Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.145
Filter
1.
Mol Med Rep ; 29(4)2024 04.
Article in English | MEDLINE | ID: mdl-38391118

ABSTRACT

Prothymosin α (ProT), a highly acidic nuclear protein with multiple cellular functions, has shown potential neuroprotective properties attributed to its anti­necrotic and anti­apoptotic activities. The present study aimed to investigate the beneficial effect of ProT on neuroplasticity after ischemia­reperfusion injury and elucidate its underlying mechanism of action. Primary cortical neurons were either treated with ProT or overexpressing ProT by gene transfection and exposed to oxygen­glucose deprivation for 2 h in vitro. Immunofluorescence staining for ProT and MAP­2 was performed to quantify ProT protein expression and assess neuronal arborization. Mice treated with vehicle or ProT (100 µg/kg) and ProT overexpression in transgenic mice received middle cerebral artery occlusion for 50 min to evaluate the effect of ProT on neuroplasticity­associated protein following ischemia­reperfusion injury. The results demonstrated that in cultured neurons ProT significantly increased neurite lengths and the number of branches, accompanied by an upregulation mRNA level of brain­derived neurotrophic factor. Furthermore, ProT administration improved the protein expressions of synaptosomal­associated protein, 25 kDa and postsynaptic density protein 95 after ischemic­reperfusion injury in vivo. These findings suggested that ProT can potentially induce neuroplasticity effects following ischemia­reperfusion injury.


Subject(s)
Reperfusion Injury , Thymosin , Thymosin/analogs & derivatives , Mice , Animals , Mice, Transgenic , Protein Precursors/genetics , Protein Precursors/metabolism , Up-Regulation , Thymosin/genetics , Thymosin/pharmacology , Thymosin/metabolism , Reperfusion Injury/drug therapy
2.
Biomolecules ; 12(9)2022 08 31.
Article in English | MEDLINE | ID: mdl-36139050

ABSTRACT

Prothymosin α (PTMA) is a phylogenetically conserved polypeptide in male gonads of Vertebrates. In Mammals, it is a ubiquitous protein, and, possessing a random-coil structure, it interacts with many other partners, in both cytoplasmic and nuclear compartments. PTMA has been widely studied during cell progression in different types of cancer because of its anti-apoptotic and proliferative properties. Here, we provided the first evidence of PTMA expression and localization in human testis and in two testicular cancers (TC): classic seminoma (CS) and Leydig cell tumor (LCT). Data showed that its protein level, together with that of proliferating cell nuclear antigen (PCNA), a cell cycle progression marker, increased in both CS and LCT samples, as compared to non-pathological (NP) tissue. Moreover, in the two-cancer tissue, a decreased apoptotic rate and an increased autophagic flux was also evidenced. Results confirmed the anti-apoptotic action of PTMA, also suggesting that it can act as a switcher from apoptosis to autophagy, to favor the survival of testicular cancer cells when they develop in adverse environments. Finally, the combined data, even if they need to be further validated, add new insight into the role of PTMA in human normal and pathological testicular tissue.


Subject(s)
Testicular Neoplasms , Thymosin , Animals , Humans , Male , Mammals/metabolism , Proliferating Cell Nuclear Antigen , Protein Precursors , Quaternary Ammonium Compounds , Testicular Neoplasms/genetics , Thymosin/analogs & derivatives , Thymosin/metabolism
3.
Cells ; 11(9)2022 04 22.
Article in English | MEDLINE | ID: mdl-35563721

ABSTRACT

The new and increasingly studied concept of immunogenic cell death (ICD) revealed a previously unknown perspective of the various regulated cell death (RCD) modalities, elucidating their immunogenic properties and rendering obsolete the notion that immune stimulation is solely the outcome of necrosis. A distinct characteristic of ICD is the release of danger-associated molecular patterns (DAMPs) by dying and/or dead cells. Thus, several members of the DAMP family, such as the well-characterized heat shock proteins (HSPs) HSP70 and HSP90, the high-mobility group box 1 protein and calreticulin, and the thymic polypeptide prothymosin α (proTα) and its immunoreactive fragment proTα(100-109), are being studied as potential diagnostic tools and/or possible therapeutic agents. Here, we present the basic aspects and mechanisms of both ICD and other immunogenic RCD forms; denote the role of DAMPs in ICD; and further exploit the relevance of human proTα and proTα(100-109) in ICD, highlighting their possible clinical applications. Furthermore, we present the preliminary results of our in vitro studies, which show a direct correlation between the concentration of proTα/proTα(100-109) and the levels of cancer cell apoptosis, induced by anticancer agents and γ-radiation.


