Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
2.
Fish Shellfish Immunol ; 103: 23-31, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32348884

ABSTRACT

Thymosin ß4 is a multifunctional protein in vertebrates that participates in physiological processes, such as wound healing, immune response, cell proliferation and migration. We assessed the multifarious roles of this small peptide in Pinctada fucata, an oyster commonly used in pearl culture in China. Our results showed that when P. fucata was challenged by bacterial pathogens or LPS, the relative expression level of Pfthymosin ß4 mRNA was significantly up-regulated, suggesting its involvement in immune response of the animal. Recombinant Pfthymosin ß4 (rPfthymosin ß4) was produced and showed in vitro different antibacterial activities against different pathogenic bacteria; the inhibitory effect of rPfthymosin ß4 on bacterial growth was relatively stronger in the broth culture than agar culture. The overexpression of Pfthymosin ß4 in Escherichia coli BL21(DE3) cells could improve their resistance to Cu2+, Zn2+, Cd2+, and H2O2, suggesting that Pfthymosin ß4 is likely involved with antioxidant. rPfthymosin ß4 also significantly promoted the proliferation and migration of mouse aortic vascular smooth muscle cells as indicated by MTT assay and cell scratch assay, respectively. In addition, chemically synthesized or recombinant Pfthymosin ß4 could transiently increase the circulating total hemocytes counts but down-regulated by RNAi in P. fucata. Taking together above results and previous studies suggested that Pfthymosin ß4 is potentially able to promote wound healing through enhancing antibacterial activity and antioxidant capacity, promotion of cell proliferation and migration, and increase of circulating hemocytes in P. fucata due to nucleus implantation injury. Thus, the future of recombinant Pfthymosin ß4 should be promising in the culture of pearls in P. fucata.


Subject(s)
Fish Diseases/immunology , Pinctada/immunology , Thymosin/immunology , Animals , Aquaculture , Lipopolysaccharides/pharmacology , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Streptococcus agalactiae/physiology , Vibrio/physiology , Vibrio Infections/immunology , Vibrio Infections/veterinary , Vibrio alginolyticus/physiology
3.
Curr Med Chem ; 27(29): 4840-4854, 2020.
Article in English | MEDLINE | ID: mdl-31389310

ABSTRACT

Prothymosin alpha (ProTα) is a highly acidic polypeptide, ubiquitously expressed in almost all mammalian cells and tissues and consisting of 109 amino acids in humans. ProTα is known to act both, intracellularly, as an anti-apoptotic and proliferation mediator, and extracellularly, as a biologic response modifier mediating immune responses similar to molecules termed as "alarmins". Antibodies and immunochemical techniques for ProTα have played a leading role in the investigation of the biological role of ProTα, several aspects of which still remain unknown and contributed to unraveling the diagnostic and therapeutic potential of the polypeptide. This review deals with the so far reported antibodies along with the related immunodetection methodology for ProTα (immunoassays as well as immunohistochemical, immunocytological, immunoblotting, and immunoprecipitation techniques) and its application to biological samples of interest (tissue extracts and sections, cells, cell lysates and cell culture supernatants, body fluids), in health and disease states. In this context, literature information is critically discussed, and some concluding remarks are presented.


Subject(s)
Antibodies , Protein Precursors , Thymosin/analogs & derivatives , Alarmins , Animals , Humans , Protein Precursors/immunology , Protein Precursors/physiology , Thymosin/immunology , Thymosin/physiology
4.
Article in English | MEDLINE | ID: mdl-31647987

ABSTRACT

ß-thymosin family comprise a series of heat-stable multifunctional polypeptides involved in actin regulation, anti-inflammation, wound healing, cell migration, angiogenesis, cardiac protection, antimicrobial processes and antiviral immunity. The roles of Tß12 (thymosin-ß12) in marine invertebrates is still largely unknown, especially in terms of antibacterial immunity. In this study, we cloned the Tß12 gene with an ORF of 126 bp coding 41 amino acids from Urechis unicinctus. Tissue distribution analysis by qRT-PCR used TBP as reference gene showed that Tß12 was widely expressed in all tissues, and the transcript levels were the highest in the body wall, followed by the coelomic fluid, and the lowest in the intestines and anal sacs. After LPS (lipopolysaccharides) injection, Tß12 expression in the body was first elevated significantly at 3 h (p < 0.05), indicating that the body wall was the first defense line of the innate immune system; in the coelomic fluid, the Tß12 mRNA levels increased after LPS injection, with a significant increase occurring at 6 h, showing that coelomic fluid functioned as the second defense line of the innate immune system. In the midgut and anal sacs, a significant increase in the Tß12 level occurred at 24 h, suggesting that the midgut and anal sacs may act as accessory organs for the innate immune system. Moreover, U. unicinctus Tß12 recombinants can effectively inhibit the growth of both gram-negative and gram-positive bacteria. These results indicate that U. unicinctus Tß12 plays important roles in innate antibacterial immune responses, which can deepen our understanding of Tß12 in marine invertebrates.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate , Polychaeta/immunology , Thymosin/immunology , Animals , Organ Specificity/genetics , Organ Specificity/immunology , Polychaeta/genetics , Thymosin/genetics
5.
Fish Shellfish Immunol ; 87: 371-378, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30703548

