Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Invest Dermatol ; 144(2): 378-386.e2, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37633457

ABSTRACT

Wound healing is a complex process involving phases of hemostasis, inflammation, proliferation, and remodeling. The regenerative process in the skin requires coordination between many regulators, including signaling molecules, transcription factors, and the epigenetic machinery. In this study, we show that chromatin regulators HDAC1 and LSD1, key components of the CoREST repressor complex, are upregulated in the regenerating epidermis during wound repair. We also show that corin, a synthetic dual inhibitor of the CoREST complex and HDAC1/LSD1 activities, significantly accelerates wound closure through enhanced re-epithelialization in a mouse tail wound model. Acetylated H3K9 (methylation of histone H3 at lysine 9) expression, a histone modification targeted by HDAC1, is increased in keratinocytes after topical treatment with 100 nM and 1 µM of corin. In vitro experiments demonstrate that corin promotes migration and inhibits the proliferation of human keratinocytes. Furthermore, expression levels of genes promoting keratinocyte migration, such as AREG, CD24, EPHB2, ITGAX, PTGS, SCT1, SERPINB2, SERPINE1, SLPI, SNAI2, and TWIST, increased in keratinocytes treated with corin. These data demonstrate that dual inhibition of class I histone deacetylases and LSD1 by corin may serve as a new approach for promoting wound re-epithelialization and provide a platform for further applications of corin for the treatment of chronic wounds.


Subject(s)
Re-Epithelialization , Skin , Mice , Animals , Humans , Skin/injuries , Keratinocytes/metabolism , Wound Healing/physiology , Disease Models, Animal , Histone Demethylases/genetics , Histone Demethylases/metabolism , Cell Movement
2.
J Clin Invest ; 134(6)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300709

ABSTRACT

Virtually all patients with BRAF-mutant melanoma develop resistance to MAPK inhibitors largely through nonmutational events. Although the epigenetic landscape is shown to be altered in therapy-resistant melanomas and other cancers, a specific targetable epigenetic mechanism has not been validated. Here, we evaluated the corepressor for element 1-silencing transcription factor (CoREST) epigenetic repressor complex and the recently developed bivalent inhibitor corin within the context of melanoma phenotype plasticity and therapeutic resistance. We found that CoREST was a critical mediator of the major distinct melanoma phenotypes and that corin treatment of melanoma cells led to phenotype reprogramming. Global assessment of transcript and chromatin changes conferred by corin revealed specific effects on histone marks connected to epithelial-mesenchymal transition-associated (EMT-associated) transcription factors and the dual-specificity phosphatases (DUSPs). Remarkably, treatment of BRAF inhibitor-resistant (BRAFi-R) melanomas with corin promoted resensitization to BRAFi therapy. DUSP1 was consistently downregulated in BRAFi-R melanomas, which was reversed by corin treatment and associated with inhibition of p38 MAPK activity and resensitization to BRAFi therapies. Moreover, this activity was recapitulated by the p38 MAPK inhibitor BIRB 796. These findings identify the CoREST repressor complex as a central mediator of melanoma phenotype plasticity and resistance to targeted therapy and suggest that CoREST inhibitors may prove beneficial for patients with BRAFi-resistant melanoma.


Subject(s)
Melanoma , Humans , Melanoma/drug therapy , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , Co-Repressor Proteins/genetics , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Phenotype , p38 Mitogen-Activated Protein Kinases
3.
J Neurosci ; 32(19): 6651-64, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22573687

