Contact Us / Request a Quote Download Manuals
Advanced Cell Diagnostics Advanced Cell Diagnostics

Search form

Please sign in
  • Log In
  • Register
  • How to Order
  • What to Buy
0 My Cart
X

You have no items in your shopping cart.

Menu
X
  • Products +
    RNAscope™/BaseScope™/ miRNAscope™
    +
    • Assay Selection Guide
    Target Probes
    +
    • All About Probes
    • Catalog Probes
    • Probe Sets
    • New Probe Request
    Manual Assays
    +
    RNAscope™ Chromogenic
    • Overview
    • RNAscope™ 2.5 HD Assay-Brown
    • RNAscope™ 2.5 HD Assay-Red
    • RNAscope™ 2.5 HD Duplex Assay
    RNAscope™ Multiplex Fluorescent
    • Overview
    • RNAscope™ HiPlex v2 Assay
    • RNAscope™ Multiplex Fluorescent V2
    BaseScope™
    • Overview
    • BaseScope™ Assay Red
    • BaseScope™ Duplex Assay
    miRNAscope™
    • Overview
    • miRNAscope™ Assay red
    • RNAscope™ Plus smRNA-RNA Assay
    DNAscope™
    • Overview
    • DNAscope™ Duplex Assay
    Automated Assays
    +
    For Lunaphore COMET™
    • RNAscope™ HiPlex Pro for COMET™
    For Leica systems
    • Overview
    • RNAscope™ 2.5 LS Assay-Brown
    • RNAscope™ 2.5 LS Assay-Red
    • RNAscope™ 2.5 LS Duplex Assay
    • RNAscope™ Multiomic LS Assay
    • RNAscope™ 2.5 LS Fluorescent Multiplex Assay
    • RNAscope™ 2.5 LSx Reagent Kit-BROWN
    • RNAscope™ 2.5 LSx Reagent Kit-RED
    • BaseScope™ LS Reagent Kit – RED
    • miRNAscope LS Reagent Kit Red
    • RNAscope™ Plus smRNA-RNA LS Assay
    Roche DISCOVERY ULTRA system
    • Overview
    • RNAscope™ VS Universal HRP
    • RNAscope™ VS Universal AP
    • RNAscope™ VS Duplex Assay
    • BaseScope™ VS Reagent Kit – RED
    RNA-Protein Co-Detection Assay
    +
    • RNAscope HiPlex-IMC™ Co-Detection
    • Integrated Codetection Assay
    • Sequential RNA Protein Detection
    Software
    +
    • Overview
    • Aperio RNA ISH Algorithm
    • HALO® image analysis platform
    Controls & Accessories
    +
    • RNAscope™
    • BaseScope™
    • miRNAscope™
    • Accessories
    How to Order
    +
    • Ordering Instructions
    • What to Buy
  • Services +
    Professional Assay Services
    +
    • Our Services
    • Multiomic Services
    • Biomarker Assay Development
    • Cell & Gene Therapy Services
    • Clinical Assay Development
    • Tissue Bank & Sample Procurement
    • Image Analysis
    Benefits
    +
    • Your Benefits
    • Certified Providers
    How to Order
    +
    • Ordering Process
    • Contact Services
  • Areas of Research +
    Most Popular
    +
    • COVID-19 Coronavirus
    • Single Cell Analysis
    • Whole-Mount
    • Anatomic Pathology Panels
    • Neuroscience
    • Inflammation
    • Gene Therapy/AAV
    • Stem Cell
    • Immuno-oncology
    • Liver Research
    • Cardiovascular & Skeletal Muscle Research
    Cell & Gene Therapy
    +
    • Gene Therapy
    • Gene Therapy/AAV
    • siRNA/ASO
    • Cell Therapy
    Cancer
    +
    • Breast Cancer
    • EGFRvIII Splice Variant
    • HPV Related Cancer
    • Immuno-oncology
    • Lung Cancer
    • PDx
    • Prostate Cancer
    • Point Mutation
    • CDR3 for TCR
    Viral
    +
    • COVID-19 Coronavirus
    • HIV & SIV
    • Infectious Disease
    • Zika Virus
    Pathways
    +
    • AKT
    • JAK STAT
    • WNT B-Catenin
    Neuroscience
    +
    Neuroscience
    • Neural Development
    • Neuronal Cell Types
    • Learning and Memory
    • G-protein-coupled Receptors & Ion Channels
    • Post-mortem Brain Tissue
    Other
    +
    • Circular RNA
    • Gene Fusions
    • HT Transcript Validation
    • Long Non-coding RNA
    • RNAseq Validation
    • Single Cell Analysis
    • Splice Variant
    • miRNA
    RNA & Protein
    +
    • Antibody Challenges
    • Dual ISH + IHC Methods
    • No Antibodies
    • RNA & Protein Analysis
    Customer Innovations
    +
    • Dual RNA+DNA ISH
    • Very old FFPE ISH
    • Wholemount ISH
    Animal Models
    +
    • Any Species
    • Mouse Model
    • Preclincal Safety
  • Technology +
    Overview
    +
    • How it Works
    • Data Image Gallery
    • Technology Video
    • Webinars
    RNA Detection
    +
    • Why RNA?
    • RNA ISH and IHC
    Pretreatment Options
    +
    • RNAscope™ Pretreatment
    • PretreatPro™
    Spotlights
    +
    • Researchers Spotlights
    • RNA & DNA
    • WISH
    • FFPE
    • Testimonials
    Publications, Guides & Posters
    +
    • Search publications
    • RNAscope™ Reference Guide
    • RNAscope™ Data Analysis Guide
    • Download RNAscope™ Posters
  • Support +
    Overview
    +
    • Get Started
    • How to Order
    • Distributors
    • Contact Support
    Troubleshooting
    +
    • Troubleshooting Guide
    • FAQs
    • User Manuals, SDS and Product Inserts
    • Documents and Downloads
    Imaging Resource
    +
    • Image Analysis
    • Image Registration Software
    • QuPath
    • HALO® image analysis platform
    Learn More
    +
    • Webinars
    • Training Videos
  • Partners +
    Partners
    +
    • Overview
    Partners Directory
    +
    Automation Partners
    • Leica Biosystem
    • Roche Diagnostics
    Workflow Partners
    • NanoString
    Software Partners
    • indica labs
    Become a Partner
    +
    • Learn How
  • Diagnostics +
    Diagnostics
    +
    • Diagnostics
    • Literature
    • Diagnostics ASR Probes
    • Diagnostics CE-IVD Probes
    • Diagnostics CE-IVD Detection
    • Companion Diagnostics
  • Image Calendar +
    Image Calendar
    +
    • Image Contest
    • Data Image Gallery
Search

Probes for INS

ACD can configure probes for the various manual and automated assays for INS for RNAscope Assay, or for Basescope Assay compatible for your species of interest.

  • Probes for INS (0)
  • Kits & Accessories (0)
  • Support & Documents (0)
  • Publications (1522)
  • Image gallery (0)
Refine Probe List

Content for comparison

Gene

  • (-) Remove TBD filter TBD (1413)
  • Lgr5 (151) Apply Lgr5 filter
  • SARS-CoV-2 (136) Apply SARS-CoV-2 filter
  • Gad1 (90) Apply Gad1 filter
  • vGlut2 (80) Apply vGlut2 filter
  • HPV E6/E7 (78) Apply HPV E6/E7 filter
  • Slc17a6 (77) Apply Slc17a6 filter
  • Axin2 (74) Apply Axin2 filter
  • SLC32A1 (74) Apply SLC32A1 filter
  • FOS (73) Apply FOS filter
  • Sst (65) Apply Sst filter
  • (-) Remove TH filter TH (63)
  • VGAT (58) Apply VGAT filter
  • Gad2 (54) Apply Gad2 filter
  • tdTomato (54) Apply tdTomato filter
  • DRD2 (53) Apply DRD2 filter
  • Slc17a7 (52) Apply Slc17a7 filter
  • GLI1 (51) Apply GLI1 filter
  • (-) Remove PVALB filter PVALB (47)
  • egfp (46) Apply egfp filter
  • ZIKV (46) Apply ZIKV filter
  • DRD1 (42) Apply DRD1 filter
  • GFAP (39) Apply GFAP filter
  • COL1A1 (38) Apply COL1A1 filter
  • Crh (37) Apply Crh filter
  • Chat (37) Apply Chat filter
  • V-nCoV2019-S (37) Apply V-nCoV2019-S filter
  • Pomc (34) Apply Pomc filter
  • PDGFRA (33) Apply PDGFRA filter
  • Il-6 (33) Apply Il-6 filter
  • Cre (33) Apply Cre filter
  • AGRP (32) Apply AGRP filter
  • PECAM1 (32) Apply PECAM1 filter
  • Npy (32) Apply Npy filter
  • Wnt5a (31) Apply Wnt5a filter
  • CXCL10 (31) Apply CXCL10 filter
  • GLP1R (31) Apply GLP1R filter
  • Sox9 (29) Apply Sox9 filter
  • CD68 (28) Apply CD68 filter
  • Penk (28) Apply Penk filter
  • PD-L1 (28) Apply PD-L1 filter
  • ACTA2 (27) Apply ACTA2 filter
  • SHH (27) Apply SHH filter
  • VGluT1 (27) Apply VGluT1 filter
  • OLFM4 (26) Apply OLFM4 filter
  • GFP (26) Apply GFP filter
  • Rbfox3 (25) Apply Rbfox3 filter
  • MALAT1 (24) Apply MALAT1 filter
  • SOX2 (24) Apply SOX2 filter
  • Ccl2 (24) Apply Ccl2 filter

Product

  • RNAscope (227) Apply RNAscope filter
  • TBD (149) Apply TBD filter
  • RNAscope Multiplex Fluorescent Assay (73) Apply RNAscope Multiplex Fluorescent Assay filter
  • RNAscope Fluorescent Multiplex Assay (54) Apply RNAscope Fluorescent Multiplex Assay filter
  • Basescope (10) Apply Basescope filter
  • RNAscope HiPlex v2 assay (10) Apply RNAscope HiPlex v2 assay filter
  • RNAscope 2.5 HD Brown Assay (9) Apply RNAscope 2.5 HD Brown Assay filter
  • DNAscope HD Duplex Reagent Kit (8) Apply DNAscope HD Duplex Reagent Kit filter
  • RNAscope 2.5 HD Duplex (8) Apply RNAscope 2.5 HD Duplex filter
  • RNAscope Multiplex Fluorescent v2 (8) Apply RNAscope Multiplex Fluorescent v2 filter
  • RNAscope 2.5 HD Reagent Kit - BROWN (7) Apply RNAscope 2.5 HD Reagent Kit - BROWN filter
  • RNAscope 2.5 HD Red assay (4) Apply RNAscope 2.5 HD Red assay filter
  • BASEscope Assay RED (2) Apply BASEscope Assay RED filter
  • RNAscope 2.0 Assay (2) Apply RNAscope 2.0 Assay filter
  • BaseScope Duplex Assay (1) Apply BaseScope Duplex Assay filter
  • DNAscope Duplex Assay (1) Apply DNAscope Duplex Assay filter
  • miRNAscope (1) Apply miRNAscope filter
  • RNAscope 2.5 HD Assay (1) Apply RNAscope 2.5 HD Assay filter
  • RNAscope 2.5 LS Assay (1) Apply RNAscope 2.5 LS Assay filter
  • RNAScope HiPlex assay (1) Apply RNAScope HiPlex assay filter
  • RNAscope HiPlex Image Registration Software (1) Apply RNAscope HiPlex Image Registration Software filter
  • RNAscope HiPlex12 Reagents Kit (1) Apply RNAscope HiPlex12 Reagents Kit filter

Research area

  • Neuroscience (237) Apply Neuroscience filter
  • Cancer (111) Apply Cancer filter
  • Development (58) Apply Development filter
  • Other: Methods (45) Apply Other: Methods filter
  • Inflammation (32) Apply Inflammation filter
  • Infectious (18) Apply Infectious filter
  • HIV (15) Apply HIV filter
  • Pain (14) Apply Pain filter
  • Stem Cells (13) Apply Stem Cells filter
  • HPV (12) Apply HPV filter
  • Other: Neuromuscular Disorders (10) Apply Other: Neuromuscular Disorders filter
  • Other: Heart (9) Apply Other: Heart filter
  • Other: Lung (9) Apply Other: Lung filter
  • CGT (8) Apply CGT filter
  • Covid (8) Apply Covid filter
  • Other: Metabolism (8) Apply Other: Metabolism filter
  • Infectious Disease (7) Apply Infectious Disease filter
  • Metabolism (7) Apply Metabolism filter
  • Stem cell (7) Apply Stem cell filter
  • Immunotherapy (6) Apply Immunotherapy filter
  • Other: Reproduction (6) Apply Other: Reproduction filter
  • Stress (6) Apply Stress filter
  • Aging (5) Apply Aging filter
  • Endocrinology (5) Apply Endocrinology filter
  • LncRNAs (5) Apply LncRNAs filter
  • Obesity (5) Apply Obesity filter
  • Reproduction (5) Apply Reproduction filter
  • Transcriptomics (5) Apply Transcriptomics filter
  • Cystic Fibrosis (4) Apply Cystic Fibrosis filter
  • Heart (4) Apply Heart filter
  • Itch (4) Apply Itch filter
  • lncRNA (4) Apply lncRNA filter
  • Memory (4) Apply Memory filter
  • Other: Kidney (4) Apply Other: Kidney filter
  • Other: Skin (4) Apply Other: Skin filter
  • Psychiatry (4) Apply Psychiatry filter
  • Alzheimer's Disease (3) Apply Alzheimer's Disease filter
  • diabetes (3) Apply diabetes filter
  • Immunology (3) Apply Immunology filter
  • Kidney (3) Apply Kidney filter
  • Lung (3) Apply Lung filter
  • other: Aging (3) Apply other: Aging filter
  • Other: Eyes (3) Apply Other: Eyes filter
  • Other: Transcriptomics (3) Apply Other: Transcriptomics filter
  • Other: Zoological Disease (3) Apply Other: Zoological Disease filter
  • Parkinson's Disease (3) Apply Parkinson's Disease filter
  • Regeneration (3) Apply Regeneration filter
  • Reproductive Biology (3) Apply Reproductive Biology filter
  • Skin (3) Apply Skin filter
  • Tumor microenvironment (3) Apply Tumor microenvironment filter

Category

  • Publications (1522) Apply Publications filter
Activating mGlu3 metabotropic glutamate receptors rescues schizophrenia-like cognitive deficits through metaplastic adaptations within the hippocampus.

Biological Psychiatry

2021 Mar 01

Dogra, S;Stansley, B;Xiang, Z;Qian, W;Gogliotti, R;Nicoletti, F;Lindsley, C;Niswender, C;Joffe, M;Conn, P;
| DOI: 10.1016/j.biopsych.2021.02.970

Background Polymorphisms in GRM3, the gene encoding the mGlu3 metabotropic glutamate receptor, are associated with impaired cognition and neuropsychiatric disorders such as schizophrenia. Limited availability of selective genetic and molecular tools has hindered progress in developing a clear understanding of the mechanisms through which mGlu3 receptors regulate synaptic plasticity and cognition. Methods We examined associative learning in mice with trace fear conditioning, a hippocampal-dependent learning task disrupted in patients with schizophrenia. Underlying cellular mechanisms were assessed using ex vivo hippocampal slice preparations with selective pharmacological tools and selective genetic deletion of mGlu3 receptor expression in specific neuronal subpopulations. Results mGlu3 receptor activation enhanced trace fear conditioning and reversed deficits induced by subchronic phencyclidine. Mechanistic studies revealed that mGlu3 receptor activation induced metaplastic changes, biasing afferent stimulation to induce long-term potentiation through a mGlu5 receptor-dependent, endocannabinoid-mediated, disinhibitory mechanism. Selective genetic deletion of either mGlu3 or mGlu5 from hippocampal pyramidal cells eliminated effects of mGlu3 activation, revealing a novel mechanism by which mGlu3 and mGlu5 interact to enhance cognitive function. Conclusions These data demonstrate that activation of mGlu3 receptors in hippocampal pyramidal cells enhances hippocampal-dependent cognition in control and impaired mice by inducing a novel form of metaplasticity to regulate circuit function – providing a clear mechanism through which genetic variation in GRM3 can contribute to cognitive deficits. Developing approaches to positively modulate mGlu3 receptor function represents an encouraging new avenue for treating cognitive disruption in schizophrenia and other psychiatric diseases.
Endothelium-derived semaphorin 3G attenuates ischemic retinopathy by coordinating β-catenin-dependent vascular remodeling

The Journal of clinical investigation

2021 Feb 15

Chen, DY;Sun, NH;Chen, X;Gong, JJ;Yuan, ST;Hu, ZZ;Lu, NN;Körbelin, J;Fukunaga, K;Liu, QH;Lu, YM;Han, F;
PMID: 33586674 | DOI: 10.1172/JCI135296

Abnormal angiogenesis and regression of the diseased retinal vasculature are key processes associated with ischemic retinopathies, but the underlying mechanisms that regulate vascular remodeling remain poorly understood. Here, we confirmed the specific expression of semaphorin 3G (Sema3G) in retinal endothelial cells (ECs), which was required for vascular remodeling and the amelioration of ischemic retinopathy. We found that Sema3G was elevated in the vitreous fluid of patients with proliferative diabetic retinopathy (PDR) and in the neovascularization regression phase of oxygen-induced retinopathy (OIR). Endothelial-specific Sema3G knockout mice exhibited decreased vessel density and excessive matrix deposition in the retinal vasculature. Moreover, loss of Sema3G aggravated pathological angiogenesis in mice with OIR. Mechanistically, we demonstrated that HIF-2α directly regulated Sema3G transcription in ECs under hypoxia. Sema3G coordinated the functional interaction between β-catenin and VE-cadherin by increasing β-catenin stability in the endothelium through the neuropilin-2 (Nrp2)/PlexinD1 receptor. Furthermore, Sema3G supplementation enhanced healthy vascular network formation and promoted diseased vasculature regression during blood vessel remodeling. Overall, we deciphered the endothelium-derived Sema3G-dependent events involved in modulating physiological vascular remodeling and regression of pathological blood vessels for reparative vascular regeneration. Our findings shed light on the protective effect of Sema3G in ischemic retinopathies.
Epithelium-derived Indian Hedgehog restricts stromal expression of ErbB family members that drive colonic tumor cell proliferation

Oncogene

2021 Jan 21

Westendorp, F;Karpus, ON;Koelink, PJ;Vermeulen, JLM;Meisner, S;Koster, J;Büller, NVJA;Wildenberg, ME;Muncan, V;van den Brink, GR;
PMID: 33479497 | DOI: 10.1038/s41388-020-01633-0

Indian Hedgehog (Ihh) is a morphogen expressed by epithelial cells in the small intestine and colon that signals in a paracrine manner to gp38+ stromal cells. The loss of Ihh signaling results in increased epithelial proliferation, lengthening and multiplication of intestinal crypts and the activation of a stromal cell immune response. How Ihh controls epithelial proliferation through the stroma and how it affects colorectal cancer development remains poorly defined. To study the influence of Ihh signaling on the earliest stage of colorectal carcinogenesis, we used a well characterized mouse model in which both alleles of the Adenoma Polyposis Coli (Apc) gene could be inducibly deleted, leading to instant transformation of the colonic epithelium to an adenomatous phenotype. Concurrent deletion of Ihh from the adenomatous colonic epithelium of Apc inducible double mutant mice resulted in a remarkable increase in the hyperproliferative epithelial phenotype and increased accumulation of Lgr5+ stem cells. Transcriptional profiling of sorted colonic gp38+ fibroblasts showed upregulation of three ErbB pathway ligands (EREG, BTC, and NRG1) in Apc-/-Ihh-/- double mutant mice. We found that recombinant EREG, BTC, and NRG1 but not Lgr5 ligand R-Spondin promoted growth and proliferation of Apc double mutant colonic organoids. Thus, the loss of Ihh enhances Apc-driven colonic adenomagenesis via upregulation of ErbB pathway family members in colonic stromal cells. Our findings highlight the critical role of epithelium-derived Indian Hedgehog as a stromal tumor suppressor in the intestine.
Gut-licensed IFNγ+ NK cells drive LAMP1+TRAIL+ anti-inflammatory astrocytes

