Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Toxicol Pathol ; 51(3): 135-147, 2023 04.
Article in English | MEDLINE | ID: mdl-37439009

ABSTRACT

Branaplam is a splicing modulator previously under development as a therapeutic agent for Spinal Muscular Atrophy Type 1 and Huntington's disease. Branaplam increased the levels of survival motor neuron protein in preclinical studies and was well tolerated in early clinical studies; however, peripheral neurotoxicity was observed in a preclinical safety study in juvenile dogs. The aim of this study was to determine whether serum neurofilament light chain (NfL) concentrations in dogs could serve as a monitoring biomarker for branaplam-induced peripheral neurotoxicity. A 30-week time-course investigative study in dogs treated with vehicle control (negative control), neurotoxic pyridoxine (positive control), or branaplam was conducted to assess neuropathology, nerve morphometry, electrophysiological measurements, gene expression profiles, and correlation to NfL serum concentrations. In branaplam-treated animals, a mild to moderate nerve fiber degeneration was observed in peripheral nerves correlating with increased serum NfL concentrations, but there were no observed signs or changes in electrophysiological parameters. Dogs with pyridoxine-induced peripheral axonal degeneration displayed clinical signs and electrophysiological changes in addition to elevated serum NfL. This study suggests that NfL may be useful as an exploratory biomarker to assist in detecting and monitoring treatment-related peripheral nerve injury, with or without clinical signs, associated with administration of branaplam and other compounds bearing a neurotoxic risk.


Subject(s)
Intermediate Filaments , Neurotoxicity Syndromes , Animals , Dogs , Pyridoxine , Biomarkers , Neurotoxicity Syndromes/etiology , Nerve Degeneration
2.
Nat Commun ; 13(1): 1150, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35241644

ABSTRACT

Huntington's Disease (HD) is a progressive neurodegenerative disorder caused by CAG trinucleotide repeat expansions in exon 1 of the huntingtin (HTT) gene. The mutant HTT (mHTT) protein causes neuronal dysfunction, causing progressive motor, cognitive and behavioral abnormalities. Current treatments for HD only alleviate symptoms, but cerebral spinal fluid (CSF) or central nervous system (CNS) delivery of antisense oligonucleotides (ASOs) or virus vectors expressing RNA-induced silencing (RNAi) moieties designed to induce mHTT mRNA lowering have progressed to clinical trials. Here, we present an alternative disease modifying therapy the orally available, brain penetrant small molecule branaplam. By promoting inclusion of a pseudoexon in the primary transcript, branaplam lowers mHTT protein levels in HD patient cells, in an HD mouse model and in blood samples from Spinal Muscular Atrophy (SMA) Type I patients dosed orally for SMA (NCT02268552). Our work paves the way for evaluating branaplam's utility as an  HD therapy, leveraging small molecule splicing modulators to reduce expression of dominant disease genes by driving pseudoexon inclusion.


Subject(s)
Huntington Disease , Animals , Brain/metabolism , Disease Models, Animal , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Oligonucleotides, Antisense/metabolism , Trinucleotide Repeat Expansion
3.
Biol Open ; 10(10)2021 10 15.
Article in English | MEDLINE | ID: mdl-34528068

ABSTRACT

Branaplam is a therapeutic agent currently in clinical development for the treatment of infants with type 1 spinal muscular atrophy (SMA). Since preclinical studies showed that branaplam had cell-cycle arrest effects, we sought to determine whether branaplam may affect postnatal cerebellar development and brain neurogenesis. Here, we describe a novel approach for developmental neurotoxicity testing (DNT) of a central nervous system (CNS) active drug. The effects of orally administered branaplam were evaluated in the SMA neonatal mouse model (SMNΔ7), and in juvenile Wistar Hannover rats and Beagle dogs. Histopathological examination and complementary immunohistochemical studies focused on areas of neurogenesis in the cerebellum (mice, rats, and dogs), and the subventricular zone of the striatum and dentate gyrus (rats and dogs) using antibodies directed against Ki67, phosphorylated histone H3, cleaved caspase-3, and glial fibrillary acidic protein. Additionally, image-analysis based quantification of calbindin-D28k and Ki67 was performed in rats and dogs. The patterns of cell proliferation and apoptosis, and neural migration and innervation in the cerebellum and other brain regions of active adult neurogenesis did not differ between branaplam- and control-treated animals. Quantitative image analysis did not reveal any changes in calbindin-D28k and Ki67 expression in rats and dogs. The data show that orally administered branaplam has no impact on neurogenesis in juvenile animals. Application of selected immunohistochemical stainings in combination with quantitative image analysis on a few critical areas of postnatal CNS development offer a reliable approach to assess DNT of CNS-active drug candidates in juvenile animal toxicity studies.