ABSTRACT

Thymosin hormones, which were shown to be involved in immune system development and differentiation in previous studies, have antimicrobial functions in different animals. Zebrafish are a useful model for immunology research. Although thymosin has been reported to be involved in the embryonic development of zebrafish, it is necessary to uncover the antimicrobial function of thymosin in zebrafish. In this study, we expressed thymosin ß (Tß) in zebrafish in vitro and studied its antimicrobial function. The Tß protein consists of 45 amino acids and is conserved among its family members, especially the actin-binding motif (LKKTET). Tß was expressed in all tested tissues and was highly expressed in the brain, liver and hindgut. After Aeromonas hydrophila challenge, the Tß transcript level increased in the skin, liver, kidney, spleen, thymus, foregut, gills and midgut. Purified recombinant thymosin ß (rTß) protein was used to study the antimicrobial mechanism. rTß could inhibit the growth of Staphylococcus aureus, Aeromonas hydrophila, Vibrio anguillarum, Pseudomonas aeruginosa and Klebsiella pneumoniae. rTß also binds to and agglutinates certain bacteria. Further study showed that rTß could combine with the polysaccharides from gram-negative and gram-positive bacterial walls. All results suggested that the Tß of zebrafish plays a significant role in innate antibacterial immune responses.


Subject(s)
Fish Proteins/immunology , Immunity, Innate/physiology , Thymosin/immunology , Zebrafish/immunology , Aeromonas hydrophila/physiology , Animals , Fish Diseases/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary
6.
Fish Shellfish Immunol ; 86: 516-524, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30468890

ABSTRACT

The ß-thymosin (Tß) proteins participate in numerous biological processes, such as cell proliferation and differentiation, anti-inflammatory and antimicrobial mechanism. To date, Tß proteins have been well studied in vertebrates, especially mammals. While limited Tß or Tß-like proteins have been reported in invertebrates. Moreover, rare information of Tß or Tß-like proteins is available in scallop species yet. In the present study, two Tß homologues, AiTß and CfTß, were identified and characterized from two scallop species bay scallop Argopecten irradians and Zhikong scallop Chlamys farreri. They were both 41 amino acid peptide and contained one THY domain, a highly conserved actin-binding motif and two conserved helix forming regions. Tissue distribution and expression profiles of their mRNA transcripts were roughly similar yet different in detail, while their recombinant proteins exhibited different immunomodulation activity on the downstream immune parameters. These results collectively indicated that the function of Tß family in scallop were functionally differentiated.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/genetics , Pectinidae/genetics , Pectinidae/immunology , Thymosin/genetics , Thymosin/immunology , Amino Acid Sequence , Animals , Base Sequence , Gene Expression Profiling , Phylogeny , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Thymosin/chemistry
7.
Fish Shellfish Immunol ; 84: 244-251, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30292805

ABSTRACT

Thymosins ß are actin-binding proteins that play a variety of different functions in inflammatory responses, wound healing, cell migration, angiogenesis, and stem cell recruitment and differentiation. In crayfish, thymosins participate in antiviral immunology. However, the roles of thymosin during bacterial infection in shrimp remain unclear. In the present study, four thymosins were identified from kuruma shrimp, Marsupenaeus japonicus, and named as Mjthymosin2, Mjthymosin3, Mjthymosin4, and Mjthymosin5 according the number of their thymosin beta actin-binding motifs. Mjthymosin3 was selected for further study because its expression level was the highest in hemocytes. Expression analysis showed that Mjthymosin3 was upregulated in hemocytes after challenged by Vibrio anguillarum or Staphylococcus aureus. The recombinant Mjthymosin3 protein could inhibit the growth of certain bacteria in an in vitro antibacterial test. Mjthymosins could facilitate external bacterial clearance in shrimp, and were beneficial to shrimp survival post V. anguillarum or S. aureus infection. The results suggested that Mjthymosins played important roles in the antibacterial immune response of kuruma shrimp.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/genetics , Penaeidae/genetics , Penaeidae/immunology , Thymosin/genetics , Thymosin/immunology , Amino Acid Sequence , Animals , Arthropod Proteins/chemistry , Arthropod Proteins/genetics , Arthropod Proteins/immunology , Gene Expression Profiling , Phylogeny , Sequence Alignment , Staphylococcus aureus/physiology , Thymosin/chemistry , Vibrio/physiology
8.
Fish Shellfish Immunol ; 69: 90-98, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28803959

