DHH (C23II) Human

Desert Hedgehog (C23II) Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to DHH (C23II) Human

DHH (C23II) Human is a recombinant form of the Desert Hedgehog (DHH) protein, a key member of the Hedgehog (Hh) signaling family. Produced in Escherichia coli, this 22.4–22.5 kDa protein corresponds to amino acids 23–198 of the full-length human DHH precursor, fused to a 24-amino acid His-tag at the N-terminus for purification . DHH plays critical roles in embryonic development, gonadal differentiation, and nerve sheath formation, with mutations linked to reproductive and neurological disorders .

Key Features:

PropertyDetails
Molecular Weight22.4–22.5 kDa (non-glycosylated)
Amino Acid Sequence201 residues (23–198 of native DHH + 24 His-tag)
TagN-terminal His-tag for chromatographic purification
Purity>90% by SDS-PAGE
Formulation20 mM Tris-HCl (pH 7.5–8.0), 0.15M NaCl, 1 mM DTT, 10% glycerol

Hedgehog Signaling Pathway:

  • Autocatalytic Cleavage: DHH is synthesized as a 45 kDa precursor that self-cleaves into:

    • N-terminal domain (19 kDa): Mediates signaling via Patched/Smoothened receptors .

    • C-terminal domain: Facilitates cholesterol attachment to the N-terminal fragment, localizing it to the cell membrane .

  • Roles in Development:

    • Gonadal Differentiation: Essential for Leydig cell development and spermatogenesis .

    • Neural Sheath Formation: Promotes perineurial tissue development in peripheral nerves .

Production and Purification

  • Expression System: E. coli .

  • Purification: Proprietary chromatographic techniques (e.g., immobilized metal affinity chromatography via His-tag) .

  • Stability:

    • Short-term: 4°C for 2–4 weeks .

    • Long-term: -20°C with 0.1% carrier protein (HSA/BSA) to prevent aggregation .

Experimental Findings:

  1. Gonadal Dysgenesis:

    • DHH mutations correlate with 46,XY partial gonadal dysgenesis (PGD) and minifascicular neuropathy .

    • In vitro studies show DHH induces steroidogenic factor 1 (SF1), critical for Leydig cell differentiation .

  2. Nerve Repair:

    • Upregulated in Schwann cells post-injury; promotes connective tissue sheath formation .

  3. Cancer Research:

    • Overexpression in ovarian cancer linked to poor prognosis .

Clinical Relevance

DisorderAssociation with DHH DysfunctionReferences
Partial Gonadal DysgenesisImpaired testis development, ambiguous genitalia
Minifascicular NeuropathyDefective perineurial sheath formation
Ovarian CancerProliferative role via Hedgehog pathway activation

Comparative Analysis with Hedgehog Proteins

ProteinIdentity with DHHKey Function
SHH~75%Neural tube patterning, limb development
IHH~75%Bone development, cartilage maintenance
DHH-Gonadal differentiation, nerve repair