Subject(s)
Immunogenic Cell Death , Thymosin , Alarmins/metabolism , Biomarkers , Humans , Immunity , Peptides , Protein Precursors , Thymosin/analogs & derivatives , Thymosin/pharmacology
4.
Circulation ; 145(7): 531-548, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35157519

ABSTRACT

BACKGROUND: Rheumatic heart valve disease (RHVD) is a leading cause of cardiovascular death in low- and middle-income countries and affects predominantly women. The underlying mechanisms of chronic valvular damage remain unexplored and regulators of sex predisposition are unknown. METHODS: Proteomics analysis of human heart valves (nondiseased aortic valves, nondiseased mitral valves [NDMVs], valves from patients with rheumatic aortic valve disease, and valves from patients with rheumatic mitral valve disease; n=30) followed by system biology analysis identified ProTα (prothymosin alpha) as a protein associated with RHVD. Histology, multiparameter flow cytometry, and enzyme-linked immunosorbent assay confirmed the expression of ProTα. In vitro experiments using peripheral mononuclear cells and valvular interstitial cells were performed using multiparameter flow cytometry and quantitative polymerase chain reaction. In silico analysis of the RHVD and Streptococcuspyogenes proteomes were used to identify mimic epitopes. RESULTS: A comparison of NDMV and nondiseased aortic valve proteomes established the baseline differences between nondiseased aortic and mitral valves. Thirteen unique proteins were enriched in NDMVs. Comparison of NDMVs versus valves from patients with rheumatic mitral valve disease and nondiseased aortic valves versus valves from patients with rheumatic aortic valve disease identified 213 proteins enriched in rheumatic valves. The expression of the 13 NDMV-enriched proteins was evaluated across the 213 proteins enriched in diseased valves, resulting in the discovery of ProTα common to valves from patients with rheumatic mitral valve disease and valves from patients with rheumatic aortic valve disease. ProTα plasma levels were significantly higher in patients with RHVD than in healthy individuals. Immunoreactive ProTα colocalized with CD8+ T cells in RHVD. Expression of ProTα and estrogen receptor alpha correlated strongly in circulating CD8+ T cells from patients with RHVD. Recombinant ProTα induced expression of the lytic proteins perforin and granzyme B by CD8+ T cells as well as higher estrogen receptor alpha expression. In addition, recombinant ProTα increased human leukocyte antigen class I levels in valvular interstitial cells. Treatment of CD8+ T cells with specific estrogen receptor alpha antagonist reduced the cytotoxic potential promoted by ProTα. In silico analysis of RHVD and Spyogenes proteomes revealed molecular mimicry between human type 1 collagen epitope and bacterial collagen-like protein, which induced CD8+ T-cell activation in vitro. CONCLUSIONS: ProTα-dependent CD8+ T-cell cytotoxicity was associated with estrogen receptor alpha activity, implicating ProTα as a potential regulator of sex predisposition in RHVD. ProTα facilitated recognition of type 1 collagen mimic epitopes by CD8+ T cells, suggesting mechanisms provoking autoimmunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Collagen Type I/metabolism , Estrogen Receptor alpha/metabolism , Heart Valve Diseases/etiology , Heart Valve Diseases/metabolism , Protein Precursors/metabolism , Thymosin/analogs & derivatives , Amino Acid Sequence , Collagen Type I/chemistry , Computational Biology/methods , Disease Susceptibility , Epitopes, T-Lymphocyte/immunology , Heart Valve Diseases/diagnosis , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Models, Biological , Models, Molecular , Protein Binding , Protein Precursors/chemistry , Protein Precursors/genetics , Proteome , Proteomics/methods , Rheumatic Heart Disease/diagnosis , Rheumatic Heart Disease/etiology , Rheumatic Heart Disease/metabolism , Structure-Activity Relationship , Thymosin/chemistry , Thymosin/genetics , Thymosin/metabolism
5.
Int J Oncol ; 59(5)2021 Nov.
Article in English | MEDLINE | ID: mdl-34558643