ABSTRACT

Thymosin beta belongs to the thymosin family, which consists of a series of highly conserved peptides involved in various biological processes. In teleosts, understanding of the immunological functions of thymosin beta is limited, particularly in vivo, which is essentially unknown. In the current study, we cloned and identified thymosin beta 4 from the teleost fish Golden pompano (Trachinotus ovatus), which we have named TroTß4. We investigated the expression patterns and functions of TroTß4 in both in vivo and in vitro assays. TroTß4 is composed of 44 amino acids and shares high sequence identities with known thymosin ß4 species in other teleosts, which contains a highly conserved actin-binding motif (LKKTET). The expression of TroTß4 was most abundant in immune organs, and was significantly up-regulated in response to infection bacterial with one of a number of bacteria (including Edwardsiella tarda, Vibrio harveyi, and Streptococcus agalactiae). Purified recombinant TroTß4 (rTroTß4) inhibited the growth of bacteria, as measured using an automatic growth curve analyzer, indicating that TroTß4 has antimicrobial functions. When administered in vivo, overexpression of TroTß4 in T. ovatus, bacterial colonization of tissues was significantly reduced. In contrast, when a DNA vector-based siRNA technology was used to knock down TroTß4 expression, bacterial dissemination and colonization of tissues increased significantly. Taken together, these results provide the first in vivo evidence to indicate that teleost thymosin beta 4 plays a significant role in innate antibacterial immune responses in addition to in vitro bacteriostatic activity. This provides valuable information regarding the biological functions of teleost thymosin beta.


Subject(s)
Fish Diseases/immunology , Immunity, Innate , Perciformes , Thymosin/genetics , Thymosin/immunology , Amino Acid Sequence , Animals , Base Sequence , Edwardsiella tarda/physiology , Enterobacteriaceae Infections/immunology , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Phylogeny , Sequence Alignment/veterinary , Streptococcal Infections/immunology , Streptococcus agalactiae/physiology , Thymosin/chemistry , Vibrio/physiology , Vibrio Infections/immunology
9.
PLoS One ; 11(6): e0156486, 2016.
Article in English | MEDLINE | ID: mdl-27310139

ABSTRACT

BACKGROUND: Prothymosin α (ProTα) (isoform 2: iso2) is a widely distributed, small acidic protein with intracellular and extracellular-associated functions. Recently, we identified two new ProTα variants with potent anti-HIV activity from CD8+ T cells and cervicovaginal lavage. The first is a splice variant of the ProTα gene known as isoB and the second is the product of ProTα pseudogene 7 (p7). Similarly to iso2, the anti-HIV activity of both variants is mediated by type I IFN. Here we tested whether the immunomodulatory activity of isoB and p7 are also TLR4 dependent and determined their kinetic of release in response to HIV-1 infection. METHODS: Type I, type III, TNF-α and IL-6 mRNA inducing activity was determined in macrophages from wild type and TLR4 knockout mice treated with recombinant ProTα variants. Supernatants from mock and HIV infected cells were analyzed by mass spectrometry in positive and negative modes for the presence of ProTα variants. In silico structural and functional analysis of ProTα variants were performed. RESULTS: We show that both isoB and p7 upregulate IFN-ß, IFN-λ1, IL-6, TNF-α and RANTES mRNAs in primary human macrophages. The potent stimulation of IFN-ß by the recombinant ProTα variants in human macrophages is dependent on the TLR4 pathway, whereas the induction of TNF-α and IL-6 may also occur independently of TLR4, suggesting the interaction of ProTα variants with other signaling molecules/receptors. In silico analyses confirmed that the novel isoB and p7 variants are intrinsically disordered proteins, which lack the NLS and mass spectrometry showed release of ProTα variants within minutes post HIV-1 infection. These features are consistent with the function of ProTα variants as damage associate molecular patterns (DAMPs). CONCLUSIONS: Our findings indicate that ProTα variants strongly inhibit viral replication mainly, but not exclusively, through TLR4 signaling and that they are released within minutes of viral infection suggesting that they may function as DAMPs.