Product Specs

Introduction
Desert Hedgehog (DHH) is a member of the Hedgehog family, which encodes signaling molecules crucial for regulating morphogenesis. DHH protein undergoes autocatalytic cleavage, resulting in an N-terminal fragment containing the signaling activity and a C-terminal fragment involved in precursor processing. The C-terminal fragment facilitates the attachment of a cholesterol moiety to the N-terminal fragment, restricting it to the cell surface and preventing its free diffusion. DHH mutations are linked to partial gonadal dysgenesis (PGD) with minifascicular polyneuropathy. DHH is essential for male gonadal differentiation, perineurial development, and acts as a spermatocyte survival factor in the testes, playing a critical role in testes development.
Description
Recombinant human DHH (C23II) is produced in E. coli. It is a single, non-glycosylated polypeptide chain comprising 177 amino acids with a molecular weight of 19.9 kDa. The purification of DHH (C23II) is achieved using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized powder.
Formulation
The product is lyophilized from a 0.2 µm filtered solution concentrated in 1x PBS with a pH of 7.4.
Solubility
For reconstitution, it is recommended to dissolve the lyophilized DHH (C23II) in sterile 18 MΩ·cm H₂O at a concentration of at least 100 µg/ml. This solution can be further diluted with other aqueous solutions.
Stability
Lyophilized DHH (C23II) remains stable at room temperature for up to 3 weeks. However, it is recommended to store the lyophilized product desiccated at a temperature below -18°C. After reconstitution, DHH (C23II) can be stored at 4°C for 2-7 days. For prolonged storage, it is advisable to store the reconstituted solution at a temperature below -18°C with the addition of a carrier protein (0.1% HSA or BSA). Avoid repeated freeze-thaw cycles.
Purity
The purity of the product is greater than 95.0% as determined by: (a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis and (b) SDS-polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Biological Activity
The biological activity of DHH (C23II) is assessed by its ability to stimulate alkaline phosphatase production in C3H/10T1/2 (CCL-226) cells. The expected half-maximal effective concentration (ED₅₀) for this effect is in the range of 15-45 µg/ml.
Synonyms
HHG-3, Desert Hedgehog homolog, MGC35145, Desert hedgehog protein, DHH.
Source
Escherichia Coli.
Amino Acid Sequence
IIGPGRGPVG RRRYARKQLV PLLYKQFVPG VPERTLGASG PAEGRVARGS ERFRDLVPNY NPDIIFKDEE NSGADRLMTE RCKERVNALA IAVMNMWPGV RLRVTEGWDE DGHHAQDSLH YEGRALDITT SDRDRNKYGL LARLAVEAGF DWVYYESRNH VHVSVKADNS LAVRAGG.

Q&A

What is DHH (C23II) Human and what are its key molecular characteristics?

DHH (C23II) Human refers to a recombinant form of human Desert Hedgehog protein, specifically comprising amino acids 23-198 of the full sequence. When produced for research purposes, it is typically expressed in E. coli as a single, non-glycosylated polypeptide chain with a molecular mass of approximately 22.4-22.5 kDa . The recombinant protein commonly includes a His-tag fusion at the N-terminus (24 amino acids) to facilitate purification through proprietary chromatographic techniques .

The DHH protein belongs to the Hedgehog family, which encodes signaling molecules involved in regulating morphogenesis. In its natural form, DHH is synthesized as a precursor that undergoes autocatalytic cleavage. The N-terminal portion (contained in the C23II construct) is soluble and contains the signaling activity, while the C-terminal portion is involved in precursor processing and attaches a cholesterol moiety to the N-terminal product. This modification restricts the N-terminal product to the cell surface and prevents it from freely diffusing throughout the organism .

PropertySpecificationReference
Amino acid sequence23-198 (human DHH)
Molecular mass22.4-22.5 kDa
Expression systemE. coli
Tag24 amino acid His-tag (N-terminus)
Physical appearanceSterile filtered clear solution
Purity>90% by SDS-PAGE

How does DHH differ functionally from other Hedgehog family proteins?

The mammalian Hedgehog family consists of three members - Sonic Hedgehog (SHH), Indian Hedgehog (IHH), and Desert Hedgehog (DHH). While these proteins share structural similarities and signaling mechanisms, they exhibit distinct expression patterns and biological functions:

DHH has a more restricted expression pattern compared to other family members. It plays essential roles in:

  • Male gonadal differentiation and testes development

  • Perineurial development in the peripheral nervous system

  • Spermatocyte survival

  • Intercellular signaling required for specific developmental patterning events

DHH defects have been specifically associated with partial gonadal dysgenesis (PGD) accompanied by minifascicular polyneuropathy . Unlike SHH, which has been extensively implicated in various cancers through both paracrine signaling and cancer stem cell (CSC) maintenance, DHH's role in cancer biology is less characterized .

When designing experiments to investigate DHH-specific functions, researchers should carefully consider:

  • The appropriate cellular context (gonadal or nerve cells often provide relevant physiological responses)

  • The potential for functional redundancy with other Hedgehog family members

  • The signaling components present in the experimental system (receptor expression, pathway components)

What are the optimal storage and handling conditions for DHH (C23II) Human protein?

