Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell ; 163(2): 324-39, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26451483

ABSTRACT

Neurodegenerative diseases have been linked to inflammation, but whether altered immunomodulation plays a causative role in neurodegeneration is not clear. We show that lack of cytokine interferon-ß (IFN-ß) signaling causes spontaneous neurodegeneration in the absence of neurodegenerative disease-causing mutant proteins. Mice lacking Ifnb function exhibited motor and cognitive learning impairments with accompanying α-synuclein-containing Lewy bodies in the brain, as well as a reduction in dopaminergic neurons and defective dopamine signaling in the nigrostriatal region. Lack of IFN-ß signaling caused defects in neuronal autophagy prior to α-synucleinopathy, which was associated with accumulation of senescent mitochondria. Recombinant IFN-ß promoted neurite growth and branching, autophagy flux, and α-synuclein degradation in neurons. In addition, lentiviral IFN-ß overexpression prevented dopaminergic neuron loss in a familial Parkinson's disease model. These results indicate a protective role for IFN-ß in neuronal homeostasis and validate Ifnb mutant mice as a model for sporadic Lewy body and Parkinson's disease dementia.


Subject(s)
Interferon-beta/metabolism , Neurons/metabolism , Receptor, Interferon alpha-beta/metabolism , Animals , Autophagy , Disease Models, Animal , Genetic Therapy , Interferon-beta/genetics , Interferon-beta/therapeutic use , Lewy Body Disease/metabolism , Lewy Body Disease/pathology , Mice , Mice, Inbred C57BL , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Receptor, Interferon alpha-beta/genetics , Signal Transduction , Transcriptome , alpha-Synuclein/metabolism
2.
Mol Cell Proteomics ; 23(6): 100782, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705386

ABSTRACT

Cellular communication within the brain is imperative for maintaining homeostasis and mounting effective responses to pathological triggers like hypoxia. However, a comprehensive understanding of the precise composition and dynamic release of secreted molecules has remained elusive, confined primarily to investigations using isolated monocultures. To overcome these limitations, we utilized the potential of TurboID, a non-toxic biotin ligation enzyme, to capture and enrich secreted proteins specifically originating from human brain pericytes in spheroid cocultures with human endothelial cells and astrocytes. This approach allowed us to characterize the pericyte secretome within a more physiologically relevant multicellular setting encompassing the constituents of the blood-brain barrier. Through a combination of mass spectrometry and multiplex immunoassays, we identified a wide spectrum of different secreted proteins by pericytes. Our findings demonstrate that the pericytes secretome is profoundly shaped by their intercellular communication with other blood-brain barrier-residing cells. Moreover, we identified substantial differences in the secretory profiles between hypoxic and normoxic pericytes. Mass spectrometry analysis showed that hypoxic pericytes in coculture increase their release of signals related to protein secretion, mTOR signaling, and the complement system, while hypoxic pericytes in monocultures showed an upregulation in proliferative pathways including G2M checkpoints, E2F-, and Myc-targets. In addition, hypoxic pericytes show an upregulation of proangiogenic proteins such as VEGFA but display downregulation of canonical proinflammatory cytokines such as CXCL1, MCP-1, and CXCL6. Understanding the specific composition of secreted proteins in the multicellular brain microvasculature is crucial for advancing our knowledge of brain homeostasis and the mechanisms underlying pathology. This study has implications for the identification of targeted therapeutic strategies aimed at modulating microvascular signaling in brain pathologies associated with hypoxia.


Subject(s)
Cell Hypoxia , Coculture Techniques , Pericytes , Spheroids, Cellular , Pericytes/metabolism , Humans , Spheroids, Cellular/metabolism , Secretome/metabolism , Endothelial Cells/metabolism , Astrocytes/metabolism , Proteomics/methods , Cell Communication , Blood-Brain Barrier/metabolism , Cells, Cultured , Brain/metabolism , Mass Spectrometry , Signal Transduction
3.
Stroke ; 55(3): 558-568, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38323422

