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 (183)
  • Image gallery (0)
Refine Probe List

Content for comparison

Gene

  • TBD (1413) Apply TBD filter
  • (-) Remove Lgr5 filter Lgr5 (151)
  • 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
  • TH (63) Apply TH filter
  • 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
  • PVALB (47) Apply PVALB filter
  • 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
  • (-) Remove AGRP filter AGRP (32)
  • 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 Multiplex Fluorescent Assay (28) Apply RNAscope Multiplex Fluorescent Assay filter
  • RNAscope 2.5 HD Red assay (26) Apply RNAscope 2.5 HD Red assay filter
  • RNAscope 2.0 Assay (20) Apply RNAscope 2.0 Assay filter
  • RNAscope Fluorescent Multiplex Assay (18) Apply RNAscope Fluorescent Multiplex Assay filter
  • RNAscope (14) Apply RNAscope filter
  • RNAscope 2.5 LS Assay (12) Apply RNAscope 2.5 LS Assay filter
  • RNAscope 2.5 HD Brown Assay (10) Apply RNAscope 2.5 HD Brown Assay filter
  • RNAscope 2.5 HD Reagent Kit - BROWN (8) Apply RNAscope 2.5 HD Reagent Kit - BROWN filter
  • RNAscope 2.5 HD Duplex (3) Apply RNAscope 2.5 HD Duplex filter
  • RNAscope 2.0 HD Assay - Chromogenic (1) Apply RNAscope 2.0 HD Assay - Chromogenic filter
  • RNAscope Fluorescent Multiplex Reagent kit (1) Apply RNAscope Fluorescent Multiplex Reagent kit filter
  • RNAscope Multiplex Fluorescent Assay v2 (1) Apply RNAscope Multiplex Fluorescent Assay v2 filter
  • RNAscope Multiplex Fluorescent v2 (1) Apply RNAscope Multiplex Fluorescent v2 filter

Research area

  • Cancer (61) Apply Cancer filter
  • Stem Cells (59) Apply Stem Cells filter
  • Development (24) Apply Development filter
  • Neuroscience (23) Apply Neuroscience filter
  • Stem cell (16) Apply Stem cell filter
  • Other (11) Apply Other filter
  • Inflammation (8) Apply Inflammation filter
  • Metabolism (6) Apply Metabolism filter
  • Developmental (3) Apply Developmental filter
  • Cancer Stem Cells (2) Apply Cancer Stem Cells filter
  • Obesity (2) Apply Obesity filter
  • Other: Endocrinology (2) Apply Other: Endocrinology filter
  • Weight Loss (2) Apply Weight Loss filter
  • Cell transcriptomics (1) Apply Cell transcriptomics filter
  • Chronic gastritis (1) Apply Chronic gastritis filter
  • Colitis (1) Apply Colitis filter
  • diabetes (1) Apply diabetes filter
  • Diet (1) Apply Diet filter
  • Endocrinology (1) Apply Endocrinology filter
  • Feeding Behavior (1) Apply Feeding Behavior filter
  • Gastro (1) Apply Gastro filter
  • Gut Microbiota (1) Apply Gut Microbiota filter
  • Human intestinal organoids (1) Apply Human intestinal organoids filter
  • Hunger (1) Apply Hunger filter
  • Hypertension (1) Apply Hypertension filter
  • hypothalamic regulation of glucose (1) Apply hypothalamic regulation of glucose filter
  • Infectious Disease (1) Apply Infectious Disease filter
  • Inflammatory Bowel Disease (1) Apply Inflammatory Bowel Disease filter
  • Keratin (1) Apply Keratin filter
  • lncRNA (1) Apply lncRNA filter
  • Nutrition (1) Apply Nutrition filter
  • Organoid (1) Apply Organoid filter
  • Organoids (1) Apply Organoids filter
  • Other: Blood Vessels (1) Apply Other: Blood Vessels filter
  • Other: Hair Growth (1) Apply Other: Hair Growth filter
  • Other: Heart (1) Apply Other: Heart filter
  • Other: Immunity (1) Apply Other: Immunity filter
  • Other: Metabolism (1) Apply Other: Metabolism filter
  • Other: Prostate (1) Apply Other: Prostate filter
  • Other: Sodium intake (1) Apply Other: Sodium intake filter
  • Other:Hypertension (1) Apply Other:Hypertension filter
  • Pulmonology (1) Apply Pulmonology filter
  • Radiation enteritis (1) Apply Radiation enteritis filter
  • Radiotherapy (1) Apply Radiotherapy filter
  • Regeneration (1) Apply Regeneration filter
  • Reproduction (1) Apply Reproduction filter
  • scRNAseq (1) Apply scRNAseq filter
  • Signalling (1) Apply Signalling filter
  • therapeutics (1) Apply therapeutics filter
  • Tumourigenesis (1) Apply Tumourigenesis filter