ABSTRACT

Differentiation of oligodendrocyte progenitor cells (OPCs) into mature oligodendrocytes is regulated by the interplay between extrinsic signals and intrinsic epigenetic determinants. In this study, we analyze the effect that the extracellular ligands sonic hedgehog (Shh) and bone morphogenetic protein 4 (BMP4), have on histone acetylation and gene expression in cultured OPCs. Shh treatment favored the progression toward oligodendrocytes by decreasing histone acetylation and inducing peripheral chromatin condensation. BMP4 treatment, in contrast, inhibited the progression toward oligodendrocytes and favored astrogliogenesis by favoring global histone acetylation and retaining euchromatin. Pharmacological treatment or silencing of histone deacetylase 1 (Hdac1) or histone deacetylase 2 (Hdac2) in OPCs did not affect BMP4-dependent astrogliogenesis, while it prevented Shh-induced oligodendrocyte differentiation and favored the expression of astrocytic genes. Transcriptional profiling of treated OPCs, revealed that BMP4-inhibition of oligodendrocyte differentiation was accompanied by increased levels of Wnt (Tbx3) and Notch-target genes (Jag1, Hes1, Hes5, Hey1, and Hey2), decreased recruitment of Hdac and increased histone acetylation at these loci. Similar upregulation of Notch-target genes and increased histone acetylation were observed in the corpus callosum of mice infused with BMP4 during cuprizone-induced demyelination. We conclude that Shh and Bmp4 differentially regulate histone acetylation and chromatin structure in OPCs and that BMP4 acts as a potent inducer of gene expression, including Notch and Wnt target genes, thereby enhancing the crosstalk among signaling pathways that are known to inhibit myelination and repair.


Subject(s)
Bone Morphogenetic Protein 4/physiology , Hedgehog Proteins/physiology , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Histones/metabolism , Oligodendroglia/physiology , Transcriptome/genetics , Acetylation , Animals , Animals, Newborn , Cells, Cultured , Female , Gene Silencing , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/genetics , Histones/antagonists & inhibitors , Histones/genetics , Mice , Mice, Inbred C57BL , Oligodendroglia/metabolism , Rats
4.
Epigenomics ; 15(3): 167-187, 2023 02.
Article in English | MEDLINE | ID: mdl-37020393

ABSTRACT

Epigenetics encompasses heritable, reversible gene expression patterns that do not arise from mutations in genomic DNA but, rather, are regulated by DNA methylation, histone modifications, RNA modifications and ncRNAs; and epigenetic dysregulation is increasingly recognized as a mechanism of neoplastic disease progression as well as resistance to cancer therapy. This review article focuses on epigenetic modifications implicated in the progression and therapeutic resistance of common cutaneous malignancies, including basal cell carcinoma, squamous cell carcinoma, T-cell lymphoma and malignant melanoma, with an emphasis on therapeutic strategies that may be used to target such disease-associated alterations.


Epigenetics involves the study of how genes can be turned on or off by factors that affect how these genes are packaged and regulated. In cancer, there are often epigenetic changes that contribute to the formation of tumors. Many of these epigenetic changes, some of which can be passed down through generations, increase the risk of skin cancers such as basal cell carcinoma, squamous cell carcinoma, T-cell lymphoma and malignant melanoma. Emerging therapies designed to target these epigenetic changes may be effective treatments for these types of skin cancers. Researchers are currently investigating how to best use these therapies to help the ever-increasing number of people with skin cancer.


Subject(s)
Carcinoma, Basal Cell , Melanoma , Skin Neoplasms , Humans , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Skin Neoplasms/pathology , Melanoma/genetics , Epigenesis, Genetic , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , DNA Methylation
5.
medRxiv ; 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37333415

ABSTRACT

Background: To date, it is still controversial whether tau phosphorylation plays a role in Huntington's disease (HD), as previous studies demonstrated either no alterations or increases in phosphorylated tau (pTau) in HD post-mortem brain and mouse models. Objectives: The goal of this study was to determine whether total tau and pTau levels are altered in HD. Methods: Immunohistochemistry, cellular fractionations, and western blots were used to measure tau and pTau levels in a large cohort of HD and control post-mortem prefrontal cortex (PFC). Furthermore, western blots were performed to assess tau, and pTau levels in HD and control isogenic embryonic stem cell (ESC)-derived cortical neurons and neuronal stem cells (NSCs). Similarly, western blots were used to assess tau and pTau in Htt Q111 and transgenic R6/2 mice. Lastly, total tau levels were assessed in HD and healthy control plasma using Quanterix Simoa assay. Results: Our results revealed that, while there was no difference in tau or pTau levels in HD PFC compared to controls, tau phosphorylated at S396 levels were increased in PFC samples from HD patients 60 years or older at time of death. Additionally, tau and pTau levels were not changed in HD ESC-derived cortical neurons and NSCs. Similarly, tau or pTau levels were not altered in Htt Q111 and transgenic R6/2 mice compared to wild-type littermates. Lastly, tau levels were not changed in plasma from a small cohort of HD patients compared to controls. Conclusion: Together these findings demonstrate that pTau-S396 levels increase significantly with age in HD PFC.