ABSTRACT

The present study aimed to explore the role of the long noncoding RNA cytoskeleton regulator (CYTOR) in non­small cell lung cancer (NSCLC) radiosensitivity by manipulating the microRNA (miR)­206/prothymosin α (PTMA) axis. First, 58 pairs of NSCLC and paracancerous tissues, normal human lung epithelial cells and NSCLC cells were collected to analyze CYTOR expression and the relationship between CYTOR and NSCLC prognosis. Subsequently, CYTOR expression in radioresistant cells was assessed. Radioresistant cells with low CYTOR expression and parental cells with high CYTOR expression were established. Functional assays were then performed to assess changes in cell radiosensitivity after irradiation treatment. Subsequently, the downstream mechanism of CYTOR was explored. The binding interactions between CYTOR and miR­206 and between miR­206 and PTMA were predicted and certified. Xenograft transplantation was applied to confirm the role of CYTOR in the radiosensitivity of NSCLC. CYTOR was overexpressed in NSCLC and was associated with poor prognosis. CYTOR was further upregulated in NSCLC cells with radioresistance. CYTOR knockdown enhanced the radiosensitivity of NSCLC cells, while overexpression of CYTOR led to the opposite result. Mechanistically, CYTOR specifically bound to miR­206 and silencing CYTOR promoted miR­206 to enhance the radiosensitivity of NSCLC cells. PTMA is a target of miR­206 and silencing CYTOR inhibited PTMA expression via miR­206, thus promoting radiosensitivity of NSCLC cells. CYTOR knockdown also enhanced NSCLC cell radiosensitivity in vivo. CYTOR was highly expressed in NSCLC, while silencing CYTOR potentiated NSCLC cell radiosensitivity by upregulating miR­206 and suppressing PTMA. The present study preliminarily revealed the role of CYTOR in radiotherapy sensitivity of NSCLC and provided a novel potential target for the clinical treatment of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Cytoskeleton/physiology , Lung Neoplasms/radiotherapy , MicroRNAs/physiology , Protein Precursors/physiology , RNA, Long Noncoding/physiology , Radiation Tolerance , Thymosin/analogs & derivatives , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Down-Regulation , Humans , Lung Neoplasms/genetics , Mice , Mice, Inbred BALB C , Prognosis , Thymosin/physiology
6.
Biomolecules ; 11(8)2021 08 09.
Article in English | MEDLINE | ID: mdl-34439840

ABSTRACT

Motifs within proteins help us categorize their functions. Intrinsically disordered proteins (IDPs) are rich in short linear motifs, conferring them many different roles. IDPs are also frequently highly charged and, therefore, likely to interact with ions. Canonical calcium-binding motifs, such as the EF-hand, often rely on the formation of stabilizing flanking helices, which are a key characteristic of folded proteins, but are absent in IDPs. In this study, we probe the existence of a calcium-binding motif relevant to IDPs. Upon screening several carefully selected IDPs using NMR spectroscopy supplemented with affinity quantification by colorimetric assays, we found calcium-binding motifs in IDPs which could be categorized into at least two groups-an Excalibur-like motif, sequentially similar to the EF-hand loop, and a condensed-charge motif carrying repetitive negative charges. The motifs show an affinity for calcium typically in the ~100 µM range relevant to regulatory functions and, while calcium binding to the condensed-charge motif had little effect on the overall compaction of the IDP chain, calcium binding to Excalibur-like motifs resulted in changes in compaction. Thus, calcium binding to IDPs may serve various structural and functional roles that have previously been underreported.