Category

  • Publications (183) Apply Publications filter
Intestinal Apc-inactivation induces HSP25 dependency

EMBO molecular medicine

2022 Nov 02

van Neerven, SM;Smit, WL;van Driel, MS;Kakkar, V;de Groot, NE;Nijman, LE;Elbers, CC;Léveillé, N;Heijmans, J;Vermeulen, L;
PMID: 36321561 | DOI: 10.15252/emmm.202216194

The majority of colorectal cancers (CRCs) present with early mutations in tumor suppressor gene APC. APC mutations result in oncogenic activation of the Wnt pathway, which is associated with hyperproliferation, cytoskeletal remodeling, and a global increase in mRNA translation. To compensate for the increased biosynthetic demand, cancer cells critically depend on protein chaperones to maintain proteostasis, although their function in CRC remains largely unexplored. In order to investigate the role of molecular chaperones in driving CRC initiation, we captured the transcriptomic profiles of murine wild type and Apc-mutant organoids during active transformation. We discovered a strong transcriptional upregulation of Hspb1, which encodes small heat shock protein 25 (HSP25). We reveal an indispensable role for HSP25 in facilitating Apc-driven transformation, using both in vitro organoid cultures and mouse models, and demonstrate that chemical inhibition of HSP25 using brivudine reduces the development of premalignant adenomas. These findings uncover a hitherto unknown vulnerability in intestinal transformation that could be exploited for the development of chemopreventive strategies in high-risk individuals.
Central anorexigenic actions of bile acids are mediated by TGR5

Nature metabolism

2021 May 01

Perino, A;Velázquez-Villegas, LA;Bresciani, N;Sun, Y;Huang, Q;Fénelon, VS;Castellanos-Jankiewicz, A;Zizzari, P;Bruschetta, G;Jin, S;Baleisyte, A;Gioiello, A;Pellicciari, R;Ivanisevic, J;Schneider, BL;Diano, S;Cota, D;Schoonjans, K;
PMID: 34031591 | DOI: 10.1038/s42255-021-00398-4

Bile acids (BAs) are signalling molecules that mediate various cellular responses in both physiological and pathological processes. Several studies report that BAs can be detected in the brain1, yet their physiological role in the central nervous system is still largely unknown. Here we show that postprandial BAs can reach the brain and activate a negative-feedback loop controlling satiety in response to physiological feeding via TGR5, a G-protein-coupled receptor activated by multiple conjugated and unconjugated BAs2 and an established regulator of peripheral metabolism3-8. Notably, peripheral or central administration of a BA mix or a TGR5-specific BA mimetic (INT-777) exerted an anorexigenic effect in wild-type mice, while whole-body, neuron-specific or agouti-related peptide neuronal TGR5 deletion caused a significant increase in food intake. Accordingly, orexigenic peptide expression and secretion were reduced after short-term TGR5 activation. In vitro studies demonstrated that activation of the Rho-ROCK-actin-remodelling pathway decreases orexigenic agouti-related peptide/neuropeptide Y (AgRP/NPY) release in a TGR5-dependent manner. Taken together, these data identify a signalling cascade by which BAs exert acute effects at the transition between fasting and feeding and prime the switch towards satiety, unveiling a previously unrecognized role of physiological feedback mediated by BAs in the central nervous system.
The Tankyrase Inhibitor OM-153 Demonstrates Antitumor Efficacy and a Therapeutic Window in Mouse Models

