Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 346
Filtrar
1.
Small ; 20(3): e2303916, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37705134

RESUMEN

The induction of anti-drug antibody (ADA) is a formidable challenge for protein-based therapy. Trichosanthin (TCS) as a class of ribosome-inactivating proteins is widely studied in tumor treatment. However, the immunogenicity can induce the formation of ADA, which can cause hypersensitivity reactions and neutralize the efficacy of TCS, thus limiting its clinical application in cancer therapy. Here, a promising solution to this issue is presented by co-administration of the rapamycin nanoparticles and TCS. PEGylated rapamycin amphiphilic molecule is designed and synthesized as a prodrug and a delivery carrier, which can self-assemble into a nanoparticle system with encapsulation of free rapamycin, a hydrophobic drug. It is found that co-injection of the PEGylated rapamycin nanoparticles and TCS could mitigate the formation of anti-TCS antibody via inducing durable immunological tolerance. Importantly, the combination of TCS and the rapamycin nanoparticles has an enhanced effect on inhibit the growth of breast cancer. This work provides a promising approach for protein toxin-based anticancer therapy and for promoting the clinical translation.


Asunto(s)
Nanopartículas , Tricosantina , Humanos , Sirolimus/farmacología , Sirolimus/uso terapéutico , Formación de Anticuerpos , Tricosantina/farmacología , Tricosantina/uso terapéutico , Anticuerpos , Polietilenglicoles
2.
Int Immunopharmacol ; 125(Pt A): 111081, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37862724

RESUMEN

Obliterative bronchiolitis (OB) is one of the main complications affecting long-term survival of post-lung transplantation patients. In this study, we evaluated the efficacy of Tk-PQ (a peptide derived from trichosanthin) in alleviating OB in a mouse ectopic tracheal transplant model. We found that post-transplantation treatment of Tk-PQ significant ameliorated OB symptoms including luminal occlusion, epithelial cells loss and fibrosis in the allograft. In addition, Tk-PQ promoted immune suppressive environment by inducing Th2 polarization and increasing Treg population which in turn led to elevated levels of anti-inflammatory cytokines IL-4, IL-10, IL-33 and decreased levels of pro-inflammatory IL-1ß. Mechanistically, we used transcriptome analysis of splenic T cells from allografted mice to show that Tk-PQ treatment down-regulated the PI3K-Akt signaling pathway. Indeed, the immune suppression phenotypes of Tk-PQ was recapitulated by a PI3K inhibitor LY294002. Taken together, Tk-PQ regulates post-transplantation immuno-rejection by modulating the balance of T cell response via the PI3K-Akt pathway, making it a promising peptide based immune rejection suppressant for patients receiving allotransplant.


Asunto(s)
Bronquiolitis Obliterante , Tricosantina , Humanos , Ratones , Animales , Tricosantina/farmacología , Tricosantina/uso terapéutico , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Citocinas/metabolismo , Péptidos/farmacología , Péptidos/uso terapéutico , Inmunosupresores/farmacología
3.
Int J Biol Sci ; 19(6): 1698-1712, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37063415

RESUMEN

Alleviating immunosuppression of the tumor microenvironment is an important strategy to improve immune checkpoint therapy. It is an urgent but unmet need to develop adjuvant therapeutics for assisting the mainstay immunotherapies. Trichosanthin is an approved gynecology drug in China and its immunomodulatory effects have drawn much attention as an old drug for new applications in cancer. In this work, a recombinant cell-penetrating trichosanthin (rTCS-LMWP) was prepared via genetic fusion of a cell-penetrating peptide sequence (LMWP) to trichosanthin aiming to overcome the intratumoral penetration and intracellular delivery challenges. The potential of trichosanthin as an adjuvant therapy was explored, including its effects on tumor cells, antigen-presenting cells, tumor immune microenvironment, and the synergistic effect in combination with anti-PD-1. The results revealed that rTCS-LMWP can stimulate the maturation of dendritic cells via activating the STING-TBK1-IRF3 pathway, repolarize the protumor M2-type macrophages, and upregulate the pro-inflammatory cytokine expression. Moreover, rTCS-LMWP can enhance anti-PD-1 therapeutic efficacy in a CT26-bearing mouse model. The synergistic effect involved the induction of immunogenic cell death in the tumors, the proliferation and functionalization of cytotoxic T cells, and the suppression of the immunosuppressive regulatory T cells. These findings indicate that trichosanthin can be developed as an immunomodulator to facilitate cancer immunotherapy.


Asunto(s)
Neoplasias Colorrectales , Tricosantina , Animales , Ratones , Células Presentadoras de Antígenos , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Citocinas , Tricosantina/farmacología , Tricosantina/uso terapéutico , Microambiente Tumoral
4.
Int J Mol Sci ; 24(2)2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36674931

RESUMEN

Trichosanthin (TCS) is a type I ribosome-inactivating protein extracted from the tuberous root of the plant Trichosanthes. TCS shows promising potential in clinical drug abortion, anti-tumor and immunological regulation. However, the molecular mechanisms of its anti-tumor and immune regulation properties are still not well discovered. In the present study, we investigated the anti-tumor activity of TCS in hepatocellular carcinoma (HCC), both in vitro and in vivo. Both HCC cell lines and xenograft tumor tissues showed considerable growth inhibition after they were treated with TCS. TCS provoked caspase-mediated apoptosis in HCC cells and xenograft tumor tissues. The recruitment of CD8+ T cells to HCC tissues and the expression of chemokines, CCL2 and CCL22, were promoted upon TCS treatment. In addition, TCS induced an upregulation of Granzyme B (GrzB), TNF-α and IFN-γ in HCC tissues, which are the major cytotoxic mediators produced by T cells. Furthermore, TCS also resulted in an increase of mannose-6-phosphate receptor (M6PR), the major receptor of GrzB, in HCC tissues. In summary, these results suggest that TCS perhaps increases T-cell immunity via promoting the secretion of chemokines and accelerating the entry of GrzB to HCC cells, which highlights the potential role of TCS in anti-tumor immunotherapy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Tricosantina , Humanos , Tricosantina/farmacología , Tricosantina/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Linfocitos T CD8-positivos/metabolismo , Granzimas , Neoplasias Hepáticas/tratamiento farmacológico , Quimiocinas/farmacología
5.
Int J Biol Macromol ; 223(Pt A): 1485-1494, 2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36395942

RESUMEN

The development of cancer vaccines based on tumor-associated antigens is hurdled by lack of an efficient adjuvant and insufficient efficacy. To improve the efficacy of vaccines, a genetically-engineered method was employed in this work to achieve the codelivery of antigen and adjuvant to enhance immune responses. Trichosanthin is a plant-derived protein that possesses cancer immune stimulation function. A genetically engineered protein vaccine composed of trichosanthin (adjuvant) and legumain domain (a peptidic antigen) was constructed, which was further chemically modified with mannose for targeting dendritic cells (DCs). The method is facile and ready for scaling up for massive production. Such a "two-in-one" vaccine is advantageous for codelivery for augmenting the immune responses. The vaccine inhibited the tumors by triggering a robust cytotoxic T lymphocyte response in the orthotopic-breast-tumor mice. Furthermore, the vaccine was loaded into the temperature-sensitive hydrogel based on Pluronic F127 for implanting use in the post-surgical site. The sustained-released vaccine from the hydrogel inhibited not only the tumor recurrence but also the lung metastases of breast cancer. These findings demonstrated that it was a safe and effective vaccination for breast cancer immunotherapy in a prophylactical and therapeutical manner for remodeling the tumor immune microenvironment and arresting tumor growth.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Tricosantina , Ratones , Animales , Hidrogeles/farmacología , Células Dendríticas , Tricosantina/farmacología , Adyuvantes Inmunológicos/farmacología , Microambiente Tumoral
6.
Protein Expr Purif ; 200: 106147, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35917982

RESUMEN

Trichosanthin (TCS), as a type 1 ribosome-inactivating protein, has a very high cytoplasmic activity in vitro and can quickly kill cancer cells. However, it is easily filtered and cleared by the kidney, which results in the short half-life and severely limits its application. In this study, we constructed several recombinant proteins by fusing the albumin binding domain mutant ABD035(abbreviated as ABD) to the N- or C-terminus of TCS to endow the recombinant TCS fusion protein with a longer half-life property binding with endogenous human serum albumin (HSA) via ABD to effectively exert its anti-tumor activity in vivo. Pull down, Dynamic light scattering and ELISA assays all showed that TCS fused with two ABD sequences at the C-terminus of TCS, has stronger binding capacity to HSA in vitro than TCS with one ABD. In vivo studies in BALB/C mice were performed and the elimination half-life of TCS-ABD-ABD is about 15-fold longer compared to TCS and anti-tumor activity is about 30% higher than that of TCS alone in BALB/C mouse experiments. Moreover, we found that TCS with two ABDs in tandem have the highest soluble expression level, more than 5 times higher than that of TCS, and the yield of purified protein of TCS-ABD-ABD was as high as 68.9 mg/L culture solution, which was about 7-fold higher than that of TCS. Furthermore, MTT assay showed that the anti-tumor activity of TCS-ABD-ABD was significantly higher than TCS fused with only one ABD sequence, indicating that the repeated ABD sequences facilitated the biological activity of TCS. In this paper, the fusion of the albumin-binding domain in tandem with TCS can effectively improve its stability in vivo and also significantly increase its soluble expression, expanding the application of the albumin-binding domain in the high soluble expression and stability of protein drugs.


Asunto(s)
Neoplasias , Tricosantina , Albúminas , Animales , Humanos , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Saporinas , Albúmina Sérica Humana , Tricosantina/genética , Tricosantina/farmacología
7.
Molecules ; 27(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35268705

RESUMEN

Trichosanthin (TCS) is a traditional Chinese herbal medicine used to treat some gynecological diseases. Its effective component has diverse biological functions, including antineoplastic activity. The human trophoblast cell line BeWo was chosen as an experimental model for in vitro testing of a drug screen for anticancer properties of TCS. The MTT method was used in this study to get a primary screen result. The result showed that 100 mM had the best IC50 value. Proteomics analysis was then performed for further investigation of the drug effect of TCS on the BeWo cell line. In this differential proteomic expression analysis, the total proteins extracted from the BeWo cell line and their protein expression level after the drug treatment were compared by 2DE. Then, 24 unique three-fold differentially expressed proteins (DEPs) were successfully identified by MALDI-TOF/TOF MS. Label-free proteomics was run as a complemental method for the same experimental procedure. There are two proteins that were identified in both the 2DE and label-free methods. Among those identified proteins, bioinformatics analysis showed the importance of pathway and signal transduction and gives us the potential possibility for the disease treatment hypothesis.


Asunto(s)
Antineoplásicos , Tricosantina , Antineoplásicos/farmacología , Línea Celular , Humanos , Proteínas , Proteómica/métodos , Tricosantina/farmacología
8.
Toxins (Basel) ; 14(3)2022 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-35324675

RESUMEN

Tian Hua Fen, a herbal powder extract that contains trichosanthin (TCS), was used as an abortifacient in traditional Chinese medicine. In 1972, TCS was purified to alleviate the side effects. Because of its clinical applications, TCS became one of the most active research areas in the 1960s to the 1980s in China. These include obtaining the sequence information in the 1980s and the crystal structure in 1995. The replication block of TCS on human immunodeficiency virus in lymphocytes and macrophages was found in 1989 and started a new chapter of its development. Clinical studies were subsequently conducted. TCS was also found to have the potential for gastric and colorectal cancer treatment. Studies on its mechanism showed TCS acts as an rRNA N-glycosylase (EC 3.2.2.22) by hydrolyzing and depurinating A-4324 in α-sarcin/ricin loop on 28S rRNA of rat ribosome. Its interaction with acidic ribosomal stalk proteins was revealed in 2007, and its trafficking in mammalian cells was elucidated in the 2000s. The adverse drug reactions, such as inducing immune responses, short plasma half-life, and non-specificity, somehow became the obstacles to its usage. Immunotoxins, sequence modification, or coupling with polyethylene glycerol and dextran were developed to improve the pharmacological properties. TCS has nicely shown the scientific basis of traditional Chinese medicine and how its research and development have expanded the knowledge and applications of ribosome-inactivating proteins.