Maintaining protein stability is critical for successful DHH experiments. For DHH (C23II) Human, researchers should follow these evidence-based storage and handling recommendations:

Short-term storage (1-2 weeks):

  • Store at +4°C

  • Maintain in the original buffer formulation

Long-term storage:

  • Aliquot upon receipt to minimize freeze-thaw cycles

  • Store at -20°C or preferably -70°C

  • Avoid more than 2-3 freeze-thaw cycles per aliquot

Buffer composition:
Standard formulations include:

  • 20mM Tris-HCl buffer (pH 7.5-8.0)

  • 0.15M NaCl

  • 10% glycerol (cryoprotectant)

  • Optional components may include 1mM DTT or 0.4M Urea

Handling recommendations:

  • Thaw aliquots slowly on ice rather than at room temperature

  • Centrifuge briefly after thawing to collect all material

  • For dilution, use buffers matched to the original formulation when possible

  • Validate protein activity after extended storage using functional assays

Improper storage and handling represent common sources of experimental variability. Researchers should document storage conditions and freeze-thaw cycles when reporting DHH experiments to facilitate reproducibility.

What functional assays can be used to assess DHH (C23II) Human activity?

Several established assays can quantitatively measure DHH functional activity:

1. GLI1 Reporter Assays:
The most direct approach involves measuring activation of the Hedgehog signaling pathway using cells transfected with GLI-responsive luciferase reporter constructs. This assay provides quantitative data on pathway activation and can establish dose-response relationships .

Methodology overview:

  • Responsive cells (e.g., NIH3T3) are transfected with a GLI-responsive luciferase construct

  • Cells are treated with varying concentrations of DHH (C23II) Human (typically 10-500 ng/mL)

  • Luciferase activity is measured after 24-48 hours

  • Results are normalized to internal controls and compared to untreated cells

2. C3H10T1/2 Differentiation Assay:
This assay measures the ability of DHH to induce osteoblast differentiation of C3H10T1/2 cells, providing a physiologically relevant functional readout .

Methodology overview:

  • C3H10T1/2 cells are cultured in appropriate medium

  • Cells are treated with DHH (C23II) Human for 3-7 days

  • Osteoblast differentiation is assessed by measuring alkaline phosphatase activity or by staining for calcification

  • This assay is particularly valuable for structure-function studies

3. Target Gene Expression Analysis:
Quantification of endogenous Hedgehog target gene expression using qRT-PCR provides another measure of pathway activation.

Methodology overview:

  • Treat responsive cells with DHH (C23II) Human

  • Extract RNA after appropriate time points (24-72 hours)

  • Measure expression of canonical Hedgehog target genes (PTCH1, GLI1, GLI2)

  • This approach can be especially useful for studying tissue-specific responses

When performing these assays, researchers should include appropriate controls:

  • Positive control: Known Hedgehog pathway activator (e.g., SAG or recombinant SHH)

  • Negative control: Buffer-only treatment

  • Dose-response analysis to establish potency (EC50)

How can researchers validate antibody specificity for DHH detection?

Antibody validation is essential for reliable DHH detection, particularly given the structural similarities between Hedgehog family members. A comprehensive validation strategy should include:

1. Cross-reactivity Assessment:

  • Test the antibody against all three purified Hedgehog proteins (DHH, SHH, IHH)

  • Quantify relative binding to each protein via ELISA or Western blot

  • Document any observed cross-reactivity (e.g., the Mouse Desert Hedgehog/Dhh N-Terminus Antibody shows approximately 20% cross-reactivity with other Hedgehog proteins)

2. Knockout/Knockdown Controls:

  • Use cells or tissues with confirmed DHH knockout/knockdown as negative controls

  • Compare staining patterns in wild-type versus knockout samples

  • This approach provides the most stringent validation of specificity

3. Peptide Competition Assays:

  • Pre-incubate the antibody with excess purified DHH protein or specific peptide epitopes

  • If the antibody is specific, this should abolish or significantly reduce signal

  • Include graded concentrations of competing peptide to establish dose-dependent inhibition

4. Multiple Antibody Validation:

  • Use multiple antibodies targeting different epitopes of DHH

  • Consistent results with different antibodies increase confidence in specificity

  • Compare monoclonal and polyclonal antibodies when available

5. Recombinant Expression Systems:

  • Test antibodies on cells transfected with DHH expression constructs versus empty vector controls

  • This creates a controlled system with defined DHH expression

For immunohistochemical applications specifically, researchers should optimize:

  • Fixation conditions (different fixatives may affect epitope accessibility)

  • Antigen retrieval methods (if required)

  • Antibody concentration and incubation conditions

The Mouse Desert Hedgehog/Dhh N-Terminus Antibody has been validated for detecting DHH in perfusion fixed frozen sections of mouse brain and spinal cord, with specific staining localized to neuronal cell bodies and processes .

How can researchers study the role of DHH in cancer models?

While SHH has been more extensively studied in cancer contexts, emerging research suggests DHH may play important roles in specific tumor types. To investigate DHH in cancer models, researchers can employ several approaches:

1. Paracrine Signaling Studies:

  • Co-implantation models (e.g., HT-29/MEF) can evaluate tumor-stromal interactions mediated by Hedgehog signaling

  • DHH (C23II) Human can be applied exogenously to mimic paracrine signaling

  • Changes in tumor growth, invasion, and stromal remodeling can be assessed

  • These models are particularly valuable for testing Hedgehog pathway inhibitors, as demonstrated in studies showing improved responses to carboplatin in models with Hedgehog inhibition

2. Genetic Modulation Approaches:

  • CRISPR/Cas9-mediated knockout or knockdown of DHH in cancer cell lines

  • Overexpression studies to assess oncogenic potential

  • Analysis of pathway activation in response to genetic manipulation

  • Correlation of DHH expression with clinical outcomes in patient databases

3. Cancer Stem Cell (CSC) Investigations:
Although evidence for DHH's specific role in CSC maintenance is limited compared to SHH, researchers can explore potential effects through:

  • Spheroid formation assays following DHH treatment or manipulation

  • Analysis of stem cell marker expression

  • In vivo limiting dilution assays to assess tumor-initiating capacity

  • It's important to note that, unlike SHH, DHH may not significantly impact CSC frequency or growth in some cancer types

4. Combination Therapy Studies:

  • Evaluate whether DHH pathway modulation affects sensitivity to standard chemotherapeutics

  • Test potential synergistic effects between DHH inhibition and other targeted therapies

  • Assess impact on resistance mechanisms

When designing cancer-related DHH experiments, researchers should carefully consider:

  • The specific cancer type and its known association with Hedgehog signaling

  • The presence of autocrine versus paracrine signaling mechanisms

  • The expression of pathway components in both tumor and stromal cells

  • Appropriate readouts for different aspects of cancer biology (proliferation, invasion, angiogenesis, etc.)

What experimental approaches can be used to study DHH mutations and their functional consequences?

DHH mutations have been associated with conditions such as partial gonadal dysgenesis and peripheral neuropathies. To study these mutations and their functional impacts, researchers can employ several complementary approaches:

1. Recombinant Protein Variants:

  • Generate DHH (C23II) Human proteins containing specific disease-associated mutations

  • Compare activity of wild-type versus mutant proteins in functional assays:

    • GLI reporter activation

    • C3H10T1/2 differentiation

    • Receptor binding studies

  • Assess protein stability and processing of mutant variants

  • This approach directly addresses the functional consequences of mutations on protein activity

2. Structural Studies:

  • Use computational modeling to predict the impact of mutations on protein structure

  • When feasible, determine crystal structures of wild-type and mutant proteins

  • Assess changes in protein-protein interactions through techniques like surface plasmon resonance

  • These approaches provide mechanistic insights into how mutations affect protein function

3. Cellular Models:

  • Generate isogenic cell lines with CRISPR/Cas9-introduced mutations

  • Compare signaling pathway activation in wild-type versus mutant cells

  • Assess transcriptional responses through RNA-seq or targeted gene expression analysis

  • Evaluate phenotypic consequences in relevant cell types (e.g., Sertoli cells for gonadal dysgenesis)

4. Animal Models:

  • Generate knock-in mouse models with specific DHH mutations

  • Characterize developmental and physiological consequences

  • Perform detailed analysis of affected tissues (gonads, peripheral nerves)

  • Test potential therapeutic interventions

5. Patient-Derived Resources:

  • Analyze primary cells from patients with DHH mutations when available

  • Generate induced pluripotent stem cells (iPSCs) from patient samples

  • Differentiate iPSCs into relevant cell types to study disease mechanisms

  • These approaches provide direct insights into human disease manifestations

When studying DHH mutations, researchers should consider both:

  • Loss-of-function effects (reduced signaling capacity)

  • Potential gain-of-function or dominant-negative effects

  • Context-dependent consequences in different cell types

  • Interactions with genetic modifiers that may influence phenotypic expression

How does DHH signaling interact with other developmental and oncogenic pathways?

DHH signaling does not function in isolation but interacts with multiple other signaling networks. Understanding these interactions is crucial for comprehending DHH's roles in development and disease:

1. Wnt/β-catenin Pathway:

  • Bidirectional regulation between Hedgehog and Wnt signaling occurs in many developmental contexts

  • These pathways can either cooperate or antagonize each other depending on cellular context

  • Key points of intersection include:

    • GSK3β activity (regulates both pathways)

    • β-catenin stability and localization

    • Shared target genes in some contexts

2. TGF-β/BMP Signaling:

  • Cooperative interactions often occur during developmental patterning

  • DHH can influence SMAD activation in some cell types

  • Coordinated regulation of target genes through composite enhancer elements

  • These interactions are particularly important in gonadal development

3. Notch Pathway:

  • Reciprocal regulation between Hedgehog and Notch signaling

  • Important for stem cell maintenance and fate decisions

  • Key intersections through shared transcriptional regulators

  • These interactions help establish developmental boundaries

4. Growth Factor Signaling:

  • Cross-talk with pathways like EGFR, PDGFR, and FGFR

  • Can involve direct protein interactions or transcriptional regulation

  • Often results in enhanced MAPK or PI3K/AKT activation

  • These interactions may be particularly relevant in cancer contexts

Experimental approaches to study pathway interactions:

  • Multi-pathway reporter systems to measure simultaneous pathway activation

  • Pharmacological inhibition of one pathway while monitoring the other

  • Proteomic analysis to identify physical interactions between pathway components

  • Transcriptomic analysis to identify shared target genes

  • Single-cell approaches to resolve cell-type specific interaction patterns

Researchers investigating DHH pathway interactions should:

  • Consider the temporal dynamics of pathway activation

  • Account for cell type-specific interaction patterns

  • Design appropriate controls for pathway modulators

  • Validate key findings with multiple complementary approaches

What are common challenges in DHH (C23II) Human experiments and how can they be addressed?

Working with DHH (C23II) Human presents several technical challenges. Here are evidence-based solutions to the most common issues:

1. Protein Activity Loss:

  • Challenge: Decreased or variable activity after storage or handling

  • Solutions:

    • Validate protein activity immediately upon receipt

    • Minimize freeze-thaw cycles by using smaller aliquots

    • Add stabilizing agents like 10% glycerol to storage buffer

    • Consider adding reducing agents (e.g., 1mM DTT) if oxidation is a concern

    • Perform functional assays before critical experiments to confirm activity

2. Cell Responsiveness Issues:

  • Challenge: Variable or absent responses to DHH treatment

  • Solutions:

    • Verify expression of key pathway components (PTCH1, SMO, GLI factors) in cell lines

    • Use positive control cells with known Hedgehog responsiveness

    • Optimize cell density (confluent cultures may respond differently)

    • Reduce serum concentration during treatment (serum can contain pathway inhibitors)

    • Consider using Hedgehog pathway sensitizers (e.g., SAG at sub-optimal concentrations)

3. Antibody Specificity Concerns:

  • Challenge: Cross-reactivity with other Hedgehog family members

  • Solutions:

    • Include recombinant SHH and IHH as controls in immunodetection experiments

    • Use antibodies with documented specificity and cross-reactivity profiles

    • Perform validation studies in systems with defined expression of Hedgehog proteins

    • Consider using multiple antibodies targeting different epitopes

    • Include appropriate negative controls (knockouts or knockdowns)

4. Experimental Design Issues:

  • Challenge: Inappropriate controls or inconsistent results

  • Solutions:

    • Include both negative controls (buffer-only) and positive controls (known pathway activators)

    • Perform dose-response analyses to establish optimal concentrations

    • Conduct time-course experiments to determine optimal treatment duration

    • Use multiple readouts to assess pathway activation (reporter assays, target gene expression)

    • Document all experimental variables that might affect outcomes

5. Buffer Compatibility Problems:

  • Challenge: Interference from buffer components in experimental systems

  • Solutions:

    • Test buffer-only controls at equivalent volumes

    • Consider buffer exchange if components might affect the assay

    • For dilutions, use buffers matched to the original formulation

    • Be aware that high glycerol concentrations may affect some cellular assays

By systematically addressing these common challenges, researchers can significantly improve the reproducibility and reliability of DHH (C23II) Human experiments.

How can researchers design comparative studies between DHH and other Hedgehog proteins?

Comparative studies between DHH and other Hedgehog family members (SHH, IHH) require careful experimental design to yield meaningful results:

1. Protein Standardization:

  • Use recombinant proteins with comparable:

    • Production systems (same expression host)

    • Purification methods

    • Tag configurations

    • Quality control metrics (purity, endotoxin levels)

  • Confirm activity of all proteins using the same functional assay

  • Determine protein concentration using the same method (e.g., Bradford assay)

2. Dose-Response Analysis:

  • Generate parallel dose-response curves for DHH, SHH, and IHH

  • Test a wide concentration range (e.g., 0.1-1000 ng/mL)

  • Calculate and compare:

    • EC50 values (potency)

    • Maximum response (efficacy)

    • Hill slopes (cooperativity)

  • These parameters provide quantitative measures for comparing protein activities

3. Receptor Binding Studies:

  • Compare binding to PTCH1 and PTCH2 receptors

  • Methods include:

    • Surface plasmon resonance

    • Cell-based binding assays with fluorescently labeled proteins

    • Competition binding assays

  • These studies directly address molecular recognition differences

4. Cell Type Panel Analysis:

  • Test activity across multiple cell types with varying:

    • Receptor expression profiles

    • Pathway component levels

    • Developmental origins

  • This approach can reveal cell type-specific preferences for different Hedgehog proteins

5. Transcriptional Response Comparison:

  • Perform RNA-seq or targeted gene expression analysis

  • Compare:

    • Common versus unique target genes

    • Magnitude of regulation for shared targets

    • Temporal dynamics of gene expression changes

  • This provides insights into signaling output differences

6. Controls and Validation:

  • Include pathway inhibitors (e.g., cyclopamine or GANT61) to confirm specificity

  • Use knockdown/knockout models of specific receptors to test dependency

  • Validate key findings with multiple methodological approaches

When designing comparative studies, researchers should:

  • Use the same experimental conditions for all proteins being compared

  • Include internal controls for normalization

  • Perform statistical analyses appropriate for comparative studies

  • Consider both quantitative differences (magnitude) and qualitative differences (pattern)

What are promising emerging applications for DHH (C23II) Human in research?

Several innovative research directions hold particular promise for advancing our understanding of DHH biology and its applications:

1. Stem Cell Differentiation and Regenerative Medicine:

  • DHH's role in development suggests potential applications in directed differentiation protocols

  • Specific applications may include:

    • Peripheral nerve regeneration, leveraging DHH's role in perineurial development

    • Male germ cell differentiation from pluripotent stem cells

    • Creating in vitro models of gonadal development

  • These approaches could provide both research tools and potential therapeutic strategies

2. Precision Medicine for DHH-Associated Disorders:

  • Functional characterization of patient-specific DHH mutations

  • Development of personalized therapeutic approaches based on mutation mechanism

  • Creation of patient-derived cellular models for drug screening

  • This personalized approach may benefit patients with conditions like partial gonadal dysgenesis

3. DHH Pathway Modulation for Therapeutic Applications:

  • Development of DHH pathway-specific modulators (agonists and antagonists)

  • Potential applications in:

    • Peripheral nerve injuries

    • Specific cancer subtypes

    • Gonadal disorders

  • Advantages over current Hedgehog pathway modulators that target SMO would include greater specificity and potentially fewer side effects

4. Organoid and Microphysiological Systems:

  • Integration of DHH signaling in complex 3D culture systems

  • Applications include:

    • Testis organoids for reproductive biology studies

    • Peripheral nerve organoids for developmental and disease modeling

    • Tumor-stroma interaction models for cancer research

  • These systems bridge the gap between traditional cell culture and animal models

5. Single-Cell Analysis of DHH Signaling Networks:

  • Application of single-cell transcriptomics and proteomics to understand:

    • Cell-type specific responses to DHH

    • Heterogeneity in pathway activation

    • Temporal dynamics of signaling

  • These approaches can reveal previously unrecognized complexity in DHH signaling

When pursuing these emerging directions, researchers should consider:

  • Combining multiple cutting-edge technologies for comprehensive analysis

  • Validating findings across different experimental systems

  • Translating basic discoveries toward clinical applications where appropriate

  • Collaborating across disciplines to address complex research questions

Product Science Overview

Introduction

Desert Hedgehog (DHH) is a member of the highly conserved Hedgehog family of proteins, which play crucial roles in various developmental processes. The Hedgehog family includes three proteins in mammals: Sonic Hedgehog (SHH), Indian Hedgehog (IHH), and Desert Hedgehog (DHH). These proteins are involved in cell signaling pathways that regulate embryonic development, tissue patterning, and stem cell maintenance.

Structure and Function

Desert Hedgehog (DHH) is a secreted protein that undergoes autocatalytic cleavage, resulting in two functional domains: the N-terminal signaling domain and the C-terminal autoprocessing domain. The N-terminal domain is responsible for the protein’s signaling activity, while the C-terminal domain facilitates the cleavage and attachment of cholesterol to the N-terminal domain, which is essential for its proper functioning .

The recombinant form of Desert Hedgehog (C23II) is produced in E. coli and consists of a single, non-glycosylated polypeptide chain containing 177 amino acids. This recombinant protein is biologically active and has been shown to induce alkaline phosphatase production in specific cell lines .

Biological Activity

Desert Hedgehog (C23II) plays a significant role in various biological processes, including:

  • Embryonic Development: DHH is crucial for the development of the gonads and peripheral nerves. It is involved in the differentiation of Sertoli cells in the testes and Schwann cells in the peripheral nervous system.
  • Tissue Patterning: DHH signaling is essential for the proper patterning of tissues during embryogenesis. It helps in the formation of various organs and structures by regulating cell proliferation, differentiation, and apoptosis.
  • Stem Cell Maintenance: DHH is involved in the maintenance of stem cell populations in certain tissues, ensuring the proper regeneration and repair of tissues throughout an organism’s life .
Applications

Recombinant Desert Hedgehog (C23II) is widely used in research to study its role in developmental biology and disease. Some of its applications include:

  • Cell Signaling Studies: Researchers use recombinant DHH to investigate the mechanisms of Hedgehog signaling pathways and their impact on cell behavior.
  • Developmental Biology: DHH is used to study the processes of embryonic development and tissue patterning, providing insights into congenital disorders and developmental abnormalities.
  • Stem Cell Research: DHH is utilized to explore its role in stem cell maintenance and differentiation, contributing to advancements in regenerative medicine and tissue engineering .

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.