Cancer Research Communications

2022 Apr 20

Brinch, S;Amundsen-Isaksen, E;Espada, S;Hammarström, C;Aizenshtadt, A;Olsen, P;Holmen, L;Høyem, M;Scholz, H;Grødeland, G;Sowa, S;Galera-Prat, A;Lehtiö, L;Meerts, I;Leenders, R;Wegert, A;Krauss, S;Waaler, J;
| DOI: 10.1158/2767-9764.crc-22-0027

The catalytic enzymes tankyrase 1 and 2 (TNKS1/2) alter protein turnover by poly-ADP-ribosylating target proteins, which earmark them for degradation by the ubiquitin-proteasomal system. Prominent targets of the catalytic activity of TNKS1/2 include AXIN proteins, resulting in TNKS1/2 being attractive biotargets for addressing of oncogenic WNT/β-catenin signaling. Although several potent small molecules have been developed to inhibit TNKS1/2, there are currently no TNKS1/2 inhibitors available in clinical practice. The development of tankyrase inhibitors has mainly been disadvantaged by concerns over biotarget-dependent intestinal toxicity and a deficient therapeutic window. Here we show that the novel, potent, and selective 1,2,4-triazole-based TNKS1/2 inhibitor OM-153 reduces WNT/β-catenin signaling and tumor progression in COLO 320DM colon carcinoma xenografts upon oral administration of 0.33-10 mg/kg twice daily. In addition, OM-153 potentiates anti-programmed cell death protein 1 (anti-PD-1) immune checkpoint inhibition and antitumor effect in a B16-F10 mouse melanoma model. A 28-day repeated dose mouse toxicity study documents body weight loss, intestinal damage, and tubular damage in the kidney after oral-twice daily administration of 100 mg/kg. In contrast, mice treated oral-twice daily with 10 mg/kg show an intact intestinal architecture and no atypical histopathologic changes in other organs. In addition, clinical biochemistry and hematologic analyses do not identify changes indicating substantial toxicity. The results demonstrate OM-153-mediated antitumor effects and a therapeutic window in a colon carcinoma mouse model ranging from 0.33 to at least 10 mg/kg, and provide a framework for using OM-153 for further preclinical evaluations. Significance: This study uncovers the effectiveness and therapeutic window for a novel tankyrase inhibitor in mouse tumor models.
Correlation of LGR5 expression and clinicopathological features in intrahepatic cholangiocarcinoma

Pathology - Research and Practice

2022 Mar 01

Yoshizawa, T;Uehara, T;Iwaya, M;Asaka, S;Kobayashi, S;Nakajima, T;Kinugawa, Y;Nagaya, T;Kamakura, M;Shimizu, A;Kubota, K;Notake, T;Masuo, H;Hosoda, K;Sakai, H;Hayashi, H;Umemura, K;Kamachi, A;Goto, T;Tomida, H;Yamazaki, S;Ota, H;Soejima, Y;
| DOI: 10.1016/j.prp.2022.153832

Leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) is a known cancer stem cell marker. However, there are no reported analyses of LGR5 mRNA expression in normal liver and liver cancer tissues. Here, we evaluated LGR5 expression by RNAscope, a newly developed RNA in situ hybridization technique, using a tissue microarray consisting of 25 samples of intrahepatic cholangiocarcinoma (ICC) selected from the medical archives at our hospital. LGR5 expression levels were divided into high and low expression groups by the five-grade scoring system, and clinicopathological features were analyzed. Low LGR5 expression was identified in some normal hepatocytes and bile duct cells. In addition, LGR5 expression was identified in all bile duct cancer samples except one case. Well-differentiated to moderately-differentiated adenocarcinoma tended to show higher LGR5 expression than poorly-differentiated adenocarcinoma (P=0.0561), and the large duct type showed significantly higher LGR5 expression levels than the small duct type (P=0.0225). Patients in the high LGR5 expression group tended to have good overall survival (OS) (P=0.0623). The Cox proportional hazard regression model revealed that the high LGR5 expression group showed independently better OS for ICC (P = 0.0285). High LGR5 expression is possibly a good prognosis factor in ICC. However, the detailed mechanism of LGR5 in this disease remains unclear, and further analysis is warranted.
A conditional transgenic mouse line for targeted expression of the stem cell marker LGR5

Dev Biol. 2015 May 20.

Norum HJ, Bergström Å, Andersson BA, Kuiper RV, Hoelzl MA, Sørlie T, Toftgård R.
PMID: 25990088 | DOI: canprevres.0090.2015.

LGR5 is a known marker of embryonic and adult stem cells in several tissues. In a mouse model, Lgr5+ cells have shown tumour-initiating properties, while in human cancers, such as basal cell carcinoma and colon cancer, LGR5 expression levels are increased: however, the effect of increased LGR5 expression is not fully understood. To study the effects of elevated LGR5 expression levels we generated a novel tetracycline-responsive, conditional transgenic mouse line expressing human LGR5, designated TRELGR5. In this transgenic line, LGR5 expression can be induced in any tissue depending on the expression pattern of the chosen transcriptional regulator. For the current study, we used transgenic mice with a tetracycline-regulated transcriptional transactivator linked to the bovine keratin 5 promoter (K5tTA) to drive expression of LGR5 in the epidermis. As expected, expression of human LGR5 was induced in the skin of double transgenic mice (K5tTA;TRELGR5). Inducing LGR5 expression during embryogenesis and early development resulted in macroscopically and microscopically detectable phenotypic changes, including kink tail, sparse fur coat and enlarged sebaceous glands. The fur and sebaceous gland phenotypes were reversible upon discontinued expression of transgenic LGR5, but this was not observed for the kink tail phenotype. There were no apparent phenotypic changes if LGR5 expression was induced at three weeks of age. The results demonstrate that increased expression of LGR5 during embryogenesis and the neonatal period alter skin development and homeostasis.
Selective and reversible suppression of intestinal stem cell differentiation by pharmacological inhibition of BET bromodomains.

Sci Rep.

2016 Feb 09

Nakagawa A, Adams CE, Huang Y, Hamarneh SR, Liu W, Von Alt KN, Mino-Kenudson M, Hodin RA, Lillemoe KD, Fernández-Del Castillo C, Warshaw AL, Liss AS.
PMID: 26856877 | DOI: 10.1038/srep20390

Absorptive and secretory cells of the small intestine are derived from a single population of Lgr5-expressing stem cells. While key genetic pathways required for differentiation into specific lineages have been defined, epigenetic programs contributing to this process remain poorly characterized. Members of the BET family of chromatin adaptors contain tandem bromodomains that mediate binding to acetylated lysines on target proteins to regulate gene expression. In this study, we demonstrate that mice treated with a small molecule inhibitor of BET bromodomains, CPI203, exhibit greater than 90% decrease in tuft and enteroendocrine cells in both crypts and villi of the small intestine, with no changes observed in goblet or Paneth cells. BET bromodomain inhibition did not alter the abundance of Lgr5-expressing stem cells in crypts, but rather exerted its effects on intermediate progenitors, in part through regulation of Ngn3 expression. When BET bromodomain inhibition was combined with the chemotherapeutic gemcitabine, pervasive apoptosis was observed in intestinal crypts, revealing an important role for BET bromodomain activity in intestinal homeostasis. Pharmacological targeting of BET bromodomains defines a novel pathway required for tuft and enteroendocrine differentiation and provides an important tool to further dissect the progression from stem cell to terminally differentiated secretory cell.

Post-fast refeeding enhances intestinal stem cell-mediated regeneration and tumourigenesis through mTORC1-dependent polyamine synthesis

Research square

2023 Jan 10

Imada, S;Shin, H;Khawaled, S;Meckelmann, S;Whittaker, C;Correa, R;Pradhan, D;Calibasi, G;Nascentes, LN;Allies, G;Wittenhofer, P;Schmitz, O;Roper, J;Vinolo, M;Cheng, CW;Tasdogan, A;Yilmaz, ÃM;
PMID: 36711807 | DOI: 10.21203/rs.3.rs-2320717/v1

For more than a century, fasting regimens have improved health, lifespan, and tissue regeneration in diverse organisms, including humans. However, how fasting and post-fast refeeding impact adult stem cells and tumour formation has yet to be explored in depth. Here, we demonstrate that post-fast refeeding increases intestinal stem cell (ISC) proliferation and tumour formation: Post-fast refeeding augments the regenerative capacity of Lgr5+ intestinal stem cells (ISCs), and loss of the tumour suppressor Apc in ISCs under post-fast refeeding leads to a higher tumour incidence in the small intestine and colon than in the fasted or ad libitum (AL) fed states. This demonstrates that post-fast refeeding is a distinct state. Mechanistically, we discovered that robust induction of mTORC1 in post-fast-refed ISCs increases protein synthesis via polyamine metabolism to drive these changes, as inhibition of mTORC1, polyamine metabolite production, or protein synthesis abrogates the regenerative or tumourigenic effects of post-fast refeeding. Thus, fast-refeeding cycles must be carefully considered when planning diet-based strategies for regeneration without increasing cancer risk, as post-fast refeeding leads to a burst not only in stem cell-driven regeneration but also in tumourigenicity.
Human distal lung maps and lineage hierarchies reveal a bipotent progenitor

Nature

2022 Apr 01

Kadur Lakshminarasimha Murthy, P;Sontake, V;Tata, A;Kobayashi, Y;Macadlo, L;Okuda, K;Conchola, AS;Nakano, S;Gregory, S;Miller, LA;Spence, JR;Engelhardt, JF;Boucher, RC;Rock, JR;Randell, SH;Tata, PR;
PMID: 35355018 | DOI: 10.1038/s41586-022-04541-3

Mapping the spatial distribution and molecular identity of constituent cells is essential for understanding tissue dynamics in health and disease. We lack a comprehensive map of human distal airways, including the terminal and respiratory bronchioles (TRBs), which are implicated in respiratory diseases1-4. Here, using spatial transcriptomics and single-cell profiling of microdissected distal airways, we identify molecularly distinct TRB cell types that have not-to our knowledge-been previously characterized. These include airway-associated LGR5+ fibroblasts and TRB-specific alveolar type-0 (AT0) cells and TRB secretory cells (TRB-SCs). Connectome maps and organoid-based co-cultures reveal that LGR5+ fibroblasts form a signalling hub in the airway niche. AT0 cells and TRB-SCs are conserved in primates and emerge dynamically during human lung development. Using a non-human primate model of lung injury, together with human organoids and tissue specimens, we show that alveolar type-2 cells in regenerating lungs transiently acquire an AT0 state from which they can differentiate into either alveolar type-1 cells or TRB-SCs. This differentiation programme is distinct from that identified in the mouse lung5-7. Our study also reveals mechanisms that drive the differentiation of the bipotent AT0 cell state into normal or pathological states. In sum, our findings revise human lung cell maps and lineage trajectories, and implicate an epithelial transitional state in primate lung regeneration and disease.
Loss of SUV420H2-dependent chromatin compaction drives right-sided colon cancer progression

Gastroenterology

2022 Nov 16

Boonsanay, V;Mosa, MH;Looso, M;Weichenhan, D;Ceteci, F;Pudelko, L;Lechel, A;Michel, CS;Künne, C;Farin, HF;Plass, C;Greten, FR;
PMID: 36402192 | DOI: 10.1053/j.gastro.2022.10.036

Epigenetic processes regulating gene expression contribute markedly to epithelial cell plasticity in colorectal carcinogenesis. The lysine methyltransferase SUV420H2 comprises an important regulator of epithelial plasticity and is primarily responsible for trimethylation of H4K20 (H4K20me3). Loss of H4K20me3 has been suggested as a hallmark of human cancer due to its interaction with DNMT1. However, the role of Suv4-20h2 in colorectal cancer is unknown.We examined the alterations in histone modifications in patient-derived colorectal cancer organoids. Patient-derived colorectal cancer organoids and mouse intestinal organoids were genetically manipulated for functional studies in PDX and orthotopic transplantation. Gene expression profiling, micrococcal nuclease assay and chromatin immunoprecipitation were performed to understand epigenetic regulation of chromatin states and gene expression in patient-derived and mouse intestinal organoids.Here, we found that reduced H4K20me3 levels occurred predominantly in right-sided patient-derived colorectal cancer organoids, which was associated with an increased chromatin accessibility. Re-compaction of chromatin by Methylstat, a histone demethylase inhibitor, resulted in reduced growth selectively in subcutaneously grown tumors derived from right-sided cancers. Using mouse intestinal organoids, we confirmed that Suv4-20h2-mediated H4K20me3 is required for maintaining heterochromatin compaction and to prevent R-loop formation. Cross-species comparison of Suv4-20h2 depleted murine organoids with right-sided colorectal cancer organoids revealed a large overlap of gene signatures involved in chromatin silencing, DNA methylation and stemness/Wnt signaling.Loss of Suv4-20h2-mediated H4K20me3 drives right-sided colorectal colorectal tumorigenesis through an epigenetically controlled mechanism of chromatin compaction. Our findings unravel a conceptually novel approach for subtype specific therapy of this aggressive form of colorectal cancer.
Does human endometrial LGR5 gene expression suggest the existence of another hormonally regulated epithelial stem cell niche?