Subject(s)
Calcium/metabolism , Intrinsically Disordered Proteins , Protein Precursors/chemistry , Sodium-Hydrogen Exchanger 1/chemistry , Thymosin/analogs & derivatives , alpha-Synuclein/chemistry , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Protein Binding , Protein Domains , Protein Structure, Secondary , Thymosin/chemistry
7.
Mitochondrion ; 59: 123-134, 2021 07.
Article in English | MEDLINE | ID: mdl-33872798

ABSTRACT

Colorectal cancer (CRC) affects millions of people worldwide. Chemoresistance seriously impairs the therapeutic effects. Lipid droplets (LDs) abnormally accumulate in CRC supported chemoresistance. Exploring the mechanism of LD-induced chemoresistance is extremely important for improving prognosis of CRC patients. The expression of PTMA was increased in both CRC tissues and cells, which was positively correlated with LD production. PTMA facilitated chemoresistance to gemcitabine by inducing LD production in CRC cells. PTMA enhanced LD biogenesis and chemoresistance to gemcitabine by promoting SREBP-1-mediated lipogenesis and STAT3 activation in CRC.


Subject(s)
Colonic Neoplasms/metabolism , Drug Resistance, Neoplasm , Protein Precursors/genetics , Protein Precursors/metabolism , STAT3 Transcription Factor/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Thymosin/analogs & derivatives , Acetylation , Caco-2 Cells , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Lipid Droplets/metabolism , Lipogenesis , Prognosis , Sterol Regulatory Element Binding Protein 1/genetics , Thymosin/genetics , Thymosin/metabolism , Up-Regulation
8.
Sci Rep ; 11(1): 6499, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33753796

ABSTRACT

The adrenal gland responds to heat stress by epinephrine and glucocorticoid release to alleviate the adverse effects. This study investigated the effect of acute heat stress on the protein profile and histone modification in the adrenal gland of layer-type country chickens. A total of 192 roosters were subject to acute heat stress and thereafter classified into a resistant or susceptible group according to body temperature change. The iTRAQ analysis identified 80 differentially expressed proteins, in which the resistant group had a higher level of somatostatin and hydroxy-δ-5-steroid dehydrogenase but a lower parathymosin expression in accordance with the change of serum glucocorticoid levels. Histone modification analysis identified 115 histone markers. The susceptible group had a higher level of tri-methylation of histone H3 lysine 27 (H3K27me3) and showed a positive crosstalk with K36me and K37me in the H3 tails. The differential changes of body temperature projected in physiological regulation at the hypothalamus-pituitary-adrenal axis suggest the genetic heterogeneity in basic metabolic rate and efficiency for heat dissipation to acclimate to thermal stress and maintain body temperature homeostasis. The alteration of adrenal H3K27me3 level was associated with the endocrine function of adrenal gland and may contribute to the thermotolerance of chickens.


Subject(s)
Adrenal Glands/metabolism , Chickens/metabolism , Heat-Shock Response , Histone Code , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Chickens/genetics , Hydroxysteroid Dehydrogenases/genetics , Hydroxysteroid Dehydrogenases/metabolism , Male , Thymosin/analogs & derivatives , Thymosin/genetics , Thymosin/metabolism
9.
Nat Commun ; 12(1): 84, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33398012

ABSTRACT

The disruption in blood supply due to myocardial infarction is a critical determinant for infarct size and subsequent deterioration in function. The identification of factors that enhance cardiac repair by the restoration of the vascular network is, therefore, of great significance. Here, we show that the transcription factor Zinc finger E-box-binding homeobox 2 (ZEB2) is increased in stressed cardiomyocytes and induces a cardioprotective cross-talk between cardiomyocytes and endothelial cells to enhance angiogenesis after ischemia. Single-cell sequencing indicates ZEB2 to be enriched in injured cardiomyocytes. Cardiomyocyte-specific deletion of ZEB2 results in impaired cardiac contractility and infarct healing post-myocardial infarction (post-MI), while cardiomyocyte-specific ZEB2 overexpression improves cardiomyocyte survival and cardiac function. We identified Thymosin ß4 (TMSB4) and Prothymosin α (PTMA) as main paracrine factors released from cardiomyocytes to stimulate angiogenesis by enhancing endothelial cell migration, and whose regulation is validated in our in vivo models. Therapeutic delivery of ZEB2 to cardiomyocytes in the infarcted heart induces the expression of TMSB4 and PTMA, which enhances angiogenesis and prevents cardiac dysfunction. These findings reveal ZEB2 as a beneficial factor during ischemic injury, which may hold promise for the identification of new therapies.