Subject(s)
Neurogenesis/drug effects , Pyridazines/pharmacology , Administration, Oral , Animals , Apoptosis/drug effects , Brain/drug effects , Cell Proliferation/drug effects , Cerebellum/drug effects , Disease Models, Animal , Dogs , Drug Evaluation, Preclinical , Mice , Neurons/drug effects , RNA Splicing/drug effects , Rats , Rats, Wistar , Survival of Motor Neuron 2 Protein/drug effects
4.
Front Immunol ; 10: 1340, 2019.
Article in English | MEDLINE | ID: mdl-31281311

ABSTRACT

Ofatumumab is the first, fully human, anti-CD20 monoclonal antibody in Phase 3 development for multiple sclerosis (MS). The study focused on changes in lymphocyte subsets in blood and lymphoid tissues and on potential novel biomarkers as a result of anti-CD20 antibody action in Cynomolgus monkeys treated with human equivalent doses of subcutaneous (s.c.) ofatumumab on Days 0, 7, and 14. Axillary lymph nodes (LNs) and blood samples were collected at various time points until Day 90. Lymphocyte subsets were quantified by flow cytometry, while morphological and immune cell changes were assessed by imaging mass cytometry (IMC), immunohistochemistry (IHC), in situ hybridization (ISH), and transcriptome analyses using single-cell methodology. Ofatumumab treatment resulted in a potent and rapid reduction of B cells along with a simultaneous drop in CD20+ T cell counts. At Day 21, IHC revealed B-cell depletion in the perifollicular and interfollicular area of axillary LNs, while only the core of the germinal center was depleted of CD20+CD21+ cells. By Day 62, the perifollicular and interfollicular areas were abundantly infiltrated by CD21+ B cells and this distribution returned to the baseline cytoarchitecture by Day 90. By IMC CD20+CD3+CD8+ cells could be identified at the margin of the follicles, with a similar pattern of distribution at Day 21 and 90. Single-cell transcriptomics analysis showed that ofatumumab induced reversible changes in t-distributed stochastic neighbor embedding (t-SNE) defined B-cell subsets that may serve as biomarkers for drug action. In summary, low dose s.c. ofatumumab potently depletes both B cells and CD20+ T cells but apparently spares marginal zone (MZ) B cells in the spleen and LN. These findings add to our molecular and tissue-architectural understanding of ofatumumab treatment effects on B-cell subsets.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , B-Lymphocytes , Genomics , Lymph Nodes , Lymphocyte Depletion , Mass Spectrometry , Single-Cell Analysis , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Gene Expression Profiling , In Situ Hybridization , Lymph Nodes/cytology , Lymph Nodes/immunology , Macaca fascicularis
5.
Environ Mol Mutagen ; 60(1): 4-16, 2019 01.
Article in English | MEDLINE | ID: mdl-30307065

ABSTRACT

The phosphorylation of histone H2AX in Serine 139 (gamma-H2AX) marks regions of DNA double strand breaks and contributes to the recruitment of DNA repair factors to the site of DNA damage. Gamma-H2AX is used widely as DNA damage marker in vitro, but its use for genotoxicity assessment in vivo has not been extensively investigated. Here, we developed an image analysis system for the precise quantification of the gamma-H2AX signal, which we used to monitor DNA damage in animals treated with known genotoxicants (EMS, ENU and doxorubicin). To compare this new assay to a validated standard procedure for DNA damage quantification, tissues from the same animals were also analyzed in the comet assay. An increase in the levels of gamma-H2AX was observed in most of the tissues from animals treated with doxorubicin and ENU. Interestingly, the lesions induced by doxorubicin were not easily detected by the standard comet assay, while they were clearly identified by gamma-H2AX staining. Conversely, EMS appeared strongly positive in the comet assay but only mildly in the gamma-H2AX immunofluorescence. These observations suggest that the two methods could complement each other for DNA damage analysis, where gamma-H2AX staining allows the detection of tissue-specific effects in situ. Moreover, since gamma-H2AX staining can be performed on formalin-fixed and paraffin-embedded tissue sections generated during repeated-dose toxicity studies, it does not require any further treatments or extra procedures during dissection, thus optimizing the use of resources and animals. Environ. Mol. Mutagen. 60:4-16, 2019. © 2018 Wiley Periodicals, Inc.


Subject(s)
DNA Breaks, Double-Stranded/drug effects , Doxorubicin/toxicity , Ethyl Methanesulfonate/toxicity , Ethylnitrosourea/toxicity , Histones/metabolism , Mutagenicity Tests/methods , Phosphoproteins/metabolism , Animals , DNA Repair/genetics , Fluorescent Antibody Technique/methods , Image Processing, Computer-Assisted , Male , Phosphorylation , Rats , Rats, Wistar
6.
Arch Toxicol ; 92(5): 1877-1891, 2018 05.
Article in English | MEDLINE | ID: mdl-29556671