ABSTRACT

BACKGROUND: Blood-based biomarkers have the potential to reflect cerebrovascular signaling after microvascular injury; yet, the detection of cell-specific signaling has proven challenging. Microvesicles retain parental cell surface antigens allowing detection of cell-specific signaling encoded in their cargo. In ischemic stroke, the progression of pathology involves changes in microvascular signaling whereby brain pericytes, perivascular cells wrapping the microcapillaries, are one of the early responders to the ischemic insult. Intercepting the pericyte signaling response peripherally by isolating pericyte-derived microvesicles may provide not only diagnostic information on microvascular injury but also enable monitoring of important pathophysiological mechanisms. METHODS: Plasma samples were collected from patients with acute ischemic stroke (n=39) at 3 time points after stroke onset: 0 to 6 hours, 12 to 24 hours, and 2 to 6 days, and compared with controls (n=39). Pericyte-derived microvesicles were isolated based on cluster of differentiation 140b expression and quantified by flow cytometry. The protein content was evaluated using a proximity extension assay, and vascular signaling pathways were examined using molecular signature hallmarks and gene ontology. RESULTS: In this case-control study, patients with acute ischemic stroke showed significantly increased numbers of pericyte-derived microvesicles (median, stroke versus controls) at 12 to 24 hours (1554 versus 660 microvesicles/µL; P=0.0041) and 2 to 6 days after stroke (1346 versus 660 microvesicles/µL; P=0.0237). Their proteome revealed anti-inflammatory properties mediated via downregulation of Kirsten rat sarcoma virus and IL (interleukin)-6/JAK/STAT3 signaling at 0 to 6 hours, but proangiogenic as well as proinflammatory signals at 12 to 24 hours. Between 2 and 6 days, proteins were mainly associated with vascular remodeling as indicated by activation of Hedgehog signaling in addition to proangiogenic signals. CONCLUSIONS: We demonstrate that the plasma of patients with acute ischemic stroke reflects (1) an early and time-dependent increase of pericyte-derived microvesicles and (2) changes in the protein cargo of microvesicles over time indicating cell signaling specifically related to inflammation and vascular remodeling.


Subject(s)
Ischemic Stroke , Stroke , Humans , Ischemic Stroke/pathology , Pericytes/pathology , Vascular Remodeling , Case-Control Studies , Hedgehog Proteins/metabolism , Brain/pathology , Stroke/pathology , Signal Transduction , Biomarkers/metabolism
4.
Int J Mol Sci ; 24(6)2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36982744

ABSTRACT

The brain needs sufficient oxygen in order to function normally. This is achieved by a large vascular capillary network ensuring that oxygen supply meets the changing demand of the brain tissue, especially in situations of hypoxia. Brain capillaries are formed by endothelial cells and perivascular pericytes, whereby pericytes in the brain have a particularly high 1:1 ratio to endothelial cells. Pericytes not only have a key location at the blood/brain interface, they also have multiple functions, for example, they maintain blood-brain barrier integrity, play an important role in angiogenesis and have large secretory abilities. This review is specifically focused on both the cellular and the molecular responses of brain pericytes to hypoxia. We discuss the immediate early molecular responses in pericytes, highlighting four transcription factors involved in regulating the majority of transcripts that change between hypoxic and normoxic pericytes and their potential functions. Whilst many hypoxic responses are controlled by hypoxia-inducible factors (HIF), we specifically focus on the role and functional implications of the regulator of G-protein signaling 5 (RGS5) in pericytes, a hypoxia-sensing protein that is regulated independently of HIF. Finally, we describe potential molecular targets of RGS5 in pericytes. These molecular events together contribute to the pericyte response to hypoxia, regulating survival, metabolism, inflammation and induction of angiogenesis.


Subject(s)
Endothelial Cells , Pericytes , Humans , Pericytes/metabolism , Endothelial Cells/metabolism , Brain/metabolism , Hypoxia/metabolism , Blood-Brain Barrier/metabolism , Oxygen/metabolism
5.
J Neurosci Res ; 98(5): 826-842, 2020 05.
Article in English | MEDLINE | ID: mdl-31758600