Subject(s)
Alarmins/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Intrinsically Disordered Proteins/pharmacology , Protein Precursors/pharmacology , Thymosin/analogs & derivatives , Toll-Like Receptor 4/immunology , Alarmins/genetics , Alarmins/immunology , Amino Acid Sequence , Animals , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Gene Expression Regulation , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/growth & development , HIV-1/immunology , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Interferons , Interleukin-6/genetics , Interleukin-6/immunology , Interleukins/genetics , Interleukins/immunology , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/virology , Mice , Mice, Knockout , Primary Cell Culture , Protein Binding , Protein Interaction Mapping , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/pharmacology , Protein Precursors/genetics , Protein Precursors/immunology , Sequence Alignment , Signal Transduction , Thymosin/genetics , Thymosin/immunology , Thymosin/pharmacology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
Cell Physiol Biochem ; 38(6): 2230-8, 2016.
Article in English | MEDLINE | ID: mdl-27189760

ABSTRACT

BACKGROUND/AIMS: Neuroinflammation mediated by activated microglia may play a pivotal role in a variety of central nervous system (CNS) pathologic conditions, including ethanol-induced neurotoxicity. The purpose of this study was to investigate the function of Tß4 in ethanol-induced microglia activation. METHODS: Quantitative real-time PCR was conducted to assess the expression of Tß4 and miR-339-5p. Western blot analysis was used to measure the expression of Tß4, phosphorylated p38, ERK, JNK, Akt, and NF-x03BA;B p65. The concentration of TNF-α and IL-1ß was determined using ELISA. NO concentration was measured using a nitric oxide colorimetric BioAssay Kit. Double immunofluorescence was performed to determine Tß4 expression, in order to assess microglial activation in neonatal mouse FASD model. RESULTS: Increased Tß4 expression was observed in ethanol treated microglia. Knockdown of Tß4 enhanced ethanol-induced inflammatory mediators tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) and nitric oxide (NO) in BV-2 cells was performed. Exogenous Tß4 treatment significantly inhibited expression and secretion of these inflammatory mediators. Tß4 treatment attenuated p38, ERK MAPKs, and nuclear factor-kappa B (NF-x03BA;B) pathway activation, and enhanced miR-339-5p expression induced by ethanol exposure in microglia. A neonatal mouse fetal alcohol spectrum disorders (FASD) model showed that Tß4 expression in the microglia of the hippocampus was markedly enhanced, while Tß4 treatment effectively blocked the ethanol-induced increase in inflammatory mediators, to the level expressed in vehicle-treated control animals. CONCLUSION: This study is the first to demonstrate the function of Tß4 in ethanol-induced microglia activation, thus contributing to a more robust understanding of the role of Tß4 treatment in CNS disease.


Subject(s)
Ethanol/adverse effects , Inflammation/chemically induced , Inflammation/immunology , Microglia/drug effects , Microglia/immunology , Thymosin/immunology , Animals , Cell Line , Female , Inflammation/genetics , Interleukin-1beta/immunology , Mice, Inbred C57BL , Microglia/metabolism , NF-kappa B/immunology , Nitric Oxide/immunology , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction , Thymosin/genetics , Tumor Necrosis Factor-alpha/immunology , Up-Regulation
11.
BMC Physiol ; 16: 2, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26932824

ABSTRACT

BACKGROUND: The cell type, cell status and specific localization of Prothymosin α (PTMA) within cells seemingly determine its function. PTMA undergoes 2 types of protease proteolytic modifications that are useful in elucidating its interactions with other molecules; a factor that typifies its roles. Preferably a nuclear protein, PTMA has been shown to function in the cytoplasm and extracellularly with much evidence leaning on pathognomonic status. As such, determination of its cellular distribution under normal physiological context while utilizing varied techniques is key to illuminating prospective validation of its distinct functions in different tissues. Differential distribution insights at normal physiology would also portent better basis for further clarification of its interactions and proteolytic modifications under pathological conditions like numerous cancer, ischemic stroke and immunomodulation. We therefore raised an antibody against the C terminal of PTMA to use in tandem with available antibody against the N terminal in a murine model to explicate the differences in its distribution in brain cell types and major peripheral organs through western blotting and immunohistochemical approaches. RESULTS: The newly generated antibody was applied against the N-terminal antibody to distinguish truncated versions of PTMA or deduce possible masking of the protein by other interacting molecules. Western blot analysis indicated presence of a truncated form of the protein only in the thymus, while immunohistochemical analysis showed that in brain hippocampus the full-length PTMA was stained prominently in the nucleus whereas in the stomach full-length PTMA staining was not observed in the nucleus but in the cytoplasm. CONCLUSION: Truncated PTMA could not be detected by western blotting when both antibodies were applied in all tissues examined except the thymus. However, immunohistochemistry revealed differential staining by these antibodies suggesting possible masking of epitopes by interacting molecules. The differential localization patterns observed in the context of nucleic versus cytoplasmic presence as well as punctate versus diffuse pattern in tissues and cell types, warrant further investigations as to the forms of PTMA interacting partners.


