DHH Human

Desert Hedgehog Human Recombinant
Shipped with Ice Packs
In Stock

Description

DHH Human Recombinant produced in E.Coli is a single, non-glycosylated, polypeptide chain containing 197 amino acids (23-198) and having a molecular mass of 22 kDa. DHH is fused to His-tag (20 a.a.) at N-terminus and is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Desert Hedgehog (DHH) is a member of the Hedgehog signaling pathway, crucial for regulating morphogenesis during embryonic development. DHH undergoes autocatalytic cleavage, producing an N-terminal signaling molecule and a C-terminal fragment involved in cholesterol modification. This modification tethers the active N-terminal to the cell surface, restricting its diffusion. Mutations in DHH have been linked to partial gonadal dysgenesis (PGD) and minifascicular polyneuropathy, highlighting its importance in gonadal and neuronal development. Specifically, DHH is crucial for male gonadal differentiation, perineurial development, and acts as a survival factor for spermatocytes within the testes.
Description
Recombinant human DHH protein, expressed in E. coli, is a non-glycosylated polypeptide chain consisting of 197 amino acids (residues 23-198). This protein has a molecular weight of 22 kDa and includes an N-terminal His-tag (20 amino acids) for purification purposes. Purification is achieved using proprietary chromatographic methods.
Physical Appearance
The product is a clear, colorless, and sterile filtered solution.
Formulation
The DHH protein solution is formulated in a buffer containing 20mM MES (pH 5.5), 0.5mM DTT (dithiothreitol), and 20% glycerol.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For long-term storage, it is recommended to store the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for extended storage. Avoid repeated freeze-thaw cycles to maintain protein stability.
Purity
The purity of the DHH protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
HHG-3, Desert Hedgehog homolog, MGC35145, Desert hedgehog protein, DHH.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MCGPGRGPVG RRRYARKQLV PLLYKQFVPG VPERTLGASG PAEGRVARGS ERFRDLVPNY NPDIIFKDEE NSGADRLMTE RCKERVNALA IAVMNMWPGV RLRVTEGWDE DGHHAQDSLH YEGRALDITT SDRDRNKYGL LARLAVEAGF DWVYYESRNH VHVSVKADNS LAVRAGG.

Q&A

What is Desert hedgehog (DHH) protein and what are its primary functions in human development?

Desert hedgehog (DHH) is one of three highly conserved hedgehog genes in mammals, alongside Sonic hedgehog (SHH) and Indian hedgehog (IHH) . As a crucial signaling molecule, DHH plays several vital roles in human development, particularly in reproductive and nervous system tissues. Unlike SHH, DHH proteins primarily function in cell-cell contact-mediated juxtacrine signaling, creating unique spatial signaling dynamics in developing tissues .

The primary developmental functions of DHH include regulating proper development of myoid cells in the peritubular region and fetal Leydig cells in the interstitial region . It specifies fetal Leydig cell fate during testis organogenesis and contributes to the differentiation of peritubular myoid cells, which are essential for testis cord formation . Additionally, DHH is required for the formation of adult-type Leydig cells and normal development of peritubular cells and seminiferous tubules in the testis .

At the molecular level, DHH, like other hedgehog proteins, binds to the receptor Patched (PTCH1), initiating signal transduction via derepression of the co-receptor Smoothened (SMO) . This activation triggers downstream signaling cascades that regulate cell fate and tissue patterning during development.

What disorders and conditions are associated with DHH abnormalities in humans?

Several significant clinical conditions have been linked to DHH abnormalities:

DHH mutations or dysregulation have been implicated in 46,XY complete pure gonadal dysgenesis, a condition characterized by sex reversal and impaired gonadal development . Patients with certain DHH mutations may also develop minifascicular neuropathy, suggesting DHH's dual role in both reproductive and nervous system development .

In oncology, DHH has been associated with the progression of stomach adenocarcinoma through its regulation of the Hedgehog signaling pathway . This finding highlights the importance of developmental pathways in cancer biology and positions DHH as a potential therapeutic target.

Recent research has also revealed a preliminary association between DHH serum levels and autism spectrum disorder, though further investigation is needed to establish a causal relationship . Furthermore, as DHH acts as a negative regulator of CD44-CD25+ double negative T lymphocytes during thymic differentiation, its dysfunction may impact immune system development .

Methodological approaches to studying these conditions typically involve genetic screening for DHH mutations, quantitative protein assessment using ELISA techniques, and functional studies using cell and animal models to elucidate pathogenic mechanisms.

How do research methodologies differ when investigating DHH signaling in various tissue contexts?

The investigation of DHH signaling across different tissue contexts requires tailored methodological approaches due to tissue-specific microenvironments and signaling dynamics. In gonadal tissue research, methodologies often focus on spatiotemporal expression patterns using techniques like in situ hybridization and immunohistochemistry to visualize DHH distribution relative to developing structures .

For quantitative analysis of DHH in human samples, sandwich ELISA represents a gold standard approach with high sensitivity (0.061ng/mL) and a practical detection range (0.156-10ng/mL) . This method is particularly valuable for analyzing DHH levels in serum, plasma, and cell culture supernatants, enabling both clinical and research applications .

When studying DHH's interaction with its receptor PTCH1, investigators typically employ binding assays such as surface plasmon resonance or co-immunoprecipitation. The downstream effects of this interaction can be assessed using reporter gene assays measuring Gli transcription factor activation, a key indicator of hedgehog pathway activation .

For neurological contexts, where DHH has been implicated in minifascicular neuropathy, methodologies expand to include electrophysiological assessments and specialized histopathological techniques for peripheral nerve analysis. These approaches help elucidate how DHH signaling contributes to myelin sheath development and maintenance.

Cross-tissue comparative analyses are particularly valuable, as they can reveal how the same molecular pathway produces different outcomes depending on cellular context and the presence of tissue-specific cofactors.

What are the current challenges in differentiating between direct and indirect effects of DHH in human developmental pathways?

Differentiating between direct and indirect effects of DHH in developmental pathways presents several methodological challenges for researchers. The interconnected nature of signaling networks means that DHH may initiate cascades with multiple downstream branches, making it difficult to isolate primary effects from secondary consequences .

One significant challenge is the temporal dynamics of development. DHH initiates processes that unfold over time, with early effects potentially triggering secondary developmental programs that become independent of continued DHH signaling. Addressing this challenge requires time-series experiments with inducible genetic systems that allow precise temporal control of DHH expression or activity.

Another complication arises from the cross-talk between hedgehog family members. Since SHH, IHH, and DHH all bind to PTCH1, researchers must carefully design experiments to distinguish DHH-specific effects from those potentially mediated by other hedgehog proteins . This often requires the use of:

  • Specific blocking antibodies against individual hedgehog proteins

  • CRISPR/Cas9-mediated gene editing to create selective knockouts

  • Domain-swapping experiments to identify protein-specific functional regions

  • Cell type-specific conditional knockouts in animal models

The tissue-specific effects of DHH further complicate analysis. For example, DHH's role in specifying fetal Leydig cell fate in testis organogenesis may involve different molecular partners than its role in peripheral nerve development . This necessitates parallel investigations across different tissue contexts with careful attention to cell-type specific responses.

Advanced techniques combining lineage tracing with single-cell transcriptomics are emerging as powerful approaches to address these challenges. These methods allow researchers to track cell populations over time while simultaneously monitoring gene expression changes, providing a more comprehensive picture of direct versus indirect DHH effects.

What are the most reliable methods for detecting and quantifying DHH protein in human samples?

The quantification of DHH protein in human samples requires sensitive and specific detection methods. Based on current research tools, the sandwich ELISA technique represents the most reliable standardized approach for DHH quantification . Commercial DHH ELISA kits offer the following performance specifications:

ParameterSpecification
Assay TypeSandwich
Detection MethodELISA
ReactivityHuman
Detection Range0.156-10 ng/mL
Sensitivity0.061 ng/mL
Intra-assay CV5.1%
Inter-assay CV9.9%

For researchers implementing DHH detection workflows, a comprehensive methodological approach involves:

  • Sample preparation: Optimize collection and processing to preserve protein integrity, including appropriate protease inhibitors and cold-chain management.

  • Quantification procedure: Follow standardized ELISA protocols with attention to temperature control, incubation times, and washing steps to ensure reproducibility.

  • Data analysis: Generate standard curves using recombinant DHH protein standards (available commercially in E. coli-expressed systems ) and apply appropriate curve-fitting models.

  • Validation: Confirm results using secondary methods such as Western blotting for molecular weight verification or mass spectrometry for peptide identification.

For localization studies, immunohistochemistry and immunofluorescence provide valuable spatial information about DHH distribution in tissues. These approaches can be complemented by in situ hybridization to detect DHH mRNA, offering insights into sites of DHH production versus action.

Researchers investigating DHH in specific developmental contexts may benefit from combining these protein detection methods with functional assays measuring downstream pathway activation, such as Gli transcription factor activity or target gene expression.

How can researchers effectively design experiments to investigate the role of DHH in human testicular development?

Designing experiments to elucidate DHH's role in human testicular development requires a multifaceted approach that addresses both mechanistic questions and translational implications. Based on current understanding, DHH/Patched1 signaling specifies fetal Leydig cell fate and regulates peritubular myoid cell development .