6.
J Huntingtons Dis ; 12(3): 267-281, 2023.
Article in English | MEDLINE | ID: mdl-37694372

ABSTRACT

BACKGROUND: To date, it is still controversial whether tau phosphorylation plays a role in Huntington's disease (HD), as previous studies demonstrated either no alterations or increases in phosphorylated tau (pTau) in HD postmortem brain and mouse models. OBJECTIVE: The goal of this study was to determine whether total tau and pTau levels are altered in HD. METHODS: Immunohistochemistry, cellular fractionations, and western blots were used to measure total tau and pTau levels in a large cohort of HD and control postmortem prefrontal cortex (PFC). Furthermore, western blots were performed to assess tau, and pTau levels in HD and control isogenic embryonic stem cell (ESC)-derived cortical neurons and neuronal stem cells (NSCs). Similarly, western blots were used to assess tau and pTau levels in HttQ111 and transgenic R6/2 mice. Lastly, total tau levels were assessed in HD and healthy control plasma using Quanterix Simoa assay. RESULTS: Our results revealed that, while there was no difference in total tau or pTau levels in HD PFC compared to controls, the levels of tau phosphorylated at S396 were increased in PFC samples from HD patients 60 years or older at time of death. Additionally, tau and pTau levels were not changed in HD ESC-derived cortical neurons and NSCs. Similarly, total tau or pTau levels were not altered in HttQ111 and transgenic R6/2 mice compared to wild-type littermates. Lastly, tau levels were not changed in plasma from a small cohort of HD patients compared to controls. CONCLUSIONS: Together these findings demonstrate that pTau-S396 levels increase significantly with age in HD PFC.


Subject(s)
Huntington Disease , Mice , Animals , Humans , Huntington Disease/metabolism , Phosphorylation , Serine/metabolism , Mice, Transgenic , Prefrontal Cortex/metabolism , Disease Models, Animal
7.
JID Innov ; 2(2): 100090, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35199090

ABSTRACT

This past decade has seen tremendous advances in understanding the molecular pathogenesis of melanoma and the development of novel effective therapies for melanoma. Targeted therapies and immunotherapies that extend survival of patients with advanced disease have been developed; however, the vast majority of patients experience relapse and therapeutic resistance over time. Moreover, cellular plasticity has been demonstrated to be a driver of therapeutic resistance mechanisms in melanoma and other cancers, largely functioning through epigenetic mechanisms, suggesting that targeting of the cancer epigenetic landscape may prove a worthwhile endeavor to ensure durable treatment responses and cures. Here, we review the epigenetic alterations that characterize melanoma development, progression, and resistance to targeted therapies as well as epigenetic therapies currently in use and under development for melanoma and other cancers. We further assess the landscape of epigenetic therapies in clinical trials for melanoma and provide a framework for future advances in epigenetic therapies to circumvent the development of therapeutic resistance in melanoma.

8.
Clin Rev Allergy Immunol ; 63(3): 447-471, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36346551

ABSTRACT

Epigenetics is the study of heritable, reversible gene expression patterns that do not originate from alterations in the DNA sequence. Epigenetic modifications influence gene expression patterns and include DNA methylation, histone modifications, and gene regulation via non-coding RNAs. While the study of epigenetics has been most broadly applied to neoplastic diseases, the role of the epigenome in a wide range of disease processes including autoimmune, allergic, and inflammatory processes is increasingly being recognized. Recent advances in the study of the epigenome have led to novel insights into the pathogenesis and potential therapeutic targets of various pathologic entities including inflammatory diseases. In this review, we examine the nature of epigenetic modifications in several well-studied autoimmune, allergic, and/or inflammatory disorders of the skin including systemic lupus erythematosus, vitiligo, systemic sclerosis, alopecia areata, pemphigus, psoriasis, atopic dermatitis, keloidal scarring, and hidradenitis suppurativa with the aim to determine how such epigenetic changes may be targeted for therapeutic benefit.


Subject(s)
Alopecia Areata , Psoriasis , Humans , Epigenomics , Epigenesis, Genetic , Skin , Psoriasis/genetics
9.
BME Front ; 2021: 9860123, 2021.
Article in English | MEDLINE | ID: mdl-37849907

ABSTRACT

Objective and Impact Statement. Molecular signatures are needed for early diagnosis and improved treatment of metastatic melanoma. By high-resolution multimodal chemical imaging of human melanoma samples, we identify a metabolic reprogramming from pigmentation to lipid droplet (LD) accumulation in metastatic melanoma. Introduction. Metabolic plasticity promotes cancer survival and metastasis, which promises to serve as a prognostic marker and/or therapeutic target. However, identifying metabolic alterations has been challenged by difficulties in mapping localized metabolites with high spatial resolution. Methods. We developed a multimodal stimulated Raman scattering and pump-probe imaging platform. By time-domain measurement and phasor analysis, our platform allows simultaneous mapping of lipids and pigments at a subcellular level. Furthermore, we identify the sources of these metabolic signatures by tracking deuterium metabolites at a subcellular level. By validation with mass spectrometry, a specific fatty acid desaturase pathway was identified. Results. We identified metabolic reprogramming from a pigment-containing phenotype in low-grade melanoma to an LD-rich phenotype in metastatic melanoma. The LDs contain high levels of cholesteryl ester and unsaturated fatty acids. Elevated fatty acid uptake, but not de novo lipogenesis, contributes to the LD-rich phenotype. Monounsaturated sapienate, mediated by FADS2, is identified as an essential fatty acid that promotes cancer migration. Blocking such metabolic signatures effectively suppresses the migration capacity both in vitro and in vivo. Conclusion. By multimodal spectroscopic imaging and lipidomic analysis, the current study reveals lipid accumulation, mediated by fatty acid uptake, as a metabolic signature that can be harnessed for early diagnosis and improved treatment of metastatic melanoma.

10.
Glia ; 57(11): 1204-15, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19170181

ABSTRACT

Multiple sclerosis (MS) is a demyelinating autoimmune disease characterized by infiltration of T cells into the central nervous system (CNS) after compromise of the blood-brain barrier. A model used to mimic the disease in mice is experimental autoimmune encephalomyelitis (EAE). In this report, we examine the clinical and histopathological course of EAE in eNOS-deficient (eNOS-/-) mice to determine the role of nitric oxide (NO) derived from this enzyme in the disease progression. We find that eNOS-/- mice exhibit a delayed onset of EAE that correlates with delayed BBB breakdown, thus suggesting that NO production by eNOS underlies the T cell infiltration into the CNS. However, the eNOS-/- mice also eventually exhibit more severe EAE and delayed recovery, indicating that NO undertakes dual roles in MS/EAE, one proinflammatory that triggers disease onset, and the other neuroprotective that promotes recovery from disease exacerbation events.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Multiple Sclerosis/physiopathology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide/metabolism , Animals , Blood-Brain Barrier , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/physiology , Myelin Sheath/physiology , Severity of Illness Index , T-Lymphocytes/physiology , Time Factors
11.
Cancer Res ; 79(10): 2649-2661, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30910803

ABSTRACT

Histone modifications, largely regulated by histone acetyltransferases (HAT) and histone deacetylases, have been recognized as major regulatory mechanisms governing human diseases, including cancer. Despite significant effort and recent advances, the mechanism by which the HAT and transcriptional coactivator p300 mediates tumorigenesis remains unclear. Here, we use a genetic and chemical approach to identify the microphthalmia-associated transcription factor (MITF) as a critical downstream target of p300 driving human melanoma growth. Direct transcriptional control of MITF by p300-dependent histone acetylation within proximal gene regulatory regions was coupled to cellular proliferation, suggesting a significant growth regulatory axis. Further analysis revealed forkhead box M1 (FOXM1) as a key effector of the p300-MITF axis driving cell growth that is selectively activated in human melanomas. Targeted chemical inhibition of p300 acetyltransferase activity using a potent and selective catalytic p300/CBP inhibitor demonstrated significant growth inhibitory effects in melanoma cells expressing high levels of MITF. Collectively, these data confirm the critical role of the p300-MITF-FOXM1 axis in melanoma and support p300 as a promising novel epigenetic therapeutic target in human melanoma. SIGNIFICANCE: These results show that MITF is a major downstream target of p300 in human melanoma whose expression is predictive of melanoma response to small-molecule inhibition of p300 HAT activity.