Subject(s)
Protein Precursors/metabolism , Thymosin/analogs & derivatives , Animals , Antibodies/immunology , Blotting, Western/methods , Cell Nucleus/metabolism , Female , Immunohistochemistry/methods , Male , Mice , Protein Precursors/immunology , Rats , Thymosin/immunology , Thymosin/metabolism
12.
Expert Opin Biol Ther ; 15 Suppl 1: S51-8, 2015.
Article in English | MEDLINE | ID: mdl-26098878

ABSTRACT

Thymosin α1 (Tα1), an epithelial cell (EC)-derived cytokine, has the strong ability to modulate signals delivered through innate immune receptors on dendritic cells (DCs), thus instructing the initiation of appropriate immune responses to T cells. In its ability to activate indoleamine 2,3-dioxygenase 1-dependent tolerogenic programs in DCs, Tα1 pivotally contributes to the maintenance of self-tolerance by regulating the function of regulatory T (Treg) cells. How Tα1 may contribute to the Treg cell ontogeny is not known. The transcriptional regulator autoimmune regulator (AIRE) is known to control central and peripheral tolerance. AIRE is highly expressed in thymic medullary ECs where it controls the ectopic expression of tissue restricted antigens for negative selection. The absence of AIRE-induced tissue-specific antigens in the thymus can lead to autoimmunity in the antigen-expressing target organ. Recently, AIRE protein has been detected in peripheral lymphoid organs, suggesting that peripheral AIRE may play a complementary role. We have addressed the possible relationship between AIRE and Tα1 and discovered an intricate crosstalk, whereby AIRE may promote prothymosin cleavage to Tα1, and Tα1 in turn transcriptionally regulates AIRE expression. Thus, similar to other members of thymic stromal poietins, Tα1 expressed within the thymus and peripheral tissues regulates the EC/DC crosstalk required for salutary immune homeostasis.


Subject(s)
Thymosin/analogs & derivatives , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , Autoimmunity/immunology , Dendritic Cells/immunology , Humans , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Thymalfasin , Thymosin/biosynthesis , Thymosin/immunology , Transcription Factors/biosynthesis , Transcription Factors/immunology , AIRE Protein
13.
Dev Comp Immunol ; 50(1): 29-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25596145

ABSTRACT

Prothymosin alpha (ProTα) and thymosin beta (Tß) belong to thymosin family, which consists of a series of highly conserved peptides involved in stimulating immune responses. ProTα b and Tß are still poorly studied in teleost. Here, the full-length cDNAs of ProTα b and Tß-like (Tß-l) were cloned and identified in common carp (Cyprinus carpio L.). The expressions of carp ProTα b and Tß-l exhibited rise-fall pattern and then trended to be stable during early development. After spring viraemia of carp virus (SVCV) infection, the carp ProTα b and Tß-l transcripts were significantly up-regulated in some immune-related organs. When transiently over-expressed carp ProTα b and Tß-l in zebrafish, these two proteins up-regulated the expressions of T lymphocytes-related genes (Rag 1, TCR-γ, CD4 and CD8α). These results suggest that carp ProTα b and Tß may ultimately enhance the immune response during viral infection and modulate the development of T lymphocytes in teleost.


Subject(s)
Carps/genetics , Fish Proteins/genetics , Protein Precursors/genetics , Thymosin/analogs & derivatives , Ubiquitins/genetics , Animals , Carps/immunology , Fish Proteins/immunology , Protein Precursors/immunology , T-Lymphocytes/immunology , Thymosin/genetics , Thymosin/immunology , Ubiquitins/immunology , Zebrafish/genetics
14.
J Innate Immun ; 6(1): 72-88, 2014.
Article in English | MEDLINE | ID: mdl-23797159

ABSTRACT

Thymosin α1 (Tα1) is a naturally occurring thymic peptide used worldwide in clinical trials for the treatment of infectious diseases and cancer. The immunomodulatory activity of Tα1 on innate immunity effector cells has been extensively described, but its mechanism of action is not completely understood. We report that Tα1-exposed human monocyte-derived macrophages (MDMs) assume the typical activated morphology also exhibited by lipopolysaccharide-activated MDMs, but show a comparatively higher ability of internalizing fluorescent beads and zymosan particles. Tα1 exposure also promptly and dramatically stimulates MDM phagocytosis and killing of Aspergillus niger conidia starting as soon as 30 min after challenge. The effect is dose dependent and early coupled to low transcription of the proinflammatory cytokines tumor necrosis factor α and interleukin-6 and unmodified Toll-like receptor expression. The Tα1-stimulated phagocytosis is strictly dependent on the integrity of the microtubule network and protein kinase C activity and occurs by a variation in the classic zipper model, with recruitment of vinculin and actin at the phagosome exhibiting a punctate distribution. These findings indicate that, in human mature MDMs, Tα1 implements pathogen internalization and killing via the stimulation of the complement receptor-mediated phagocytosis. Our observations document that Tα1 is an early and potent activator of innate immunity and reinforce the concept of its pleiotropy.


Subject(s)
Aspergillus niger/immunology , Complement System Proteins/metabolism , Macrophages/immunology , Phagocytosis , Thymosin/analogs & derivatives , Actins/metabolism , Cells, Cultured , Complement Activation , Cytotoxicity, Immunologic , Down-Regulation , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Lipopolysaccharides/immunology , Phagocytosis/immunology , Thymalfasin , Thymosin/immunology , Thymosin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vinculin/metabolism , Zymosan/metabolism
15.
BMC Immunol ; 14: 43, 2013 Sep 22.
Article in English | MEDLINE | ID: mdl-24053720

ABSTRACT

BACKGROUND: Active cancer immunotherapies are beginning to yield clinical benefit, especially those using peptide-pulsed dendritic cells (DCs). Different adjuvants, including Toll-like receptor (TLR) agonists, commonly co-administered to cancer patients as part of a DC-based vaccine, are being widely tested in the clinical setting. However, endogenous DCs in tumor-bearing individuals are often dysfunctional, suggesting that ex vivo educated DCs might be superior inducers of anti-tumor immune responses. We have previously shown that prothymosin alpha (proTα) and its immunoreactive decapeptide proTα(100-109) induce the maturation of human DCs in vitro. The aim of this study was to investigate whether proTα- or proTα(100-109)-matured DCs are functionally competent and to provide preliminary evidence for the mode of action of these agents. RESULTS: Monocyte-derived DCs matured in vitro with proTα or proTα(100-109) express co-stimulatory molecules and secrete pro-inflammatory cytokines. ProTα- and proTα(100-109)-matured DCs pulsed with HER-2/neu peptides induce TH1-type immune responses, prime autologous naïve CD8-positive (+) T cells to lyse targets expressing the HER-2/neu epitopes and to express a polyfunctional profile, and stimulate CD4+ T cell proliferation in an HER-2/neu peptide-dependent manner. DC maturation induced by proTα and proTα(100-109) is likely mediated via TLR-4, as shown by assessing TLR-4 surface expression and the levels of the intracellular adaptor molecules TIRAP, MyD88 and TRIF. CONCLUSIONS: Our results suggest that proTα and proTα(100-109) induce both the maturation and the T cell stimulatory capacity of DCs. Although further studies are needed, evidence for a possible proTα and proTα(100-109) interaction with TLR-4 is provided. The initial hypothesis that proTα and the proTα-derived immunoactive decapeptide act as "alarmins", provides a rationale for their eventual use as adjuvants in DC-based anti-cancer immunotherapy.


Subject(s)
Epitopes/immunology , Peptides/immunology , Protein Precursors/immunology , Receptor, ErbB-2/immunology , Th1 Cells/immunology , Thymosin/analogs & derivatives , Adaptor Proteins, Vesicular Transport/immunology , Adaptor Proteins, Vesicular Transport/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes/metabolism , Flow Cytometry , Humans , Immunoblotting , Immunotherapy, Adoptive/methods , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Peptides/pharmacology , Protein Precursors/chemistry , Protein Precursors/pharmacology , Receptor, ErbB-2/metabolism , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Th1 Cells/drug effects , Th1 Cells/metabolism , Thymosin/chemistry , Thymosin/immunology , Thymosin/pharmacology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
16.
J Immunol Methods ; 395(1-2): 54-62, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-23831611

