DUSP22 Human

Dual Specificity Phosphatase 22 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions and Mechanisms

DUSP22 regulates multiple signaling pathways through dephosphorylation:

SubstratePathway AffectedBiological Outcome
AKT (Serine/Threonine)PI3K/AKT/mTORTumor suppression in NSCLC
FAK (Tyrosine Y397)ERK1/2, NF-κBInhibition of NASH-HCC progression
EGFR (Tyrosine)EGFR/c-MetReduced lung tumorigenesis
Lck (Tyrosine Y394)TCR SignalingImmune response modulation

DUSP22 also suppresses STAT3 activation in T-cell lymphomas, altering cytokine production and tumor microenvironment dynamics .

Cancer

  • Non-Small Cell Lung Cancer (NSCLC): Low DUSP22 expression correlates with advanced TNM stages and poor prognosis. Exogenous DUSP22 expression reduces colony formation and xenograft growth by inhibiting EGFR phosphorylation .

  • Hepatocellular Carcinoma (HCC): Hepatic DUSP22 deficiency accelerates lipid deposition, inflammation, and fibrosis in NASH, promoting HCC. Gene therapy restoring DUSP22 mitigates these effects .

  • Anaplastic Large Cell Lymphoma (ALCL): DUSP22 rearrangements (e.g., t(6;7)(p25.3;q32.3)) occur in 28% of ALK-negative ALCL cases, associated with distinct morphology and immunophenotype but variable clinical outcomes .

Immune Regulation

DUSP22 knockout enhances T-cell activation and TCR signaling, exacerbating autoimmune responses. In ALCL, DUSP22 rearrangements reduce STAT3 activity and PD-L1 expression, altering immune evasion mechanisms .

Clinical and Therapeutic Implications

  • Prognostic Marker: Low DUSP22 expression in lung adenocarcinoma predicts shorter disease-free survival (HR = 1.8, P < 0.001) .

  • Therapeutic Target:

    • Gene Therapy: AAV-mediated DUSP22 delivery suppresses NASH-HCC in murine models .

    • Drug Sensitivity: DUSP22 knockdown increases EGFR inhibitor (gefitinib) efficacy in EGFR-mutant lung cancer .

Cancer TypeTherapeutic StrategyOutcome
NASH-HCCDUSP22 overexpression↓ Fibrosis, ↓ Tumor growth
ALCLJAK/STAT3 inhibitionSynergy with DUSP22-R status

Research Gaps and Future Directions

  • Paralog Interference: The 16p11.2 paralog complicates genetic analyses, necessitating improved detection methods .

  • Clinical Heterogeneity: While DUSP22-rearranged ALCL shows unique molecular profiles, its prognostic significance remains debated .

  • Mechanistic Depth: Further studies are needed to elucidate DUSP22’s role in non-canonical pathways, such as oxidative stress responses in fatty liver disease .

Product Specs

Introduction
Dual Specificity Phosphatase 22, also known as DUSP22, belongs to the protein-tyrosine phosphatase family. This phosphatase is characterized by a tyrosine-protein phosphatase domain. DUSP22 plays a role in activating the Jnk signaling pathway. It dephosphorylates and deactivates both p38 and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK). Notably, DUSP22 interacts with MAPK1 and MAPK8.
Description
Recombinant DUSP22, derived from humans and produced in E. coli, is a single, non-glycosylated polypeptide chain. This chain consists of 207 amino acids (specifically, amino acids 1 through 184) and has a molecular mass of 23.3 kDa. The DUSP22 is fused to a 23 amino acid His-tag located at the N-terminus. Purification is achieved using proprietary chromatographic methods.
Physical Appearance
A clear and colorless solution that has been sterilized through filtration.
Formulation
The DUSP22 protein solution is provided at a concentration of 0.5 mg/ml. It is formulated in a buffer consisting of 20mM Tris-HCl (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
For optimal storage, keep the solution at 4°C if it will be used within 2 to 4 weeks. For longer storage durations, freeze the solution at -20°C. When storing long-term, it is advisable to add a carrier protein such as 0.1% HSA or BSA. It is important to avoid repeated cycles of freezing and thawing.
Purity
Purity levels exceed 85.0%, as determined by SDS-PAGE analysis.
Synonyms
Dual specificity protein phosphatase 22, DUSP22, JNK-stimulatory phosphatase-1, JSP-1, Low molecular weight dual specificity phosphatase 2, LMW-DSP2, Mitogen-activated protein kinase phosphatase x, MAP kinase phosphatase x, MKP-x, LMWDSP2, MKPX, JKAP, JSP1, LMWDSP2, MKPX, VHX.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMGNGMNK ILPGLYIGNF KDARDAEQLS KNKVTHILSV HDSARPMLEG VKYLCIPAAD SPSQNLTRHF KESIKFIHEC RLRGESCLVH CLAGVSRSVT LVIAYIMTVT DFGWEDALHT VRAGRSCANP NVGFQRQLQE FEKHEVHQYR QWLKEEYGES PLQDAEEAKN ILAAPGILKF WAFLRRL.

Q&A

What is DUSP22 and what are its main functions in human cells?

DUSP22 is an atypical phosphatase consisting of 184 amino acid residues that is widely expressed across various human tissues. It belongs to the dual-specificity phosphatase family, with the ability to dephosphorylate both tyrosine and serine/threonine residues. DUSP22 is also known as JKAP (JNK pathway-associated phosphatase) due to its role in regulating the JNK pathway .

The main functions of DUSP22 include:

  • Dephosphorylation of multiple substrates including P38/MAPK, JNK/MAPK, estrogen receptor (ER), focal adhesion kinases (FAK), lymphocyte-specific protein tyrosine kinase, and signal transducer and activator of transcription 3 (STAT3)

  • Regulation of T-cell receptor signaling

  • Modulation of muscle cell differentiation and myofiber development

  • Control of endothelial-to-mesenchymal transition (EndMT)

What signaling pathways are regulated by DUSP22?

DUSP22 interacts with and regulates several major signaling pathways:

  • MAPK Signaling Pathway: DUSP22 dephosphorylates both P38/MAPK and JNK/MAPK, serving as a negative regulator of these pathways . In skeletal muscle, this regulation appears critical for preventing atrophy through a DUSP22-JNK-FOXO3a signaling axis .

  • Smad2/3 Pathway: In endothelial cells, DUSP22 regulates the Smad2/3 pathway, which plays a crucial role in EndMT .

  • FOXO3a Signaling: DUSP22 targeting downregulates genes linked to FOXO3a signaling, including key atrophy genes atrogin-1 and MuRF-1, as shown by RNA sequencing analysis of targeted aged muscle .

  • PI3K-Akt Pathway: Pharmacological targeting of DUSP22 has been shown to affect genes linked to the PI3K-Akt pathway, suggesting regulatory influence in this critical growth and survival pathway .

How is DUSP22 expression altered in pathological conditions?

DUSP22 expression undergoes significant alterations in several pathological conditions:

  • Skeletal Muscle Wasting: DUSP22 is upregulated in various muscle wasting conditions including:

    • Age-related sarcopenia

    • Dexamethasone-induced muscle atrophy

    • Immobilization-induced muscle atrophy, where DUSP22 expression inversely correlates with muscle mass

  • Endothelial Dysfunction: In the transforming growth factor-β-induced EndMT model using human umbilical vein endothelial cells (HUVECs), DUSP22 expression is downregulated . This downregulation appears to contribute to EndMT progression, while DUSP22 overexpression ameliorates EndMT .

  • Other Diseases: DUSP22 dysregulation has been linked to T-cell lymphoma and Alzheimer's disease, though the exact mechanisms require further investigation .

How does DUSP22 modulate skeletal muscle atrophy at the molecular level?

DUSP22 modulates skeletal muscle atrophy through several interconnected molecular mechanisms:

  • JNK-FOXO3a Signaling Axis: DUSP22 regulates a DUSP22-JNK-FOXO3a signaling axis. Pharmacological targeting or knockdown of DUSP22 inhibits this pathway, leading to downregulation of atrophy-related genes .

  • Atrogene Regulation: DUSP22 targeting downregulates key atrogenes:

    • Atrogin-1 (MAFbx): A muscle-specific ubiquitin ligase upregulated during atrophy

    • MuRF-1: A muscle RING-finger protein involved in ubiquitin-mediated proteolysis

  • Mitochondrial Function Modulation: DUSP22 overexpression affects mitochondrial function by:

    • Downregulating PGC-1α, a master regulator of mitochondrial biogenesis

    • Upregulating UCP-3, a mitochondrial uncoupling protein

  • Autophagy Pathway Activation: DUSP22 overexpression upregulates autophagy genes, including:

    • LC-3B (microtubule-associated proteins 1A/1B light chain 3B)

    • Ctsl (cathepsin L1)

  • Ubiquitin-Proteasome System (UPS) Regulation: DUSP22 overexpression upregulates UBR2 (ubiquitin protein ligase E3 component N-recognin 2), a component of the UPS .

What transcriptional changes occur following DUSP22 pharmacological targeting in aged muscle?

RNA sequencing analysis of tibialis anterior (TA) muscle in geriatric mice (27 months-old) revealed significant transcriptional changes following DUSP22 pharmacological targeting with BML-260:

  • Global Gene Expression Changes:

    • 350 differentially expressed genes (DEGs) were identified

    • Principal component analysis showed consistent effects of BML-260 on gene expression in aged mice

  • FOXO3a Signaling:

    • Downregulation of genes linked to FOXO3a signaling, including atrogin-1 and MuRF-1

  • Myokine Expression:

    • Upregulation of musclin (OSTN), an exercise-induced myokine that protects against muscle wasting

  • Muscle Cell Differentiation and Development:

    • Upregulation of genes linked to muscle cell differentiation and muscle fiber development

    • Increased expression of Myh4 (myosin heavy chain 2b), predominant in fast-twitch muscle

    • Upregulation of Six1 and Six4, which activate fast-type muscle gene programs

  • PI3K-Akt Pathway:

    • Downregulation of genes linked to the PI3K-Akt pathway

  • Gene Set Enrichment Analysis (GSEA):

    • Under-representation of genes involved in negative regulation of hypertrophy and response to inactivity

    • Enrichment of genes involved in skeletal muscle cell differentiation and proliferation

How does DUSP22 influence endothelial-to-mesenchymal transition in cardiovascular disease models?

DUSP22 plays a regulatory role in endothelial-to-mesenchymal transition (EndMT), a process implicated in cardiovascular diseases such as cardiac hypertrophy, myofibrosis, cardiac remodeling, and heart failure:

  • Expression Patterns During EndMT:

    • DUSP22 expression is downregulated during TGF-β-induced EndMT in human umbilical vein endothelial cells (HUVECs)

    • This downregulation correlates with phenotypic changes characteristic of EndMT

  • Effect on Endothelial and Mesenchymal Markers:

    • During DUSP22 downregulation, expression of endothelial cell markers (CD31, VE-cadherin, E-cadherin, VEGF) decreases

    • Simultaneously, mesenchymal cell markers (α-SMA, vimentin, N-cadherin, FSP1, fibronectin) increase

  • Signaling Pathway Regulation:

    • DUSP22 regulates EndMT through both the Smad2/3 and MAPK signaling pathways

    • Verification using signaling pathway inhibitors confirmed these regulatory mechanisms

  • Functional Consequences:

    • DUSP22 deficiency aggravates EndMT

    • Conversely, DUSP22 overexpression ameliorates EndMT

    • DUSP22 downregulation enhances HUVEC migration rates following TGF-β induction

What is the mechanism of action of BML-260 as a DUSP22 inhibitor?

BML-260 is a small molecule inhibitor that targets DUSP22 with several notable characteristics:

  • Chemical Structure and Properties:

    • Based on the rhodanine (2-thioxothiazolidin-4-one) chemical scaffold

    • Rhodanine is classified as a privileged heterocyclic compound amenable to structural modification

    • The compound has a yellow color and can interact photometrically in biological assessments

  • Molecular Docking:

    • Binding of BML-260 to human DUSP22 has been analyzed with CB-Dock2 software

    • The Vina score was calculated using human DUSP22 (PDB: 6lvq) and BML-260 (CID: 1565747)

  • Functional Effects in Skeletal Muscle:

    • BML-260 treatment increases mean body weight in dexamethasone-treated mice

    • Improves muscle performance in both acceleration and constant rotarod testing models

    • Increases myofiber cross-sectional area (CSA) and shifts myofiber area distribution toward larger sized fibers

    • Increases tibialis anterior muscle mass

    • Inhibits upregulation of atrogin-1 and MuRF-1

  • Effects in Human Skeletal Muscle Cells:

    • BML-260 treatment recovers mean myotube diameter and increases proportion of larger myotubes in dexamethasone-induced atrophy models

    • Inhibits MuRF-1 expression in human myotubes

What in vitro models are appropriate for studying DUSP22 function?

Several in vitro models have been validated for DUSP22 research:

  • Murine C2C12 Myoblast/Myotube System:

    • Growth conditions: DMEM with 10% fetal bovine serum and 1% penicillin/streptomycin

    • Differentiation protocol: Culture in DMEM with 2% horse serum for 96 hours

    • Atrophy induction: Treatment with 10 μM dexamethasone for 24 hours

    • Readouts: Myotube diameter, fusion index, differentiation index, gene expression analysis

  • Human Skeletal Myoblast System:

    • Commercial human skeletal myoblasts (available from Thermo-Fisher Scientific)

    • Differentiation: Culture in differentiation media for 72 hours

    • Atrophy induction: Treatment with 10 μM dexamethasone for 24 hours

    • Analysis: H&E staining, myotube diameter measurement via light microscopy

  • Human Umbilical Vein Endothelial Cells (HUVECs):

    • EndMT induction: Stimulation with 10 ng/ml TGF-β for 48 hours

    • Phenotypic characterization: Assessment of endothelial markers (CD31, VE-cadherin, E-cadherin, VEGF) and mesenchymal markers (α-SMA, vimentin, N-cadherin, FSP1, fibronectin)

    • Functional assessment: Cell migration assays

What genetic manipulation approaches are effective for DUSP22 studies?

Several genetic manipulation techniques have been successfully employed in DUSP22 research:

  • CRISPR-Cas9 Gene Editing:

    • Used for DUSP22 overexpression in C2C12 myoblasts

    • Confirmation of editing efficiency via qPCR

    • Enables assessment of effects on myotube formation, fusion index, and differentiation index

  • siRNA Knockdown:

    • In vitro: Effective for DUSP22 knockdown in cell culture models

    • In vivo: Intramuscular delivery using Invivofectamine 3.0 reagent

    • Protocol for aged mice: 35 μg siRNA complexes injected into TA muscle, with a second injection 4 days later, and analysis at day 10

    • Verification: qPCR confirmation of knockdown efficiency

  • Plasmid-Based Overexpression:

    • Used for DUSP22 overexpression studies in HUVECs

    • Allows assessment of EndMT phenotype reversal

What in vivo models are most effective for studying DUSP22 in disease contexts?

Multiple in vivo models have been validated for studying DUSP22's role in disease:

  • Dexamethasone-Induced Muscle Atrophy Model:

    • Dexamethasone treatment induces muscle wasting

    • Useful for testing pharmacological agents

    • Metrics: Body weight, grip strength, rotarod performance, muscle histology, gene expression

  • Aging-Related Sarcopenia Model:

    • Geriatric mice (24-27 months old)

    • Allows study of age-related muscle wasting

    • Compatible with both genetic (siRNA) and pharmacological (BML-260) interventions

    • Treatment protocol: 5 mg/kg BML-260 via intraperitoneal delivery every 24h for 4 weeks

  • Hind Limb Immobilization Model:

    • 14-week-old male C57BL/6 mice with immobilized hind limbs using plastic EP-tubes

    • Immobilization period: 14 days

    • Allows study of disuse-induced muscle atrophy

    • Compatible with pharmacological intervention (5 mg/kg BML-260)

What is the therapeutic potential of DUSP22 targeting in human diseases?

DUSP22 targeting shows promising therapeutic potential across multiple human diseases:

  • Skeletal Muscle Wasting Disorders:

    • Effective in multiple models of muscle wasting including aging-related sarcopenia, dexamethasone-induced atrophy, and immobilization-induced atrophy

    • BML-260 prevents muscle wasting via inhibition of the DUSP22-JNK-FOXO3a signaling axis

    • Efficacy demonstrated in human donor-derived skeletal muscle cells, suggesting translational potential

  • Cardiovascular Diseases:

    • DUSP22 overexpression ameliorates endothelial-to-mesenchymal transition (EndMT)

    • Potential relevance for cardiac hypertrophy, myofibrosis, cardiac remodeling, and heart failure

    • The identified DUSP22 regulatory role in EndMT may lead to new therapeutic strategies

  • Other Potential Indications:

    • Previous research has linked aberrant DUSP22 to T-cell lymphoma and Alzheimer's disease

    • May be relevant to other aging-related disorders such as osteoarthritis and type 2 diabetes

    • Potential role in depression via regulation of musclin, a myokine shown to alleviate depressive symptoms in animal models

What biomarkers can be used to assess the efficacy of DUSP22-targeted interventions?

Several biomarkers have been validated for assessing DUSP22-targeted interventions:

  • Molecular Biomarkers:

    • DUSP22 expression levels (measured by qPCR or western blot)

    • Atrogene expression (atrogin-1, MuRF-1) as indicators of muscle atrophy pathways

    • JNK and FOXO3a phosphorylation status as indicators of pathway activation

    • Musclin (OSTN) levels as indicator of anti-atrophic response

  • Functional/Physiological Biomarkers:

    • Muscle mass measurements (individual muscle weights)

    • Myofiber cross-sectional area distributions

    • Grip strength as indicator of muscle function

    • Rotarod performance (acceleration and constant models)

  • Histological Markers:

    • Myofiber type distribution (fast vs. slow twitch fibers)

    • Myotube diameter in cell culture models

    • Expression of endothelial vs. mesenchymal markers in EndMT models

What are the most promising avenues for DUSP22 drug development?

Several promising approaches for DUSP22-targeted drug development emerge from current research:

  • Rhodanine-Based Scaffold Optimization:

    • BML-260 is based on the rhodanine (2-thioxothiazolidin-4-one) chemical scaffold

    • This scaffold is amenable to structural modification and lead development

    • Further rounds of structural optimization could improve drug-like properties, biological stability, and DUSP22 targeting specificity

  • Theranostic Development:

    • Rhodanine's yellow color and photometric interaction potential in biological assessments suggests possible development as a theranostic agent

    • Could potentially serve as both a diagnostic biomarker and therapeutic drug for skeletal muscle wasting

  • Alternative JNK Inhibition Strategies:

    • DUSP22 targeting represents an indirect approach to JNK inhibition

    • May circumvent limitations encountered with direct JNK inhibitors, none of which have yet achieved clinical approval despite extensive research

  • Tissue-Specific Delivery Systems:

    • Development of targeted delivery systems for skeletal muscle or endothelial tissue could enhance efficacy while reducing off-target effects

    • Current in vivo studies use systemic (intraperitoneal) or local (intramuscular) delivery approaches

How might DUSP22's role in musclin regulation inform therapeutic approaches?

The discovery that DUSP22 targeting upregulates musclin expression opens interesting therapeutic possibilities:

  • Cardio-Sarcopenia Applications:

    • Musclin has been shown to be critical for cardiac conditioning

    • Cardiac hypertrophy and skeletal muscle wasting share similar molecular mechanisms

    • Cardio-sarcopenia is recognized as a syndrome of concern in aging

    • DUSP22 targeting might address both cardiac and skeletal muscle aspects of aging-related decline

  • Neuropsychiatric Applications:

    • Skeletal muscle-secreted musclin has recently been shown to alleviate depression in animal models

    • DUSP22 targeting might have unexplored potential in depressive disorders through musclin upregulation

    • This represents a novel link between muscle physiology and mental health

  • Mechanistic Understanding:

    • Further research is needed to determine whether musclin levels are directly regulated by DUSP22 or via downstream effects on JNK-FOXO3a inhibition

    • Understanding this regulatory mechanism could inform more targeted therapeutic approaches

Product Science Overview

Biological Properties and Functions

DUSP22 plays a crucial role in various cellular processes by participating in multiple signaling cascades. It is known to activate the JNK signaling pathway and inactivate the tyrosine kinase Lck by dephosphorylating the tyrosine-394 residue during T cell receptor signaling . Additionally, DUSP22 dephosphorylates and deactivates p38 and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK), which are involved in stress response pathways .

Expression Patterns and Tissue Distribution

DUSP22 is expressed in various tissues, with significant roles in immune cells. Dysregulation of DUSP22 has been associated with human lupus nephritis, highlighting its importance in immune regulation .

Recombinant Human DUSP22

Recombinant Human DUSP22 is typically produced in Escherichia coli (E. coli) or wheat germ expression systems. The recombinant protein often includes a purification tag, such as a His-tag or GST-tag, to facilitate purification and detection . The recombinant protein is used in various research applications, including Western Blotting (WB), Enzyme-Linked Immunosorbent Assay (ELISA), and functional studies.

Applications and Research Use

Recombinant Human DUSP22 is primarily used for research purposes to study its role in signaling pathways and its potential therapeutic applications. It is not approved for use in humans or clinical diagnosis .

Storage and Handling

Recombinant Human DUSP22 should be stored at -20°C for long-term storage and at 4°C for short-term storage. It is important to avoid repeated freeze-thaw cycles to maintain protein integrity .

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.