ABSTRACT

Scar formation after injury of the brain or spinal cord is a common event. While glial scar formation by astrocytes has been extensively studied, much less is known about the fibrotic scar, in particular after stroke. Platelet-derived growth factor receptor ß-expressing (PDGFRß+ ) pericytes have been suggested as a source of the fibrotic scar depositing fibrous extracellular matrix (ECM) proteins after detaching from the vessel wall. However, to what extent these parenchymal PDGFRß+ cells contribute to the fibrotic scar and whether targeting these cells affects fibrotic scar formation in stroke is still unclear. Here, we utilize male transgenic mice that after a permanent middle cerebral artery occlusion stroke model have a shift from a parenchymal to a perivascular location of PDGFRß+ cells due to the loss of regulator of G-protein signaling 5 in pericytes. We find that only a small fraction of parenchymal PDGFRß+ cells co-label with type I collagen and fibronectin. Consequently, a reduction in parenchymal PDGFRß+ cells by ca. 50% did not affect the overall type I collagen or fibronectin deposition after stroke. The redistribution of PDGFRß+ cells to a perivascular location, however, resulted in a reduced thickening of the vascular basement membrane and changed the temporal dynamics of glial scar maturation after stroke. We demonstrate that parenchymal PDGFRß+ cells are not the main contributor to the fibrotic ECM, and therefore targeting these cells might not impact on fibrotic scar formation after stroke.


Subject(s)
Brain/pathology , Extracellular Matrix/pathology , Gliosis/pathology , Pericytes/pathology , Stroke/pathology , Animals , Brain/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Gliosis/metabolism , Male , Mice , Pericytes/metabolism , Stroke/metabolism
6.
J Pediatr Nurs ; 38: e24-e29, 2018.
Article in English | MEDLINE | ID: mdl-28939000

ABSTRACT

PURPOSE: The induction of anesthesia is known to be the most stressful part of the day of surgery for the child. Non-pharmacologic intervention is a field of great interest. The aims of this prospective randomized study were to evaluate if (1). A preoperative visit to the operating theatre would attenuate the anxiety of the child on the day of surgery. (2). A preoperative visit to the operating theatre would attenuate the anxiety of the parent on the day of surgery. DESIGN AND METHODS: Children aged 3-12years and their parents were randomly assigned to the intervention group visiting the operating theatre before surgery and the control group, which never visited there. Anxiety of the children in the preoperative period was measured by using the Swedish version of the modified Yale Preoperative Anxiety Scale (m-YPAS). Parent anxiety was measured by the State-Trait Anxiety Inventory (STAI) instrument. RESULTS: Both the children and their parents showed an increase in anxiety during the day of surgery up to the induction of anesthesia. Children in the intervention group showed no reduction in anxiety compared to the control group. There were no differences in anxiety between the parents in the intervention and the control groups. CONCLUSIONS: Though a preoperative visit to the surgery department and extensive information and therapeutic play does not seem to decrease the anxiety of the children scheduled for surgery and their parents it might be very important as information is highly wanted. Non-pharmacological interventions still need investigation in larger studies.


Subject(s)
Anxiety/prevention & control , Patient Education as Topic/methods , Stress, Psychological/prevention & control , Surgical Procedures, Operative/psychology , Adaptation, Psychological , Adult , Age Factors , Analysis of Variance , Child , Child, Preschool , Female , Humans , Male , Operating Rooms , Parents/psychology , Preoperative Period , Prospective Studies , Reference Values , Risk Assessment , Sex Factors , Statistics, Nonparametric , Stress, Psychological/etiology , Surgical Procedures, Operative/methods , Sweden , Treatment Outcome
7.
J Neurosci ; 33(35): 14231-45, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23986257

ABSTRACT

Glioblastoma multiforme (GBM) is the most aggressive form of brain tumor. In general, tumor growth requires disruption of the tissue microenvironment, yet how this affects glioma progression is unknown. We studied program death-ligand (PD-L)1 in neurons and gliomas in tumors from GBM patients and associated the findings with clinical outcome. Remarkably, we found that upregulation of PD-L1 by neurons in tumor-adjacent brain tissue (TABT) associated positively with GBM patient survival, whereas lack of neuronal PD-L1 expression was associated with high PD-L1 in tumors and unfavorable prognosis. To understand the molecular mechanism of PD-L1 signaling in neurons, we investigated PD-L1 function in cerebellar and cortical neurons and its impact on gliomas. We discovered that neuronal PD-L1-induced caspase-dependent apoptosis of glioma cells. Because interferon (IFN)-ß induces PD-L1 expression, we studied the functional consequences of neuronal Ifnb gene deletion on PD-L1 signaling and function. Ifnb-/- neurons lacked PD-L1 and were defective in inducing glioma cell death; this effect was reversed on PD-L1 gene transfection. Ifnb-/- mice with intracerebral isografts survived poorly. Similar to the observations in GBM patients, better survival in wild-type mice was associated with high neuronal PD-L1 in TABT and downregulation of PD-L1 in tumors, which was defective in Ifnb-/- mice. Our data indicated that neuronal PD-L1 signaling in brain cells was important for GBM patient survival. Reciprocal PD-L1 regulation in TABT and tumor tissue could be a prognostic biomarker for GBM. Understanding the complex interactions between tumor and adjacent stromal tissue is important in designing targeted GBM therapies.