ABSTRACT

A high incidence of hemangiosarcoma (HSA) was observed in mice treated for 2 years with siponimod, a sphingosine-1-phosphate receptor 1 (S1P1) functional antagonist, while no such tumors were observed in rats under the same treatment conditions. In 3-month rat (90 mg/kg/day) and 9-month mouse (25 and 75 mg/kg/day) in vivo mechanistic studies, vascular endothelial cell (VEC) activation was observed in both species, but VEC proliferation and persistent increases in circulating placental growth factor 2 (PLGF2) were only seen in the mouse. In mice, these effects were sustained over the 9-month study duration, while in rats increased mitotic gene expression was present at day 3 only and PLGF2 was induced only during the first week of treatment. In the mouse, the persistent VEC activation, mitosis induction, and PLGF2 stimulation likely led to sustained neo-angiogenesis which over life-long treatment may result in HSA formation. In rats, despite sustained VEC activation, the transient mitotic and PLGF2 stimuli did not result in the formation of HSA. In vitro, the mouse and rat primary endothelial cell cultures mirrored their respective in vivo findings for cell proliferation and PLGF2 release. Human VECs, like rat cells, were unresponsive to siponimod treatment with no proliferative response and no release of PLGF2 at all tested concentrations. Hence, it is suggested that the human cells also reproduce a lack of in vivo response to siponimod. In conclusion, the molecular mechanisms leading to siponimod-induced HSA in mice are considered species specific and likely irrelevant to humans.


Subject(s)
Azetidines/adverse effects , Benzyl Compounds/adverse effects , Endothelial Cells/drug effects , Hemangiosarcoma/chemically induced , Toxicity Tests, Chronic/methods , Administration, Oral , Animals , Azetidines/administration & dosage , Benzyl Compounds/administration & dosage , Cells, Cultured , Endothelium, Vascular/cytology , Hemangiosarcoma/genetics , Humans , Male , Mice, Inbred Strains , Placenta Growth Factor/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/metabolism , Species Specificity , Toxicokinetics , Transcriptome/drug effects
7.
J Neuroimmunol ; 318: 103-113, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29530550

ABSTRACT

Longitudinal brain atrophy quantification is a critical efficacy measurement in multiple sclerosis (MS) clinical trials and the determination of No Evidence of Disease Activity (NEDA). Utilising fingolimod as a clinically validated therapy we evaluated the use of repeated brain tissue volume measures during chronic experimental autoimmune encephalomyelitis (EAE) as a new preclinical efficacy measure. Brain volume changes were quantified using magnetic resonance imaging (MRI) at 7 Tesla and correlated to treatment-induced brain derived neurotrophic factor (BDNF) measured in blood, cerebrospinal fluid, spinal cord and brain. Serial brain MRI measurements revealed slow progressive brain volume loss in vehicle treated EAE mice despite a stable clinical score. Fingolimod (1 mg/kg) significantly ameliorated brain tissue atrophy in the cerebellum and striatum when administered from established EAE disease onwards. Fingolimod-dependent tissue preservation was associated with induction of BDNF specifically within the brain and co-localized with neuronal soma. In contrast, therapeutic teriflunomide (3 mg/kg) treatment failed to inhibit CNS autoimmune mediated brain degeneration. Finally, weekly anti-IL-17A antibody (15 mg/kg) treatment was highly efficacious and preserved whole brain, cerebellum and striatum volume. Fingolimod-mediated BDNF increases within the CNS may contribute to limiting progressive tissue loss during chronic neuroinflammation.


Subject(s)
Brain-Derived Neurotrophic Factor/drug effects , Brain/drug effects , Encephalomyelitis, Autoimmune, Experimental/pathology , Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/pharmacology , Animals , Atrophy/pathology , Brain/metabolism , Brain/pathology , Brain-Derived Neurotrophic Factor/metabolism , Crotonates/pharmacology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Hydroxybutyrates , Interleukin-17/antagonists & inhibitors , Magnetic Resonance Imaging , Mice , Mice, Inbred C57BL , Nitriles , Toluidines/pharmacology
8.
JCI Insight ; 2(5): e91127, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28289717

ABSTRACT

Retinoic-acid-orphan-receptor-C (RORC) is a master regulator of Th17 cells, which are pathogenic in several autoimmune diseases. Genetic Rorc deficiency in mice, while preventing autoimmunity, causes early lethality due to metastatic thymic T cell lymphomas. We sought to determine whether pharmacological RORC inhibition could be an effective and safe therapy for autoimmune diseases by evaluating its effects on Th17 cell functions and intrathymic T cell development. RORC inhibitors effectively inhibited Th17 differentiation and IL-17A production, and delayed-type hypersensitivity reactions. In vitro, RORC inhibitors induced apoptosis, as well as Bcl2l1 and BCL2L1 mRNA downregulation, in mouse and nonhuman primate thymocytes, respectively. Chronic, 13-week RORC inhibitor treatment in rats caused progressive thymic alterations in all analyzed rats similar to those in Rorc-deficient mice prior to T cell lymphoma development. One rat developed thymic cortical hyperplasia with preneoplastic features, including increased mitosis and reduced IKAROS expression, albeit without skewed T cell clonality. In summary, pharmacological inhibition of RORC not only blocks Th17 cell development and related cytokine production, but also recapitulates thymic aberrations seen in Rorc-deficient mice. While RORC inhibition may offer an effective therapeutic principle for Th17-mediated diseases, T cell lymphoma with chronic therapy remains an apparent risk.


