Noggin Mouse

Noggin Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Neural and Skeletal Development

  • Neural Tube Patterning: Noggin inhibits BMP-4 to promote dorsal structures like the neural plate . Transgenic NSE-Noggin mice exhibit hyperinnervation of the enteric nervous system (ENS), leading to increased fecal output and altered gastrointestinal motility .

  • Joint Formation: Knockout models display fused joints (symphalangism) and vertebral malformations, mimicking human SYNS1/SYM1 syndromes .

Bone Homeostasis

  • Overexpression in osteoblasts reduces bone density (osteoporosis), while deletion causes osteopenia via dysregulated BMP signaling .

Neuroprotection in Stroke Models

  • Infarct Reduction: NSE-Noggin mice showed 50% smaller infarct volumes post-middle cerebral artery occlusion (p < 0.01) .

  • Oligodendrocyte Progenitor Recruitment: Enhanced platelet-derived growth factor receptor-α+ cells in ischemic zones, suggesting white matter repair mechanisms .

Adipose Tissue Regulation

  • Plasma Noggin levels correlate with obesity in mice (BMI >27 equivalents). Adipose-specific depletion exacerbates weight gain, implicating Noggin in metabolic regulation .

Recombinant Mouse Noggin Products

Commercial variants differ in formulation and applications:

Product (Supplier)FormulationApplicationsActivity (ED₅₀)
R&D Systems 1967-NG Lyophilized with BSACell culture, ELISA standards0.06–0.3 µg/mL (BMP-4 inhibition)
BioLegend 787704 Carrier-free (citric acid)Stem cell maintenance, neuroprotection assays0.04–0.20 µg/mL (BMP-4 inhibition)
KACTUS NOG-MM601 HEK293-expressed, His/Flag-taggedCancer research (gastric cell proliferation)N/A

Clinical and Experimental Implications

  • Disease Models: NSE-Noggin mice are used to study gut hyperinnervation’s role in colorectal cancer and inflammation .

  • Therapeutic Potential: Noggin delivery enhances neural stem cell survival in hippocampal regions, suggesting applications in neurodegenerative diseases .

Genetic and Functional Insights

  • Mutations: Missense mutations (e.g., Val187Ile) disrupt BMP binding, causing joint fusion pathologies .

  • Transcriptional Regulation: Noggin-LacZ reporter mice reveal dynamic expression in adult cartilage, implicating BMP antagonism in osteoarthritis progression .

Product Specs

Introduction
Noggin, a secreted polypeptide encoded by the NOG gene, plays a crucial role in embryonic development and bone formation. It inhibits signaling proteins in the transforming growth factor-beta (TGF-beta) superfamily, including bone morphogenetic protein-4 (BMP4). Noggin's ability to diffuse efficiently through extracellular matrices allows it to establish morphogenic gradients. This protein exhibits pleiotropic effects throughout development. Initially identified in Xenopus, noggin restored normal dorsal-ventral body axis in embryos with UV-induced ventralization. Mouse knockout studies underscore noggin's involvement in neural tube fusion, joint formation, and other developmental processes. Notably, dominant human NOG mutations are linked to proximal symphalangism (SYM1) and multiple synostoses syndrome (SYNS1), both characterized by multiple joint fusions. These syndromes map to chromosome 17q22, the same region as NOG. Importantly, all NOG mutations affect evolutionarily conserved amino acid residues. Human noggin shares significant homology with its Xenopus, rat, and mouse counterparts.
Description
Recombinant Mouse Noggin, produced in E. coli, is a non-glycosylated protein with two polypeptide chains linked by disulfide bonds. Each chain comprises 206 amino acids, resulting in a total molecular weight of approximately 46.4 kDa (23.2 kDa per chain).
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
Lyophilized from a 0.2µm filtered solution containing 30% acetonitrile and 0.1% TFA.
Solubility
Before opening, briefly centrifuge the product to collect contents at the bottom. Reconstitute in 10mM HAc to achieve a concentration of 0.1-1.0 mg/ml. For further dilutions, use appropriate buffered solutions.
Stability
Lyophilized Mouse Noggin remains stable at room temperature for 3 weeks. However, it is recommended to store desiccated below -18°C. Upon reconstitution, store Mouse Noggin at 4°C for 2-7 days or below -18°C for future use. For long-term storage, add a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
Purity
Determined by SDS-PAGE, purity is greater than 95.0%.
Biological Activity
The ED50, determined by inhibiting BMP-4-induced alkaline phosphatase production in murine ATDC5 cells, is less than 2ng/ml. This translates to a specific activity exceeding 5.0 x 10^5 IU/mg in the presence of 5ng/ml BMP-4.
Synonyms
Noggin, SYM1, SYNS1, NOG.
Source
Escherichia Coli.
Amino Acid Sequence
MQHYLHIRPAPSDNLPLVDLIEHPDPIFDPKEKDLNETLLRSLLGGHYD
PGFMATSPPEDRPGGGGGPAGGAEDLAELDQLLRQRPSGAMPSEIKG
LEFSEGLAQGKKQRLSKKLRRKLQMWLWSQTFCPVLYAWNDLGSRF
WPRYVKVGSCFSKRSCSVPEGMVCKPSKSVHLTVLRWRCQRRGQR
CGWIPIQYPIISECKCSC.