Subject(s)
Ischemia/pathology , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Dependovirus/metabolism , Gene Expression Regulation , Humans , Ischemia/genetics , Mice, Knockout , Models, Biological , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic/genetics , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thymosin/analogs & derivatives , Thymosin/genetics , Thymosin/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics
10.
Cell Mol Neurobiol ; 41(8): 1817-1828, 2021 Nov.
Article in English | MEDLINE | ID: mdl-32856232

ABSTRACT

Prothymosin alpha (ProTα) is involved in multiple cellular processes. Upon serum-free stress, ProTα lacking a signal peptide sequence is non-classically released from C6 glioma cells as a complex with Ca2+-binding cargo protein S100A13. Thus, ProTα and S100A13 are conceived to be members of damage-associated molecular patterns (DAMPs)/alarmins. However, it remains to be determined whether stress-induced release of ProTα and S100A13 involves SNARE proteins in the mechanisms underlying membrane tethering of the multiprotein complex. In the present study, we used C6 glioma cells as a model of ProTα release. In pull-down assay, p40 synaptotagmin-1 (Syt-1), a vesicular SNARE, formed a hetero-oligomeric complex with homodimeric S100A13 in a Ca2+-dependent manner. The interaction between p40 Syt-1 and S100A13 was also Ca2+-dependent in surface plasmon resonance (SPR). Immunoprecipitation using conditioned medium (CM) revealed that p40 Syt-1 was co-released with ProTα and S100A13 upon serum-free stress. In in situ proximity ligation assay (PLA), Syt-1 interacted with S100A13 upon serum-free stress in C6 glioma cells. The intracellular delivery of anti-Syt-1 IgG blocked serum free-induced release of ProTα and S100A13. Serum free-induced ProTα-EGFP release was significantly blocked by botulinum neurotoxin/C1 (BoNT/C1), which cleaves target SNARE syntaxin-1 (Stx-1). In immunocytochemistry, the cellular loss of ProTα-EGFP, S100A13, and Syt-1 was also blocked by BoNT/C1. Furthermore, the intracellular delivery of anti-Stx-1 IgG or Stx-1 siRNA treatment blocked Syt-1, S100A13 and ProTα release from C6 glioma cells. All these findings suggest that SNARE proteins play roles in stress-induced non-classical release of DAMPs/alarmins proteins, ProTα and S100A13 from C6 glioma cells.


Subject(s)
Alarmins/metabolism , Protein Precursors/metabolism , S100 Proteins/metabolism , SNARE Proteins/metabolism , Thymosin/analogs & derivatives , Animals , Calcium/metabolism , Cell Line, Tumor , Protein Binding/physiology , Rats , Thymosin/metabolism
11.
Nat Commun ; 11(1): 5736, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33184256