ABSTRACT

Apoptosis is characterized by a series of discrete biochemical events, among which is the truncation of the nuclear polypeptide prothymosin alpha (proTα) by activated caspase-3. This early apoptotic event results in the generation of a carboxy-terminal fragment of proTα, the immunoactive decapeptide proTα(100-109). We hypothesized that the detection of increased levels of proTα(100-109) in serum can be directly correlated with the induction of massive cell apoptosis, resulting from a severe bacterial infection. Thus, using high-affinity-purified polyclonal antibodies (Abs), raised in rabbits and a prototype antibody-capture system, we developed a highly sensitive and specific competitive ELISA for proTα(100-109). The sensitivity of the ELISA (0.1ng/mL to 10µg/mL) is acceptable for the quantification of the decapeptide in serum samples. To assess our initial hypothesis, we determined the concentration of proTα(100-109) in the sera of mice infected with the bacterium Streptococcus pyogenes over the course of the infection. We show that serum concentration of proTα(100-109) was marginal to undetectable before infection, increased over time and peaked at 72h postinfection. In silico analysis suggests that the Abs generated are unlikely to cross-react with any other unrelated mouse or bacterial protein. Further validation of our ELISA using serum samples from humans, infected with bacteria, may provide a useful tool to differentiate the causative agent of a potentially lethal septic infection.


Subject(s)
Apoptosis/immunology , Enzyme-Linked Immunosorbent Assay/methods , Peptide Fragments/blood , Peptide Fragments/immunology , Protein Precursors/blood , Protein Precursors/immunology , Streptococcal Infections/blood , Thymosin/analogs & derivatives , Amino Acid Sequence , Animals , Antibody Specificity , Computer Simulation , Enzyme-Linked Immunosorbent Assay/statistics & numerical data , Epitopes/chemistry , Epitopes/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptide Fragments/genetics , Protein Precursors/genetics , Rabbits , Streptococcal Infections/immunology , Streptococcal Infections/pathology , Streptococcus pyogenes , Thymosin/blood , Thymosin/genetics , Thymosin/immunology
17.
Cancer Lett ; 337(2): 237-47, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23684552

ABSTRACT

In the present study, the immunomodulatory and synergistic anti-tumor activity of thymosin α1-thymopentin fusion peptide (Tα1-TP5) was investigated in vivo. In addition, the potential receptor of Tα1-TP5 was investigated by surface plasmon resonance (SPR) binding studies. It was found that Tα1-TP5 (305 µg/kg) alleviated immunosuppression induced by hydrocortisone (HC). Tα1-TP5 (305 µg/kg) combined with cyclophosphamide (CY) had a better tumor growth inhibitory effect than CY alone. Furthermore, Tα1-TP5 had a higher affinity (KD=6.84 µmol/L) to toll-like receptor 2 (TLR2) than Tα1 (K(D)=35.4 µmol/L), but its affinity was not significantly different from that of TP5. The results of our present work indicate that Tα1-TP5 can possibly be developed as a new immunomodulatory agent.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immunologic Factors/pharmacology , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Thymopentin/pharmacology , Thymosin/analogs & derivatives , Toll-Like Receptor 2/metabolism , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Atrophy , B7-2 Antigen/metabolism , Cyclophosphamide/pharmacology , Drug Synergism , Histocompatibility Antigens Class I/metabolism , Hydrocortisone/pharmacology , Immunologic Factors/metabolism , Immunosuppressive Agents/pharmacology , Interferon-gamma/blood , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Binding , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Surface Plasmon Resonance , Thymalfasin , Thymocytes/drug effects , Thymocytes/immunology , Thymocytes/pathology , Thymopentin/immunology , Thymopentin/metabolism , Thymosin/immunology , Thymosin/metabolism , Thymosin/pharmacology , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/pathology , Time Factors , Tumor Burden/drug effects
18.
Immunotherapy ; 5(5): 497-511, 2013 May.
Article in English | MEDLINE | ID: mdl-23638745

ABSTRACT

Toll-like receptor (TLR) agonists possess remarkable properties, particularly with regard to dendritic cell activation, promoting Th1-type cytokine production and optimizing cytotoxic T-cell responses. Preclinical and clinical studies conducted to date show that TLR agonists can improve currently applied anticancer vaccination protocols. Although these have resulted in the US FDA approval of three TLR agonists for use in humans, their abundant application encounters limitations, principally due to dose-limiting toxicity evoking from systemic cytokine production. Here, using selected examples of clinical studies, we provide a concise review regarding the knowledge acquired thus far on the adjuvant use of TLR agonists as cancer vaccine components. We also provide evidence on the exploitation of a novel TLR agonist, prothymosin-α, which enhances the efficacy of tumor-reactive effectors without causing severe adverse effects.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Neoplasms/prevention & control , Toll-Like Receptors/agonists , Animals , Clinical Trials as Topic , Humans , Immunotherapy/methods , Neoplasms/immunology , Protein Precursors/immunology , Rats , Thymosin/analogs & derivatives , Thymosin/immunology , Treatment Outcome
19.
Ann N Y Acad Sci ; 1270: 105-11, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23050825

ABSTRACT

Antibodies against thymosin ß4 are available from various sources and have been used in immunohistochemistry, ELISA, and Western blot analyses. None of these antibodies have been fully characterized for specificity and influence of fixation techniques. This presents a difficulty because many tissues express more than one member of the ß-thymosin family; in addition, highly homologous sequences are typical elements of ß-thymosins. It is also important to scrutinize the influence of fixatives on the antibody-binding capability. Fixatives such as formaldehyde are well known as cross-linking reagents. Chemical modifications within the thymosin ß4 molecule might change the putative epitope recognized by the antibody. These considerations suggest that investigations on thymosin ß4 antibodies available to the scientific community are important and necessary before any experiment can be performed to exclude cross-reactivity with other ß-thymosins that are coexistent in the examined tissue and to prove antibody binding after fixation steps.


Subject(s)
Antibodies/immunology , Fixatives/adverse effects , Thymosin/immunology , Cross Reactions/drug effects , Formaldehyde/adverse effects , Humans
20.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 20(3): 564-70, 2012 Jun.
Article in Chinese | MEDLINE | ID: mdl-22739156

ABSTRACT

This study was purposed to evaluate the safety and curative effect of autologous cytokine induced killer cells (CIK) combined with low-dose IL-2 regimen containing immune enhancement of thymic peptide on elderly patients with B-cell chronic lymphocytic leukemia (B-CLL). Thymic peptide α1 was subcutaneously given as the immunoenhancement agent at a dose of 1.6 mg/d, 14 days as one cycle. Peripheral blood mononuclear cells (PBMNC) from 5 patients with B-CLL were isolated once a week to induce ex vivo CIK cells through culture in the context of interferon (IFN)-γ, interleukin (IL)-2 and anti-CD3 monoclonal antibody. The PBMNC were separated from patients before and after 14 days as one cycle of thymic peptide α1 administration. Parameters of amplification ability, effector cells quantity, lymphocyte subgroups percentage and antitumor cytotoxicity were compared before and after thymic peptide administration. The 5 patients were treated with CIK cells combined with low-dose IL-2 regimen immediately after injection of thymic peptide α1. The CIK cells plus low-dose IL-2 regimen containing thymic peptide enhancement was defined as: thymic peptide α1 1.6 mg/d was subcutaneously administered once every other day; (4 - 6) ×10(9) of CIK cells were transfused followed by IL-2 subcutaneous administration of 1 mU/d on days 1-10, 28 days as one cycle. Clinical evaluation parameters including cellular immunity function, CLL related biomarkers, disease state and infectious frequency and degree were investigated before and after CIK cells infusion puls IL-2. The results showed that the amount of amplified CIK cells, the percentage and amplification times of effector cells and antitumor cytotoxicity more significantly increased after thymic peptide α1 treatment than before its use (P < 0.05). The total 46 cycles of CIK cells infusion plus IL-2 were completed in the 5 CLL patients. No adverse reaction was observed. After treatment of CIK cells plus IL-2, the general conditions of 5 CLL patients were to different extent improved. Simultaneously, percentages of CD3(+), CD3(+)CD8(+), and CD3(+)CD56(+) cells in peripheral blood remarked by raised (P < 0.05), the serum level of ß2 microglobulin was significantly declined (P < 0.05), and the frequency and degree of infection was also decreased (P < 0.05). Following CIK cells plus IL-2 therapy, the transformation of disease state from partial remission (PR) to complete remission was seen in 3 patients, from stable disease (SD) to PR in 1 patient, and from progress of disease to SD in 1 patient. It is concluded that the regimen of autologous CIK cells combined with low-dose IL-2 containing immune enhancement of thymic peptide is safety and effective for the treatment of elderly patients with B-CLL.


Subject(s)
Cytokine-Induced Killer Cells/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Thymosin/immunology , Aged , Aged, 80 and over , Humans , Interleukin-2/administration & dosage , Interleukin-2/therapeutic use , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...