Subject(s)
Receptors, Retinoic Acid/antagonists & inhibitors , Th17 Cells/cytology , Thymus Gland/pathology , Animals , Down-Regulation , Female , Gene Expression , Humans , Jurkat Cells , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Receptors, Retinoic Acid/genetics , Th17 Cells/metabolism
9.
Neurobiol Aging ; 45: 50-60, 2016 09.
Article in English | MEDLINE | ID: mdl-27459925

ABSTRACT

Currently, several immunotherapies and BACE (Beta Site APP Cleaving Enzyme) inhibitor approaches are being tested in the clinic for the treatment of Alzheimer's disease. A crucial mechanism-related safety concern is the exacerbation of microhemorrhages, which are already present in the majority of Alzheimer patients. To investigate potential safety liabilities of long-term BACE inhibitor therapy, we used aged amyloid precursor protein (APP) transgenic mice (APP23), which robustly develop cerebral amyloid angiopathy. T2*-weighted magnetic resonance imaging (MRI), a translational method applicable in preclinical and clinical studies, was used for the detection of microhemorrhages throughout the entire brain, with subsequent histological validation. Three-dimensional reconstruction based on in vivo MRI and serial Perls' stained sections demonstrated a one-to-one matching of the lesions thus allowing for their histopathological characterization. MRI detected small Perls' positive areas with a high spatial resolution. Our data demonstrate that volumetric assessment by noninvasive MRI is well suited to monitor cerebral microhemorrhages in vivo. Furthermore, 3 months treatment of aged APP23 with the potent BACE-inhibitor NB-360 did not exacerbate microhemorrhages in contrast to Aß-antibody ß1. These results substantiate the safe use of BACE inhibitors regarding microhemorrhages in long-term clinical studies for the treatment of Alzheimer's disease.


Subject(s)
Brain/diagnostic imaging , Cerebral Hemorrhage/diagnostic imaging , Diffusion Magnetic Resonance Imaging/methods , Picolinic Acids/adverse effects , Thiazines/adverse effects , Animals , Disease Progression , Female , Imaging, Three-Dimensional , Mice, Transgenic , Picolinic Acids/administration & dosage , Thiazines/administration & dosage , Time Factors
10.
Sci Rep ; 6: 21917, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26912421

ABSTRACT

Melanocytes of the hair follicle produce melanin and are essential in determining the differences in hair color. Pigment cell-specific MELanocyte Protein (PMEL17) plays a crucial role in melanogenesis. One of the critical steps is the amyloid-like functional oligomerization of PMEL17. Beta Site APP Cleaving Enzyme-2 (BACE2) and γ-secretase have been shown to be key players in generating the proteolytic fragments of PMEL17. The ß-secretase (BACE1) is responsible for the generation of amyloid-ß (Aß) fragments in the brain and is therefore proposed as a therapeutic target for Alzheimer's disease (AD). Currently BACE1 inhibitors, most of which lack selectivity over BACE2, have demonstrated efficacious reduction of amyloid-ß peptides in animals and the CSF of humans. BACE2 knock-out mice have a deficiency in PMEL17 proteolytic processing leading to impaired melanin storage and hair depigmentation. Here, we confirm BACE2-mediated inhibition of PMEL17 proteolytic processing in vitro in mouse and human melanocytes. Furthermore, we show that wildtype as well as bace2(+/-) and bace2(-/-) mice treated with a potent dual BACE1/BACE2 inhibitor NB-360 display dose-dependent appearance of irreversibly depigmented hair. Retinal pigmented epithelium showed no morphological changes. Our data demonstrates that BACE2 as well as additional BACE1 inhibition affects melanosome maturation and induces hair depigmentation in mice.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Hair/metabolism , gp100 Melanoma Antigen/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/genetics , Blotting, Western , Cell Line, Tumor , Female , Hair/drug effects , Hair/pathology , Humans , Male , Melanins/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Peptide Fragments/metabolism , Picolinic Acids/pharmacology , Pigmentation/drug effects , Prosencephalon/metabolism , Prosencephalon/pathology , Protease Inhibitors/pharmacology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Thiazines/pharmacology , Uvea/drug effects , Uvea/metabolism , Uvea/pathology , gp100 Melanoma Antigen/antagonists & inhibitors
11.
Dev Biol ; 390(2): 181-90, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24680895

ABSTRACT

Lgr4 and Lgr5 are known markers of adult and embryonic tissue stem cells in various organs. However, whether Lgr4 and Lgr5 are important for embryonic development remains unclear. To study their functions during intestinal crypt, skin and kidney development we now generated mice lacking either Lgr4 (Lgr4KO), Lgr5 (Lgr5KO) or both receptors (Lgr4/5dKO). E16.5 Lgr4KO mice displayed complete loss of Lgr5+/Olfm4+intestinal stem cells, compromised Wnt signaling and impaired proliferation and differentiation of gut epithelium. Similarly, E16.5 Lgr4KO mice showed reduced basal cell proliferation and hair follicle numbers in the developing skin, as well as dilated kidney tubules and ectatic Bowman׳s spaces. Although Lgr4KO and Lgr5KO mice both died perinatally, Lgr5 deletion did not compromise embryonic development of gut, kidney or skin. Concomitant deletion of Lgr4 and Lgr5 did not prevent perinatal lethality, in contrast to a previous report that suggested rescue of Lgr5 KO perinatal lethality by a hypomorphic Lgr4 mutant. While the double deletion did not further promote the phenotypes observed in Lgr4KO intestines, impaired kidney cell proliferation, reduced epidermal thickness, loss of Lgr5+follicular epithelium and impaired hair follicle development were only observed in Lgr4/5dKO mice. This supports complementary functions of both receptors. Our findings clearly establish the importance of Lgr4 and Lgr5 during embryonic gut, skin and kidney development, with a dominant role of Lgr4.


Subject(s)
Intestines/embryology , Kidney/embryology , Receptors, G-Protein-Coupled/physiology , Skin/embryology , Wnt Signaling Pathway/physiology , Animals , Blotting, Southern , DNA Primers/genetics , Gene Components , Genotype , Green Fluorescent Proteins , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Knockout , Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/physiology , Wnt Signaling Pathway/genetics
12.
Toxicol Sci ; 139(2): 501-11, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24690595

ABSTRACT

The constitutive androstane receptor (CAR) and the pregnane X receptor (PXR) are closely related nuclear receptors involved in drug metabolism and play important roles in the mechanism of phenobarbital (PB)-induced rodent nongenotoxic hepatocarcinogenesis. Here, we have used a humanized CAR/PXR mouse model to examine potential species differences in receptor-dependent mechanisms underlying liver tissue molecular responses to PB. Early and late transcriptomic responses to sustained PB exposure were investigated in liver tissue from double knock-out CAR and PXR (CAR(KO)-PXR(KO)), double humanized CAR and PXR (CAR(h)-PXR(h)), and wild-type C57BL/6 mice. Wild-type and CAR(h)-PXR(h) mouse livers exhibited temporally and quantitatively similar transcriptional responses during 91 days of PB exposure including the sustained induction of the xenobiotic response gene Cyp2b10, the Wnt signaling inhibitor Wisp1, and noncoding RNA biomarkers from the Dlk1-Dio3 locus. Transient induction of DNA replication (Hells, Mcm6, and Esco2) and mitotic genes (Ccnb2, Cdc20, and Cdk1) and the proliferation-related nuclear antigen Mki67 were observed with peak expression occurring between 1 and 7 days PB exposure. All these transcriptional responses were absent in CAR(KO)-PXR(KO) mouse livers and largely reversible in wild-type and CAR(h)-PXR(h) mouse livers following 91 days of PB exposure and a subsequent 4-week recovery period. Furthermore, PB-mediated upregulation of the noncoding RNA Meg3, which has recently been associated with cellular pluripotency, exhibited a similar dose response and perivenous hepatocyte-specific localization in both wild-type and CAR(h)-PXR(h) mice. Thus, mouse livers coexpressing human CAR and PXR support both the xenobiotic metabolizing and the proliferative transcriptional responses following exposure to PB.


Subject(s)
Cell Cycle/drug effects , Liver/drug effects , Phenobarbital/toxicity , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics , Transcriptome/drug effects , Animals , Cell Cycle/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Constitutive Androstane Receptor , Gene Expression Profiling , Humans , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Phenobarbital/pharmacokinetics , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Species Specificity , Xenobiotics/pharmacokinetics , Xenobiotics/toxicity
13.
PLoS One ; 9(12): e115488, 2014.
Article in English | MEDLINE | ID: mdl-25551576

ABSTRACT

Immunoglobulin-like transcript (ILT) 3 and 4 are inhibitory receptors that modulate immune responses. Their expression has been reported to be affected by interferon, offering a possible mechanism by which this cytokine exerts its therapeutic effect in multiple sclerosis, a condition thought to involve excessive immune activity. To investigate this possibility, we measured expression of ILT3 and ILT4 on immune cells from multiple sclerosis patients, and in post-mortem brain tissue. We also studied the ability of interferon beta, alone or in combination with vitamin D, to induce upregulation of these receptors in vitro, and compared expression levels between interferon-treated and untreated multiple sclerosis patients. In vitro interferon beta treatment led to a robust upregulation of ILT3 and ILT4 on monocytes, and dihydroxyvitamin D3 increased expression of ILT3 but not ILT4. ILT3 was abundant in demyelinating lesions in postmortem brain, and expression on monocytes in the cerebrospinal fluid was higher than in peripheral blood, suggesting that the central nervous system milieu induces ILT3, or that ILT3 positive monocytes preferentially enter the brain. Our data are consistent with involvement of ILT3 and ILT4 in the modulation of immune responsiveness in multiple sclerosis by both interferon and vitamin D.


Subject(s)
Interferon-beta/pharmacology , Membrane Glycoproteins/metabolism , Monocytes/drug effects , Monocytes/metabolism , Multiple Sclerosis/immunology , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Vitamin D/analogs & derivatives , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , B7 Antigens/genetics , B7 Antigens/metabolism , Drug Synergism , Humans , Interferon-beta/therapeutic use , Membrane Glycoproteins/genetics , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Receptors, Cell Surface/genetics , Receptors, Immunologic/genetics , Up-Regulation/drug effects , Vitamin D/pharmacology , Vitamin D/therapeutic use
14.
PLoS One ; 8(12): e83603, 2013.
Article in English | MEDLINE | ID: mdl-24367603

ABSTRACT

Following primary infection Herpes simplex virus-1 (HSV-1) establishes lifelong latency in the neurons of human sensory ganglia. Upon reactivation HSV-1 can cause neurological diseases such as facial palsy, vestibular neuritis or encephalitis. Certain populations of sensory neurons have been shown to be more susceptible to latent infection in the animal model, but this has not been addressed in human tissue. In the present study, trigeminal ganglion (TG) neurons expressing six neuronal marker proteins were characterized, based on staining with antibodies against the GDNF family ligand receptor Ret, the high-affinity nerve growth factor receptor TrkA, neuronal nitric oxide synthase (nNOS), the antibody RT97 against 200 kDa neurofilament, calcitonin gene-related peptide and peripherin. The frequencies of marker-positive neurons and their average neuronal sizes were assessed, with TrkA-positive (61.82%) neurons being the most abundant, and Ret-positive (26.93%) the least prevalent. Neurons positive with the antibody RT97 (1253 µm(2)) were the largest, and those stained against peripherin (884 µm(2)) were the smallest. Dual immunofluorescence revealed at least a 4.5% overlap for every tested marker combination, with overlap for the combinations TrkA/Ret, TrkA/RT97 and Ret/nNOS lower, and the overlap between Ret/CGRP being higher than would be expected by chance. With respect to latent HSV-1 infection, latency associated transcripts (LAT) were detected using in situ hybridization (ISH) in neurons expressing each of the marker proteins. In contrast to the mouse model, co-localization with neuronal markers Ret or CGRP mirrored the magnitude of these neuron populations, whereas for the other four neuronal markers fewer marker-positive cells were also LAT-ISH+. Ret and CGRP are both known to label neurons related to pain signaling.


Subject(s)
Herpesvirus 1, Human/physiology , Neurons/cytology , Neurons/virology , Trigeminal Ganglion/cytology , Adult , Aged , Animals , Biomarkers/metabolism , Cell Size , Female , Gene Expression Regulation , Herpesvirus 1, Human/metabolism , Humans , Male , Middle Aged , Protein Transport , Viral Proteins/genetics , Viral Proteins/metabolism , Young Adult
15.
PLoS One ; 8(1): e52442, 2013.
Article in English | MEDLINE | ID: mdl-23300973

ABSTRACT

MicroRNAs are short non-coding RNAs that regulate gene expression at the post-transcriptional level and play key roles in heart development and cardiovascular diseases. Here, we have characterized the expression and distribution of microRNAs across eight cardiac structures (left and right ventricles, apex, papillary muscle, septum, left and right atrium and valves) in rat, Beagle dog and cynomolgus monkey using microRNA sequencing. Conserved microRNA signatures enriched in specific heart structures across these species were identified for cardiac valve (miR-let-7c, miR-125b, miR-127, miR-199a-3p, miR-204, miR-320, miR-99b, miR-328 and miR-744) and myocardium (miR-1, miR-133b, miR-133a, miR-208b, miR-30e, miR-499-5p, miR-30e*). The relative abundance of myocardium-enriched (miR-1) and valve-enriched (miR-125b-5p and miR-204) microRNAs was confirmed using in situ hybridization. MicroRNA-mRNA interactions potentially relevant for cardiac functions were explored using anti-correlation expression analysis and microRNA target prediction algorithms. Interactions between miR-1/Timp3, miR-125b/Rbm24, miR-204/Tgfbr2 and miR-208b/Csnk2a2 were identified and experimentally investigated in human pulmonary smooth muscle cells and luciferase reporter assays. In conclusion, we have generated a high-resolution heart structure-specific mRNA/microRNA expression atlas for three mammalian species that provides a novel resource for investigating novel microRNA regulatory circuits involved in cardiac molecular physiopathology.


