propose a hypothesis stating that loss of microglia and microglial neuroprotective functions could, at least in part, account for aging-related neurodegeneration. In the current review, we sum up the many aspects that characterize microglial activation and compare them to those that characterize microglial senescence and degeneration. We also consider the possible role of oxidative stress as a cause of microglial degeneration. We finish up by discussing the role microglial cells play in terms of amyloid clearance and degradation with the underlying idea that removal of amyloid constitutes a microglial neuroprotective function, which may become compromised during aging.


Subject(s)
Brain/pathology , Microglia/pathology , Nerve Degeneration/pathology , Neurons/pathology , Aging , Alzheimer Disease/pathology , Animals , Brain/growth & development , Brain Injuries/pathology , Cellular Senescence , Cytosol/pathology , Humans , Huntington Disease/pathology , Hydrogen Peroxide/metabolism , Lipofuscin/metabolism , Models, Animal , Parkinson Disease/pathology
20.
Neuron Glia Biol ; 3(3): 245-53, 2007 Aug.
Article in English | MEDLINE | ID: mdl-18634615

ABSTRACT

Neuroinflammation resulting from chronic reactive microgliosis is thought to contribute to age-related neurodegeneration, as well as age-related neurodegenerative diseases, specifically Alzheimer's disease (AD). Support of this theory comes from studies reporting a progressive, age-associated increase in microglia with an activated phenotype. Although the underlying cause(s) of this microglial reactivity is idiopathic, an accepted therapeutic strategy for the treatment of AD is inhibition of microglial activation using anti-inflammatory agents. Although the effectiveness of anti-inflammatory treatment for AD remains equivocal, microglial inhibition is being tested as a potential treatment for additional neurodegenerative disorders including amyotrophic lateral sclerosis and Parkinson's disease. Given the important and necessary functions of microglia in normal brain, careful evaluation of microglial function in the aged brain is a necessary first step in targeting more precise treatment strategies for aging-related neurodegenerative diseases. Studies from our laboratory have shown multiple age-related changes in microglial morphology and function that are suggestive of cellular senescence. In this manuscript, we review current knowledge of microglia in the aging brain and present new, unpublished work that further supports the theory that microglia experience an age-related decline in proliferative function as a result of cellular senescence.

SELECTION OF CITATIONS
SEARCH DETAIL
...