Hum Reprod.

2018 Apr 10

Tempest N, Baker AM, Wright NA, Hapangama DK.
PMID: 29648645 | DOI: 10.1093/humrep/dey083

Abstract

STUDY QUESTION:

Is human endometrial leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) gene expression limited to the postulated epithelial stem cell niche, stratum basalis glands, and is it hormonally regulated?

SUMMARY ANSWER:

LGR5 expressing cells are not limited to the postulated stem cell niche but LGR5 expression is hormonally regulated.

WHAT IS KNOWN ALREADY:

The human endometrium is a highly regenerative tissue; however, endometrial epithelial stem cell markers are yet to be confirmed. LGR5 is a marker of stem cells in various epithelia.

STUDY DESIGN, SIZE, DURATION:

The study was conducted at a University Research Institute. Endometrial samples from 50 healthy women undergoing benign gynaecological surgery with no endometrial pathology at the Liverpool Women's hospital were included and analysed in the following six sub-categories; proliferative, secretory phases of menstrual cycle, postmenopausal, those using oral and local progestagens and samples for in vitro explant culture.

PARTICIPANTS/MATERIALS, SETTING, METHODS:

In this study, we used the gold standard method, in situ hybridisation (ISH) along with qPCR and a systems biology approach to study the location of LGR5 gene expression in full thickness human endometrium and Fallopian tubes. The progesterone regulation of endometrial LGR5 was examined in vivo and in short-term cultured endometrial tissue explants in vitro. LGR5 expression was correlated with epithelial proliferation (Ki67), and expression of previously reported epithelia progenitor markers (SOX9 and SSEA-1) immunohistochemistry (IHC).

MAIN RESULTS AND THE ROLE OF CHANCE:

LGR5 gene expression was significantly higher in the endometrial luminal epithelium than in all other epithelial compartments in the healthy human endometrium, including the endometrial stratum basalis (P < 0.05). The strongest SSEA-1 and SOX9 staining was observed in the stratum basalis glands, but the general trend of SOX9 and SSEA-1 expression followed the same cyclical pattern of expression as LGR5. Stratum functionalis epithelial Ki67-LI and LGR5 expression levels correlated significantly (r = 0.74, P = 0.01), however, they did not correlate in luminal and stratum basalis epithelium (r = 0.5 and 0.13, respectively). Endometrial LGR5 demonstrates a dynamic spatiotemporal expression pattern, suggesting hormonal regulation. Oral and local progestogens significantly reduced endometrial LGR5 mRNA levels compared with women not on hormonal treatment (P < 0.01). Our data were in agreement with in silico analysis of published endometrial microarrays.

LARGE SCALE DATA:

We did not generate our own large scale data but interrogated publically available large scale data sets.

LIMITATIONS, REASONS FOR CAUTION:

In the absence of reliable antibodies for human LGR5 protein and validated lineage markers for the various epithelial populations that potentially exist within the endometrium, our study does not formally characterise or examine the functional ability of the resident LGR5+ cells as multipotent.

WIDER IMPLICATIONS OF THE FINDINGS:

These data will facilitate future lineage tracing studies in the human endometrial epithelium; to identify the location of stem cells and further complement the in vitro functional studies, to confirm if the LGR5 expressing epithelial cells indeed represent the epithelial stem cell population.

The 26RFa (QRFP)/GPR103 neuropeptidergic system in mice relays insulin signalling into the brain to regulate glucose homeostasis

Diabetologia

2022 Apr 27

El Mehdi, M;Takhlidjt, S;Devère, M;Arabo, A;Le Solliec, MA;Maucotel, J;Bénani, A;Nedelec, E;Duparc, C;Lefranc, B;Leprince, J;Anouar, Y;Prévost, G;Chartrel, N;Picot, M;
PMID: 35476025 | DOI: 10.1007/s00125-022-05706-5

26RFa (pyroglutamilated RFamide peptide [QRFP]) is a biologically active peptide that regulates glucose homeostasis by acting as an incretin and by increasing insulin sensitivity at the periphery. 26RFa is also produced by a neuronal population localised in the hypothalamus. In this study we investigated whether 26RFa neurons are involved in the hypothalamic regulation of glucose homeostasis.26Rfa+/+, 26Rfa-/- and insulin-deficient male C57Bl/6J mice were used in this study. Mice received an acute intracerebroventricular (i.c.v.) injection of 26RFa, insulin or the 26RFa receptor (GPR103) antagonist 25e and were subjected to IPGTTs, insulin tolerance tests, acute glucose-stimulated insulin secretion tests and pyruvate tolerance tests (PTTs). Secretion of 26RFa by hypothalamic explants after incubation with glucose, leptin or insulin was assessed. Expression and quantification of the genes encoding 26RFa, agouti-related protein, the insulin receptor and GPR103 were evaluated by quantitative reverse transcription PCR and RNAscope in situ hybridisation.Our data indicate that i.c.v.-injected 26RFa induces a robust antihyperglycaemic effect associated with an increase in insulin production by the pancreatic islets. In addition, we found that insulin strongly stimulates 26Rfa expression and secretion by the hypothalamus. RNAscope experiments revealed that neurons expressing 26Rfa are mainly localised in the lateral hypothalamic area, that they co-express the gene encoding the insulin receptor and that insulin induces the expression of 26Rfa in these neurons. Concurrently, the central antihyperglycaemic effect of insulin is abolished in the presence of a GPR103 antagonist and in 26RFa-deficient mice. Finally, our data indicate that the hypothalamic 26RFa neurons are not involved in the central inhibitory effect of insulin on hepatic glucose production, but mediate the central effects of the hormone on its own peripheral production.We have identified a novel mechanism in the hypothalamic regulation of glucose homeostasis, the 26RFa/GPR103 system, and we provide evidence that this neuronal peptidergic system is a key relay for the central regulation of glucose metabolism by insulin.
Suppression of intestinal tumorigenesis in Apc mutant mice by Musashi-1 deletion.

J Cell Sci.

2017 Jan 12

Wolfe AR, Ernlund A, McGuinness W, Lehmann C, Carl K, Balmaceda N, Neufeld KL.
PMID: 28082422 | DOI: 10.1242/jcs.197574

Therapeutic strategies based on a specific oncogenic target are better justified when elimination of that particular oncogene reduces tumorigenesis in a model organism. One such oncogene, Musashi 1 (Msi-1), regulates translation of target mRNAs and is implicated in promoting tumorigenesis in colon and other tissues. Msi-1 targets include the tumor suppressor Adenomatous Polyposis Coli (Apc), a Wnt pathway antagonist lost in ∼80% of all colorectal cancers. Cell culture experiments have established that Msi-1 is a Wnt target, thus positioning Msi-1 and Apc as mutual antagonists in a double negative feedback loop. Here, we report that intestines from mice lacking Msi-1 display aberrant Apc/Msi-1 double negative feedback, reduced Wnt and Notch signaling, decreased proliferation, and changes in stem cell populations, features predicted to suppress tumorigenesis. Indeed, ApcMin and Apc1322T mice have a dramatic reduction in intestinal polyp number when Msi-1 is deleted. Together, these results provide genetic evidence that Msi-1 contributes to intestinal tumorigenesis driven by Apc loss, and validate the pursuit of Msi-1 inhibitors as chemo-prevention agents to reduce tumor burden.

Pages

  • « first
  • ‹ previous
  • 1
  • 2
  • 3
  • 4
  • 5
  • 6
  • 7
  • 8
  • 9
  • …
  • 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?