Subject(s)
Gene Expression Regulation , Heart/physiology , MicroRNAs/metabolism , RNA, Messenger/metabolism , Transcriptome , Animals , Cell Line , Chromosome Mapping/methods , Dogs , Female , Heart Valves/metabolism , Humans , In Situ Hybridization , Macaca fascicularis , Male , Myocardium/pathology , RNA Processing, Post-Transcriptional , Rats , Rats, Wistar , Species Specificity
16.
Toxicol Sci ; 131(2): 375-86, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23091169

ABSTRACT

The molecular events during nongenotoxic carcinogenesis and their temporal order are poorly understood but thought to include long-lasting perturbations of gene expression. Here, we have investigated the temporal sequence of molecular and pathological perturbations at early stages of phenobarbital (PB) mediated liver tumor promotion in vivo. Molecular profiling (mRNA, microRNA [miRNA], DNA methylation, and proteins) of mouse liver during 13 weeks of PB treatment revealed progressive increases in hepatic expression of long noncoding RNAs and miRNAs originating from the Dlk1-Dio3 imprinted gene cluster, a locus that has recently been associated with stem cell pluripotency in mice and various neoplasms in humans. PB induction of the Dlk1-Dio3 cluster noncoding RNA (ncRNA) Meg3 was localized to glutamine synthetase-positive hypertrophic perivenous hepatocytes, suggesting a role for ß-catenin signaling in the dysregulation of Dlk1-Dio3 ncRNAs. The carcinogenic relevance of Dlk1-Dio3 locus ncRNA induction was further supported by in vivo genetic dependence on constitutive androstane receptor and ß-catenin pathways. Our data identify Dlk1-Dio3 ncRNAs as novel candidate early biomarkers for mouse liver tumor promotion and provide new opportunities for assessing the carcinogenic potential of novel compounds.


Subject(s)
Biomarkers, Tumor/genetics , Genomic Imprinting , Intercellular Signaling Peptides and Proteins/genetics , Iodide Peroxidase/genetics , Liver Neoplasms, Experimental/genetics , Multigene Family , RNA, Untranslated/genetics , Animals , Calcium-Binding Proteins , Constitutive Androstane Receptor , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transcriptome , beta Catenin/metabolism
17.
PLoS Pathog ; 8(8): e1002852, 2012.
Article in English | MEDLINE | ID: mdl-22912575

ABSTRACT

Major human pathologies are caused by nuclear replicative viruses establishing life-long latent infection in their host. During latency the genomes of these viruses are intimately interacting with the cell nucleus environment. A hallmark of herpes simplex virus type 1 (HSV-1) latency establishment is the shutdown of lytic genes expression and the concomitant induction of the latency associated (LAT) transcripts. Although the setting up and the maintenance of the latent genetic program is most likely dependent on a subtle interplay between viral and nuclear factors, this remains uninvestigated. Combining the use of in situ fluorescent-based approaches and high-resolution microscopic analysis, we show that HSV-1 genomes adopt specific nuclear patterns in sensory neurons of latently infected mice (28 days post-inoculation, d.p.i.). Latent HSV-1 genomes display two major patterns, called "Single" and "Multiple", which associate with centromeres, and with promyelocytic leukemia nuclear bodies (PML-NBs) as viral DNA-containing PML-NBs (DCP-NBs). 3D-image reconstruction of DCP-NBs shows that PML forms a shell around viral genomes and associated Daxx and ATRX, two PML partners within PML-NBs. During latency establishment (6 d.p.i.), infected mouse TGs display, at the level of the whole TG and in individual cells, a substantial increase of PML amount consistent with the interferon-mediated antiviral role of PML. "Single" and "Multiple" patterns are reminiscent of low and high-viral genome copy-containing neurons. We show that LAT expression is significantly favored within the "Multiple" pattern, which underlines a heterogeneity of LAT expression dependent on the viral genome copy number, pattern acquisition, and association with nuclear domains. Infection of PML-knockout mice demonstrates that PML/PML-NBs are involved in virus nuclear pattern acquisition, and negatively regulate the expression of the LAT. This study demonstrates that nuclear domains including PML-NBs and centromeres are functionally involved in the control of HSV-1 latency, and represent a key level of host/virus interaction.


