scispace - formally typeset
Search or ask a question

How to reconstitute TGF-β1? 


Best insight from top research papers

To reconstitute TGF-β1, a scalable purification process from virally inactivated human platelets can be employed, where platelet lysates are treated with solvent/detergent, clarified, and chromatographed on an anion-exchange column to obtain a fraction enriched in TGF-β1 . Additionally, a complex involving a TGF-β1 inhibitory peptide and cyclodextrin has been developed for therapeutic purposes . The interaction between TGF-β1 and its receptors, T beta R-I and T beta R-II, has been extensively studied, showing that T beta R-II binds TGF-β1, leading to the recruitment and phosphorylation of T beta R-I, essential for downstream signaling . Furthermore, for abdominal aortic aneurysms, TGF-β1 has been encapsulated in PLGA nanoparticles to induce elastogenesis, demonstrating sustained release and bio-functionality of the protein .

Answers from top 5 papers

More filters
Papers (5)Insight
Reconstitute TGF-β1 by activating lyophilized TGF-β1 with acidic buffer containing BSA, followed by reconstitution in PBS with BSA before use, as detailed in the study.
TGF-β1 can be reconstituted by forming a complex between receptors TβR-I and TβR-II, where TβR-II phosphorylates TβR-I, initiating signal propagation to downstream substrates.
TGF-β1 can be reconstituted by purifying it from virally inactivated human platelet lysates using a scalable chromatographic process, yielding a fraction enriched in TGF-β1 for potential therapeutic applications.
Not addressed in the paper.
Not addressed in the paper.

Related Questions

How does TGF-β-Neutralizing Antibody 1D11 work?5 answersThe TGF-β-neutralizing antibody 1D11 works by targeting the TGF-β signaling pathway to exert its effects in various conditions. In metastatic breast cancer, 1D11 enhances the CD8+ T-cell-mediated antitumor immune response, increases infiltration of natural killer cells and T cells at metastatic sites, and enhances expression of cytotoxic effectors on CD8+ T cells. In osteogenesis imperfecta, 1D11 treatment increases trabecular bone volume and strength, suppresses serum biomarkers of bone turnover, and affects osteoblast and osteoclast numbers. In cancer therapy, specifically in colon cancer and melanoma models, blocking TGF-β1 with 1D11 enhances the efficacy of immunotherapies by increasing infiltration of activated CD8 T cells and delaying tumor progression through a tumor-intrinsic effect. Additionally, in T. cruzi infection, 1D11 reduces cardiomyocyte invasion and parasite load, improving cardiac performance and reducing fibrosis.
What is a TGF-B ?4 answersTransforming growth factor-β (TGF-β) is a multifunctional cytokine involved in cancer progression, immune regulation, and tissue homeostasis. In cancer, TGF-β exhibits dual roles, acting as a tumor suppressor in early stages by inducing apoptosis and cell cycle arrest, while promoting tumor growth, invasion, and metastasis in advanced stages. TGF-β also plays a crucial role in immune suppression within the tumor microenvironment, limiting T cell activation and promoting resistance to immune checkpoint inhibitors. Moreover, TGF-β can modulate the behavior of pericytes in glioblastoma, leading to blood-brain barrier disruption. Additionally, TGF-β-producing B cells have been implicated in regulating immune responses in cancer, autoimmune disorders, and inflammatory diseases. Overall, TGF-β is a key player in various physiological and pathological processes, making it a potential target for therapeutic interventions.
How TGF beta 1 protein purified from e.coli?4 answersTransforming growth factor beta 1 (TGF-β1) protein can be purified from Escherichia coli (E. coli) using various methods. One approach involves the use of fusion tags to increase solubility and facilitate purification. For example, the fusion of TGF-β1 with thioredoxin (Trx) or superfolder GFP (sfGFP) has been successful in producing soluble forms of the protein. After expression in E. coli, the fusion protein is typically found in insoluble form as inclusion bodies. These inclusion bodies can be solubilized and refolded using redox systems such as glutathione or glutathione redox-coupled systems. The fusion protein can then be purified using affinity chromatography, such as Ni-NTA agarose, to separate the carrier protein from the target protein. Additional purification steps, such as cation-exchange chromatography, can be employed to separate monomeric molecules from noncovalently bound oligomers. The purified TGF-β1 protein can be confirmed for its structure and biological activity using techniques like circular dichroism analysis and bioassays.
What is the role of TGF-β in NET formation and function?4 answersTGF-β is a potent immunosuppressive cytokine that affects the function of NK cells. It can alter the expression of effector molecules, chemokine receptors, and activate the conversion of NK cells to less cytolytic ILC1s. TGF-β signaling in lymphatic endothelial cells maintains the structure of lymphatic vessels and promotes tumor lymphatic metastasis. TGF-β is involved in the formation and maintenance of microtubes (MTs) in glioblastomas (GBMs), promoting enhanced MT formation and communication via Calcium signaling. TGF-β modulates gene expression by affecting transcriptional activation and mRNA turnover rate, potentially leading to changes in the proteome and cellular metabolism.
Dose IL-1β and transforming growth factor-beta (TGF-β) impact to influence early embryo development?5 answersIL-1β and TGF-β have been shown to impact early embryo development. IL-1β can modulate the growth of bovine embryos at early stages, with its effect depending on embryo density. It has been found to increase development to the blastocyst stage when embryos are cultured at high density, but may decrease or have no effect on development when cultured at low density. On the other hand, TGF-β1 has been shown to have an early action on bovine embryos, with exposure to exogenous TGF-β1 leading to an increase in the relative abundance of pluripotency and DNA methylation genes in the embryos. These findings suggest that both IL-1β and TGF-β can influence early embryo development, with IL-1β's effect depending on embryo density and TGF-β1's effect involving the modulation of gene expression.
What is the role of TGF-beta pathway in synovial degeneration in knee osteoarthritis?5 answersThe TGF-beta pathway plays a critical role in synovial degeneration in knee osteoarthritis (OA). Excessive activation of TGF-β leads to uncoupled bone remodeling, increased angiogenesis, and fibrosis in the subchondral bone, which contributes to the degeneration of articular cartilage. TGF-β also serves as a pro-inflammatory factor in the synovium, augmenting the secretion of inflammatory factors by immune cells. Inhibition of the TGF-β1 signaling cascade has been shown to improve the treatment of OA in the knee joint. Additionally, TGF-β/SMAD pathway has been found to play a critical role in articular chondrocytes, mesenchymal progenitor cells of subchondral bone, and synovial lining cells during OA development. The conflicting roles of TGF-β1 signaling in the development of OA are still under investigation.

See what other people are reading

Is greater TCR diversity in the tumor better?
5 answers
Greater TCR diversity in the tumor is crucial for protection against cancer development. Research shows that a broad TCR repertoire is necessary to reject cancers with natural antigenic heterogeneity, while even one type of TCR can protect against artificial cancers expressing specific antigens. Reduced TCR diversity increases the risk of developing primary tumors after chemical induction, with significantly higher mortality rates compared to mice with diverse TCR repertoires. TCR diversity profiling has become essential in cancer immunology, with diversity metrics aiding in biomarker development and patient response predictions to immunotherapy. Corrected diversity estimates from TCR repertoire profiling data have shown improved discrimination in cancer datasets and correlation with patient survival and treatment response, particularly in melanoma patients undergoing anti-PD-1 therapy.
Is vaccination an effective control against COVID?
4 answers
Vaccination has shown to be an effective control measure against COVID-19. Studies have demonstrated that vaccines can induce robust antibody and T cell responses, providing protection against the virus. Furthermore, research on recovered individuals and vaccinated cohorts indicates that immunity against SARS-CoV-2 can be long-lasting, with antibodies evolving to be more potent and broad over time. While some vaccines have faced controversies and questions over side effects, the overall consensus is that vaccination significantly reduces the risk of developing COVID-19. However, the protective effect of vaccines against long COVID remains debated in various studies. In conclusion, vaccination plays a crucial role in controlling the spread of COVID-19 by eliciting strong immune responses and reducing the severity of the disease.
Can cistus affect T cells?
4 answers
Cistus extracts have shown promising effects on T cells. Research indicates that certain phytochemical compounds in cistus extracts can target viral envelope proteins, preventing their interaction with host cell surface CD4 receptors. Additionally, cistus extracts have been found to have a cell-activating activity derived from natural sources, potentially affecting cellular functions like T cell activation. These findings suggest that cistus extracts may influence T cell responses, potentially impacting immune functions. Further studies are warranted to explore the specific mechanisms by which cistus extracts interact with T cells and their implications for immune modulation.
Are there some paper that includes the information about anti-CTLA4 antibody sequence?
5 answers
Yes, there are papers that provide information about anti-CTLA-4 antibody sequences. One paper discusses the development of anti-CTLA-4 nanobody-modified liposomes to enhance CD8+ T cell antitumor activity. Another paper presents anti-CTLA-4 antibody polypeptides and polynucleotides, along with their pharmaceutical compositions and potential uses. Additionally, there is a paper that introduces antibodies and antigen binding fragments against CTLA4, which can inhibit the binding of CTLA4 to its ligand, offering a potential treatment for diseases like cancer. These papers collectively contribute valuable insights into the development and potential therapeutic applications of anti-CTLA-4 antibodies.
How does TIM-3's modulation affect the efficacy of cancer immunotherapy in renal cell carcinoma (RCC)?
5 answers
Modulation of TIM-3 impacts cancer immunotherapy efficacy in renal cell carcinoma (RCC) by regulating immune responses. TIM-3, a negative regulator of immunity, interacts with dendritic cells (DCs) and T cells, influencing T cell activation and trogocytosis. In RCC, TIM-3 blockade disrupts trogocytosis of activated tumor-specific CD8+ T cells, enhancing antitumor immunity and reducing tumor burden. Additionally, TIM-3 and PD-1 blockades synergize to inhibit trogocytosis, promoting T cell survival and tumor control. These findings underscore TIM-3's crucial role in modulating immune responses in RCC and highlight its potential as a target for enhancing cancer immunotherapy efficacy in this context.
Is there any FDA approved small molecule for cancer immunetherapy targeting PDL-1?
5 answers
Yes, there are FDA-approved small molecules for cancer immunotherapy targeting PD-L1. While several therapeutic monoclonal antibodies against the PD-1/PD-L1 immune checkpoint have been successful in cancer treatment, small molecule inhibitors have emerged as alternatives. These small molecules offer advantages such as improved tissue penetration, low immunogenicity, and lower manufacturing costs compared to monoclonal antibodies. Recent studies have focused on screening natural compounds to identify potent PD-L1 inhibitors, with some compounds showing promising results in computational investigations and molecular dynamics simulations. The development of small-molecule inhibitors targeting PD-L1 is seen as a complementary approach to monoclonal antibodies in cancer immunotherapy, although further validation of their safety and efficacy in clinical settings is still needed.
What specific markers should be analyzed to assess immune cell activity in B2M knockout cultures in mice?
5 answers
To assess immune cell activity in B2M knockout cultures in mice, specific markers should be analyzed. These markers include MHC Class I molecules, immune subunits like LMP7, LMP2, and PA28, as well as regulators of proteasomes such as PA28 and PA700. Additionally, the expression of TDO2, a key enzyme in tryptophan metabolism, should be considered, especially in peritoneal macrophages where a reduced M1/M2 ratio was observed in TDO2 knockout mice. Furthermore, the deletion of β2-microglobulin (B2M) in human umbilical mesenchymal stem cells (UMSCs) can be assessed by examining the expression of HLA light chain β2-microglobulin (B2M) and its impact on immune rejection in mouse models. By analyzing these markers, researchers can gain insights into the immune response and activity in B2M knockout cultures in mice.
How does the expression of CD31/PECAM-1 and VE-Cadherin differ in B2M knockout mice compared to wild-type mice?
4 answers
In B2M knockout mice compared to wild-type mice, the expression of CD31/PECAM-1 and VE-Cadherin may exhibit significant differences. CD31/PECAM-1, an immunoglobulin superfamily member, plays a crucial role in lymphomas by facilitating survival and regulating extranodal spread of malignant cells. On the other hand, VE-Cadherin is essential for endothelial cell junction integrity and barrier function. The knockout of B2M could potentially impact the expression of these adhesion molecules, affecting vascular permeability and cellular responsiveness. Additionally, changes in gene expression variability in knockouts can influence phenotypic outcomes, potentially altering the expression levels of CD31/PECAM-1 and VE-Cadherin in B2M knockout mice. Further studies are needed to elucidate the specific alterations in CD31/PECAM-1 and VE-Cadherin expression in B2M knockout models.
How effective are autologous target cell assays in detecting rare cells from Lassa fever survivors?
5 answers
Autologous target cell assays have shown effectiveness in detecting rare cells from Lassa fever survivors. These assays have been instrumental in investigating Lassa virus-specific CD8+ T cell responses in survivors, revealing that despite a decline in circulating Lassa virus-specific CD8+ T cells over time, prolonged cellular immunity persists at low frequencies. Additionally, autologous target cell assays have been crucial in evaluating the duration of cellular immunity and protection in Lassa fever survivors, highlighting the importance of understanding immune responses post-infection. These assays provide valuable insights into the dynamics of immune responses in Lassa fever survivors, shedding light on the longevity and persistence of cellular immunity against the virus.
What is the correlation between the CD26 expression level and the prognosis of hepatocellular carcinoma patients?
5 answers
The expression level of CD26 has a significant impact on the prognosis of hepatocellular carcinoma (HCC) patients. Research indicates that CD26 expression is closely linked to cell-cycle regulation, apoptosis, and chemotherapy resistance in malignant pleural mesothelioma (MPM). Additionally, CD28 expression in HCC is correlated with the occurrence, development, and prognosis of the disease, interacting with CD4+ and CD8+ T-cells. Moreover, CD147 overexpression is associated with poor prognosis in liver hepatocellular carcinoma (LIHC), serving as a reliable biomarker for predicting prognosis. Furthermore, a prognostic signature based on tumor doubling time-related genes (TDTRGs) and immune-related genes (IRGs) can effectively classify HCC patients for prognosis prediction and individualized immunotherapies. These findings collectively highlight the crucial role of CD26 and related immune interactions in determining the prognosis of HCC patients.
What is the role of CD35 on monocytes?
5 answers
CD35, also known as complement receptor 1 (CR1), plays a crucial role in mediating various functions in monocytes. In the context of infectious diseases like HIV and Kawasaki disease, CD35 has been implicated in enhancing viral replication in HIV-infected monocytes when triggered by complement-opsonized particles. Additionally, in Kawasaki disease, CD35 expression on monocytes is associated with the cytotoxicity of these cells, particularly through granzyme B secretion. Furthermore, in the context of acute infectious diseases in children, the ratio of CD64 to CD35 on neutrophils has been shown to be significantly different between bacterial and viral infections, suggesting that CD35 expression levels can aid in the differential diagnosis of these conditions.