Nature

2021 Jan 06

Sanmarco, LM;Wheeler, MA;Gutiérrez-Vázquez, C;Polonio, CM;Linnerbauer, M;Pinho-Ribeiro, FA;Li, Z;Giovannoni, F;Batterman, KV;Scalisi, G;Zandee, SEJ;Heck, ES;Alsuwailm, M;Rosene, DL;Becher, B;Chiu, IM;Prat, A;Quintana, FJ;
PMID: 33408417 | DOI: 10.1038/s41586-020-03116-4

Astrocytes are glial cells that are abundant in the central nervous system (CNS) and that have important homeostatic and disease-promoting functions1. However, little is known about the homeostatic anti-inflammatory activities of astrocytes and their regulation. Here, using high-throughput flow cytometry screening, single-cell RNA sequencing and CRISPR-Cas9-based cell-specific in vivo genetic perturbations in mice, we identify a subset of astrocytes that expresses the lysosomal protein LAMP12 and the death receptor ligand TRAIL3. LAMP1+TRAIL+ astrocytes limit inflammation in the CNS by inducing T cell apoptosis through TRAIL-DR5 signalling. In homeostatic conditions, the expression of TRAIL in astrocytes is driven by interferon-γ (IFNγ) produced by meningeal natural killer (NK) cells, in which IFNγ expression is modulated by the gut microbiome. TRAIL expression in astrocytes is repressed by molecules produced by T cells and microglia in the context of inflammation. Altogether, we show that LAMP1+TRAIL+ astrocytes limit CNS inflammation by inducing T cell apoptosis, and that this astrocyte subset is maintained by meningeal IFNγ+ NK cells that are licensed by the microbiome.
April Stroke Literature Synopses (Preclinical)

Stroke

2023 Apr 01

Branyan, T;Sohrabji, F;
| DOI: 10.1161/STROKEAHA.123.042326

The blood-brain barrier (BBB) consists of endothelial cells, astrocytic end feet, and pericytes to form a barrier that minimizes the entry of circulating proteins and cells into the brain. However, stroke is known to cause significant damage to the BBB, causing the barrier to become permeable, which allows immune cells and other substances to be extravasated into the brain parenchyma. Preservation of the BBB is associated with better ischemic stroke outcomes; therefore, this synopsis summarizes 3 new studies that aim to characterize specific mechanisms of BBB damage and identify potential therapeutic pathways to preserve barrier integrity. CD36 (cluster of differentiation 36) is a glycoprotein expressed by monocytes and macrophages, as well as by endothelial cells. Previous studies have shown that global knockout of CD36 prevents stroke-induced damage, and Kim et al in 2023 published a study in the Journal of Cerebral Blood Flow and Metabolism titled “Endothelial Cell CD36 Mediates Stroke-Induced Brain Injury via BBB Dysfunction and Monocyte Infiltration in Normal and Obese Conditions,” in which they explore the role of CD36 specifically in endothelial cells. Conditional deletion of CD36 in endothelial cells improved stroke outcomes, as indicated by reduced infarct size and hemispheric swelling. Moreover, this deletion improved survival and motor function. Additionally, CD36 deletion in endothelial cells reduced IgG expression in the brain, indicating improved vascular integrity. There was reduced monocyte infiltration into the brain and reduced MCP-1 (monocyte chemoattractant protein-1) and CCR2 (chemokine receptor type 2) expression in the mice with endothelial cell deletion of CD36. This reduced monocyte trafficking persisted even when normalized for infarct size, suggesting that vascular integrity is maintained independent of cell loss. Intriguingly, endothelial cell-specific deletion of CD36 also made mice resistant to developing an obesity phenotype, providing a potential molecular cause for obesity as a stroke risk factor. In the Proceedings of the National Academy for Science in a publication titled “Myeloid-Derived MIF Drives RIPK1-Mediated Cerebromicrovascular Endothelial Cell Death to Exacerbate Ischemic Brain Injury,” Li et al in 2023 describe how macrophage MIF (migration inhibitory factor) exacerbates endothelial cell death and increases BBB permeability after middle cerebral artery occlusion (MCAo). By treating endothelial cells with MIF and subjecting them to oxygen-glucose deprivation followed by reoxygenation, the authors demonstrated that MIF promotes endothelial cell death specifically by activating RIPK1 (receptor-interacting protein kinase 1). Surgical trauma in both mice and humans increases circulating MIF, and the authors use a perioperative ischemic stroke model to see how this surgically induced increase in MIF affects outcomes after distal MCAo. Two-photon imaging showed that perioperative ischemic stroke mice showed increased adhesion of myeloid cells to ischemic microvascular endothelial cells, and RNAscope analysis showed that MIF expression was increased in microglia surrounding endothelial cells in perioperative ischemic stroke mice. Perioperative ischemic stroke mice also exhibited larger infarct volumes and exacerbated BBB damage. The authors then used CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9) to delete MIF in myeloid cells, which resulted in reduced expression of necrosis markers phosphorylated RIPK1, phosphorylated RIPK3, and CC3 (cleaved caspase 3) in endothelial cells. Mice with myeloid-deleted MIF showed reduced zonulin-1 loss, a marker of endothelial tight junctions, and sensorimotor deficits after distal MCAo. Peripheral blood mononuclear cells from mice with myeloid-deleted MIF were given after MCAo and resulted in smaller infarct volume and reduced IgG extravasation as compared with mice who were given peripheral blood mononuclear cells from wild types. Endothelial cells were cocultured with peripheral blood mononuclear cells from mice with myeloid-deleted MIF and wild types, and zonulin-1 expression was preserved in endothelial cocultured with peripheral blood mononuclear cells from myeloid-deleted MIF mice. Administration of MIF inhibitor before stroke reduced infarct volume, prevented IgG extravasation, preserved tight junction integrity, and prevented endothelial cell death. Collectively, these data show that myeloid-derived MIF is detrimental to BBB integrity after stroke and deleting this source of MIF can improve outcomes through preserving BBB health. Finally, Li et al in 2023 show in ACS Nano in a publication titled “Inducible Pluripotent Stem Cell-Derived Small Extracellular Vesicles Rejuvenate Senescent Blood-Brain Barrier to Protect Against Ischemic Stroke in Aged Mice” that small extracellular vesicles (sEVs) from induced pluripotent stem cells (iPSCs) are able to restore BBB function in old mice by reversing cellular senescence. Mice treated with iPSC-sEVs showed reduced senescence-associated β-galactosidase, p16, p53, p21, and γ-H2AX (histone family member X), all of which are markers associated with cellular senescence. Pretreatment with iPSC-sEVs before MCAo reduced infarct volume in the aged mice, improved neurological score, and reduced sensorimotor deficits, indicating improved stroke outcomes. Moreover, these mice showed decreased leakage of Evans blue dye into the brain parenchyma and preservation of tight junction proteins, indicating that these sEVs preserve BBB integrity after stroke. Mice treated with sEVs showed decreased immune cell infiltration after stroke and attenuated expression of tumor necrosis factor-α, IL (interleukin)-17, IL-6, and IL-1β, Moreover, sEV treatment reduced ischemia-induced apoptosis of oligodendrocytes and neurons. Reversal of the senescent phenotype of the BBB was tested in vitro, by chemically inducing senescence using D-galactose in endothelial cell cultures and subsequent treatment with iPSC-sEVs. sEV treatment reduced senescence markers and prevented loss of tight junction proteins. Oxygen-glucose deprivation was then used to mimic stroke conditions in these cultures, and sEV treatment preserved angiogenic properties of endothelial cells and reduced dextran leakage in a transwell assay. These experiments affirm the in vivo findings that sEV treatment reverses BBB senescence. The 3 studies summarized in this synopsis show 3 different potential pathways that may serve as a target for preserving BBB function after stroke. Deleting endothelial cell CD36, deleting myeloid-derived MIF, or reversing BBB senescence using iPSC-sEVs resulted in improved vascular integrity and overall better stroke outcomes.
Identification of a spontaneous mutation in Adiponectin receptor 1 in a complement factor H knockout mouse

Investigative Ophthalmology & Visual Science

2021 Jan 01

YANG, J;Banks, A;Buchanan, NM;Liao, S;Lima, E;Rangaswamy, N;Arellano, I;Hanks, S;Vrouvlianis, J;Qiu, Y;Rice, DS;

Purpose : A subset of mice in our Cfh knockout (Cfh-/-) colony exhibited rapid retinal degeneration, suggesting a spontaneous mutation occurred on mouse chromosome 1 (Chr 1). The retinal phenotype was similar to that in AdipoR1 knockout (AdipoR1-/-) mice, whose gene is located near the Cfh locus on Chr 1. We attempted to determine if a mutation in AdipoR1 occurred on the Cfh-/- background. Methods : We performed an allele complementation test with a cross between a Cfh-/- mouse with retinal degeneration and an AdipoR1-/- mouse with retinal degeneration. RNA-seq, in situ hybridization, immunohistochemistry and protein analysis were used to profile Cfh-/- and AdipoR1-/- mice. Results : About 50% of Cfh-/- mice exhibited retinal degeneration. Cfh-/- mice, regardless of retinal phenotype, demonstrated elevated complement activation in the eye compared to littermate controls. All offspring from the complementation test exhibited retinal degeneration, implying that AdipoR1 mutant alleles were responsible for the retinal degeneration. AdipoR1 protein, normally present in the RPE apical microvilli, was notably absent in Cfh-/- mice with retinal degeneration. Cfh-/- mice with normal retinal anatomy expressed AdipoR1 protein and were comparable to littermates[QY1] . Adipor1 mRNA expression levels were comparable across the colony as measured by RNAscope and RNA-seq. Analysis of AdipoR1 mRNA sequence revealed a transversion at position c.841 C>T in the Adipor1 gene concordant with mice exhibiting retinal degeneration. Further breeding identified mutant AdipoR1 mRNA in two Cfh+/- mice and one Cfh+/+ mice with early onset retinal degeneration, implying a recent cross over on Chr 1. This missense mutation results in a proline to serine conversion (P281S) in the fifth transmembrane domain of AdipoR1 and is predicted to be detrimental to the AdipoR1 protein. Conclusions : The spontaneous mutation in the AdipoR1 gene results in early onset retinal degeneration in a subset of our Cfh-/- mouse colony. Elevated complement activity in the eye does not appear to affect the course of retinal degeneration, as gene signatures are comparable across all mice with retinal degeneration, regardless of Cfh genotype.
Cellular Diversity in Human Subgenual Anterior Cingulate and Dorsolateral Prefrontal Cortex by Single-Nucleus RNA-sequencing

The Journal of neuroscience : the official journal of the Society for Neuroscience

2023 Apr 10

Kim, B;Kim, D;Schulmann, A;Patel, Y;Caban-Rivera, C;Kim, P;Jambhale, A;Johnson, KR;Feng, N;Xu, Q;Kang, SJ;Mandal, A;Kelly, M;Akula, N;McMahon, FJ;Lipska, B;Marenco, S;Auluck, PK;
PMID: 37037607 | DOI: 10.1523/JNEUROSCI.0830-22.2023

Regional cellular heterogeneity is a fundamental feature of the human neocortex; however, details of this heterogeneity are still undefined. We utilized single-nucleus RNA-sequencing (snRNA-seq) to examine cell-specific transcriptional features in the dorsolateral prefrontal cortex (DLPFC) and the subgenual anterior cingulate cortex (sgACC)-regions implicated in major psychiatric disorders. Droplet-based nuclei-capture and library preparation were performed on replicate samples from eight male donors without history of psychiatric or neurological disorder. Unsupervised clustering identified major neural cell classes. Subsequent iterative clustering of neurons further revealed 20 excitatory and 22 inhibitory subclasses. Inhibitory cells were consistently more abundant in the sgACC and excitatory neuron subclusters exhibited considerable variability across brain regions. Excitatory cell subclasses also exhibited greater within-class transcriptional differences between the two regions. We utilized these molecular definitions to determine which cell classes might be enriched in loci carrying a genetic signal in genome-wide association studies (GWAS) or for differentially expressed genes (DEGs) in mental illness. We found that the heritable signals of psychiatric disorders were enriched in neurons and that while the gene expression changes detected in bulk-RNA-sequencing studies was dominated by glial cells, some alterations could be identified in specific classes of excitatory and inhibitory neurons. Intriguingly, only two excitatory cell classes exhibited concomitant region-specific enrichment for both GWAS loci and transcriptional dysregulation. In sum, by detailing the molecular and cellular diversity of the DLPFC and sgACC, we were able generate hypotheses on regional and cell-specific dysfunctions that may contribute to the development of mental illness.SIGNIFICANCE STATEMENT:Dysfunction of the subgenual anterior cingulate cortex (sgACC) has been implicated in mood disorders, particularly major depressive disorder, and the dorsolateral prefrontal cortex (DLPFC)-a subsection of the Prefrontal cortex involved in executive functioning-has been implicated in schizophrenia. Understanding the cellular composition of these regions is critical to elucidating the neurobiology underlying psychiatric and neurological disorders. We studied cell type diversity of the sgACC and DLPFC of humans with no neuropsychiatric illness utilizing a clustering analysis of single-nuclei RNA-sequencing (snRNA-seq) data. Defining the transcriptomic profile of cellular subpopulations in these cortical regions is a first step to demystifying the cellular and molecular pathways involved in psychiatric disorders.
Arcuate Angiotensin II increases arterial pressure via coordinated increases in sympathetic nerve activity and vasopressin secretion

eNeuro

2021 Dec 17

Shi, Z;Stornetta, DS;Stornetta, RL;Brooks, VL;
PMID: 34937769 | DOI: 10.1523/ENEURO.0404-21.2021

The arcuate nucleus (ArcN) is an integrative hub for the regulation of energy balance, reproduction, and arterial pressure (AP), all of which are influenced by Angiotensin II (AngII); however, the cellular mechanisms and downstream neurocircuitry are unclear. Here we show that ArcN AngII increases AP in female rats via two phases, both of which are mediated via activation of AngII type 1 receptors (AT1aR): initial vasopressin-induced vasoconstriction, followed by slowly developing increases in sympathetic nerve activity (SNA) and heart rate (HR). In male rats, ArcN AngII evoked a similarly slow increase in SNA, but the initial pressor response was variable. In females, the effects of ArcN AngII varied during the estrus cycle, with significant increases in SNA, HR, and AP occurring during diestrus and estrus, but only increased AP during proestrus. Pregnancy markedly increased the expression of AT1aR in the ArcN with parallel substantial AngII-induced increases in SNA and MAP. In both sexes, the sympathoexcitation relied on suppression of tonic ArcN sympathoinhibitory Neuropeptide Y inputs, and activation of pro-opiomelanocortin (POMC) projections, to the paraventricular nucleus (PVN). Few or no NPY or POMC neurons expressed the AT1aR, suggesting that AngII increases AP and SNA at least in part indirectly via local interneurons, which express tyrosine hydroxylase (TH) and VGat (i.e. GABAergic). ArcN TH neurons release GABA locally, and central AT1aR and TH neurons mediate stress responses; therefore, we propose that TH AT1aR neurons are well situated to locally coordinate the regulation of multiple modalities within the ArcN in response to stress.SIGNIFICANCEThe arcuate nucleus (ArcN) is an integrative hub for the regulation of energy balance, reproduction, and arterial pressure (AP), all of which are influenced by Angiotensin II (AngII). Here we show that ArcN AngII activates AT1aR to increase AP in male and female rats by slowly increasing sympathetic nerve activity. In females, ArcN AngII also evoked an initial pressor response mediated by vasopressin-induced vasoconstriction. Pregnant and estrus females responded more than males, in association with higher ArcN AT1aR expression. AT1aR were identified in ArcN interneurons that express tyrosine hydroxylase (TH) and GABA. Since brain AT1aR and TH mediate stress responses, ArcN AT1aR TH neurons are well situated to locally coordinate autonomic, hormonal, and behavioral responses to stress.
Notum Influences Trachealis Muscle Organization in a Mouse Model of Tracheal Stenosis

TP77. TP077 PEDIATRIC CONGENITAL LUNG LESIONS

2021 May 01

Sinner, D;Burra, K;Bottasso-Arias, N;Schablein, R;Hilvano, N;Leesman, L;
| DOI: 10.1164/ajrccm-conference.2021.203.1_MeetingAbstracts.A3448

Diseases of the airway such as Tracheobronchomalacia (TBM) and Complete tracheal rings (CTR) are prevalent conditions associated with abnormal patterning of the trachealis muscle and cartilage. However, the underlying mechanisms of tracheal patterning are poorly understood. We have demonstrated that Wnt signaling via Wls plays an essential role in determining the mesenchymal patterning of the trachea. Notum, a direct target of Wnt signaling, encodes an enzyme that inactivates Wnt ligands, thus attenuating Wnt signaling's strength in developing trachea. In Notum deficient mice, chondroblasts and smooth muscle cells of the trachea were specified properly. Meanwhile, deletion of Notum impaired and delayed mesenchymal condensations of chondroblasts causing abnormal cartilage and stenosis. Further, tracheal muscle organization was disrupted. We hypothesize that chondrocyte condensation influences the trachealis muscle organization during tracheal tubulogenesis. Methods: We utilized genetically modified mice wherein Notum, Wnt5a, and Ror2 were deleted in the germline or conditionally ablated in tracheal mesenchyme. Tracheal smooth muscle cells were genetically labeled using γSMAeGFP mice. RNA scope, whole-mount stain, immunofluorescence, and fluorescent microscopy were utilized to visualize changes in trachealis muscle cell and cartilage organization induced by in vivo gene deletion and ex vivo treatments. Results: Analysis of muscle cells of control tracheas at E12, E14, and E16 showed progressive trachealis muscle organization with myocytes oriented perpendicular to the elongation axis of the trachea. In contrast, Notum deficient tracheas showed disordered orientation of the muscle cells with changes in the cytoskeleton. The anomalous muscle cell arrangement was increasingly observed after E14 after mesenchymal condensations were completed, and cartilage formed. Ex vivo studies with ABC99, a pharmacological inhibitor of Notum affected trachealis muscle organization, recapitulating the in vivo data. The Planar Cell Polarity (PCP) branch of the non-canonical Wnt signaling pathway mediates organization and functioning of the trachealis muscle. Mesenchymal deletion of the non-canonical Wnt5a and its receptor Ror2 impaired trachealis muscle cell orientation, causing changes in myocytes' cytoskeletal organization similar to changes observed in Notum deficient trachea. Different from Notum deletion, changes in myocyte organization preceded cartilaginous mesenchymal condensation. Despite the abnormal muscle organization in Wnt5a, cartilaginous mesenchymal condensations occurred, although reduced in number. We conclude that tracheal chondrogenesis affects trachealis muscle formation by constraining the expansion and altering the cytoskeletal organization of tracheal myocytes after mesenchymal condensation takes place. Thus, delayed tracheal chondrogenesis may partially underlie the pathology of tracheal stenosis. These studies were partially supported by NIH-NHLBI R01HL144774-01A1 to DS.
Persistence of Human Bocavirus 1 in Tonsillar Germinal Centers and Antibody-Dependent Enhancement of Infection

mBio

2021 Feb 02

Xu, M;Perdomo, MF;Mattola, S;Pyöriä, L;Toppinen, M;Qiu, J;Vihinen-Ranta, M;Hedman, K;Nokso-Koivisto, J;Aaltonen, LM;Söderlund-Venermo, M;
PMID: 33531399 | DOI: 10.1128/mBio.03132-20

Human bocavirus 1 (HBoV1), a nonenveloped single-stranded DNA parvovirus, causes mild to life-threatening respiratory tract infections, acute otitis media, and encephalitis in young children. HBoV1 often persists in nasopharyngeal secretions for months, hampering diagnosis. It has also been shown to persist in pediatric palatine and adenoid tonsils, which suggests that lymphoid organs are reservoirs for virus spread; however, the tissue site and host cells remain unknown. Our aim was to determine, in healthy nonviremic children with preexisting HBoV1 immunity, the adenotonsillar persistence site(s), host cell types, and virus activity. We discovered that HBoV1 DNA persists in lymphoid germinal centers (GCs), but not in the corresponding tonsillar epithelium, and that the cell types harboring the virus are mainly naive, activated, and memory B cells and monocytes. Both viral DNA strands and both sides of the genome were detected, as well as infrequent mRNA. Moreover, we showed, in B-cell and monocyte cultures and ex vivo tonsillar B cells, that the cellular uptake of HBoV1 occurs via the Fc receptor (FcγRII) through antibody-dependent enhancement (ADE). This resulted in viral mRNA transcription, known to occur exclusively from double-stranded DNA in the nucleus, however, with no detectable productive replication. Confocal imaging with fluorescent virus-like particles moreover disclosed endocytosis. To which extent the active HBoV1 GC persistence has a role in chronic inflammation or B-cell maturation disturbances, and whether the virus can be reactivated, will be interesting topics for forthcoming studies.IMPORTANCE Human bocavirus 1 (HBoV1), a common pediatric respiratory pathogen, can persist in airway secretions for months hampering diagnosis. It also persists in tonsils, providing potential reservoirs for airway shedding, with the exact location, host cell types, and virus activity unknown. Our study provides new insights into tonsillar HBoV1 persistence. We observed HBoV1 persistence exclusively in germinal centers where immune maturation occurs, and the main host cells were B cells and monocytes. In cultured cell lines and primary tonsillar B cells, we showed the virus uptake to be significantly enhanced by HBoV1-specific antibodies, mediated by the cellular IgG receptor, leading to viral mRNA synthesis, but without detectable productive replication. Possible implications of such active viral persistence could be tonsillar inflammation, disturbances in immune maturation, reactivation, or cell death with release of virus DNA, explaining the long-lasting HBoV1 airway shedding.
Structure and Function of Neuronal Circuits Linking Ventrolateral Preoptic Nucleus and Lateral Hypothalamic Area

The Journal of neuroscience : the official journal of the Society for Neuroscience

2023 May 31

Prokofeva, K;Saito, YC;Niwa, Y;Mizuno, S;Takahashi, S;Hirano, A;Sakurai, T;
PMID: 37117013 | DOI: 10.1523/JNEUROSCI.1913-22.2023

To understand how sleep-wakefulness cycles are regulated, it is essential to disentangle structural and functional relationships between the preoptic area (POA) and lateral hypothalamic area (LHA), since these regions play important yet opposing roles in the sleep-wakefulness regulation. GABA- and galanin (GAL)-producing neurons in the ventrolateral preoptic nucleus (VLPO) of the POA (VLPOGABA and VLPOGAL neurons) are responsible for the maintenance of sleep, while the LHA contains orexin-producing neurons (orexin neurons) that are crucial for maintenance of wakefulness. Through the use of rabies virus-mediated neural tracing combined with in situ hybridization (ISH) in male and female orexin-iCre mice, we revealed that the vesicular GABA transporter (Vgat, Slc32a1)- and galanin (Gal)-expressing neurons in the VLPO directly synapse with orexin neurons in the LHA. A majority (56.3 ± 8.1%) of all VLPO input neurons connecting to orexin neurons were double-positive for Vgat and Gal Using projection-specific rabies virus-mediated tracing in male and female Vgat-ires-Cre and Gal-Cre mice, we discovered that VLPOGABA and VLPOGAL neurons that send projections to the LHA received innervations from similarly distributed input neurons in many brain regions, with the POA and LHA being among the main upstream areas. Additionally, we found that acute optogenetic excitation of axons of VLPOGABA neurons, but not VLPOGAL neurons, in the LHA of male Vgat-ires-Cre mice induced wakefulness. This study deciphers the connectivity between the VLPO and LHA, provides a large-scale map of upstream neuronal populations of VLPO→LHA neurons, and reveals a previously uncovered function of the VLPOGABA→LHA pathway in the regulation of sleep and wakefulness.SIGNIFICANCE STATEMENT We identified neurons in the ventrolateral preoptic nucleus (VLPO) that are positive for vesicular GABA transporter (Vgat) and/or galanin (Gal) and serve as presynaptic partners of orexin-producing neurons in the lateral hypothalamic area (LHA). We depicted monosynaptic input neurons of GABA- and galanin-producing neurons in the VLPO that send projections to the LHA throughout the entire brain. Their input neurons largely overlap, suggesting that they comprise a common neuronal population. However, acute excitatory optogenetic manipulation of the VLPOGABA→LHA pathway, but not the VLPOGAL→LHA pathway, evoked wakefulness. This study shows the connectivity of major components of the sleep/wake circuitry in the hypothalamus and unveils a previously unrecognized function of the VLPOGABA→LHA pathway in sleep-wakefulness regulation. Furthermore, we suggest the existence of subpopulations of VLPOGABA neurons that innervate LHA.
Nicotinic acetylcholine receptors expressed by striatal interneurons inhibit striatal activity and control striatal-dependent behaviors

The Journal of neuroscience : the official journal of the Society for Neuroscience

2022 Feb 11

Abbondanza, A;Ribeiro Bas, I;Modrak, M;Capek, M;Minich, J;Tyshkevich, A;Naser, S;Rangotis, R;Houdek, P;Sumova, A;Dumas, S;Bernard, V;Janickova, H;
PMID: 35165173 | DOI: 10.1523/JNEUROSCI.1627-21.2022

Acetylcholine is an important modulator of striatal activity and it is vital to controlling striatal-dependent behaviors, including motor and cognitive functions. Despite this significance, the mechanisms determining how acetylcholine impacts striatal signaling are still not fully understood. In particular, little is known about the role of nicotinic acetylcholine receptors (nAChRs) expressed by striatal interneurons. In the present study, we used fluorescent in situ hybridization (FISH) to determine which neuronal types express the most prevalent beta2 nicotinic subunit in the mouse striatum. Our data support a common view that nAChR expression is mostly restricted to striatal interneurons. Surprisingly though, cholinergic interneurons (CINs) were identified as a population with the highest expression of beta2 nicotinic subunit. To investigate the functional significance of beta2-containing nAChRs in striatal interneurons, we deleted them by injecting the AAV-Cre vector into the striatum of beta2-flox/flox male mice. The deletion led to alterations in several behavioral domains, namely to an increased anxiety-like behavior, decrease in sociability ratio, deficit in discrimination learning and increased amphetamine-induced hyperlocomotion and c-Fos expression in mice with beta2 deletion. Further colocalization analysis showed that the increased c-Fos expression was present in both medium spiny neurons and presumed striatal interneurons. The present study concludes, that despite being relatively rare, beta2-containing nAChRs are primarily expressed in striatal neurons by CINs and play a significant role in behavior.SIGNIFICANCE STATEMENTA large variety of nicotinic acetylcholine receptors are expressed in the striatum, a brain region that is crucial in the control of behavior. The complexity of receptors with different functions is hindering our understanding of mechanisms through which striatal acetylcholine modulates behavior. We focused on the role of a small population of beta2-containing nicotinic acetylcholine receptors. We identified neuronal types expressing these receptors and determined their impact in the control of explorative behavior, anxiety-like behavior, learning and sensitivity to stimulants. Additional experiments showed that these alterations were associated with an overall increased activity of striatal neurons. Thus, the small population of nicotinic receptors represents an interesting target for a modulation of response to stimulant drugs and other striatal-based behavior.

Pages

  • « first
  • ‹ previous
  • …
  • 24
  • 25
  • 26
  • 27
  • 28
  • 29
  • 30
  • 31
  • 32
  • …
  • next ›
  • last »
X
Description
sense
Example: Hs-LAG3-sense
Standard probes for RNA detection are in antisense. Sense probe is reverse complent to the corresponding antisense probe.
Intron#
Example: Mm-Htt-intron2
Probe targets the indicated intron in the target gene, commonly used for pre-mRNA detection
Pool/Pan
Example: Hs-CD3-pool (Hs-CD3D, Hs-CD3E, Hs-CD3G)
A mixture of multiple probe sets targeting multiple genes or transcripts
No-XSp
Example: Hs-PDGFB-No-XMm
Does not cross detect with the species (Sp)
XSp
Example: Rn-Pde9a-XMm
designed to cross detect with the species (Sp)
O#
Example: Mm-Islr-O1
Alternative design targeting different regions of the same transcript or isoforms
CDS
Example: Hs-SLC31A-CDS
Probe targets the protein-coding sequence only
EnEmProbe targets exons n and m
En-EmProbe targets region from exon n to exon m
Retired Nomenclature
tvn
Example: Hs-LEPR-tv1
Designed to target transcript variant n
ORF
Example: Hs-ACVRL1-ORF
Probe targets open reading frame
UTR
Example: Hs-HTT-UTR-C3
Probe targets the untranslated region (non-protein-coding region) only
5UTR
Example: Hs-GNRHR-5UTR
Probe targets the 5' untranslated region only
3UTR
Example: Rn-Npy1r-3UTR
Probe targets the 3' untranslated region only
Pan
Example: Pool
A mixture of multiple probe sets targeting multiple genes or transcripts

Enabling research, drug development (CDx) and diagnostics

Contact Us
  • Toll-free in the US and Canada
  • +1877 576-3636
  • 
  • 
  • 
Company
  • Overview
  • Leadership
  • Careers
  • Distributors
  • Quality
  • News & Events
  • Webinars
  • Patents
Products
  • RNAscope or BaseScope
  • Target Probes
  • Controls
  • Manual assays
  • Automated Assays
  • Accessories
  • Software
  • How to Order
Research
  • Popular Applications
  • Cancer
  • Viral
  • Pathways
  • Neuroscience
  • Other Applications
  • RNA & Protein
  • Customer Innovations
  • Animal Models
Technology
  • Overview
  • RNA Detection
  • Spotlight Interviews
  • Publications & Guides
Assay Services
  • Our Services
  • Biomarker Assay Development
  • Cell & Gene Therapy Services
  • Clinical Assay Development
  • Tissue Bank & Sample Procurement
  • Image Analysis
  • Your Benefits
  • How to Order
Diagnostics
  • Diagnostics
  • Companion Diagnostics
Support
  • Getting started
  • Contact Support
  • Troubleshooting Guide
  • FAQs
  • Manuals, SDS & Inserts
  • Downloads
  • Webinars
  • Training Videos

Visit Bio-Techne and its other brands

  • bio-technie
  • protein
  • bio-spacific
  • rd
  • novus
  • tocris
© 2025 Advanced Cell Diagnostics, Inc.
  • Terms and Conditions of Sale
  • Privacy Policy
  • Security
  • Email Preferences
  • 
  • 
  • 

For Research Use Only. Not for diagnostic use. Refer to appropriate regulations. RNAscope is a registered trademark; and HybEZ, EZ-Batch and DNAscope are trademarks of Advanced Cell Diagnostics, Inc. in the United States and other countries. All rights reserved. ©2025 Advanced Cell Diagnostics, Inc.

 

Contact Us / Request a Quote
Download Manuals
Request a PAS Project Consultation
Order online at
bio-techne.com
OK
X
Contact Us

Complete one of the three forms below and we will get back to you.

For Quote Requests, please provide more details in the Contact Sales form below

  • Contact Sales
  • Contact Support
  • Contact Services
  • Offices

Advanced Cell Diagnostics

Our new headquarters office starting May 2016:

7707 Gateway Blvd.  
Newark, CA 94560
Toll Free: 1 (877) 576-3636
Phone: (510) 576-8800
Fax: (510) 576-8798

 

Bio-Techne

19 Barton Lane  
Abingdon Science Park
Abingdon
OX14 3NB
United Kingdom
Phone 2: +44 1235 529449
Fax: +44 1235 533420

 

Advanced Cell Diagnostics China

20F, Tower 3,
Raffles City Changning Office,
1193 Changning Road, Shanghai 200051

021-52293200
info.cn@bio-techne.com
Web: www.acdbio.com/cn

For general information: Info.ACD@bio-techne.com
For place an order: order.ACD@bio-techne.com
For product support: support.ACD@bio-techne.com
For career opportunities: hr.ACD@bio-techne.com

See Distributors
×

You have already Quick ordered an Item in your cart . If you want to add a new item , Quick ordered Item will be removed form your cart. Do You want to continue?

OK Cancel
Need help?

How can we help you?