ABSTRACT

Highly charged intrinsically disordered proteins can form complexes with very high affinity in which both binding partners fully retain their disorder and dynamics, exemplified by the positively charged linker histone H1.0 and its chaperone, the negatively charged prothymosin α. Their interaction exhibits another surprising feature: The association/dissociation kinetics switch from slow two-state-like exchange at low protein concentrations to fast exchange at higher, physiologically relevant concentrations. Here we show that this change in mechanism can be explained by the formation of transient ternary complexes favored at high protein concentrations that accelerate the exchange between bound and unbound populations by orders of magnitude. Molecular simulations show how the extreme disorder in such polyelectrolyte complexes facilitates (i) diffusion-limited binding, (ii) transient ternary complex formation, and (iii) fast exchange of monomers by competitive substitution, which together enable rapid kinetics. Biological polyelectrolytes thus have the potential to keep regulatory networks highly responsive even for interactions with extremely high affinities.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Polyelectrolytes/chemistry , Kinetics , Magnetic Resonance Spectroscopy , Molecular Chaperones/chemistry , Molecular Dynamics Simulation , Protein Binding , Protein Interaction Domains and Motifs , Protein Precursors/chemistry , Staining and Labeling , Thymosin/analogs & derivatives
12.
Life Sci Alliance ; 3(12)2020 12.
Article in English | MEDLINE | ID: mdl-33087487

ABSTRACT

This study reports that parathymosin (PTMS) is secreted by hypothalamic stem/progenitor cells (htNSC) to inhibit senescence of recipient cells such as fibroblasts. Upon release, PTMS is rapidly transferred into the nuclei of various cell types, including neuronal GT1-7 cells and different peripheral cells, and it is effectively transferred into neuronal nuclei in various brain regions in vivo. Notably, brain neurons also produce and release PTMS, and because neuronal populations are large, they are important for maintaining PTMS in the cerebrospinal fluid which is further transferable into the blood. Compared with several other brain regions, the hypothalamus is stronger for long-distance PTMS transfer, supporting a key hypothalamic role in this function. In physiology, aging is associated with declines in PTMS production and transfer in the brain, and ptms knockdown in the hypothalamus versus hippocampus were studied showing different contributions to neurobehavioral physiology. In conclusion, the brain is an endocrine organ through secretion and nuclear transfer of PTMS, and the hypothalamus-brain orchestration of this function is protective in physiology and counteractive against aging-related disorders.


Subject(s)
Bodily Secretions/metabolism , Hypothalamus/metabolism , Thymosin/analogs & derivatives , Animals , Brain/metabolism , Endocrine Glands/metabolism , Fibroblasts/metabolism , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Stem Cells/metabolism , Thymosin/metabolism , Thymosin/physiology
13.
Clin Sci (Lond) ; 134(18): 2435-2445, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32909608

ABSTRACT

High-risk pregnancies, such as pregnancies with gestational diabetes mellitus (GDM), are becoming more common and as such, have become important public health issues worldwide. GDM increases the risks of macrosomia, premature infants, and preeclampsia. Although placental dysfunction, including fibrosis is associated with the development of GDM, factors that link these observations remain unknown. Prothymosin α (ProTα) is expressed in the placenta and is involved in cell proliferation and immunomodulation. It also plays an important role in insulin resistance and fibrosis. However, the role of ProTα in GDM is still unclear. In the present study, we found that fibrosis-related protein expressions, such as type I collagen (Col-1) were significantly increased in the placentae of ProTα transgenic mice. With elevated fibrosis-related protein expressions, placental weights significantly increased in GDM group. In addition, placental and circulating ProTα levels were significantly higher in patients with GDM (n=39), compared with the healthy group (n=102), and were positively correlated with Col-1 expression. Mice with streptozotocin (STZ)-induced GDM had increased ProTα, fasting blood glucose, Col-1, and placental weight, whereas plasma insulin levels were decreased. ProTα overexpression enhanced nuclear factor κB (NFκB) activation to increase fibrosis-related protein expressions in 3A-Sub-E trophoblasts, while treatment with an NFκB inhibitor reversed the effect of ProTα on fibrosis-related protein expressions. We further investigated whether ProTα is regulated by hyperglycemia-induced reactive oxygen species (ROS). In conclusion, ProTα increases the amount of placental connective tissue and thus contributes to the pathogenesis of placental fibrosis in GDM. Therefore, ProTα may be a novel therapeutic target for GDM.


Subject(s)
Collagen Type I/metabolism , Diabetes, Gestational/metabolism , Diabetes, Gestational/pathology , Placenta/pathology , Protein Precursors/metabolism , Thymosin/analogs & derivatives , Adult , Animals , