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 CSF1

ACD can configure probes for the various manual and automated assays for CSF1 for RNAscope Assay, or for Basescope Assay compatible for your species of interest.

ACD’s data images for CSF1 gene.

  • Expression of CSF1 in Human Lymphoma sample using RNAscope™ 2.5 HD Assay Brown

  • Expression of CSF1 in Human meningioma sample using RNAscope™ 2.5 HD Assay Brown

  • Expression of CSF1 in Human ovarian cancer sample using RNAscope™ 2.5 HD Assay Brown

  • Probes for CSF1 (318)
  • Kits & Accessories (0)
  • Support & Documents (0)
  • Publications (14)
  • Image gallery (0)
Refine Probe List

Content for comparison

Gene

  • CSF1 (11) Apply CSF1 filter
  • CSF1R (3) Apply CSF1R filter
  • TBD (2) Apply TBD filter
  • Gal (1) Apply Gal filter
  • Fgfr3 (1) Apply Fgfr3 filter
  • Sox11 (1) Apply Sox11 filter
  • CD14 (1) Apply CD14 filter
  • CCKBR (1) Apply CCKBR filter
  • Rspo3 (1) Apply Rspo3 filter
  • NTRK2 (1) Apply NTRK2 filter
  • EREG (1) Apply EREG filter
  • FOLR2 (1) Apply FOLR2 filter
  • Spp1 (1) Apply Spp1 filter
  • MCAM (1) Apply MCAM filter
  • Npy (1) Apply Npy filter
  • Atf3 (1) Apply Atf3 filter
  • Ntrk3 (1) Apply Ntrk3 filter
  • PPIB (1) Apply PPIB filter
  • MYH11 (1) Apply MYH11 filter
  • Sprr1a (1) Apply Sprr1a filter
  • TUBB3 (1) Apply TUBB3 filter
  • CX3CL1 (1) Apply CX3CL1 filter
  • CXCL12 (1) Apply CXCL12 filter
  • Gpr151 (1) Apply Gpr151 filter
  • Id4 (1) Apply Id4 filter
  • Serpina3n (1) Apply Serpina3n filter
  • Nts (1) Apply Nts filter
  • Ecel1 (1) Apply Ecel1 filter
  • NCAM1 (1) Apply NCAM1 filter
  • HLA-G (1) Apply HLA-G filter
  • EFNB1 (1) Apply EFNB1 filter
  • DapB (1) Apply DapB filter
  • Cd36 (1) Apply Cd36 filter
  • Cxcl16 (1) Apply Cxcl16 filter
  • CXCR6 (1) Apply CXCR6 filter
  • MMP11 (1) Apply MMP11 filter
  • Dpep1 (1) Apply Dpep1 filter
  • LZTS1 (1) Apply LZTS1 filter
  • Chad (1) Apply Chad filter
  • Steap4 (1) Apply Steap4 filter
  • Ptprs (1) Apply Ptprs filter
  • CLEC3B (1) Apply CLEC3B filter
  • MEOX1 (1) Apply MEOX1 filter
  • LGI4 (1) Apply LGI4 filter
  • EphB6 (1) Apply EphB6 filter
  • FNDC1 (1) Apply FNDC1 filter
  • FHL5 (1) Apply FHL5 filter
  • Polr2ra (1) Apply Polr2ra filter

Product

  • RNAscope (2) Apply RNAscope filter
  • RNAscope 2.5 HD Duplex (2) Apply RNAscope 2.5 HD Duplex filter
  • RNAscope 2.0 Assay (1) Apply RNAscope 2.0 Assay filter
  • RNAscope 2.5 HD Red assay (1) Apply RNAscope 2.5 HD Red assay filter
  • RNAscope 2.5 LS Assay (1) Apply RNAscope 2.5 LS Assay filter
  • RNAscope 2.5 VS Assay (1) Apply RNAscope 2.5 VS Assay filter
  • RNAscope Fluorescent Multiplex Assay (1) Apply RNAscope Fluorescent Multiplex Assay filter
  • RNAscope Multiplex Fluorescent Assay (1) Apply RNAscope Multiplex Fluorescent Assay filter
  • TBD (1) Apply TBD filter

Research area

  • Cancer (6) Apply Cancer filter
  • Development (2) Apply Development filter
  • Heart: Aortic Valve Disease (1) Apply Heart: Aortic Valve Disease filter
  • Infectious Disease (1) Apply Infectious Disease filter
  • Inflammation (1) Apply Inflammation filter
  • Neuroscience (1) Apply Neuroscience filter
  • Other: Tenosynovial giant cell tumor (1) Apply Other: Tenosynovial giant cell tumor filter
  • Reproductive Biology (1) Apply Reproductive Biology filter

Category

  • Publications (14) Apply Publications filter
Diagnostic utility of CSF1 immunohistochemistry in tenosynovial giant cell tumor for differentiating from giant cell-rich tumors and tumor-like lesions of bone and soft tissue

Diagnostic pathology

2022 Nov 01

Sugita, S;Takenami, T;Kido, T;Aoyama, T;Hosaka, M;Segawa, K;Sugawara, T;Fujita, H;Shimizu, J;Murahashi, Y;Emori, M;Hasegawa, T;
PMID: 36320082 | DOI: 10.1186/s13000-022-01266-9

Tenosynovial giant cell tumor (TSGCT) is a benign fibrohistiocytic tumor that affects the synovium of joints, bursa, and tendon sheaths and is categorized into localized TSGCT (LTSGCT) and diffuse TSGCT (DTSGCT). LTSGCT and DTSGCT are characterized by recurrent fusions involving the colony-stimulating factor 1 (CSF1) gene and its translocation partner collagen type VI alpha 3 chain. The fusion gene induces intratumoral overexpression of CSF1 mRNA and CSF1 protein. CSF1 expression is a characteristic finding of TSGCT and detection of CSF1 mRNA and CSF1 protein may be useful for the pathological diagnosis. Although there have been no effective anti-CSF1 antibodies to date, in situ hybridization (ISH) for CSF1 mRNA has been performed to detect CSF1 expression in TSGCT. We performed CSF1 immunohistochemistry (IHC) using anti-CSF1 antibody (clone 2D10) in cases of TSGCT, giant cell-rich tumor (GCRT), and GCRT-like lesion and verified its utility for the pathological diagnosis of TSGCT.We performed CSF1 IHC in 110 cases including 44 LTSGCTs, 20 DTSGCTs, 1 malignant TSGCT (MTSGCT), 10 giant cell tumors of bone, 2 giant cell reparative granulomas, 3 aneurysmal bone cysts, 10 undifferentiated pleomorphic sarcomas, 10 leiomyosarcomas, and 10 myxofibrosarcomas. We performed fluorescence ISH (FISH) for CSF1 rearrangement to confirm CSF1 expression on IHC in TSGCTs. We considered the specimens to have CSF1 rearrangement if a split signal was observed in greater than 2% of the tumor cells.Overall, 50 of 65 TSGCT cases, including 35 of the 44 LTSGCTs and 15 of the 20 DTSGCTs, showed distinct scattered expression of CSF1 in the majority of mononuclear tumor cells. MTSGCT showed no CSF1 expression. Non-TSGCT cases were negative for CSF1. FISH revealed CSF1 rearrangement in 6 of 7 CSF1-positive cases on IHC. On the other hand, FISH detected no CSF1 rearrangement in all CSF1-negative cases on IHC. Thus, the results of IHC corresponded to those of FISH.We revealed characteristic CSF1 expression on IHC in cases of TSGCT, whereas the cases of non-TSGCT exhibited no CSF1 expression. CSF1 IHC may be useful for differentiating TSGCTs from histologically mimicking GCRTs and GCRT-like lesions.
RNAscope CSF1 Chromogenic in situ Hybridization: A Potentially Useful Tool in the Differential Diagnosis of Tenosynovial Giant Cell Tumors

Human pathology

2021 May 28

Thangaiah, JJ;Koepplin, JW;Folpe, AL;
PMID: 34058245 | DOI: 10.1016/j.humpath.2021.05.010

Colony Stimulating Factor-1 (CSF1) up regulation and CSF1/Colony-stimulating factor 1 receptor (CSF1R) signaling pathway is central to the tumorigenesis of tenosynovial giant cell tumors (TGCT) of both localized (LTGCT) and diffuse (DTGCT) types, and has been demonstrated in a small number of malignant tumors (MTGCT) as well. In situ hybridization for CSF1 mRNA has been shown to be potentially useful in the diagnosis of TGCT, although only a relatively small number of cases have been studied. We studied CSF1 mRNA expression using RNAscope chromogenic in situ hybridization (CISH) in standard tissue sections from 31 TGCT and 26 non-TGCT, and in tumor microarray slides (Pantomics normal MN0341, Pantomics tumor MTU391, Pantomics melanoma MEL961). Among normal tissues, CSF1 mRNA expression was invariably present in synovium (10/10, 100%) and absent in all other normal tissues. All LTGCT and DTGCT were positive (24/24, 100%), exclusively in large, eosinophilic synoviocytes. MTGCT contained large clusters of CSF1-positive malignant synoviocytes (8/8, 100%); malignant spindled cells were also positive. Among non-TGCT, CSF1 CISH was less often positive with high specificity (90%). CSF1-positive cases included leiomyosarcoma, giant cell tumor of bone and of soft parts, pulmonary carcinoma and others. The sensitivity and specificity of RNAscope CSF1 mRNA CISH for the diagnosis of TGCT were 100% and 90%, respectively. We conclude that RNAscope CSF1 CISH may be a valuable adjunct for the diagnosis of TGCT of all types, especially those with atypical or malignant morphologic features. Detection of CSF1 mRNA expression may also have predictive significance in cases where use of the CSF1 inhibitor pexidartinib is considered.
Activated microglia drive demyelination via CSF1R signaling

Glia

2021 Feb 23

Marzan, DE;Brügger-Verdon, V;West, BL;Liddelow, S;Samanta, J;Salzer, JL;
PMID: 33620118 | DOI: 10.1002/glia.23980

Microgliosis is a prominent pathological feature in many neurological diseases including multiple sclerosis (MS), a progressive auto-immune demyelinating disorder. The precise role of microglia, parenchymal central nervous system (CNS) macrophages, during demyelination, and the relative contributions of peripheral macrophages are incompletely understood. Classical markers used to identify microglia do not reliably discriminate between microglia and peripheral macrophages, confounding analyses. Here, we use a genetic fate mapping strategy to identify microglia as predominant responders and key effectors of demyelination in the cuprizone (CUP) model. Colony-stimulating factor 1 (CSF1), also known as macrophage colony-stimulating factor (M-CSF) - a secreted cytokine that regulates microglia development and survival-is upregulated in demyelinated white matter lesions. Depletion of microglia with the CSF1R inhibitor PLX3397 greatly abrogates the demyelination, loss of oligodendrocytes, and reactive astrocytosis that results from CUP treatment. Electron microscopy (EM) and serial block face imaging show myelin sheaths remain intact in CUP treated mice depleted of microglia. However, these CUP-damaged myelin sheaths are lost and robustly phagocytosed upon-repopulation of microglia. Direct injection of CSF1 into CNS white matter induces focal microgliosis and demyelination indicating active CSF1 signaling can promote demyelination. Finally, mice defective in adopting a toxic astrocyte phenotype that is driven by microglia nevertheless demyelinate normally upon CUP treatment implicating microglia rather than astrocytes as the primary drivers of CUP-mediated demyelination. Together, these studies indicate activated microglia are required for and can drive demyelination directly and implicate CSF1 signaling in these events.
Structure-Guided Blockade of CSF1R Kinase in Tenosynovial Giant-Cell Tumor.

N Engl J Med. 2015 Jul 30;373(5):428-37.

Tap WD, Wainberg ZA, Anthony SP, Ibrahim PN, Zhang C, Healey JH, Chmielowski B, Staddon AP, Cohn AL, Shapiro GI, Keedy VL, Singh AS, Puzanov I, Kwak EL, Wagner AJ, Von Hoff DD, Weiss GJ, Ramanathan RK, Zhang J, Habets G, Zhang Y, Burton EA, Visor G, Sanft
PMID: 26222558 | DOI: 10.1056/NEJMoa1411366.

 BACKGROUND:
 Expression of the colony-stimulating factor 1 (CSF1) gene is elevated in most tenosynovial giant-cell tumors. This observation has led to the discovery and clinical development of therapy targeting the CSF1 receptor (CSF1R).
 METHODS:
 Using x-ray co-crystallography to guide our drug-discovery research, we generated a potent, selective CSF1R inhibitor, PLX3397, that traps the kinase in the autoinhibited conformation. We then conducted a multicenter, phase 1 trial in two parts to analyze this compound. In the first part, we evaluated escalations in the dose of PLX3397 that was administered orally in patients with solid tumors (dose-escalation study). In the second part, we evaluated PLX3397 at the chosen phase 2 dose in an extension cohort of patients with tenosynovial giant-cell tumors (extension study). Pharmacokinetic and tumor responses in the enrolled patients were assessed, and CSF1 in situ hybridization was performed to confirm the mechanism of action of PLX3397 and that the pattern of CSF1 expression was consistent with the pathological features of tenosynovial giant-cell tumor.
 RESULTS:
 A total of 41 patients were enrolled in the dose-escalation study, and an additional 23 patients were enrolled in the extension study. The chosen phase 2 dose of PLX3397 was 1000 mg per day. In the extension study, 12 patients with tenosynovial giant-cell tumors had a partial response and 7 patients had stable disease. Responses usually occurred within the first 4 months of treatment, and the median duration of response exceeded 8 months. The most common adverse events included fatigue, change in hair color, nausea, dysgeusia, and periorbital edema; adverse events rarely led to discontinuation of treatment.
 CONCLUSIONS:
 Treatment of tenosynovial giant-cell tumors with PLX3397 resulted in a prolonged regression in tumor volume in most patients. (Funded by Plexxikon; ClinicalTrials.gov number, NCT01004861.).

Increased Microglial CSF1R Expression in the SIV/Macaque Model of HIV CNS Disease

J Neuropathol Exp Neurol.

2018 Jan 08

Knight AC, Brill SA, Queen SE, Tarwater PM, Mankowski JL.
PMID: 29319809 | DOI: 10.1093/jnen/nlx115

Chronic microglial activation and associated neuroinflammation are key factors in neurodegenerative diseases including HIV-associated neurocognitive disorders. Colony stimulating factor 1 receptor (CSF1R)-mediated signaling is constitutive in cells of the myeloid lineage, including microglia, promoting cell survival, proliferation, and differentiation. In amyotrophic lateral sclerosis and Alzheimers disease, CSF1R is upregulated. Inhibiting CSF1R signaling in animal models of these diseases improved disease outcomes. In our studies, CNS expression of the CSF1R ligand, colony-stimulating factor 1 (CSF1) was significantly increased in a SIV/macaque model of HIV CNS disease. Using a Nanostring nCounter immune panel, we found CSF1 overexpression was strongly correlated with upregulation of microglial genes involved in antiviral and oxidative stress responses. Using in situ hybridization, we found that CSF1R mRNA was only present in Iba-1 positive microglia. By ELISA and immunostaining with digital image analysis, SIV-infected macaques had significantly higher CSF1R levels in frontal cortex than uninfected macaques (p = 0.018 and p = 0.02, respectively). SIV-infected macaques treated with suppressive ART also had persistently elevated CSF1R similar to untreated SIV-infected macaques. Coordinate upregulation of CSF1 and CSF1R expression implicates this signaling pathway in progressive HIV CNS disease.

Does CSF1 over-expression or rearrangement influence biological behaviour in tenosynovial giant cell tumours of the knee

Histopathology.

2018 Aug 28

Mastboom MJL, Hoek DM, Bovee JVMG, van de Sande MAJ, Szuhai K.
PMID: 30152874 | DOI: 10.1111/his.13744

Abstract

INTRODUCTION:

Localized- and diffuse-type tenosynovial giant cell tumours (TGCT) are regarded different clinical and radiological TGCT-types. However, genetically and histopathologically they seem indistinguishable. We aimed to correlate CSF1-expression and CSF1-rearrangement with the biological behaviour of different TGCT-types with clinical outcome (recurrence).

METHODS:

Along a continuum of extremes, therapy naïve knee TGCT patients with >3 year follow-up, mean age 43(range 6-71)years and 56% female were selected. Nine localized-(two recurrences), 16 diffuse-type(nine recurrences) and four synovitis as control were included. Rearrangement of the CSF1-locus was evaluated with split-apart Fluorescence In Situ Hybridization (FISH) probes. Regions were selected to score after identifying CSF1-expressing regions, using mRNA ISH with the help of digital correlative microscopy. CSF1-rearrangement was considered positive in samples containing >2 split signals/100 nuclei.

RESULTS:

Irrespective of TGCT-subtype, all cases showed CSF1-expression and in 76% CSF1-rearrangement was detected. Quantification of CSF1-expressing cells was not informative, due to the extensive intra tumour heterogeneity. Of the four synovitis cases, two also showed CSF1-expression, without CSF1-rearrangement. No correlation between CSF1-expression or rearrangement with clinical subtype and local recurrence was detected. Both localized- and diffuse-TGCT cases showed a scattered distribution in the tissue of CSF1-expressing cells.

CONCLUSION:

In diagnosing TGCT, CSF1 mRNA-ISH in combination with CSF1 split-apart FISH; using digital correlative microscopy, is an auxiliary diagnostic tool to identify rarely occurring neoplastic cells. This combined approach allowed us to detect CSF1-rearrangement in 76% of the TGCT-cases. Neither CSF1-expression nor presence of CSF1-rearrangement could be associated with the difference in biological behaviour of TGCT. 

Plexiform fibrohistiocytic tumor: a clinicopathological and immunohistochemical study of 39 tumors, with evidence for a CSF1-producing "null cell" population

Virchows Archiv : an international journal of pathology

2022 Sep 07

Thangaiah, JJ;Dashti, NK;Agaimy, A;Fritchie, K;Folpe, AL;
PMID: 36071257 | DOI: 10.1007/s00428-022-03408-2

Plexiform fibrohistiocytic tumor (PFHT) is a mesenchymal tumor of intermediate malignancy, typically occurring in the superficial soft tissues of young patients and displaying a biphasic pattern, with nodules of histiocytoid cells surrounded by fascicles of myofibroblastic spindled cells. The pathogenesis of PHFT is unknown. We comprehensively studied 39 PFHT, occurring in 25 females (66%) and 13 males (34%), ranging from 2 to 55 years of age (median 21 years). The tumors most often occurred in the upper extremity (n = 16, 41%) and ranged from 0.4 to 6.1 cm in size (median 1.5 cm). One patient with known neurofibromatosis type 1 presented with metachronous tumors of the finger and back. Clinical follow-up (29 patients; range 5-168 months; median 60 months) showed 3 tumors to have recurred locally; none was metastasized. One patient died of an unrelated cause; all others were alive without disease at the time of last follow-up. Immunohistochemistry showed the histiocytoid nodules of all cases to contain CD163/CD11c-positive histiocytes and cells negative for both markers ("null cells"). CSF1 expression was present in "null cells" in 7/10 cases (RNAscope chromogenic in situ hybridization). The Ki-67 labeling index was very low (< 5%); Ki-67-positive cells within histiocytoid nodules appeared to represent "null cells." All tested cases were negative for significant mutations or fusion events (TruSight Mutation Panel, TruSight Fusion Panel, Mayo Clinic Melanoma Targeted Gene Panel). We conclude that PHFT may be even more indolent than has been appreciated, although classification as an "intermediate" tumor is correct. We hypothesize that the CSF1-producing "null cells" of PHFT may represent the neoplastic element, with the bulk of the tumor masses comprising recruited and reactive cell populations.
CD206+ tendon resident macrophages and their potential crosstalk with fibroblasts and the ECM during tendon growth and maturation

Frontiers in Physiology

2023 Feb 22

Bautista, C;Srikumar, A;Tichy, E;Qian, G;Jiang, X;Qin, L;Mourkioti, F;Dyment, N;
| DOI: 10.3389/fphys.2023.1122348

Resident macrophages exist in a variety of tissues, including tendon, and play context-specific roles in their tissue of residence. In this study, we define the spatiotemporal distribution and phenotypic profile of tendon resident macrophages and their crosstalk with neighboring tendon fibroblasts and the extracellular matrix (ECM) during murine tendon development, growth, and homeostasis. Fluorescent imaging of cryosections revealed that F4/80+ tendon resident macrophages reside adjacent to Col1a1-CFP+ Scx-GFP+ fibroblasts within the tendon fascicle from embryonic development (E15.5) into adulthood (P56). Through flow cytometry and qPCR, we found that these tendon resident macrophages express several well-known macrophage markers, including Adgre1 (F4/80), Mrc1 (CD206), Lyve1, and Folr2, but not Ly-6C, and express the Csf1r-EGFP (“MacGreen”) reporter. The proportion of Csf1r-EGFP+ resident macrophages in relation to the total cell number increases markedly during early postnatal growth, while the density of macrophages per mm2 remains constant during this same time frame. Interestingly, proliferation of resident macrophages is higher than adjacent fibroblasts, which likely contributes to this increase in macrophage proportion. The expression profile of tendon resident macrophages also changes with age, with increased pro-inflammatory and anti-inflammatory cytokine expression in P56 compared to P14 macrophages. In addition, the expression profile of limb tendon resident macrophages diverges from that of tail tendon resident macrophages, suggesting differential phenotypes across anatomically and functionally different tendons. As macrophages are known to communicate with adjacent fibroblasts in other tissues, we conducted ligand-receptor analysis and found potential two-way signaling between tendon fibroblasts and resident macrophages. Tendon fibroblasts express high levels of Csf1, which encodes macrophage colony stimulating factor (M-CSF) that acts on the CSF1 receptor (CSF1R) on macrophages. Importantly, Csf1r-expressing resident macrophages preferentially localize to Csf1-expressing fibroblasts, supporting the “nurturing scaffold” model for tendon macrophage patterning. Lastly, we found that tendon resident macrophages express high levels of ECM-related genes, including Mrc1 (mannose receptor), Lyve1 (hyaluronan receptor), Lair1 (type I collagen receptor), Ctss (elastase), and Mmp13 (collagenase), and internalize DQ Collagen in explant cultures. Overall, our study provides insights into the potential roles of tendon resident macrophages in regulating fibroblast phenotype and the ECM during tendon growth.
Dual leucine zipper kinase is required for mechanical allodynia and microgliosis after nerve injury.

Elife.

2018 Jul 03

Wlaschin JJ, Gluski JM, Nguyen E, Silberberg H, Thompson JH, Chesler AT, Le Pichon CE.
PMID: 29968565 | DOI: 10.7554/eLife.33910

Neuropathic pain resulting from nerve injury can become persistent and difficult to treat but the molecular signaling responsible for its development remains poorly described. Here, we identify the neuronal stress sensor dual leucine zipper kinase (DLK; Map3k12) as a key molecule controlling the maladaptive pathways that lead to pain following injury. Genetic or pharmacological inhibition of DLK reduces mechanical hypersensitivity in a mouse model of neuropathic pain. Furthermore, DLK inhibition also prevents the spinal cord microgliosis that results from nerve injury and arises distant from the injury site. These striking phenotypes result from the control by DLK of a transcriptional program in somatosensory neurons regulating the expression of numerous genes implicated in pain pathogenesis, including the immune gene Csf1. Thus, activation of DLK is an early event, or even the master regulator, controlling a wide variety of pathways downstream of nerve injury that ultimately lead to chronic pain.

Therapeutic targeting of macrophages enhances chemotherapy efficacy by unleashing type I interferon response

Nat Cell Biol

2019 Mar 18

Salvagno C, Ciampricotti M, Tuit S, Hau CS, van Weverwijk A, Coffelt SB, Kersten K, Vrijland K, Kos K, Ulas T, Song JY, Ooi CH, Ruttinger D, Cassier PA, Jonkers J, Schultze JL, Ries CH and de Visser KE
PMID: 30886344 | DOI: 10.1038/s41556-019-0298-1

Recent studies have revealed a role for macrophages and neutrophils in limiting chemotherapy efficacy; however, the mechanisms underlying the therapeutic benefit of myeloid-targeting agents in combination with chemotherapy are incompletely understood. Here, we show that targeting tumour-associated macrophages by colony-stimulating factor-1 receptor (CSF-1R) blockade in the K14cre;Cdh1(F/F);Trp53(F/F) transgenic mouse model for breast cancer stimulates intratumoural type I interferon (IFN) signalling, which enhances the anticancer efficacy of platinum-based chemotherapeutics. Notably, anti-CSF-1R treatment also increased intratumoural expression of type I IFN-stimulated genes in patients with cancer, confirming that CSF-1R blockade is a powerful strategy to trigger an intratumoural type I IFN response. By inducing an inflamed, type I IFN-enriched tumour microenvironment and by further targeting immunosuppressive neutrophils during cisplatin therapy, antitumour immunity was activated in this poorly immunogenic breast cancer mouse model. These data illustrate the importance of breaching multiple layers of immunosuppression during cytotoxic therapy to successfully engage antitumour immunity in breast cancer.
Single-cell transcriptomics reveal cellular diversity of aortic valve and the immunomodulation by PPARγ during hyperlipidemia

Nature communications

2022 Sep 17

Lee, SH;Kim, N;Kim, M;Woo, SH;Han, I;Park, J;Kim, K;Park, KS;Kim, K;Shim, D;Park, SE;Zhang, JY;Go, DM;Kim, DY;Yoon, WK;Lee, SP;Chung, J;Kim, KW;Park, JH;Lee, SH;Lee, S;Ann, SJ;Lee, SH;Ahn, HS;Jeong, SC;Kim, TK;Oh, GT;Park, WY;Lee, HO;Choi, JH;
PMID: 36115863 | DOI: 10.1038/s41467-022-33202-2

Valvular inflammation triggered by hyperlipidemia has been considered as an important initial process of aortic valve disease; however, cellular and molecular evidence remains unclear. Here, we assess the relationship between plasma lipids and valvular inflammation, and identify association of low-density lipoprotein with increased valvular lipid and macrophage accumulation. Single-cell RNA sequencing analysis reveals the cellular heterogeneity of leukocytes, valvular interstitial cells, and valvular endothelial cells, and their phenotypic changes during hyperlipidemia leading to recruitment of monocyte-derived MHC-IIhi macrophages. Interestingly, we find activated PPARγ pathway in Cd36+ valvular endothelial cells increased in hyperlipidemic mice, and the conservation of PPARγ activation in non-calcified human aortic valves. While the PPARγ inhibition promotes inflammation, PPARγ activation using pioglitazone reduces valvular inflammation in hyperlipidemic mice. These results show that low-density lipoprotein is the main lipoprotein accumulated in the aortic valve during hyperlipidemia, leading to early-stage aortic valve disease, and PPARγ activation protects the aortic valve against inflammation.
GPR151 in nociceptors modulates neuropathic pain via regulating P2X3 function and microglial activation

Brain : a journal of neurology

2021 Jul 09

Xia, LP;Luo, H;Ma, Q;Xie, YK;Li, W;Hu, H;Xu, ZZ;
PMID: 34244727 | DOI: 10.1093/brain/awab245

Neuropathic pain is a major health problem that affects up to 7-10% of the population worldwide. Currently, neuropathic pain is difficult to treat due to its elusive mechanisms. Here we report that orphan G protein-coupled receptor 151 (GPR151) in nociceptive sensory neurons controls neuropathic pain induced by nerve injury. GPR151 was mainly expressed in nonpeptidergic C-fiber dorsal root ganglion (DRG) neurons and highly upregulated after nerve injury. Importantly, conditional knockout of Gpr151 in adult nociceptive sensory neurons significantly alleviated chronic constriction injury (CCI)-induced neuropathic pain-like behavior but did not affect basal nociception. Moreover, GPR151 in DRG neurons was required for CCI-induced neuronal hyperexcitability and upregulation of colony-stimulating factor 1 (CSF1), which is necessary for microglial activation in the spinal cord after nerve injury. Mechanistically, GPR151 coupled with P2X3 ion channels and promoted their functional activities in neuropathic pain-like hypersensitivity. Knockout of Gpr151 suppressed P2X3-mediated calcium elevation and spontaneous pain behavior in CCI mice. Conversely, overexpression of Gpr151 significantly enhanced P2X3-mediated calcium elevation and DRG neuronal excitability. Furthermore, knockdown of P2X3 in DRGs reversed CCI-induced CSF1 upregulation, spinal microglial activation, and neuropathic pain-like behavior. Finally, the co-expression of GPR151 and P2X3 was confirmed in small-diameter human DRG neurons, indicating the clinical relevance of our findings. Together, our results suggest that GPR151 in nociceptive DRG neurons plays a key role in the pathogenesis of neuropathic pain and could be a potential target for treating neuropathic pain.

Pages

  • 1
  • 2
  • 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?