Q&A

What is Noggin and what are its primary functions in mouse development?

Noggin is a secreted glycoprotein that functions as a BMP antagonist by binding to BMPs (particularly BMP2, BMP4, and BMP7) and preventing their interaction with receptors. In mouse development, Noggin plays crucial roles in neural induction, skeletal development, and organogenesis. It functions by inhibiting BMP signaling, which creates balanced gradient signals necessary for proper tissue patterning and cell fate decisions. Noggin's activity is particularly important in neural development, where it promotes neurogenesis by antagonizing BMP-mediated astrocytic differentiation . Additionally, Noggin is involved in tooth development, prostate morphogenesis, and enteric nervous system formation .

What are the main types of Noggin mouse models available for research?

Several Noggin mouse models have been developed for research purposes:

  • Noggin knockout mice (Noggin-/-): These mice completely lack Noggin expression and exhibit severe developmental defects, including failure of neural tube closure and abnormal skeletal development .

  • Conditional Noggin knockout mice: These models allow tissue-specific deletion of Noggin using Cre-loxP technology.

  • Noggin overexpression models: Includes mice with tissue-specific Noggin overexpression, such as NSE-Noggin mice (neuron-specific enolase promoter driving Noggin) .

  • Reporter mouse lines: Noggin-LacZ knockin mice that allow visualization of Noggin expression patterns .

  • DMP1-Cre/pMes-Nog mice: These mice overexpress Noggin in differentiated odontoblasts and osteocytes, useful for studying tooth development .

Each model provides different advantages depending on the research question being investigated.

How does Noggin expression vary across different tissues in adult mice?

Noggin expression in adult mice shows tissue-specific patterns that often reflect its continued role in tissue homeostasis. According to studies using Noggin-LacZ reporter mice, Noggin is expressed in:

  • Articular cartilage: Particularly in joints, where it helps maintain cartilage integrity .

  • Skeletal elements: Including sutures in the skull and ribs .

  • Neural tissues: Including specific populations of neurons .

  • Dental tissues: In odontoblasts and dental pulp .

  • Prostate: In the mesenchyme surrounding the urogenital sinus .

  • Intervertebral discs and pubic symphysis: Suggesting roles in maintaining these specialized cartilage tissues .

The expression pattern indicates that Noggin continues to regulate BMP signaling in adult tissues, particularly in those undergoing constant remodeling or containing stem cell populations.

How should I design experiments to study neurogenesis using Noggin mouse models?

When designing experiments to study neurogenesis using Noggin mouse models, consider the following approach:

  • Select appropriate model: For enhanced neurogenesis, use NSE-Noggin mice or devise a method to deliver exogenous Noggin. For reduced neurogenesis, use Noggin conditional knockouts or heterozygotes.

  • Age considerations: Select appropriate age points based on your research question. For adult neurogenesis studies, 3-6 month old mice are typically used, while developmental studies require specific embryonic or early postnatal timepoints.

  • Tissue preparation: For neural stem cell analysis, both in vivo and ex vivo approaches can be used:

    • In vivo: BrdU/EdU labeling followed by immunohistochemistry for cell proliferation and differentiation markers

    • Ex vivo: Isolation of neural stem cells for neurosphere assays

  • Analysis methods:

    • Immunohistochemistry for neural stem cell markers (Nestin, Sox2) and differentiation markers (DCX, NeuN)

    • BrdU/EdU labeling to track proliferation and differentiation

    • Behavioral testing to correlate cellular changes with functional outcomes

  • Controls: Always include age-matched wild-type littermates as controls to account for strain background effects .

What are the key considerations when designing experiments with Noggin overexpression mice?