A comprehensive experimental design strategy should include:

  • Human tissue studies:

    • Analyze DHH expression patterns in human fetal testis samples across developmental stages

    • Correlate DHH levels with structural development using immunohistochemistry and histological techniques

    • Compare DHH expression in normal versus dysgenetic testes to identify pathological alterations

  • In vitro modeling:

    • Develop organ culture systems using human fetal testicular tissue

    • Employ CRISPR/Cas9 to modify DHH expression in testicular cells

    • Use recombinant DHH protein (such as the partial active form expressed in E. coli ) to supplement culture media and observe developmental effects

  • Signaling pathway analysis:

    • Investigate downstream mediators using phosphoproteomic approaches

    • Employ pathway inhibitors to block specific steps in DHH signaling

    • Utilize reporter constructs to visualize pathway activation in real-time

  • Translational correlation:

    • Screen patients with testicular dysgenesis for DHH mutations

    • Correlate genotype with phenotype to establish causality

    • Develop functional assays for identified mutations using cell-based systems

The experimental timeline should account for the developmental sequences in testicular formation, with particular attention to the critical windows when DHH signaling initiates and executes its developmental program. Comparative studies between human and model organism systems can provide valuable insights, though careful consideration of species-specific differences is essential.

For human studies, ethical considerations and appropriate regulatory approvals must be secured, particularly when working with fetal tissues. Collaboration between basic scientists, clinical researchers, and bioethicists strengthens the experimental design process and enhances the translational value of the research.

What collaborative frameworks are most effective for interdisciplinary DHH protein research?

Effective interdisciplinary research on DHH protein benefits from structured collaborative frameworks that acknowledge the complexity of both the biological system and the research ecosystem. Drawing from collaboration research, three critical interaction types should be considered when developing DHH research teams :

First, human-human interactions form the foundation of successful collaborations. The concept of "primus inter pares" (first among equals) can be valuable in DHH research teams, where leadership is present but responsive to the expertise of all team members . This approach recognizes that developmental biology, molecular biochemistry, clinical medicine, and bioinformatics all bring essential perspectives to DHH research.

Second, human-machine/material interactions shape knowledge production in contemporary research. In DHH studies, this includes recognition that computational tools, imaging systems, and laboratory equipment are not neutral instruments but active mediators that shape findings . Research teams should acknowledge how these tools influence experimental design and interpretation.

Third, machine/material-machine/material interactions become increasingly important as research techniques grow more sophisticated. In DHH research, this might involve understanding how automated high-throughput systems interact with biological samples or how imaging algorithms affect data interpretation .

An effective collaborative framework for DHH protein research should include:

  • Regular cross-disciplinary dialogues to develop shared vocabularies

  • Clear documentation of both successful and failed methodological approaches

  • Recognition of all contributors' roles in knowledge production

  • Mechanisms for constructive dissent to drive innovation

  • Pathways for translation between basic science and clinical applications

These frameworks can help overcome common collaboration challenges such as disciplinary silos, status differences between academic and technical contributors, and the pressure for accelerated research timelines .

How can researchers address the challenges of data integration in multi-omics approaches to DHH signaling research?

Multi-omics approaches to DHH signaling research generate diverse data types—genomic, transcriptomic, proteomic, and phenotypic—that must be integrated for comprehensive understanding. This integration presents both technical and collaborative challenges that researchers must systematically address.

From a collaborative perspective, data integration challenges mirror broader research collaboration issues, including status differences between team members, accelerating research timelines, and academic structures that privilege individual contributions . Overcoming these challenges requires acknowledging that data integration is not merely a technical problem but a collaborative one requiring diverse expertise.

Methodologically, researchers can address data integration challenges through:

  • Standardized data collection protocols:

    • Implement consistent sampling procedures across research groups

    • Standardize metadata capture to facilitate cross-study comparisons

    • Develop quality control metrics specific to DHH research

  • Integrated analysis platforms:

    • Utilize computational tools designed for multi-omics integration

    • Implement machine learning approaches for pattern recognition across data types

    • Develop visualization tools that represent relationships between different data layers

  • Knowledge management systems:

    • Create shared databases documenting DHH pathway components and interactions

    • Establish ontologies specific to developmental signaling research

    • Implement version control for both data and analysis pipelines

These technical solutions must be complemented by collaborative practices that recognize how "power differences, and therefore issues of leadership and of equality" influence research outcomes . This includes creating space for methodological discussions, acknowledging the contributions of data scientists and bioinformaticians as equal partners rather than service providers, and resisting the "drive towards absolute consistency" that might silence important disciplinary perspectives .

Successful multi-omics integration in DHH research ultimately requires resisting both hierarchical thinking that privileges certain data types or disciplines and the acceleration pressures that limit thorough documentation and methodological transparency.

What emerging technologies show promise for advancing our understanding of DHH protein function?

Several cutting-edge technologies are poised to transform our understanding of DHH protein function in development and disease. Single-cell multi-omics approaches represent a particularly promising frontier, allowing researchers to correlate DHH expression with global changes in cellular state at unprecedented resolution. These technologies can reveal how individual cells respond to DHH signaling in heterogeneous tissues like developing testes or peripheral nerves .

Spatial transcriptomics and proteomics technologies are enhancing our ability to map DHH distribution and activity within tissue contexts. These approaches preserve spatial information while providing molecular profiles, helping researchers understand how DHH gradients influence cell fate decisions across developing tissues. For DHH research specifically, these methods could elucidate the spatial relationships between DHH-producing cells and their targets during testis cord formation .

In structural biology, advances in cryo-electron microscopy are improving our ability to visualize protein-protein interactions at atomic resolution. Applied to DHH research, these techniques could reveal the precise molecular interfaces between DHH and its receptor PTCH1, potentially identifying specific structural features that distinguish DHH from other hedgehog family members .

Organoid technologies and microfluidic organ-on-chip platforms are creating new opportunities to model DHH function in human development. These systems can recapitulate aspects of testicular or nerve development in controlled environments, allowing for manipulation of DHH signaling and real-time observation of developmental processes.

CRISPR-based epigenome editing represents another frontier, potentially allowing researchers to modulate DHH expression without altering the underlying genetic sequence. This approach could help distinguish between developmental and maintenance roles of DHH signaling by enabling precise temporal control of gene expression.

How might advances in understanding DHH function translate into therapeutic approaches for associated disorders?

Advancing our understanding of DHH function opens several promising therapeutic avenues for associated disorders, particularly in reproductive medicine, oncology, and neurology. For disorders of sex development linked to DHH mutations, such as 46,XY complete pure gonadal dysgenesis, therapeutic strategies could involve either replacement of DHH function or modulation of downstream pathways .

In oncology, where DHH has been implicated in stomach adenocarcinoma progression, inhibitory approaches targeting either DHH itself or its downstream signaling components represent potential therapeutic strategies . Small molecule inhibitors of SMO have already entered clinical use for other hedgehog-driven cancers, suggesting a potential pathway for translation to DHH-associated malignancies.

For minifascicular neuropathy associated with DHH dysfunction, therapeutic approaches might focus on supporting myelin maintenance and nerve function through either restoration of DHH signaling or activation of compensatory pathways. Cell-based therapies using engineered Schwann cells could potentially address the myelin abnormalities observed in these conditions.

The preliminary association between DHH and autism spectrum disorder suggests another potential area for therapeutic exploration, though causality must first be firmly established . If confirmed, serum DHH levels might serve as biomarkers to guide personalized therapeutic approaches.

Translating these possibilities into clinical realities will require interdisciplinary collaboration between basic scientists, clinicians, and pharmaceutical developers. As noted in research on collaborative practices, such translation benefits from frameworks that acknowledge the distinct perspectives of different stakeholders while creating space for productive integration of knowledge .

Product Science Overview

Structure and Function

Desert Hedgehog is a secreted protein that undergoes autocatalytic cleavage, resulting in two distinct domains: the N-terminal signaling domain and the C-terminal domain. The N-terminal domain is responsible for the protein’s signaling activity, while the C-terminal domain is involved in precursor processing. The N-terminal domain is further modified by the addition of cholesterol and a fatty acid acyl chain, which anchor it to the cell membrane .

The primary function of DHH is to bind to Patched receptors (PTCH1 and PTCH2) on the cell surface. This binding relieves the repression of Smoothened (SMO) signaling, leading to the activation of downstream signaling pathways that regulate gene expression. DHH signaling is essential for various developmental processes, including male gonadal differentiation and perineurial development .

Recombinant Human Desert Hedgehog

Recombinant human Desert Hedgehog (rhDHH) is produced using recombinant DNA technology, typically in Escherichia coli (E. coli) expression systems. The recombinant protein is a non-glycosylated polypeptide chain containing 177 amino acids, with a molecular mass of approximately 20 kDa . The production of rhDHH involves chromatographic techniques to ensure high purity and biological activity.

Applications and Research

Recombinant human Desert Hedgehog is widely used in laboratory research to study its role in developmental biology and disease. It is particularly valuable for investigating the mechanisms of Hedgehog signaling and its implications in various conditions, such as partial gonadal dysgenesis and minifascicular polyneuropathy . Additionally, rhDHH is used in cell culture and tissue culture experiments to explore its effects on cell differentiation and tissue patterning.

Storage and Stability

Recombinant human Desert Hedgehog is typically lyophilized and stored at temperatures below -70°C to maintain its stability. Upon reconstitution, the protein remains stable for up to one week at 4°C or up to three months at -20°C. To enhance stability and prevent degradation, a carrier protein such as Bovine Serum Albumin (BSA) is often added .

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.