Thymosin α1

Thymosin a1 Acetate
Shipped with Ice Packs
In Stock

Description

Introduction and Basic Properties

Tα1 is a synthetic analogue of thymosin α1, a peptide isolated from thymus extracts . Key properties include:

PropertyValueSource
CAS Number62304-98-7
Molecular FormulaC₁₂₉H₂₁₅N₃₃O₅₅
Molecular Weight3108.28 Da
Amino Acid SequenceAc-Ser-Asp-Ala-Ala-Val-Asp-Thr-Ser-Ser-Glu-Ile-Thr-Thr-Lys-Asp-Leu-Lys-Glu-Lys-Lys-Glu-Val-Val-Glu-Glu-Ala-Glu-Asn-OH
Storage ConditionsLyophilized powder stored at -18°C; reconstituted solutions at 4°C

Tα1 is synthesized chemically due to challenges in biosynthesis, including Nα-acetylation and small peptide expression .

Biological Mechanisms and Activities

Tα1 exhibits pleiotropic immune-regulatory effects through multiple pathways:

Immunomodulation

  • T Cell Activation: Enhances maturation of T cells (CD4+/CD8+) and prevents apoptosis .

  • Cytokine Modulation:

    • Upregulates IL-2, IL-12, IFN-γ, and IL-10 .

    • Downregulates pro-inflammatory cytokines (IL-1β, TNF-α) .

  • Antigen Presentation: Increases MHC-I expression and viral antigen visibility on infected cells .

Anti-Tumor Activity

  • Inhibits growth of PD-L1-expressing non-small cell lung cancer (NSCLC) and multiple myeloma cells in vitro .

  • Reduces metastatic lesions in NSCLC xenograft models .

Anti-Infective Properties

  • Acts as a Toll-like receptor (TLR-2/9) agonist, stimulating innate and adaptive immunity .

  • Enhances vaccine efficacy by boosting antibody production .

Clinical Applications and Therapeutic Uses

Tα1 is approved for hepatitis B/C in 35 countries and investigated in diverse conditions:

ConditionMechanismClinical Evidence
Hepatitis B/CReduces viral replication; restores T cell functionApproved in multiple countries
Cancer (e.g., NSCLC, Melanoma)Inhibits PD-L1+ tumor growth; adjuvant to chemotherapyPhase II trials show reduced toxicity
COVID-19Modulates cytokine storms; enhances T cell responseMixed results: reduced mortality in some studies , no benefit in others
Autoimmune DiseasesBalances Th1/Th2 cytokine profilesInvestigational

Key Clinical Trials

StudyDesignOutcomeReference
Sepsis (TESTS, 2025)Phase III, n=1,106No mortality reduction vs. placebo
COVID-19 (2021)Retrospective, n=1,026Higher non-recovery rate with Tα1
Cystic FibrosisPreclinicalReduces A. fumigatus infection severity

Product Specs

Introduction
Thymalfasin, a synthetic version of thymosin-alpha-1, is a 28-amino acid protein originating from the prothymosin-alpha protein. Known for its various immune-regulating effects, thymosin-alpha-1 promotes the maturation of murine T-cell precursors, human thymocytes, and underdeveloped cord blood lymphocytes. It also stimulates the production of IL-2, high-affinity IL-2 receptors, and B-cell growth factors by peripheral blood mononuclear cells. Thymosin's activity targets both T-helper and cytotoxic/suppressor T-cell groups. Furthermore, studies indicate that thymosin-alpha-1 enhances the effectiveness of antigen presentation by macrophages and acts as a natural regulator of alpha-thrombin activity.
Description
Thymosin α1 acetate, also called thymalfasin, possesses immune-boosting qualities that enhance the body's defense mechanisms. This protein boasts a molecular formula of C129H215N33O55, an amino acid sequence of Ac-Ser-Asp-Ala-Ala-Val-Asp-Thr-Ser-Ser-Glu-Ile-Thr-Thr-Lys-Asp-Leu-Lys-Glu-Lys-Lys-Glu-Val-Val-Glu-Glu-Ala-Glu-Asn-OH, and a molecular weight of 3108.32 Daltons.
Physical Appearance
The product appears as a sterile, white powder obtained through a freeze-drying process.
Formulation
The protein, at a concentration of 1mg/ml, underwent a freeze-drying process without any additional ingredients.
Solubility
For reconstitution, dissolve the lyophilized Thymosin α1 in sterile 18MΩ-cm H2O at a minimum concentration of 100 µg/ml. This solution can be further diluted using other aqueous solutions.
Stability
While the lyophilized Thymosin α1 remains stable at room temperature for up to 3 weeks, it is best stored in a dry environment below -18°C. After reconstitution, keep Thymalfasin refrigerated at 4°C for a maximum of 2 to 7 days. For extended storage, freeze the solution below -18°C. It's crucial to avoid repeated freezing and thawing cycles. To further enhance long-term storage, consider adding a carrier protein such as 0.1% HSA or BSA.
Purity
The purity exceeds 99.0%, as confirmed by the following analyses: (a) Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) and (b) Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE).

Q&A

What is the biological origin of Thymosin α1?

Thymosin α1 is an endogenous peptide originally identified in the thymus gland. It is the asparaginyl endopeptidase cleavage product of prothymosin α (ProTα), an acidic nuclear protein consisting of 109 amino acid residues . While highly concentrated in the thymus, particularly in thymic epithelial cells, Tα1 is also found in smaller amounts in other lymphoid tissues (spleen, lymph nodes) and non-lymphoid tissues (lungs, kidneys, brain) . The peptide was first isolated and characterized from calf thymus in 1977, identified as part of thymosin fraction 5 (TF5) .

What are the primary molecular mechanisms through which Thymosin α1 exerts its immunomodulatory effects?

Thymosin α1 functions through multiple immunomodulatory pathways:

  • It augments T-cell–mediated immune responses by stimulating T-cell differentiation and maturation

  • It activates natural killer cells

  • It stimulates dendritic cell function and maturation

  • It modulates the production of pro-inflammatory cytokines

  • It plays a role in activating indoleamine 2,3-dioxygenase 1-dependent tolerogenic programs in dendritic cells (DCs)

  • It regulates the function of regulatory T (Treg) cells, contributing to self-tolerance maintenance

  • It blocks steroid-induced apoptosis of thymocytes

This multi-faceted activity profile explains why Tα1 has been investigated across various immune-related conditions.

What is the relationship between Thymosin α1 and autoimmune regulator (AIRE)?

Recent research has revealed an intricate crosstalk between Thymosin α1 and the autoimmune regulator (AIRE). AIRE is a transcriptional regulator highly expressed in thymic medullary epithelial cells where it controls the ectopic expression of tissue-restricted antigens for negative selection . The relationship appears bidirectional:

  • AIRE may promote prothymosin cleavage to Tα1

  • Tα1 in turn transcriptionally regulates AIRE expression

This interaction suggests Tα1 plays a significant role in central and peripheral tolerance mechanisms. The absence of AIRE-induced tissue-specific antigens in the thymus can lead to autoimmunity in the antigen-expressing target organ, highlighting the importance of this regulatory pathway .

What are the optimal methods for quantifying Thymosin α1 in biological samples?

For researchers quantifying Tα1 in biological samples, several methodological approaches are validated:

  • Immunoassays: The gold standard for measuring circulating Tα1, with detection ranges capable of measuring the normal physiological range (252-1158 pg/ml in healthy adults) . These include:

    • ELISA (Enzyme-Linked Immunosorbent Assay)

    • RIA (Radioimmunoassay)

  • Mass Spectrometry: For more precise identification and quantification in complex biological matrices

  • Western Blotting: Useful for semi-quantitative analysis in tissue samples

When designing experiments to measure Tα1, researchers should consider tissue-specific variations in concentration and potential degradation in ex vivo samples. Timing of sample collection is critical since Tα1 levels may fluctuate during immune responses or with circadian rhythms.

What experimental models are most appropriate for studying Thymosin α1's effects?

Based on successful preclinical studies, several experimental models have proven valuable:

  • Murine Melanoma Models: C57BL/6 mice challenged with B16 melanoma cells have demonstrated significant survival benefits when treated with Tα1 in combination with cyclophosphamide and IFN-αβ . This model showed 20-30% of mice receiving high-dose Tα1 remained tumor-free for up to a year, compared to no tumor-free animals in other treatment groups .

  • Ex vivo T-cell and Dendritic Cell Cultures: Allow for isolated examination of cellular mechanisms

  • Immunocompromised Models: Valuable for investigating Tα1's potential to restore immune function

For meaningful translational results, it's crucial to select models that reflect the specific aspect of Tα1 biology being investigated. Researchers should consider that Tα1's effects may vary significantly between species and between in vitro and in vivo conditions.

How should researchers approach the production of Thymosin α1 for experimental use?

Researchers have multiple options for sourcing Tα1:

  • Commercial Synthetic Peptide: Currently, clinical-grade Tα1 (ZADAXIN®) is synthesized using solid-phase peptide synthesis . This approach ensures high purity and consistent quality, though at potentially higher cost.

  • Genetic Engineering Expression Systems: Recent advances allow for biological production:

    • Prokaryotic expression systems (E. coli)

    • Eukaryotic expression systems (yeast, mammalian cells)

When evaluating biologically produced Tα1, researchers should verify its activity by measuring:

  • Cytokine secretion induction

  • Lymphocyte proliferation stimulation

  • Specific receptor binding

It's essential to characterize any recombinant Tα1 thoroughly before experimental use, comparing its activity to established synthetic standards.

How does Thymosin α1 interact with traditional chemotherapeutic agents in cancer models?

The interaction between Tα1 and chemotherapeutic agents represents a complex area of research with significant clinical implications. Evidence shows:

  • Enhancement of Efficacy: In melanoma treatment, combining Tα1 with dacarbazine (DTIC) plus interferon alfa showed promising results in phase II studies. After a mean of 5.3 treatment cycles, 50% of patients demonstrated response (25% complete response, 25% partial response) .

  • Reduced Toxicity Profile: When Tα1 was added to chemotherapy regimens, researchers observed reduced hematologic toxicity compared to chemotherapy alone. In non-small-cell lung cancer patients, grade 3-4 hematologic toxicity was 0% in the Tα1 group versus 50% in the ifosfamide-only group .

  • Immune Response Activation: Mechanistically, Tα1 appears to enhance anti-tumor immunity in the context of chemotherapy, potentially through:

    • Increased CD3 and CD8 cell levels

    • Enhanced natural killer cell activity

    • Improved dendritic cell function

A detailed comparison of adverse events from a large multicenter study is presented in the table below:

System Organ ClassDIT 1.6 (n = 97)DIT 3.2 (n = 97)DIT 6.4 (n = 98)DT 3.2 (n = 98)DI (n = 95)
Blood and lymphatic system disorders20.619.614.320.411.6
Gastrointestinal disorders48.556.733.738.842.1
General disorders and administration site conditions58.863.957.136.754.7
Infections and infestations16.515.511.214.314.7
Metabolism and nutrition disorders21.719.610.217.414.7
Nervous system disorders23.714.416.313.310.5

Note: DIT = dacarbazine (DTIC) plus interferon alfa (IFN-α) and thymosin α1 (Tα1); DT = DTIC plus Tα1; DI = DTIC and IFN-α .

What are the considerations for investigating Thymosin α1's role in central versus peripheral immune tolerance?

Investigating Tα1's dual roles in central and peripheral tolerance requires sophisticated experimental approaches:

  • Central Tolerance Studies:

    • Focus on Tα1's interaction with AIRE in thymic medullary epithelial cells

    • Examine negative selection processes of self-reactive T cells

    • Investigate the transcriptional regulation between AIRE and Tα1

    • Consider thymic organoid models to study these interactions in controlled environments

  • Peripheral Tolerance Mechanisms:

    • Explore Tα1's effects on dendritic cell tolerogenic programming

    • Study regulatory T cell induction and function in peripheral tissues

    • Investigate tissue-specific expression patterns of Tα1

  • Experimental Considerations:

    • Use conditional knockout models to differentiate central versus peripheral effects

    • Employ lineage tracing for thymic-derived versus peripherally-induced Tregs

    • Apply single-cell technologies to capture cellular heterogeneity in responses

Recent findings suggest that AIRE protein has been detected in peripheral lymphoid organs, indicating peripheral AIRE may play a complementary role to its thymic function . This discovery opens new avenues for investigating how Tα1 might regulate tolerance across multiple tissue compartments.

How should researchers approach dose-response relationships in Thymosin α1 studies?

Determining optimal dosing for Tα1 experiments requires methodical approaches:

  • Preclinical Dose Finding:

    • In murine melanoma models, Tα1 at 6,000 μg/kg/d demonstrated significantly greater survival benefit than 200 or 600 μg/kg/d doses

    • Splenocytes from mice treated with the highest Tα1 dose showed markedly increased cytotoxicity against melanoma cells

  • Clinical Dosing Considerations:

    • Phase II melanoma studies employed 1-2 mg/d Tα1 administered on specific days within treatment cycles

    • In hepatocellular carcinoma studies, 900 μg/m² twice weekly for 6 months was used

    • Non-small-cell lung cancer protocols used 1 mg on days 8-11 and 15-18 of 21-day cycles

  • Experimental Design Recommendations:

    • Include multiple dose cohorts when possible

    • Consider different dosing schedules, not just absolute dose

    • Measure pharmacokinetic parameters when feasible

    • Correlate dosing with specific immunological biomarkers

For robust dose-response characterization, researchers should include time-series analysis since Tα1's immunomodulatory effects may have distinct temporal patterns, with some responses occurring rapidly and others developing over extended periods.

What approaches can researchers use to study the relationship between Thymosin α1 and the epithelial cell/dendritic cell crosstalk?

Investigating the complex crosstalk between epithelial cells (ECs), dendritic cells (DCs), and Tα1 requires specialized approaches:

  • Co-culture Systems:

    • Establish co-cultures of thymic or peripheral epithelial cells with immature dendritic cells

    • Measure changes in dendritic cell maturation markers, cytokine production, and T cell stimulatory capacity

    • Evaluate the impact of adding or neutralizing Tα1 in these systems

  • Transcriptomic Analysis:

    • Apply RNA-seq to identify gene expression changes in both ECs and DCs during their interaction

    • Focus on pathways influenced by Tα1 signaling

    • Compare wild-type with Tα1-deficient conditions

  • Signaling Pathway Investigation:

    • Map the receptors and downstream signaling molecules activated by Tα1 in both cell types

    • Use phospho-specific antibodies to track signaling cascades

    • Apply pathway inhibitors to determine critical nodes

  • In vivo Models:

    • Develop conditional knockout models for cell-specific deletion of Tα1 or its receptors

    • Use intravital microscopy to visualize EC/DC interactions in thymic and peripheral tissues

Research has established that Tα1 expressed within the thymus and peripheral tissues regulates the EC/DC crosstalk required for healthy immune homeostasis . Understanding the molecular mechanisms of this regulation could provide insights into fundamental immune tolerance processes.

How can researchers effectively study the therapeutic potential of Thymosin α1 in combination immunotherapies?

Given the complex interactions in modern immunotherapy, systematic approaches to studying Tα1 combinations include:

  • Rational Combination Design:

    • Pair Tα1 with agents targeting complementary immune pathways

    • Consider combinations with checkpoint inhibitors (anti-PD-1, anti-CTLA-4)

    • Evaluate synergy with cancer vaccines or CAR-T approaches

  • Mechanistic Studies:

    • Determine if Tα1 enhances T cell infiltration into tumors

    • Assess changes in tumor microenvironment (cytokines, immune cell composition)

    • Measure impact on dendritic cell antigen presentation and T cell priming

  • Biomarker Development:

    • Identify predictive biomarkers for Tα1 response

    • Develop pharmacodynamic markers to confirm on-target activity

    • Establish immune monitoring panels optimized for Tα1-based combinations

  • Experimental Models:

    • Use syngeneic and humanized mouse models to assess combination efficacy

    • Consider patient-derived xenografts for translational relevance

    • Implement ex vivo human tumor slice cultures for rapid screening

Historical data supports this approach, as Tα1 has shown ability to improve efficacy of dacarbazine-based regimens in melanoma, with 50% response rates in phase II studies and median survival time of 11.5 months . Similar encouraging results were observed when Tα1 was combined with DTIC and interleukin-2, yielding a 36% objective response rate .

What methodological considerations are important when studying Thymosin α1's effects on indoleamine 2,3-dioxygenase 1-dependent tolerogenic pathways?

Investigating Tα1's role in tolerogenic programming requires specific methodological considerations:

  • Enzymatic Activity Assessment:

    • Measure IDO1 activity through kynurenine/tryptophan ratios

    • Use specific IDO1 inhibitors as controls

    • Evaluate changes in IDO1 expression at transcriptional and protein levels

  • Dendritic Cell Functional Analysis:

    • Characterize tolerogenic DC phenotypes (surface markers, cytokine profiles)

    • Assess DC capacity to induce T regulatory cells

    • Examine antigen presentation capabilities under Tα1 influence

  • T Cell Response Evaluation:

    • Measure generation of FoxP3+ regulatory T cells

    • Assess suppressive function of Tregs generated under different conditions

    • Analyze cytokine production profiles

  • Tissue-Specific Considerations:

    • Compare effects in different tissue microenvironments

    • Consider tissue-resident DC populations which may respond differently

    • Evaluate the role of local Tα1 concentrations

Research has established that Tα1 contributes to self-tolerance maintenance by regulating Treg cell function through IDO1-dependent tolerogenic programming in DCs . This mechanism represents a critical aspect of immune homeostasis that may have implications for autoimmune disease and cancer immunotherapy research.

What are the key unanswered questions regarding Thymosin α1's role in immune development and homeostasis?

Despite significant advances, several fundamental questions remain:

  • Developmental Biology:

    • How does Tα1 contribute to thymic development and maturation?

    • What is the precise role of Tα1 in T cell lineage commitment?

    • How does Tα1 influence thymic involution with aging?

  • Regulatory Mechanisms:

    • What are the complete transcriptional networks regulated by Tα1?

    • How is Tα1 production and processing regulated at the molecular level?

    • What determines tissue-specific responses to Tα1?

  • Receptor Biology:

    • What is the primary receptor for Tα1?

    • How does Tα1 signaling differ between cell types?

    • What are the signaling pathways activated downstream of Tα1 binding?

  • Evolutionary Perspectives:

    • How conserved is Tα1 function across species?

    • What can comparative biology tell us about essential versus adaptive functions?

Understanding these fundamental aspects will provide crucial insights into both basic immunology and potential therapeutic applications of Tα1.

How might novel genetic engineering approaches advance Thymosin α1 research and production?

Emerging genetic technologies present opportunities to revolutionize Tα1 research:

  • Production Optimization:

    • CRISPR-engineered cell lines for enhanced Tα1 expression

    • Synthetic biology approaches to create optimized production systems

    • Novel fusion proteins to improve stability or targeting

  • Genetic Manipulation Tools:

    • Conditional knockout models to study Tα1 in specific tissues or developmental stages

    • Inducible expression systems to control timing of Tα1 activity

    • Reporter systems to track Tα1 production and activity in real-time

  • Therapeutic Development:

    • Gene therapy approaches to deliver Tα1 to specific tissues

    • Engineered cell therapies that produce Tα1 in response to specific signals

    • Modification of Tα1 sequence for enhanced pharmacokinetics or target specificity

Current research indicates that genetic engineering methods using prokaryotic or eukaryotic expression systems can produce biologically active Tα1, with demonstrated effectiveness in increasing cytokine secretion and promoting lymphocyte proliferation in vitro . This opens promising avenues for biotechnological production of Tα1 for both research and clinical applications.

Product Science Overview

Discovery and Structure

Thymosin α1 was first isolated from thymosin fraction 5, a preparation derived from the thymus gland. It was the first peptide from this fraction to be completely sequenced and synthesized . The amino acid sequence of Thymosin α1 is as follows: Ac-Ser-Asp-Ala-Ala-Val-Asp-Thr-Ser-Ser-Glu-Ile-Thr-Thr-Lys-Asp-Leu-Lys-Glu-Lys-Lys-Glu-Val-Val-Glu-Glu-Ala-Glu-Asn-OH .

Biological Function

Thymosin α1 plays a crucial role in the immune system. It enhances cell-mediated immunity by promoting the maturation of T cells, which are essential for the adaptive immune response . This peptide is believed to be a major component responsible for restoring immune function in animals lacking thymus glands .

Therapeutic Applications

Thymosin α1 has been approved in several countries for the treatment of various diseases, particularly those involving immune dysfunction. It is used to treat chronic hepatitis B and C, and it has shown potential in treating other conditions such as cystic fibrosis, septic shock, acute respiratory distress syndrome, peritonitis, acute cytomegalovirus infection, tuberculosis, severe acute respiratory syndrome, and lung infections in critically ill patients . Additionally, it has been studied for its potential use in cancer treatment, particularly in conjunction with chemotherapy .

Mechanism of Action

Thymosin α1 modulates the immune response by enhancing the function of dendritic cells and T cells. It upregulates activation markers such as CD40, CD80, and TIM-3, particularly in plasmacytoid dendritic cells (pDCs), and increases the production of TNFα . This modulation helps improve lymphocyte functionality and reduces the production of proinflammatory cytokines, making it a beneficial therapeutic alternative for various immune-related conditions .

Clinical Studies and Future Prospects

Clinical trials have demonstrated the efficacy of Thymosin α1 in enhancing immune responses and improving outcomes in patients with immune-related diseases . Its potential as an adjuvant in SARS-CoV2 treatment has also been explored, showing promising results in reducing proinflammatory cytokine production and improving T cell functionality . As research continues, Thymosin α1 may find broader applications in immunotherapy and the treatment of autoimmune disorders.

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.