When working with Noggin overexpression mice:

  • Expression verification: Always confirm Noggin overexpression using:

    • RT-PCR for mRNA levels

    • Western blot or ELISA for protein levels

    • Reporter gene expression (e.g., EGFP in NSE-Noggin-IRES-EGFP constructs)

  • Phenotype characterization: Systematically assess:

    • Gross anatomical changes

    • Cellular composition of target tissues

    • Molecular markers for altered BMP signaling (reduced phosphorylated SMAD1/5/8)

    • Functional consequences using appropriate behavioral or physiological tests

  • Dosage effects: Consider that different levels of Noggin may yield different phenotypes:

    • Use heterozygous and homozygous transgenic mice to assess dose-dependent effects

    • Compare tissue-specific versus global overexpression

  • Timing considerations:

    • For inducible systems, test different induction timepoints to distinguish developmental versus homeostatic roles

    • Include time-course analyses to capture dynamic changes

  • Environmental variables: Control for environmental factors that may influence phenotypes, including microbiota composition, which has been suggested to affect outcomes in enteric nervous system studies .

How can I effectively use Noggin to promote dopaminergic neuron differentiation from stem cells?

To effectively use Noggin for promoting dopaminergic neuron differentiation:

  • Timing of Noggin administration:

    • Early treatment (days 0-7 for mouse ESCs, days 0-9 for human ESCs) during differentiation is effective

    • Continuous treatment throughout differentiation shows even better results

  • Culture system optimization:

    • Use PA6 stromal cell co-culture method with ESCs

    • Supplement with recombinant Noggin protein (typically 100-200 ng/ml)

    • Consider combining with other factors (SHH, FGF8) for synergistic effects

  • Verification methods:

    • Immunostain for tyrosine hydroxylase (TH) to identify dopaminergic neurons

    • Quantify TH+ cell numbers and morphology

    • Assess dopamine production using HPLC or ELISA

    • Evaluate electrophysiological properties of differentiated neurons

  • Transplantation considerations:

    • Pre-treat cells with Noggin before transplantation

    • Use immunosuppression (e.g., cyclosporin) for xenotransplantation

    • Evaluate functional recovery using behavioral tests (e.g., apomorphine-induced rotation)

How do I interpret contradictory phenotypes between different Noggin mouse models?

When encountering contradictory phenotypes:

  • Genetic background effects:

    • Different mouse strains may have modifier genes affecting Noggin-related phenotypes

    • Backcross to a common genetic background for direct comparisons

    • Use littermate controls to minimize background effects

  • Expression level considerations:

    • Verify actual levels of Noggin expression/inhibition in your specific model

    • Different promoters drive different expression patterns and levels

    • Document protein levels in relevant tissues using Western blot or ELISA

  • Compensatory mechanisms:

    • Assess expression of other BMP antagonists (Chordin, Gremlin1) that might compensate for Noggin alterations

    • Examine BMP receptor and downstream signaling activation states

  • Developmental timing:

    • Phenotypes may differ depending on when Noggin function is altered

    • Early embryonic loss may trigger different compensatory mechanisms than adult-onset loss

  • Environmental variables:

    • Housing conditions, diet, and microbiome can affect phenotypes

    • The NSE-Noggin mouse, for example, showed different enteric neuronal density outcomes in different laboratory environments

What phenotypic changes should I expect in the dentition of Noggin overexpression mice?

In Noggin overexpression mice targeting dental tissues (e.g., DMP1-Cre/pMes-Nog mice), expect the following changes:

A detailed quantitative comparison is presented in the table below:

ParameterWild-typeNoggin Overexpression% Change
Total tooth volumeBaselineRelatively unchanged~0%
Dentin volumeBaselineReduced-12%
Dentin volume/tooth volumeBaselineReduced-8%
Pulp volumeBaselineIncreased+20%
Apparent densityBaselineReducedSignificant
Material densityBaselineUnchanged~0%

How does Noggin affect neural stem cell behavior in aging and neurodegenerative models?

Noggin has significant effects on neural stem cell (NSC) behavior in aging and neurodegenerative contexts:

  • Aging effects:

    • In the SAMP8 senescence-accelerated mouse model, an age-related increase in BMP6 correlates with premature NSC loss

    • Noggin treatment rescues the NSC pool in aged SAMP8 mice, indicating BMP signaling mediates age-related stem cell depletion

  • Neurogenesis regulation:

    • Noggin treatment restores neurogenesis in aged mice

    • Treatment normalizes behavioral phenotypes in SAMP8 mice to match control SAMR1 animals

  • Cellular mechanisms:

    • Noggin blocks the age-related shift from neurogenesis to astroglial differentiation

    • It prevents premature NSC pool depletion by maintaining NSC self-renewal capacity

    • Noggin counteracts enhanced canonical BMP signaling seen in aged mice

  • Potential therapeutic implications:

    • Noggin or similar BMP antagonists may protect hippocampal function during aging

    • These findings suggest therapeutic strategies for neurodegenerative conditions like Alzheimer's disease, where hippocampal dysfunction is prominent

What are the most effective methods to quantify changes in BMP signaling in Noggin mouse models?

To effectively quantify BMP signaling changes:

  • Phosphorylated SMAD1/5/8 analysis:

    • Western blot analysis of pSMAD1/5/8 levels (primary readout of canonical BMP signaling)

    • Immunohistochemistry to visualize spatial distribution of pSMAD1/5/8

    • Flow cytometry for single-cell quantification in dissociated tissues

  • Transcriptional targets:

    • qRT-PCR for BMP target genes (ID1, ID2, ID3, MSX1/2)

    • RNA-seq for genome-wide transcriptional changes

    • In situ hybridization for spatial resolution of target gene expression

  • Reporter systems:

    • BRE-lacZ or BRE-GFP reporter mice (containing BMP responsive elements)

    • Cross these reporters with your Noggin mouse model

  • Protein-level analyses:

    • ELISA or mass spectrometry to quantify BMP ligand levels

    • Co-immunoprecipitation to assess Noggin-BMP binding

    • Proximity ligation assays to visualize protein interactions in situ

  • Functional readouts:

    • Cell proliferation assays (BrdU/EdU incorporation)

    • Differentiation marker analysis

    • Apoptosis assessment (TUNEL, cleaved caspase-3)

How can I establish the optimal dosage of Noggin for experimental applications?

To establish optimal Noggin dosage:

  • In vitro dose-response experiments:

    • Test recombinant Noggin protein at concentrations ranging from 50-500 ng/ml

    • Measure outcomes including:

      • Cell proliferation

      • Differentiation marker expression

      • BMP signaling inhibition (pSMAD1/5/8 levels)

    • Plot dose-response curves to identify minimum effective concentration

  • In vivo administration optimization:

    • For local delivery, test 100-1000 ng/μl range in initial studies

    • For systemic delivery, begin with 0.5-5 mg/kg range

    • Consider delivery methods:

      • Osmotic minipumps for continuous delivery

      • Viral vectors for sustained local expression

      • Direct injection for acute effects

  • Timing considerations:

    • Test both early (e.g., days 0-7 in mouse ESCs) and continuous treatment regimens

    • Include washout periods to assess persistence of effects

  • Biological indicators of appropriate dosing:

    • Reduction in pSMAD1/5/8 levels by 50-80%

    • Rescue of phenotypes in BMP overexpression models

    • Absence of non-specific effects on other signaling pathways

  • Model-specific considerations:

    • Cell culture: 100-200 ng/ml is typical for neural differentiation protocols

    • For transplantation studies: pre-treatment of cells with Noggin before transplantation enhances outcomes

What techniques can I use to study the interaction between Noggin and other BMP antagonists?

To study interactions between Noggin and other BMP antagonists:

  • Co-expression analysis:

    • Dual immunofluorescence for Noggin and other antagonists (Chordin, Gremlin1, etc.)

    • Single-cell RNA-seq to identify co-expressing cell populations

    • Reporter mice comparison (e.g., Noggin-LacZ vs. Gremlin1-LacZ) to map overlapping expression domains

  • Functional interaction studies:

    • Generate compound mutants (e.g., Noggin+/- × Chordin+/-)

    • Use combined treatment with multiple recombinant antagonists

    • Develop and test compensatory responses by knocking down one antagonist and measuring changes in others

  • Biochemical interaction analysis:

    • Co-immunoprecipitation to detect protein-protein interactions

    • Surface plasmon resonance to measure binding affinities

    • Proximity ligation assays to visualize protein proximity in situ

  • Competitive binding assays:

    • ELISA-based competition assays with labeled BMPs

    • Assess differential affinities for various BMP ligands

    • Measure changes in downstream signaling with combinations of antagonists

  • Spatial analysis in tissues:

    • 3D reconstruction of expression domains using confocal microscopy

    • Create BMP signaling maps using pSMAD1/5/8 immunostaining

    • Correlate with expression of multiple antagonists

Why might I observe different phenotypes than previously reported in my Noggin mouse strain?

Several factors can explain phenotypic differences:

  • Genetic background drift:

    • Mouse strains can undergo genetic drift over generations

    • Background mutations may accumulate and modify phenotypes

    • Solution: Regularly backcross to refresh genetic background

  • Environmental variables:

    • Microbiome composition can substantially affect outcomes, especially in enteric nervous system studies

    • Housing conditions, diet, and stress levels impact phenotypes

    • Solution: Standardize housing conditions and report detailed environmental parameters

  • Transgene expression variations:

    • Transgene silencing can occur over generations

    • Copy number variations affect expression levels

    • Solution: Regularly verify transgene expression levels with qPCR and protein assays

  • Methodology differences:

    • Differences in tissue preparation, fixation, antibodies

    • Quantification approaches may vary between labs

    • Solution: Replicate original methods precisely or acknowledge methodological differences

  • Age and sex differences:

    • Phenotypes may be age and sex-dependent

    • Solution: Match age and sex in experimental groups and include both sexes in studies

How can I address compensatory mechanisms when studying Noggin knockout or overexpression models?

To address compensatory mechanisms:

  • Measure expression of other BMP antagonists:

    • Assess changes in Chordin, Gremlin1/2, Follistatin, and other antagonists

    • Use qRT-PCR, Western blot, and immunohistochemistry to detect compensatory upregulation

  • Temporal analysis:

    • Conduct time-course studies from early embryonic stages through adulthood

    • Identify when compensatory mechanisms begin to emerge

    • Consider inducible models to bypass developmental compensation

  • Compound genetic approaches:

    • Generate double knockouts or combined overexpression models

    • Example: Noggin+/- × Bmp4+/- mice show partial phenotypic rescue compared to Noggin-/- alone

    • Create conditional knockouts of multiple antagonists in the same tissue

  • Pathway analysis beyond SMAD signaling:

    • Assess non-canonical BMP signaling pathways (MAPK, PI3K/AKT)

    • Examine cross-talk with other signaling pathways (WNT, Hedgehog, FGF)

  • Single-cell approaches:

    • Use single-cell RNA-seq to identify cell-specific compensatory mechanisms

    • Perform single-cell signaling analyses to detect heterogeneous responses

What controls are essential when performing transplantation studies with Noggin-treated cells?

For transplantation studies with Noggin-treated cells, include these essential controls:

  • Cell preparation controls:

    • Untreated cells from the same preparation

    • Cells treated with heat-inactivated Noggin protein

    • Cells treated with an irrelevant protein of similar size

    • Pre-transplantation quality assessment (viability, purity, marker expression)

  • Surgical controls:

    • Sham surgery (procedure without cell delivery)

    • Vehicle-only injections

    • Transplantation of non-neuronal cells or fibroblasts

  • Immunosuppression controls:

    • If using immunosuppression (e.g., cyclosporin), include both immunosuppressed and non-immunosuppressed groups

    • Monitor immune response around grafts with microglial/macrophage markers

  • Functional assessment controls:

    • Baseline behavioral measurements pre-surgery

    • Include both lesioned/diseased non-transplanted animals and healthy controls

    • For apomorphine-induced rotation tests in Parkinson's models, include dose-response measures

  • Analytical controls:

    • Stereological counting with appropriate sampling parameters

    • Double-blind assessment of behavioral and histological outcomes

    • Multiple timepoints post-transplantation to assess survival dynamics

Product Science Overview

Structure and Function

Noggin is a disulfide-linked homodimer, meaning it consists of two identical subunits connected by disulfide bonds . It functions primarily as an antagonist to Bone Morphogenetic Proteins (BMPs), particularly BMP-4, but also modulates the activities of other BMPs such as BMP-2, BMP-7, BMP-13, and BMP-14 . By binding to these BMPs, Noggin prevents them from interacting with their receptors, thereby inhibiting their signaling pathways .

Role in Development

Noggin’s ability to inhibit BMP signaling is critical during embryonic development. It helps regulate the formation of the neural tube, somites, and other structures by controlling the balance between BMP signaling and other pathways. This balance is essential for proper tissue differentiation and organogenesis .

Recombinant Mouse Noggin

Recombinant Mouse Noggin is produced using a mouse myeloma cell line (NS0) and is typically purified to a high degree of purity (>90%) using SDS-PAGE under reducing conditions . The recombinant protein is often used in research to study its effects on cell differentiation, tissue culture, and various bioassays.

Applications

Recombinant Mouse Noggin is widely used in scientific research due to its ability to inhibit BMP-induced processes. Some of its applications include:

  • Cell Culture: Used to study the effects of BMP inhibition on various cell types.
  • Bioassays: Employed in assays to measure its ability to inhibit BMP-induced alkaline phosphatase production in chondrogenic cells .
  • Tissue Engineering: Investigated for its potential in promoting tissue regeneration and repair by modulating BMP signaling.
Storage and Stability

Recombinant Mouse Noggin is typically lyophilized and can be reconstituted in sterile PBS. It is stable for up to 12 months when stored at -20 to -70°C and for shorter periods at 2 to 8°C after reconstitution .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.