Subject(s)
E1A-Associated p300 Protein/genetics , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , Acetylation , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cellular Senescence/genetics , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic/genetics , Histone Acetyltransferases/genetics , Humans
12.
Cancer Cell ; 36(5): 528-544.e10, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31631026

ABSTRACT

H3K27M mutations resulting in epigenetic dysfunction are frequently observed in diffuse intrinsic pontine glioma (DIPGs), an incurable pediatric cancer. We conduct a CRISPR screen revealing that knockout of KDM1A encoding lysine-specific demethylase 1 (LSD1) sensitizes DIPG cells to histone deacetylase (HDAC) inhibitors. Consistently, Corin, a bifunctional inhibitor of HDACs and LSD1, potently inhibits DIPG growth in vitro and in xenografts. Mechanistically, Corin increases H3K27me3 levels suppressed by H3K27M histones, and simultaneously increases HDAC-targeted H3K27ac and LSD1-targeted H3K4me1 at differentiation-associated genes. Corin treatment induces cell death, cell-cycle arrest, and a cellular differentiation phenotype and drives transcriptional changes correlating with increased survival time in DIPG patients. These data suggest a strategy for treating DIPG by simultaneously inhibiting LSD1 and HDACs.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Stem Neoplasms/drug therapy , Glioma/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/mortality , Brain Stem Neoplasms/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Chromatin/metabolism , DNA Methylation/drug effects , DNA Methylation/genetics , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glioma/genetics , Glioma/mortality , Glioma/pathology , Histone Code/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/metabolism , Humans , Mice , Mutation , Pons/pathology , RNA-Seq , Xenograft Model Antitumor Assays
13.
Mol Diagn Ther ; 22(2): 203-218, 2018 04.
Article in English | MEDLINE | ID: mdl-29411301

ABSTRACT

Earlier identification of aggressive melanoma remains a goal in the field of melanoma research. With new targeted and immune therapies that have revolutionized the care of patients with melanoma, the ability to predict progression and monitor or predict response to therapy has become the new focus of research into biomarkers in melanoma. In this review, promising biomarkers are highlighted. These biomarkers have been used to diagnose melanoma as well as predict progression to advanced disease and response to therapy. The biomarkers take various forms, including protein expression at the level of tissue, genetic mutations of cancer cells, and detection of circulating DNA. First, a brief description is provided about the conventional tissue markers used to stage melanoma, including tumor depth. Next, protein biomarkers, which provide both diagnostic and prognostic information, are described. This is followed by a discussion of important genetic mutations, microRNA, and epigenetic modifications that can provide therapeutic and prognostic material. Finally, emerging serologic biomarkers are reviewed, including circulating melanoma cells and exosomes. Overall the goal is to identify biomarkers that aid in the earlier identification and improved treatment of aggressive melanoma.


Subject(s)
Biomarkers, Tumor/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Biomarkers, Tumor/blood , Humans , Melanoma/diagnosis , Melanoma/genetics , Melanoma/pathology , Molecular Targeted Therapy , Prognosis , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Treatment Outcome , Melanoma, Cutaneous Malignant
14.
Nat Commun ; 9(1): 53, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29302039

ABSTRACT

Here we report corin, a synthetic hybrid agent derived from the class I HDAC inhibitor (entinostat) and an LSD1 inhibitor (tranylcypromine analog). Enzymologic analysis reveals that corin potently targets the CoREST complex and shows more sustained inhibition of CoREST complex HDAC activity compared with entinostat. Cell-based experiments demonstrate that corin exhibits a superior anti-proliferative profile against several melanoma lines and cutaneous squamous cell carcinoma lines compared to its parent monofunctional inhibitors but is less toxic to melanocytes and keratinocytes. CoREST knockdown, gene expression, and ChIP studies suggest that corin's favorable pharmacologic effects may rely on an intact CoREST complex. Corin was also effective in slowing tumor growth in a melanoma mouse xenograft model. These studies highlight the promise of a new class of two-pronged hybrid agents that may show preferential targeting of particular epigenetic regulatory complexes and offer unique therapeutic opportunities.


Subject(s)
Benzamides/pharmacology , Co-Repressor Proteins/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Melanoma/drug therapy , Nerve Tissue Proteins/antagonists & inhibitors , Pyridines/pharmacology , Tranylcypromine/pharmacology , Aged , Animals , Antineoplastic Agents , Cell Line, Tumor , Cell Proliferation , Co-Repressor Proteins/metabolism , Drug Design , Drug Screening Assays, Antitumor , Female , Histone Deacetylases/chemistry , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Skin Neoplasms/drug therapy , Xenograft Model Antitumor Assays
15.
BMC Immunol ; 8: 10, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-17634104

ABSTRACT

BACKGROUND: Myelin Oligodendrocyte Glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) is the most commonly used mouse model for multiple sclerosis (MS). During the of progression of EAE, microglia, the immunocompetent cells of the brain, become activated and accumulate around demyelinated lesions. Microglial activation is mediated by the extracellular protease tissue Plasminogen Activator (tPA), and mice lacking tPA display altered EAE progression. In this study, we have used pharmacological inhibitors and stimulators of microglial/macrophage activation to examine the temporal requirement for microglial activation in EAE progression and to determine whether such approaches might potentially be of therapeutic value. RESULTS: Intervention using the tripeptide macrophage/microglia inhibitory factor MIF (TKP) and the tetrapeptide macrophage/microglial stimulator tuftsin (TKPR) attenuated EAE symptoms and revealed that the timing of macrophage/microglial activation is critical for the clinical outcome of EAE. We show that the disease progression can potentially be manipulated favorably at early stages by altering the timing of microglial activation, which in turn alters the systemic immune response to favor upregulation of T helper cell 2 genes that promote recovery from EAE. CONCLUSION: Preventative and therapeutic modulation of macrophage/microglial activity significantly alters the outcome of EAE at symptomatic stages. Specific molecular targets have been identified that represent potential avenues of exploration for the treatment and prevention of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Macrophage Migration-Inhibitory Factors/administration & dosage , Macrophages/drug effects , Microglia/drug effects , Myelin Sheath/drug effects , Tuftsin/administration & dosage , Animals , Cell Proliferation , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Immunization/methods , Immunologic Factors , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Myelin Proteins , Myelin Sheath/metabolism , Myelin Sheath/pathology , Myelin-Associated Glycoprotein/chemistry , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Tuftsin/metabolism
16.
PLoS One ; 7(4): e34933, 2012.
Article in English | MEDLINE | ID: mdl-22529957

ABSTRACT

Multiple sclerosis (MS) is a demyelinating autoimmune disease mediated by infiltration of T cells into the central nervous system after compromise of the blood-brain barrier. We have previously shown that administration of tuftsin, a macrophage/microglial activator, dramatically improves the clinical course of experimental autoimmune encephalomyelitis (EAE), a well-established animal model for MS. Tuftsin administration correlates with upregulation of the immunosuppressive Helper-2 T cell (Th2) cytokine transcription factor GATA-3. We now show that tuftsin-mediated microglial activation results in shifting microglia to an anti-inflammatory phenotype. Moreover, the T cell phenotype is shifted towards immunoprotection after exposure to tuftsin-treated activated microglia; specifically, downregulation of pro-inflammatory Th1 responses is triggered in conjunction with upregulation of Th2-specific responses and expansion of immunosuppressive regulatory T cells (Tregs). Finally, tuftsin-shifted T cells, delivered into animals via adoptive transfer, reverse the pathology observed in mice with established EAE. Taken together, our findings demonstrate that tuftsin decreases the proinflammatory environment of EAE and may represent a therapeutic opportunity for treatment of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Immunologic Factors/administration & dosage , Tuftsin/administration & dosage , Adoptive Transfer , Animals , Culture Media, Conditioned , Demyelinating Diseases/immunology , Demyelinating Diseases/therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Forkhead Transcription Factors/metabolism , Immunosuppression Therapy , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Phenotype , STAT1 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL