SHH Mouse

Sonic Hedgehog Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Isoforms

SHH is synthesized as a 437-amino-acid precursor protein that undergoes autocatalytic cleavage into two fragments:

  • N-terminal peptide (Shh-N): The biologically active fragment (~19–20 kDa) responsible for signaling .

  • C-terminal peptide (Shh-C): Mediates precursor cleavage and is rapidly degraded post-processing .

Key Modifications:

  • Cholesterol addition: Enhances membrane tethering and multimerization, critical for gradient formation and signaling potency .

  • Palmitoylation: Further stabilizes Shh-N activity .

PropertyDetails
Molecular Weight (Shh-N)19–20 kDa (non-glycosylated)
IsoformsRecombinant forms (e.g., C25II mutant) with enhanced bioactivity
Expression SitesOligodendrocytes, hypothalamic neurons, floor plate, limb bud mesenchyme

Biological Functions

SHH signaling regulates:

  • Embryonic Development:

    • Neural tube ventralization and limb patterning .

    • Hypothalamic specification, with Shh knockouts causing holoprosencephaly (single forebrain ventricle) .

  • Adult Roles:

    • Oligodendrocyte-derived SHH maintains astrocyte and neuronal circuits .

    • Modulates neural stem cell proliferation and tissue repair .

Mechanism: SHH binds Patched (Ptc), relieving Smoothened (Smo) repression and activating Gli transcription factors .

Research Models and Phenotypes

Mouse models elucidate SHH’s developmental and pathological roles:

ModelPhenotypeReference
Shh global KOHoloprosencephaly, absent ventral neural tube structures, limb defects
Nkx2-1-Cre conditional KOHypothalamic hypoplasia, loss of hypocretin neurons
Pink1 KO (PD model)Ciliary defects, dysregulated SHH signaling, mitochondrial dysfunction
Oligodendrocyte-specific SHHGrid-like distribution in adult brain; regulates astrocyte-neuron crosstalk

SHH in Disease and Therapeutics

  • Parkinson’s Disease (PD): SHH signaling hyperactivity in Pink1-deficient models contributes to ciliary shortening and mitochondrial defects, reversible via SHH inhibition .

  • Cancer: Aberrant SHH activation drives tumors in breast, brain, and liver .

  • Therapeutic Targets: Recombinant SHH-N (e.g., Catalog #464-SH) induces dopaminergic neuron differentiation in vitro .

Recombinant SHH Applications

Commercial SHH proteins are vital for in vitro studies:

ProductActivityUse Cases
Recombinant Shh-N (C25II)ED₅₀: 0.05–0.25 µg/mL; induces alkaline phosphatase in C3H10T1/2 fibroblasts Stem cell differentiation
Carrier-free Shh-NMonomeric form (20.7 kDa) with >30x potency vs. competitors Neural patterning assays

Product Specs

Introduction
Recombinant Mouse Sonic Hedgehog is a member of a small family of secreted proteins crucial for development in both vertebrates and invertebrates. The three mammalian hedgehog genes (sonic, desert, and Indian) exhibit approximately 60% homology. Notably, Mouse Sonic Hedgehog shares 99% homology with its human counterpart. Sonic Hedgehog plays a vital role in guiding early embryonic development and has been identified as a primary inductive signal in the patterning of the ventral neural tube, the anterior-posterior limb axis, and ventral somites. Binding to the Patched receptor, Sonic Hedgehog, in conjunction with Smoothened, activates the transcription of target genes. In the absence of Sonic Hedgehog, the Patched receptor suppresses the inherent signaling activity of Smoothened. Furthermore, Sonic Hedgehog regulates the Gli oncogene. This intercellular signaling molecule is essential for various developmental patterning processes, including ventral cell fate induction in the neural tube and somites by signals from the notochord and anterior-posterior axis polarization in the developing limb bud. Sonic Hedgehog exhibits both floor plate- and motor neuron-inducing activities. Mutations in a long-range enhancer of Sonic Hedgehog, located within an intron of the limb region 1 gene, can lead to preaxial polydactyly.
Description
Recombinant Mouse Sonic Hedgehog, produced in E. coli, is an unglycosylated polypeptide chain composed of 176 amino acids, with a molecular weight of 19.8 kDa. It shares 99% homology with the human Sonic Hedgehog gene. The cysteine residue at position 25 has been replaced with isoleucine. Purification of Sonic Hedgehog is achieved through proprietary chromatographic methods.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
Lyophilized from a 1 mg/mL solution in 10 mM sodium phosphate buffer (pH 7.5).
Solubility
Reconstitute the lyophilized Mouse Sonic Hedgehog in sterile 18 MΩ·cm H₂O to a concentration of at least 100 µg/mL. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized Mouse Sonic Hedgehog remains stable at room temperature for up to 3 weeks. However, for long-term storage, it should be kept desiccated below -18 °C. After reconstitution, store Sonic Hedgehog at 4 °C for 2-7 days. For extended storage, freeze aliquots below -18 °C. Avoid repeated freeze-thaw cycles.
Purity
Greater than 97.0% as determined by SDS-PAGE.
Biological Activity
The biological activity is determined by the dose-dependent induction of alkaline phosphatase production in C3H/10T1/2 (CCL-226) fibroblasts and is typically 0.48-0.72 µg/mL.
Synonyms
SHH, HHG-1, HHG1, Sonic hedgehog protein.
Source
Escherichia Coli.
Amino Acid Sequence
MIIGPGRGFG KRRHPKKLTP LAYKQFIPNV AEKTLGASGR YEGKITRNSE RFKELTPNYN PDIIFKDEEN TGADRLMTQR CKDKLNALAI SVMNQWPGVK LRVTEGWDED GHHSEESLHY EGRAVDITTS DRDRSKYGML ARLAVEAGFD WVYYESKAHI HCSVKAENSV AAKSGG.

Q&A

What is Sonic Hedgehog (Shh) and what related proteins exist in mice?

Sonic Hedgehog (Shh) is a member of the Hedgehog (Hh) family of highly conserved proteins widely represented throughout the animal kingdom . In mammals, three related Hedgehog proteins exist: Sonic (Shh), Desert (Dhh), and Indian (Ihh), which share a high degree of amino acid sequence identity (Shh and Ihh are 93% identical) . Shh plays critical roles in cell growth, cell specialization, and the normal shaping (patterning) of the body . It is particularly important for development of the brain and spinal cord (central nervous system), eyes, limbs, and many other parts of the body .

The protein undergoes important post-translational modifications, including palmitoylation and cholesterol addition, which contribute to its membrane tethering and assembly into multimers . These lipid modifications significantly increase Shh signaling potency . Release from the plasma membrane occurs through the cooperative action of several proteins including DISP1, SCUBE2, and TACE/ADAM17 .

Where is Shh expressed in the adult mouse brain?

Recent research using single-molecule fluorescent in situ hybridization (smFISH) has revealed that Shh expression in the adult mouse brain is much broader than originally reported . Shh transcripts have been detected in almost all brain regions, with varying densities classified as very high (>30 dots/cell), high (10-30 dots/cell), moderate (6-9 dots/cell), low (2-5 dots/cell), or undetectable (0-1 dot/cell) .

Studies have identified Shh mRNA in HuC/D-positive neurons and in a subset of cells expressing oligodendroglial markers Olig2 and Sox10 . A previously unrecognized population of neurons co-expressing Shh transcripts and the nitrergic marker nNOS has been identified, alongside broad expression in hypothalamic nuclei . This extensive distribution pattern suggests potential new roles for Shh in the regulation of neural circuits beyond its well-established developmental functions .

How does Shh function as a morphogen in mouse neural development?

In neural development, Shh functions as a classic morphogen, creating a concentration gradient that specifies different cell fates . The protein is produced by the notochord and floor plate, establishing a ventral HIGH–dorsal LOW gradient throughout the neural tube . This gradient directly controls gene expression and cell fate decisions in the ventral neural tube .

Experimental evidence using naïve neural tissue explants demonstrated that progressive 2- to 3-fold changes in Shh concentration induce distinct neuronal subtypes . There is a strong correlation between the concentration of Shh necessary to induce each neuronal subtype and their position of generation in vivo . Neurons generated in more ventral regions of the neural tube require correspondingly higher Shh concentrations .

The direct action of Shh across long distances was confirmed through mosaic expression of dominant inhibitors of its receptor Patched (Ptc) . When signaling was blocked in specific cells, it resulted in cell-autonomous inhibition of ventral neural tube cell types . Additionally, the blockade caused more dorsally positioned cells to respond as if exposed to higher Shh concentrations, indicating that feedback mechanisms normally limit Shh spread through tissues .

How does the Shh signaling pathway function in mouse embryonic stem cells?

In mouse embryonic stem cells (mESCs), Shh signaling operates through several interconnected mechanisms that regulate proliferation, migration, and differentiation . The pathway begins with Shh disrupting adherens junctions through proteolysis by activating matrix metalloproteinases (MMPs) . This disruption leads to the release of β-catenin from adherens junctions, which then mediates cell cycle-dependent mESC proliferation .

Shh-mediated Gli1 expression leads to integrin β1 up-regulation, followed by focal adhesion kinase (FAK) and Src phosphorylation . Among Rho-GTPases, Rac1 and Cdc42 are specifically activated in a Shh-dependent manner, leading to F-actin formation essential for cell motility . This F-actin formation is suppressed when cells are transfected with Rac1 and Cdc42 siRNA, confirming the pathway specificity .

The functional importance of this signaling cascade extends to in vivo applications. In skin wound healing assays, Shh-treated mESCs increased angiogenesis and improved wound repair compared to controls . Notably, this effect was diminished when Shh-treated mESCs were transfected with integrin β1 siRNA, demonstrating the essential role of integrin β1 in mediating Shh's regenerative effects .

What role does Patched play in Shh gradient formation and signal transduction?

Patched (Ptc) serves as the primary receptor for Shh and plays a dual role in both signal transduction and morphogen gradient formation . When Shh binds to Ptc, it relieves the inhibition that Ptc exerts on Smoothened (Smo), a transmembrane protein essential for downstream signal transduction . This initiates the intracellular signaling cascade that ultimately regulates gene expression .

Beyond its role in signal reception, Ptc critically regulates Shh distribution in tissues . Binding of Shh to Ptc restricts the movement of Shh through tissue, creating a feedback mechanism that limits Shh spread . This was demonstrated through experiments where blocking Shh signaling with mutated forms of Ptc resulted in more dorsally positioned cells responding as if exposed to higher Shh concentrations .

This feedback regulation helps establish and maintain the ventral HIGH–dorsal LOW gradient of Shh that emanates from the notochord and floor plate . Through this mechanism, Ptc ensures that the precise concentration-dependent induction of different neuronal subtypes occurs at the appropriate positions along the dorsal-ventral axis of the neural tube .

What techniques are available for detecting and quantifying Shh in mouse samples?

Researchers can employ several complementary techniques to detect and quantify Shh in mouse samples:

Single-molecule Fluorescent In Situ Hybridization (smFISH):

This technique detects Shh mRNA transcripts at the single-cell level, visualizing specific transcripts as discrete spots within cells . Density can be scored as very high (>30 dots/cell), high (10-30 dots/cell), moderate (6-9 dots/cell), low (2-5 dots/cell), or undetectable (0-1 dot/cell) . The method allows multiplexing to simultaneously detect Shh and other markers such as cell-type specific genes .

Enzyme-Linked Immunosorbent Assay (ELISA):

Quantikine ELISA kits specific for mouse Shh N-terminus provide precise quantification of protein levels . These assays demonstrate excellent performance metrics:

Sample TypeAverage % RecoveryRange %
Cell Culture Samples (n=6)10196-109
Heparin Plasma (n=4)9786-108
Serum (n=4)9886-109

Precision metrics further demonstrate reliability:

Intra-Assay PrecisionInter-Assay Precision
Sample12312
Mean (pg/mL)22.754.621221.950.4
Standard Deviation1.081.3110.62.282.96
CV%4.82.4510.45.9

Functional Activity Assays:

Biological activity of Shh can be assessed through functional assays such as alkaline phosphatase production by C3H/10T1/2 (CCL-226) cells . Recombinant mouse Shh typically demonstrates an ED50 (effective dose producing 50% maximum response) of <2.0 μg/ml, corresponding to a specific activity of >500 units/mg .

How should researchers design experiments to study Shh concentration-dependent effects?

Studying Shh concentration-dependent effects requires careful experimental design:

  • Gradient Establishment: Create precise concentration gradients using purified recombinant Shh-N protein . Even 2- to 3-fold differences in concentration can induce distinct cellular responses, so accurate dilution series are essential .

  • Explant Cultures: Naïve neural tissue explants provide an excellent system for studying concentration-dependent effects on neuronal subtype specification . These cultures maintain tissue architecture while allowing precise control over morphogen exposure .

  • Time-Course Analysis: Consider both concentration and duration of Shh exposure, as both parameters contribute to cell fate decisions . Sequential sampling at multiple timepoints can reveal the dynamics of cellular responses .

  • Readout Systems: Employ multiple readouts to comprehensively assess responses:

    • Immunostaining for cell-type specific markers to identify differentiation outcomes

    • Gene expression analysis for Shh target genes

    • Protein phosphorylation for signaling pathway activation

    • Functional assays appropriate to the cell types being studied

  • Perturbation Approaches: Use pathway inhibitors, dominant-negative constructs, or siRNA/CRISPR techniques to manipulate specific components of the Shh pathway . This helps establish causal relationships rather than just correlations .

  • In vivo Validation: Confirm findings from in vitro concentration studies using appropriate in vivo models, such as conditional knockout mice or electroporation of pathway components .

How does Shh signaling contribute to tissue regeneration through mouse embryonic stem cells?

Shh signaling plays a crucial role in enhancing the regenerative potential of mouse embryonic stem cells (mESCs) through multiple mechanisms . When mESCs are treated with Shh, they exhibit improved capabilities for tissue repair, particularly in wound healing contexts .

The molecular basis for this enhancement begins with Shh-induced adherens junction disruption through MMP activation . This releases β-catenin, which promotes cell proliferation . Simultaneously, Shh-mediated Gli1 activation upregulates integrin β1, triggering FAK and Src phosphorylation . This signaling cascade activates Rac1 and Cdc42 GTPases, leading to F-actin formation critical for cell migration .

These findings suggest that Shh pretreatment could serve as a valuable strategy for enhancing stem cell-based therapies for tissue regeneration . The study authors concluded that Shh can promote tissue regeneration and regenerative medicine in multiple contexts through these mechanisms .

What is the distribution of Shh-expressing cells in the adult mouse brain and what are their phenotypes?

Recent research using single-molecule fluorescent in situ hybridization (smFISH) has revealed that Shh expression in the adult mouse brain is much more extensive than previously recognized . Shh transcripts have been detected in almost all brain regions, challenging earlier assumptions about its limited expression .

The phenotypic characterization of Shh-expressing cells has identified several distinct populations:

  • Neuronal Populations: Shh mRNA has been identified in HuC/D-positive neurons throughout multiple brain regions .

  • Oligodendroglial Cells: A subset of cells expressing the oligodendroglial markers Olig2 and Sox10 also express Shh transcripts . This includes CC1-positive mature oligodendrocytes that express Shh protein as recognized by the specific monoclonal antibody C9C5 .

  • Nitrergic Neurons: A previously unrecognized population of neurons co-expresses Shh transcripts and the nitrergic marker nNOS . This suggests potential new roles for Shh in regulating neural circuits involving nitric oxide signaling .

  • Hypothalamic Expression: Broad expression of Shh transcripts has been observed in hypothalamic nuclei, including cells expressing Gad67 in the ventromedial hypothalamus (VMH) . Between 100-225 nNOS-expressing cells were counted in the VMH and dorsomedial hypothalamus (DMH), with 42-48 cells counted in the arcuate nucleus (ARC) where nNOS transcript is less expressed .

This diverse expression pattern suggests that Shh may have previously unrecognized functions in the adult brain, potentially including roles in neural circuit maintenance, plasticity, and regulation of hypothalamic functions .

How does Shh interact with other signaling pathways in mouse development and disease?

Shh signaling integrates with multiple other pathways to regulate development and disease processes:

  • β-catenin/Wnt Pathway: Shh-induced disruption of adherens junctions releases β-catenin, which can activate Wnt target genes and promote cell proliferation . This cross-talk between Shh and Wnt signaling is critical for coordinated tissue development and regeneration .

  • Integrin Signaling: Shh-mediated Gli1 expression leads to integrin β1 upregulation, connecting Shh signaling to cell-extracellular matrix interactions . This linkage is essential for proper cell migration and tissue remodeling during development and wound healing .

  • Cytoskeletal Regulation: Shh activates Rac1 and Cdc42 GTPases, influencing actin cytoskeleton organization . This affects cell morphology, motility, and connectivity, particularly important in neural development and axon guidance .

  • Notch Pathway: Shh and Notch signaling cooperate to regulate neural progenitor maintenance and differentiation in the developing central nervous system . This interaction helps define the boundaries between different neural domains .

  • BMP/TGF-β Pathways: Shh often antagonizes BMP signaling during neural tube patterning, creating opposing gradients that precisely position different cell types along the dorsal-ventral axis .

The integration of these pathways allows for precise spatial and temporal control of developmental processes. Dysregulation of these interactions can contribute to various pathologies, including developmental disorders and cancer . Understanding these pathway interactions is critical for developing targeted therapeutic approaches for conditions involving aberrant Shh signaling .

What are common technical challenges in detecting Shh in adult mouse tissues?

Detecting Shh in adult mouse tissues presents several technical challenges that researchers must address:

  • Low Expression Levels: Shh expression in adult tissues is often lower than in embryonic tissues, requiring highly sensitive detection methods . This necessitates optimized protocols and careful attention to background signal .

  • Lipid Modifications: Shh undergoes palmitoylation and cholesterol addition that affect its solubility and detection in standard assays . These modifications require specialized extraction methods to ensure complete recovery of the protein .

  • Antibody Specificity: Ensuring antibody specificity is critical, particularly to avoid cross-reactivity with other Hedgehog family members (Dhh, Ihh) . Validation using known positive samples and Shh knockout tissues as negative controls is essential .

  • RNA vs. Protein Detection: Discrepancies between mRNA and protein expression patterns can occur due to post-transcriptional regulation . This necessitates combining techniques like smFISH for mRNA and immunohistochemistry for protein detection to obtain a complete picture .

  • Signal Quantification: Establishing consistent scoring systems for expression levels, such as the density categories used in smFISH (very high: >30 dots/cell; high: 10-30 dots/cell; moderate: 6-9 dots/cell; low: 2-5 dots/cell; undetectable: 0-1 dot/cell) .

Recent advances using single-molecule fluorescent in situ hybridization (smFISH) have helped overcome some of these challenges, revealing much broader expression of Shh transcripts in adult brain regions than previously reported . This suggests that earlier studies may have missed important aspects of Shh biology due to technical limitations.

How can researchers ensure reproducibility in Shh mouse studies?

Ensuring reproducibility in Shh mouse studies requires attention to several critical factors:

  • Standardized Protein Sources:

    • Use well-characterized recombinant proteins with defined activity metrics (e.g., ED50 <2.0 μg/ml for inducing alkaline phosphatase production)

    • Verify that natural mouse Shh samples produce dose-response curves parallel to recombinant standards

  • Validated Detection Methods:

    • ELISA assays should demonstrate consistent recovery rates (97-101%) across different sample types

    • Acceptable precision metrics include intra-assay CV% of 2.4-5.0% and inter-assay CV% of 4.9-10.4%

  • Detailed Reporting:

    • Document exact mouse strains, sex, and age

    • Report specific brain regions or tissue sampling methods with anatomical precision

    • When scoring expression, use established density categories (e.g., very high: >30 dots/cell)

  • Multi-technique Validation:

    • Combine complementary approaches (e.g., smFISH, immunohistochemistry)

    • Use both qualitative and quantitative assessments

    • Verify findings with functional assays when possible

  • Biological Replicates:

    • Use appropriate sample sizes (e.g., n=3 animals for density estimations)

    • Include inter-animal variability assessments

    • Present data as mean ± SEM to accurately represent variability

By implementing these practices, researchers can enhance the reliability and reproducibility of their findings, facilitating meaningful comparisons across different studies and laboratories.

Product Science Overview

Introduction

Sonic Hedgehog (Shh) is a highly conserved protein that plays a crucial role in embryonic development. It is part of the Hedgehog signaling pathway, which is essential for the regulation of cell growth, differentiation, and tissue patterning. The name “Sonic Hedgehog” was inspired by the popular video game character, reflecting the protein’s significant impact on developmental biology.

Discovery and Nomenclature

The Hedgehog gene was first discovered in fruit flies (Drosophila melanogaster) in the 1980s. Researchers identified three homologs in vertebrates: Desert Hedgehog (Dhh), Indian Hedgehog (Ihh), and Sonic Hedgehog (Shh). Among these, Shh is the most extensively studied due to its critical role in various developmental processes.

Structure and Function

Sonic Hedgehog is a secreted protein that undergoes autocatalytic cleavage to produce an active N-terminal fragment. This fragment is responsible for the protein’s signaling activity. Shh binds to its receptor, Patched (Ptch), which in turn regulates the activity of another protein called Smoothened (Smo). This signaling cascade ultimately influences the expression of target genes involved in cell proliferation, differentiation, and tissue patterning.

Role in Development

Shh is expressed in several embryonic tissues that are vital for the patterning of the developing central nervous system, somite, and limb . It is also involved in the development of whiskers, hair, foregut, teeth, and bones . The protein’s gradient in tissues helps establish the spatial organization of cells, guiding the formation of various structures during embryogenesis.

Recombinant Mouse Sonic Hedgehog

Recombinant Mouse Sonic Hedgehog (Shh) is produced using E. coli expression systems. The recombinant protein typically includes the N-terminal signaling domain, which retains its biological activity. It is used in various research applications to study the Hedgehog signaling pathway and its role in development and disease.

Applications in Research

Recombinant Shh is utilized in studies involving:

  • Cell Differentiation: Investigating how Shh influences the differentiation of stem cells into specific cell types.
  • Tissue Engineering: Exploring the potential of Shh in regenerating damaged tissues and organs.
  • Cancer Research: Understanding the role of aberrant Shh signaling in the development and progression of cancers, such as basal cell carcinoma and medulloblastoma.
  • Drug Development: Screening for compounds that can modulate the Hedgehog signaling pathway, offering potential therapeutic strategies for various diseases.

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.