Subject(s)
B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neurons/metabolism , Adult , Aged , Animals , Apoptosis , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnosis , Cerebellum/pathology , Cerebral Cortex/pathology , Female , Gene Deletion , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnosis , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Male , Mice , Middle Aged , Prognosis
8.
Cytokine ; 64(1): 112-21, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23972727

ABSTRACT

Interleukin-4 (IL-4) is a potent antiinflammatory cytokine. However its use in the clinic is hampered by side effects. We here describe the identification of a novel synthetic peptide, termed Ph8, derived from α-helix C of IL-4, which interacts with IL-4 receptor α (IL-4Rα). Employing various cultured genetically engineered cell lines and primary lymphocytes, surface plasmon resonance, qPCR, ELISA and immunoblotting techniques we found that Ph8 bound IL-4Rα and mimicked the anti-inflammatory effects of IL-4 by inhibiting TNF-α production by macrophages in vitro. It induced phosphorylation of STAT6 65kD but inhibited phosphorylation of STAT6 110 kD induced by IL-4 in a B-cell line that expressed the type I receptor. It also inhibited the IL-4-stimulated expression of a STAT6-inducible reporter gene in cells that expressed the type II receptor. Ph8 inhibited the proliferation of Th1/2 cells and downregulated the production of IFN-γ in stimulated Th1 cells. Moreover, Ph8 did not induce any shift in Th1/Th2 profile. This is a favorable effect and it is indicating that Ph8 could block general T cell activation and inflammatory responses without further inducing the side effects generally associated with IL-4 signaling. These data collectively show that Ph8 is only a partial agonist of IL-4 mimicking its desirable properties. In agreement, Ph8 treatment of rats with collagen-induced arthritis, a Th1- and antibody- mediated disease of joint, delayed the manifestation of chronic inflammation and reduced acute inflammation in carrageenan-induced edema. Our findings indicate that Ph8 is a promising potential drug candidate for the treatment of inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Interleukin-4 Receptor alpha Subunit/metabolism , Interleukin-4/pharmacology , Peptide Fragments/pharmacology , Animals , Arthritis, Experimental/drug therapy , Cell Proliferation/drug effects , Edema/drug therapy , HEK293 Cells , Humans , Interferon-gamma/metabolism , Interleukin-4/analogs & derivatives , Interleukin-4/chemistry , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Phosphorylation/drug effects , Protein Binding , Rats , Rats, Wistar , STAT6 Transcription Factor/metabolism , Th1 Cells/drug effects , Th1 Cells/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
9.
Transl Stroke Res ; 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37378751

ABSTRACT

The current treatment options for ischemic stroke aim to achieve reperfusion but are time critical. Novel therapeutic approaches that can be given beyond the limited time window of 3-4.5 h are still an unmet need to be addressed to improve stroke outcomes. The lack of oxygen and glucose in the area of ischemic injury initiates a pathological cascade leading to blood-brain barrier (BBB) breakdown, inflammation, and neuronal cell death, a process that may be intercepted to limit stroke progression. Pericytes located at the blood/brain interface are one of the first responders to hypoxia in stroke and therefore a potential target cell for early stroke interventions. Using single-cell RNA sequencing in a mouse model of permanent middle cerebral artery occlusion, we investigated the temporal differences in transcriptomic signatures in pericytes at 1, 12, and 24 h after stroke. Our results reveal a stroke-specific subcluster of pericytes that is present at 12 and 24 h and characterized by the upregulation of genes mainly related to cytokine signaling and immune response. This study identifies temporal transcriptional changes in the acute phase of ischemic stroke that reflect the early response of pericytes to the ischemic insult and its secondary consequences and may constitute potential future therapeutic targets.