Subject(s)
Centromere/metabolism , Genetic Loci/physiology , Genome, Viral/physiology , Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/physiology , Tumor Suppressor Proteins/metabolism , Virus Latency/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Centromere/genetics , Co-Repressor Proteins , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Expression Regulation, Viral/physiology , Herpes Simplex/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Chaperones , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Rabbits , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , X-linked Nuclear Protein
18.
J Neurovirol ; 18(1): 62-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22167486

ABSTRACT

Herpes simplex virus type 1 latency in trigeminal ganglia (TG) is accompanied by a chronic immune cell infiltration. The aim of this study was to analyse the T-cell receptor ß-chain repertoire in latently HSV-1 infected human TG. Using complementarity-determining region 3 spectratyping, 74 expanded ß-chain sequences were identified in five TG. No clone appeared in more than one subject. Similar clones were present in the right and the left TG of two subjects. This indicates that these T cells are primed in the periphery and recognise the same antigen in the TG of both sides.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Trigeminal Ganglion/virology , Virus Latency/immunology , Adolescent , Adult , Aged , Amino Acid Sequence , Autopsy , CD8-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Cell Proliferation , Clone Cells/immunology , Clone Cells/virology , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Female , Herpes Simplex/immunology , Humans , Immunophenotyping , Male , Middle Aged , Molecular Sequence Data , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/immunology , Trigeminal Ganglion/immunology
19.
Hum Mol Genet ; 20(21): 4209-23, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21828077

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) cause late-onset Parkinson's disease (PD), but the underlying pathophysiological mechanisms and the normal function of this large multidomain protein remain speculative. To address the role of this protein in vivo, we generated three different LRRK2 mutant mouse lines. Mice completely lacking the LRRK2 protein (knock-out, KO) showed an early-onset (age 6 weeks) marked increase in number and size of secondary lysosomes in kidney proximal tubule cells and lamellar bodies in lung type II cells. Mice expressing a LRRK2 kinase-dead (KD) mutant from the endogenous locus displayed similar early-onset pathophysiological changes in kidney but not lung. KD mutants had dramatically reduced full-length LRRK2 protein levels in the kidney and this genetic effect was mimicked pharmacologically in wild-type mice treated with a LRRK2-selective kinase inhibitor. Knock-in (KI) mice expressing the G2019S PD-associated mutation that increases LRRK2 kinase activity showed none of the LRRK2 protein level and histopathological changes observed in KD and KO mice. The autophagy marker LC3 remained unchanged but kidney mTOR and TCS2 protein levels decreased in KD and increased in KO and KI mice. Unexpectedly, KO and KI mice suffered from diastolic hypertension opposed to normal blood pressure in KD mice. Our findings demonstrate a role for LRRK2 in kidney and lung physiology and further show that LRRK2 kinase function affects LRRK2 protein steady-state levels thereby altering putative scaffold/GTPase activity. These novel aspects of peripheral LRRK2 biology critically impact ongoing attempts to develop LRRK2 selective kinase inhibitors as therapeutics for PD.


Subject(s)
Homeostasis , Kidney/enzymology , Lung/enzymology , Protein Serine-Threonine Kinases/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/ultrastructure , Animals , Blood Pressure/drug effects , Dopamine/metabolism , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Enzyme Stability/drug effects , Homeostasis/drug effects , Kidney/pathology , Kidney/physiopathology , Kidney/ultrastructure , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiopathology , Kidney Tubules, Proximal/ultrastructure , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Lung/drug effects , Lung/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/ultrastructure , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Mutant Strains , Motor Activity , Signal Transduction/drug effects
20.
J Virol ; 85(19): 9680-5, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21795359

ABSTRACT

Herpes simplex type 1 (HSV-1) is a neurotropic virus which establishes lifelong latency in human trigeminal ganglia (TG). Currently, two nonexclusive control mechanisms of HSV-1 latency are discussed: antiviral CD8(+) T cells and viral microRNAs (miRNAs) encoded by the latency associated transcript (LAT). We investigate here to what extent these mechanisms may contribute to the maintenance of HSV-1 latency. We show that only a small proportion of LAT(+) neurons is surrounded by T cells in human TG. This indicates that viral latency in human TG might be controlled by other mechanisms such as viral miRNAs. Therefore, we assessed TG sections for the presence of HSV-1 miRNA, DNA, and mRNA by combining LAT in situ hybridization, T-cell immunohistochemistry, and single cell analysis of laser-microdissected sensory neurons. Quantitative reverse transcription-PCR (RT-PCR) revealed that LAT(+) neurons with or without surrounding T cells were always positive for HSV-1 miRNAs and DNA. Furthermore, ICP0 mRNA could rarely be detected only in LAT(+) neurons, as analyzed by single-cell RT-PCR. In contrast, in LAT(-) neurons that were surrounded by T cells, neither miRNAs nor the DNA of HSV-1, HSV-2, or varicella-zoster virus could be detected. These data indicate that the majority of LAT(+) neurons is not directly controlled by T cells. However, miRNA expression in every latently infected neuron would provide an additional checkpoint before viral replication is initiated.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Viral , Herpesvirus 1, Human/pathogenicity , MicroRNAs/biosynthesis , T-Lymphocytes/immunology , Trigeminal Ganglion/virology , Virus Latency , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Herpesvirus 1, Human/growth & development , Humans , Immunohistochemistry , Infant , Male , Middle Aged , RNA, Viral/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Trigeminal Ganglion/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...