Asunto(s)
Tricosantina , Animales , Mamíferos , Ratas , Investigación , Proteínas Ribosómicas/química , Ribosomas , Saporinas , Tricosantina/química , Tricosantina/farmacología
9.
J Integr Med ; 19(6): 515-525, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34538767

RESUMEN

OBJECTIVE: Plant-derived cytotoxic transgene expression, such as trichosanthin (tcs), regulated by recombinant adeno-associated virus (rAAV) vector is a promising cancer gene therapy. However, the cytotoxic transgene can hamper the vector production in the rAAV producer cell line, human embryonic kidney (HEK293) cells. Here, we explored microRNA-122 (miR122) and its target sequence to limit the expression of the cytotoxic gene in the rAAV producer cells. METHODS: A miR122 target (122T) sequence was incorporated into the 3' untranslated region of the tcs cDNA sequence. The firefly luciferase (fluc) transgene was used as an appropriate control. Cell line HEK293-mir122 was generated by the lentiviral vector-mediated genome integration of the mir122 gene in parental HEK293 cells. The effects of miR122 overexpression on cell growth, transgene expression, and rAAV production were determined. RESULTS: The presence of 122T sequence significantly reduced transgene expression in the miR122-enriched Huh7 cell line (in vitro), fresh human hepatocytes (ex vivo), and mouse liver (in vivo). Also, the normal liver physiology was unaffected by delivery of 122T sequence by rAAV vectors. Compared with the parental cells, the miR122-overexpressing HEK293-mir122 cell line showed similar cell growth rate and expression of transgene without 122T, as well as the ability to produce liver-targeting rAAV vectors. Fascinatingly, the yield of rAAV vectors carrying the tcs-122T gene was increased by 77.7-fold in HEK293-mir122 cells. Moreover, the tcs-122T-containing rAAV vectors significantly reduced the proliferation of hepatocellular carcinoma cells without affecting the normal liver cells. CONCLUSION: HEK293-mir122 cells along with the 122T sequence provide a potential tool to attenuate the cytotoxic transgene expression, such as tcs, during rAAV vector production.


Asunto(s)
MicroARNs , Tricosantina , Animales , Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Células HEK293 , Humanos , Ratones , MicroARNs/genética
10.
Bioengineered ; 12(1): 2779-2790, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34233587

RESUMEN

Based on many studies, trichosanthin (TCS) has an antiviral effect that regulates immune response, and targets cancer cells to exert broad-spectrum anti-tumor pharmacological activities. It is speculated that TCS may be a potential natural active drug for preventing as well as treating cervical cancer. But the clearer impact along with underlying TCS mechanism on cervical cancer are still unclear. The purpose of this study is to investigate the function and potential mechanism of TCS in cervical cancer. We measured the viability of cervical cancer cell lines (HeLa & caski cells) using CCK-8 analysis, detected cell proliferation efficiency through Ki-67 staining, analyzed cell apoptosis rate via flow cytometry as well as annexin V-FITC/PI double staining, performed apoptosis-related protein expression through western blotting, evaluated cell migration along with invasion by wound as well as transwell assays, carried out MMP via JC-1 and Rh123 fluorescent probes, as well as detected intracellular ATP and ROS levels by flow cytometry, respectively, to evaluate the effects of TCS. We found that TCS inhibited viability along with proliferation, induced apoptosis, as well as inhibited HeLa & caski cell migration along with invasion in a time- and dose-dependent manner. Additionally, TCS also reduced MMP, and the production of adenosine triphosphate, as well as induced the increase of intracellular reactive oxygen species in cancer cell lines. In accordance with the present studies, TCS inhibits HeLa & caski cell proliferation along with migration but promotes their apoptosis, which may be mediated by regulating oxidative stress.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Tricosantina/farmacología , Neoplasias del Cuello Uterino/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Células HeLa , Humanos
11.
FEBS Lett ; 595(17): 2221-2236, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34328639

RESUMEN

The ribosome is subjected to post-translational modifications, including phosphorylation, that affect its biological activity. Among ribosomal elements, the P-proteins undergo phosphorylation within the C terminus, the element which interacts with trGTPases or ribosome-inactivating proteins (RIPs); however, the role of phosphorylation has never been elucidated. Here, we probed the function of phosphorylation on the interaction of P-proteins with RIPs using the ribosomal P1-P2 dimer. We determined the kinetic parameters of the interaction with the toxins using biolayer interferometry and microscale thermophoresis. The results present the first mechanistic insight into the function of P-protein phosphorylation, showing that introduction of a negative charge into the C terminus of P1-P2 proteins promotes α-helix formation and decreases the affinity of the P-proteins for the RIPs.


Asunto(s)
Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas Ribosómicas/química , Proteínas Ribosómicas/metabolismo , Toxinas Biológicas/metabolismo , Ácido Glutámico/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Fosfoproteínas/genética , Fosforilación , Dominios Proteicos , Proteínas Ribosómicas/genética , Ricina/química , Ricina/metabolismo , Serina/metabolismo , Toxinas Biológicas/química , Tricosantina/química , Tricosantina/metabolismo
12.
Mol Biol Rep ; 48(4): 3827-3840, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33895972

RESUMEN

Ribosome inactivating proteins (RIPs) as family of anti-cancer drugs recently received much attention due to their interesting anti-cancer mechanism. In spite of small drugs, RIPs use the large-size effect (LSE) to prevent the efflux process governed by drug resistance transporters (DRTs) which prevents inside of the cells against drug transfection. There are many clinical translation obstacles that severely restrict their applications especially their delivery approach to the tumor cells. As the main goal of this review, we will focus on trichosanthin (TCS) and gelonin (Gel) and other types, especially scorpion venom-derived RIPs to clarify that they are struggling with what types of bio-barriers and these challenges could be solved in cancer therapy science. Then, we will try to highlight recent state-of-the-arts in delivery of RIPs for cancer therapy.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteínas Inactivadoras de Ribosomas Tipo 1/toxicidad , Tricosantina/toxicidad , Animales , Antineoplásicos Fitogénicos/toxicidad , Humanos , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Venenos de Escorpión/uso terapéutico , Venenos de Escorpión/toxicidad , Bloqueadores de los Canales de Sodio/uso terapéutico , Bloqueadores de los Canales de Sodio/toxicidad , Tricosantina/uso terapéutico
13.
BMC Complement Med Ther ; 21(1): 88, 2021 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-33750370

RESUMEN

BACKGROUND: Tongue squamous cell carcinoma (TSCC) is a common type of oral cancer, with a relatively poor prognosis and low post-treatment survival rate. Various strategies and novel drugs to treat TSCC are emerging and under investigation. Trichosanthin (TCS), extracted from the root tubers of Tian-Hua-Fen, has been found to have multiple biological and pharmacological functions, including inhibiting the growth of cancer cells. Granzyme B (GrzB) is a common toxic protein secreted by natural killer cells and cytotoxic T cells. Our group has reported that TCS combined with GrzB might be a superior approach to inhibit liver tumor progression, but data relating to the use of this combination to treat TSCC remain limited. The aim of this study was to examine the effectiveness of TCS on TSCC processes and underlying mechanisms. METHODS: First, we screened the potential antitumor activity of TCS using two types of SCC cell lines. Subsequently, a subcutaneous squamous cell carcinoma xenograft model in nude mice was established. These model mice were randomly divided into four groups and treated as follows: control group, TCS treatment group, GrzB treatment group, and TCS/GrzB combination treatment group. Various tumorigenesis parameters, such as Ki67, PCNA, caspase-3, Bcl-2 and VEGFA, et al., were performed to determine the effects of these treatments on tumor development. RESULTS: Screening confirmed that the SCC25 line exhibited greater sensitivity than the SCC15 line to TCS in vitro studies. TCS or GrzB treatment significantly inhibited tumor growth compared with the inhibition seen in the control group. The TCS/GrzB combination inhibited tumor growth more than either drug alone. TCS treatment inhibited tumor proliferation by downregulating Ki67 and Bcl2 protein expression while accelerating tumor apoptosis. In the TCS/GrzB-treated group, expression of Ki67 was further downregulated, while the level of activated caspase-3 was increased, compared with their expression in either of the single drug treatment groups. CONCLUSION: These results suggest that the TCS/GrzB combination could represent an effective immunotherapy for TSCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Granzimas/uso terapéutico , Neoplasias de la Lengua/tratamiento farmacológico , Tricosantina/uso terapéutico , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Journal of Integrative Medicine ; (12): 515-525, 2021.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-922523

RESUMEN

OBJECTIVE@#Plant-derived cytotoxic transgene expression, such as trichosanthin (tcs), regulated by recombinant adeno-associated virus (rAAV) vector is a promising cancer gene therapy. However, the cytotoxic transgene can hamper the vector production in the rAAV producer cell line, human embryonic kidney (HEK293) cells. Here, we explored microRNA-122 (miR122) and its target sequence to limit the expression of the cytotoxic gene in the rAAV producer cells.@*METHODS@#A miR122 target (122T) sequence was incorporated into the 3' untranslated region of the tcs cDNA sequence. The firefly luciferase (fluc) transgene was used as an appropriate control. Cell line HEK293-mir122 was generated by the lentiviral vector-mediated genome integration of the mir122 gene in parental HEK293 cells. The effects of miR122 overexpression on cell growth, transgene expression, and rAAV production were determined.@*RESULTS@#The presence of 122T sequence significantly reduced transgene expression in the miR122-enriched Huh7 cell line (in vitro), fresh human hepatocytes (ex vivo), and mouse liver (in vivo). Also, the normal liver physiology was unaffected by delivery of 122T sequence by rAAV vectors. Compared with the parental cells, the miR122-overexpressing HEK293-mir122 cell line showed similar cell growth rate and expression of transgene without 122T, as well as the ability to produce liver-targeting rAAV vectors. Fascinatingly, the yield of rAAV vectors carrying the tcs-122T gene was increased by 77.7-fold in HEK293-mir122 cells. Moreover, the tcs-122T-containing rAAV vectors significantly reduced the proliferation of hepatocellular carcinoma cells without affecting the normal liver cells.@*CONCLUSION@#HEK293-mir122 cells along with the 122T sequence provide a potential tool to attenuate the cytotoxic transgene expression, such as tcs, during rAAV vector production.


Asunto(s)
Animales , Humanos , Ratones , Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Células HEK293 , MicroARNs/genética , Tricosantina
16.
J Biol Chem ; 295(46): 15588-15596, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32878986

RESUMEN

The principal virulence factor of human pathogenic enterohemorrhagic Escherichia coli is Shiga toxin (Stx). Shiga toxin 2a (Stx2a) is the subtype most commonly associated with severe disease outcomes such as hemorrhagic colitis and hemolytic uremic syndrome. The catalytic A1 subunit (Stx2A1) binds to the conserved elongation factor binding C-terminal domain (CTD) of ribosomal P stalk proteins to inhibit translation. Stx2a holotoxin also binds to the CTD of P stalk proteins because the ribosome-binding site is exposed. We show here that Stx2a binds to an 11-mer peptide (P11) mimicking the CTD of P stalk proteins with low micromolar affinity. We cocrystallized Stx2a with P11 and defined their interactions by X-ray crystallography. We found that the last six residues of P11 inserted into a shallow pocket on Stx2A1 and interacted with Arg-172, Arg-176, and Arg-179, which were previously shown to be critical for binding of Stx2A1 to the ribosome. Stx2a formed a distinct P11-binding mode within a different surface pocket relative to ricin toxin A subunit and trichosanthin, suggesting different ribosome recognition mechanisms for each ribosome inactivating protein (RIP). The binding mode of Stx2a to P11 is also conserved among the different Stx subtypes. Furthermore, the P stalk protein CTD is flexible and adopts distinct orientations and interaction modes depending on the structural differences between the RIPs. Structural characterization of the Stx2a-ribosome complex is important for understanding the role of the stalk in toxin recruitment to the sarcin/ricin loop and may provide a new target for inhibitor discovery.


Asunto(s)
Péptidos/metabolismo , Proteínas Ribosómicas/química , Toxina Shiga II/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Humanos , Simulación de Dinámica Molecular , Péptidos/química , Unión Proteica , Proteínas Ribosómicas/metabolismo , Ribosomas/metabolismo , Ricina/química , Ricina/metabolismo , Toxina Shiga II/química , Tricosantina/química , Tricosantina/metabolismo
17.
J Drug Target ; 28(9): 970-981, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32363946

RESUMEN

Targeted delivery of antitumor drugs is especially important for tumour therapy. Tumour targeting peptides have been shown to be very effective drug carriers for tumour therapy. Interleukin-4 receptor (IL-4R) is overexpressed on the surface of various human solid tumours. To obtain a better targeting peptide, we first designed a novel targeting peptide derived from interleukin-4 (IL-4), ILBP-b. ILBP-b contains the key high-affinity binding residue E9 of IL-4 to IL-4R. Compared with a reported targeting peptide ILBP-a (containing another key high affinity residue R88), ILBP-b was proved to be a better targeting peptide by the fluorescence experiments. Then, we further fused ILBP-b and ILBP-a to increase the multisite-binding ability of ILBP-b and got a better targeting peptide ILBP-ba. ILBP-ba showed a stronger preferential binding ability to IL-4R high-expressing cells than ILBP-a and ILBP-b. Competitive binding experiments demonstrated ILBP-ba specifically targets IL-4R. By fusing ILBP-ba with drug protein trichosanthin (TCS), in vitro drug carrying experiments showed that ILBP-ba could specifically enhance the killing effect of TCS on IL-4R high-expressing tumour cells (more than 10 folds). These results indicated that ILBP-ba has great potential for drug delivery applications targeting IL-4R and will be beneficial for the development of tumour therapeutic agents.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Péptidos/administración & dosificación , Unión Proteica/fisiología , Receptores de Interleucina-4/efectos de los fármacos , Tricosantina/farmacología , Antineoplásicos Fitogénicos/administración & dosificación , Línea Celular Tumoral , Portadores de Fármacos/administración & dosificación , Sistemas de Liberación de Medicamentos , Humanos , Interleucinas/química , Péptidos/química , Plásmidos , Tricosantina/administración & dosificación
18.
J Mol Graph Model ; 98: 107619, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32311663

RESUMEN

Alpha-momorcharin (α-MMC), trichosanthin (TCS), and momordica anti-HIV protein of 30 kD (MAP30) are potential anti-tumor drug candidates but have cytotoxicity to normal cells. The binding of these proteins to LRP1 receptor and the subsequent endocytosis are essential to their cytotoxicity, but this binding process remains largely unknown. This study, in-silico analysis of the binding patterns, was conducted via the protein-protein docking software, ZDOCK 3.0.2 package, to better understand the binding process. Specifically, α-MMC, TCS and MAP30 were selected and bound to binding subunits CR56 and CR17 of LRP1. After docking, the 10 best docking solutions are retained based on the default ZDOCK scores and used for structural assessment. Our results showed that, α-MMC bound to LRP1 stably at the amino acid residues 1-20, at which 8 residues formed 21 hydrogen bonds with 15 residues of CR56 and 10 residues formed 15 hydrogen bonds with 12 residues of CR17. In contrast, TCS and MAP30 bound mainly to LRP1 at the residues 1-57/79-150 and residues 58-102, respectively, which were functional domains of TCS and MAP30. Since residues 1-20 are outside the functional domain of α-MMC, α-MMC is considered more suitable to attenuate by mutating the receptor binding site. Thus, our analysis lays the foundation for future genetic engineering work on α-MMC, and makes important contributions to its potential clinical use in cancer treatment.


Asunto(s)
Momordica , Tricosantina , Línea Celular Tumoral , Ligandos , Proteínas Inactivadoras de Ribosomas
19.
Curr Protein Pept Sci ; 20(3): 285-295, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29932034

RESUMEN

Traditional Chinese Medicine (TCM) has been practiced in China for thousands of years. As a complementary and alternative treatment, herbal medicines that are frequently used in the TCM are the most accepted in the Western world. However, animal materials, which are equally important in the TCM practice, are not well-known in other countries. On the other hand, the Chinese doctors had documented the toxic profiles of hundreds of animals and plants thousand years ago. Furthermore, they saw the potential benefits of these materials and used their toxic properties to treat a wide variety of diseases, such as heavy pain and cancer. Since the 50s of the last century, efforts of the Chinese government and societies to modernize TCM have achieved tremendous scientific results in both laboratory and clinic. A number of toxic proteins have been isolated and their functions identified. Although most of the literature was written in Chinese, this review provide a summary, in English, regarding our knowledge of the clinical use of the toxic proteins isolated from a plant, Tian Hua Fen, and an animal, scorpion, both of which are famous toxic prescriptions in TCM.


Asunto(s)
Péptidos , Proteínas , Venenos de Escorpión/química , Tricosantina , Animales , Humanos , Medicina Tradicional China , Péptidos/química , Péptidos/farmacología , Péptidos/uso terapéutico , Plantas Medicinales , Proteínas/química , Proteínas/farmacología , Proteínas/uso terapéutico , Tricosantina/química , Tricosantina/farmacología , Tricosantina/uso terapéutico
20.
Pathol Res Pract ; 215(4): 632-638, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30567634

RESUMEN

BACKGROUND: Previous studies have indicated that Trichosanthin (TCS) exerts anti-virus, immunoregulation and a broad spectrum anti-tumor pharmacological activities. Trichosanthin is a promising agent for the treatment of cervical cancer. However, the exact effects and potential mechanism of TCS on cervical cancer are not well known. METHOD: The cell viability of TCS on cervical cancer cell lines (HeLa and caski cells) were detected by a Cell Counting Kit-8 (CCK-8) assay. Cell proliferation was measured by Ki-67 staining and cell apoptosis was detected by flow cytometry. Cell migration and invasion were detected by wound assay and transwell assay, respectively. The levels of E-cadherin, N-cadherin, Snail, Bcl-2, Caspase-3, p-STAT5, STAT5, p-C-myc, C-myc were detected by western blot. RESULTS: The present study showed that TCS inhibited the proliferation of HeLa and caski cells and reduced Ki-67 and P-C-myc expression. In addition, flow cytometric analysis showed that TCS induced the apoptosis of HeLa and caski cells. The potent effect of TCS on cell apoptosis as determined by the increase the levels of caspase-3 and decrease the levels of Bcl-2. TCS also inhibited cervical cancer cell invasion, migration and epithelial-mesenchymal transition (EMT). Furthermore, TCS treatment markedly inhibited the activation of STAT5/C-myc signaling pathway. CONCLUSION: In conclusion, the present study suggest that TCS inhibits the proliferation, migration and EMT of human cervical cancer cells, which maybe mediated by inhibiting the activation of STAT5/C-myc signaling pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/efectos de los fármacos , Tricosantina/farmacología , Neoplasias del Cuello Uterino/metabolismo , Apoptosis/efectos de los fármacos , Cadherinas/metabolismo , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Células HeLa , Humanos , Neoplasias del Cuello Uterino/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...