10.
Biol Open ; 11(10)2022 10 15.
Article in English | MEDLINE | ID: mdl-36111549

ABSTRACT

Adaptive biological mechanisms to hypoxia are crucial to maintain oxygen homeostasis, especially in the brain. Pericytes, cells uniquely positioned at the blood-brain interface, respond fast to hypoxia by expressing regulator of G-protein signalling 5 (RGS5), a negative regulator of G-protein-coupled receptors. RGS5 expression in pericytes is observed in pathological hypoxic environments (e.g. tumours and ischaemic stroke) and associated with perivascular depletion of pericytes and vessel leakage. However, the regulation of RGS5 expression and its functional role in pericytes are not known. We demonstrate that RGS5 acts as a hypoxia-responsive protein in human brain pericytes that is regulated independent of hypoxia inducible factor-1α (HIF-1α), rapidly stabilized under hypoxia, but degraded under normoxic conditions. We show that RGS5 expression desensitizes pericytes to signalling of platelet-derived growth factor-BB (PDGFBB) and sphingosine 1-phosphate (S1P), and blocks chemokinesis or chemotaxis induced by these factors. Our data imply a role for RGS5 in antagonizing pericyte recruitment and retention to blood vessels during hypoxia and support RGS5 as a target in counteracting vessel leakage under pathological hypoxic conditions. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Brain Ischemia , Pericytes , RGS Proteins , Stroke , Brain/metabolism , Brain Ischemia/metabolism , Brain Ischemia/pathology , GTP-Binding Proteins/metabolism , Humans , Hypoxia/metabolism , Oxygen , Pericytes/metabolism , Pericytes/pathology , Platelet-Derived Growth Factor/metabolism , RGS Proteins/genetics , RGS Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Stroke/metabolism
11.
Sci Rep ; 11(1): 1120, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441868

ABSTRACT

The pathological hallmark of Parkinson's disease (PD) is the formation of Lewy bodies containing aggregated alpha-synuclein (α-syn). Although PD is associated with these distinct histological changes, other pathological features such as microvascular alterations have been linked to neurodegeneration. These changes need to be investigated as they create a hostile brain microenvironment and may contribute to the development and progression of the disease. We use a human α-syn overexpression mouse model that recapitulates some of the pathological features of PD in terms of progressive aggregation of human α-syn, impaired striatal dopamine fiber density, and an age-dependent motor deficit consistent with an impaired dopamine release. We demonstrate for the first time in this model a compromised blood-brain barrier integrity and dynamic changes in vessel morphology from angiogenesis at earlier stages to vascular regression at later stages. The vascular alterations are accompanied by a pathological activation of pericytes already at an early stage without changing overall pericyte density. Our data support and further extend the occurrence of vascular pathology as an important pathophysiological aspect in PD. The model used provides a powerful tool to investigate disease-modifying factors in PD in a temporal sequence that might guide the development of new treatments.


Subject(s)
Blood-Brain Barrier/physiopathology , Corpus Striatum/blood supply , Disease Models, Animal , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Pericytes/physiology , alpha-Synuclein/genetics , Aging , Animals , Blood Vessels/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Transgenic , Motor Activity , Neurons/metabolism , Neurons/pathology , Pericytes/pathology , Recombinant Fusion Proteins/metabolism , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/metabolism
12.
Life Sci Alliance ; 3(11)2020 11.
Article in English | MEDLINE | ID: mdl-32958605

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are known to contribute to immune evasion in cancer. However, the function of the human granulocytic (G)-MDSC subset during tumor progression is largely unknown, and there are no established markers for their identification in human tumor specimens. Using gene expression profiling, mass cytometry, and tumor microarrays, we here demonstrate that human G-MDSCs occur as neutrophils at distinct maturation stages, with a disease-specific profile. G-MDSCs derived from patients with metastatic breast cancer and malignant melanoma display a unique immature neutrophil profile, that is more similar to healthy donor neutrophils than to G-MDSCs from sepsis patients. Finally, we show that primary G-MDSCs from metastatic breast cancer patients co-transplanted with breast cancer cells, promote tumor growth, and affect vessel formation, leading to myeloid immune cell exclusion. Our findings reveal a role for human G-MDSC in tumor progression and have clinical implications also for targeted immunotherapy.


Subject(s)
Breast Neoplasms/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Neutrophils/metabolism , Adult , Aged , Breast Neoplasms/immunology , Female , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Granulocytes/metabolism , Granulocytes/physiology , Humans , Immunotherapy/methods , Melanoma/metabolism , Middle Aged , Myeloid Cells/metabolism , Myeloid-Derived Suppressor Cells/physiology , Neutrophils/physiology , Transcriptome/genetics
13.
PLoS One ; 13(3): e0194146, 2018.
Article in English | MEDLINE | ID: mdl-29518129

ABSTRACT

Brain pericytes are important to maintain vascular integrity of the neurovascular unit under both physiological and ischemic conditions. Ischemic stroke is known to induce an inflammatory and hypoxic response due to the lack of oxygen and glucose in the brain tissue. How this early response to ischemia is molecularly regulated in pericytes is largely unknown and may be of importance for future therapeutic targets. Here we evaluate the transcriptional responses in in vitro cultured human brain pericytes after oxygen and/or glucose deprivation. Hypoxia has been widely known to stabilise the transcription factor hypoxia inducible factor 1-alpha (HIF1α) and mediate the induction of hypoxic transcriptional programs after ischemia. However, we find that the transcription factors Jun Proto-Oncogene (c-JUN), Nuclear Factor Of Kappa Light Polypeptide Gene Enhancer In B-Cells (NFκB) and signal transducer and activator of transcription 3 (STAT3) bind genes regulated after 2hours (hs) of omitted glucose and oxygen before HIF1α. Potent HIF1α responses require 6hs of hypoxia to substantiate transcriptional regulation comparable to either c-JUN or STAT3. Phosphorylated STAT3 protein is at its highest after 5 min of oxygen and glucose (OGD) deprivation, whereas maximum HIF1α stabilisation requires 120 min. We show that STAT3 regulates angiogenic and metabolic pathways before HIF1α, suggesting that HIF1α is not the initiating trans-acting factor in the response of pericytes to ischemia.


Subject(s)
Brain/metabolism , Glucose/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oxygen/metabolism , Pericytes/metabolism , STAT3 Transcription Factor/metabolism , Transcription, Genetic , Brain/pathology , Cell Hypoxia , Humans , Pericytes/pathology , Proto-Oncogene Mas
14.
Mol Immunol ; 39(16): 1035-43, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12749910

ABSTRACT

Mice deficient for SPI-group ETS transcription factors PU.1 or SPI-B fail to generate lymphocytes or do not mount normal antibody mediated immune responses, respectively. PU.1 expression is restricted to B-, T-lymphocytes and macrophages, while SPI-B is expressed in B- and T-lymphocytes. SPI-C is an ETS transcription factor closely related to PU.1 and SPI-B, and expressed temporarily during B-cell development and in macrophages. By deletion and mutation analysis we show that the SPI-C protein has a transactivation domain located to the N-terminus, and that the transactivation activity is reduced to that of the DNA binding domain (DBD) alone when four aspartic acid residues are mutated to alanines. PU.1 and SPI-B regulate transcription from acidic domains located to the N-terminus and by recruiting the co-activator PIP to adjacent sites in a sequence specific manner. In contrast to PU.1 and PIP, SPI-C and PIP were unable to form a distinct ternary complex on the Ig lambda light chain lambda(2-4) enhancer element, suggesting that SPI-C could act both as a positive and negative transcriptional regulator during B-lymphocyte differentiation.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Transcriptional Activation , Amino Acid Sequence , Animals , Aspartic Acid/genetics , B-Lymphocytes/immunology , Binding Sites , COS Cells , Consensus Sequence , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Interferon Regulatory Factors , Macrophages/immunology , Molecular Sequence Data , Mutation , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Stem Cells/immunology , Time Factors , Trans-Activators/genetics , Transcription Factors/metabolism
15.
Gene ; 299(1-2): 271-8, 2002 Oct 16.
Article in English | MEDLINE | ID: mdl-12459275

ABSTRACT

Erythroblast transformation-specific domain (ETS) transcription factors regulate some of the critical molecular mechanisms controlling the differentiation of multipotent haematopoietic progenitor cells into effector B-lymphocytes. The SPI-group ETS-protein transcription factors PU.1 and SPI-B play essential and, although coexpressed and binding to similar DNA sequences, unique roles in B-cell differentiation in mice. Mouse SPI-C is an SPI-group ETS protein expressed temporarily during B-cell development and in macrophages. Here we present the genomic organization of the mouse SPI-C gene, and show by rapid amplification of cDNA ends (5'-RACE) analysis that transcription of the mouse SPI-C mRNA starts at a single site producing a single processed transcript. We have also isolated a cDNA clone encoding the human SPI-C homologue, which displays 65% amino acid identity to the murine protein. In addition, we show that the genomic structure of the human and mouse genes are similar, containing a 5' non-coding exon followed by five coding exons. Human SPI-C mRNA is preferentially detected in foetal and adult spleen, lymph nodes and at lower levels in bone marrow and foetal liver. Finally a phylogenetic prediction analysis of SPI-group protein sequences suggest that the SPI-C proteins form a distinct subgroup, with human SPI-C being closest related to the mouse SPI-C protein.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Profiling , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/genetics , Exons , Genes/genetics , HeLa Cells , Humans , Introns , K562 Cells , Mice , Molecular Sequence Data , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Transcription, Genetic , Tumor Cells, Cultured
16.
Nat Med ; 20(3): 272-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24531377

ABSTRACT

The defective generation or function of regulatory T (Treg) cells in autoimmune disease contributes to chronic inflammation and tissue injury. We report the identification of FoxA1 as a transcription factor in T cells that, after ectopic expression, confers suppressive properties in a newly identified Treg cell population, herein called FoxA1(+) Treg cells. FoxA1 bound to the Pdl1 promoter, inducing programmed cell death ligand 1 (Pd-l1) expression, which was essential for the FoxA1(+) Treg cells to kill activated T cells. FoxA1(+) Treg cells develop primarily in the central nervous system in response to autoimmune inflammation, have a distinct transcriptional profile and are CD4(+)FoxA1(+)CD47(+)CD69(+)PD-L1(hi)FoxP3(-). Adoptive transfer of stable FoxA1(+) Treg cells inhibited experimental autoimmune encephalomyelitis in a FoxA1-and Pd-l1-dependent manner. The development of FoxA1(+) Treg cells is induced by interferon-ß (IFN-ß) and requires T cell-intrinsic IFN-α/ß receptor (Ifnar) signaling, as the frequency of FoxA1(+) Treg cells was reduced in Ifnb(-/-) and Ifnar(-/-) mice. In individuals with relapsing-remitting multiple sclerosis, clinical response to treatment with IFN-ß was associated with an increased frequency of suppressive FoxA1(+) Treg cells in the blood. These findings suggest that FoxA1 is a lineage-specification factor that is induced by IFN-ß and supports the differentiation and suppressive function of FoxA1(+) Treg cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-alpha/metabolism , Multiple Sclerosis, Relapsing-Remitting/metabolism , T-Lymphocytes, Regulatory/cytology , Adult , Animals , Apoptosis , B7-H1 Antigen/metabolism , Cell Differentiation , Cell Lineage , Central Nervous System/metabolism , Female , Humans , Immunosuppression Therapy , Interferon-beta/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Transcription Factors/metabolism
17.
Biochem Biophys Res Commun ; 344(4): 1155-60, 2006 Jun 16.
Article in English | MEDLINE | ID: mdl-16647686

ABSTRACT

SPI-C is a novel ETS protein that is expressed in B lymphocytes. No target gene for SPI-C has so far been defined. We have performed a yeast two-hybrid screen using SPI-C as bait in order to further analyze the functional role of this orphan transcription factor. We found that SPI-C interacted specifically with the C-terminus of STAT6 in yeast. By co-immunoprecipitation in transfected COS7 cells the physical interaction between SPI-C and STAT6 was confirmed. Furthermore, this protein-protein interaction is functional since we could demonstrate that SPI-C and STAT6 stimulated IL4 induced Iepsilon transcription synergistically but only when both proteins bound to DNA. Thus, a protein interaction between SPI-C and STAT6 is the basis for a novel mechanism for regulation of IL4 induced gene expression.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , Immunoglobulin E/genetics , STAT6 Transcription Factor/metabolism , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Germ Cells/immunology , Germ Cells/metabolism , Humans , Immunoprecipitation , Interleukin-4/metabolism , Interleukin-4/pharmacology , Molecular Sequence Data , Protein Interaction Mapping , Proto-Oncogene Proteins c-ets , Transcription, Genetic/drug effects , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL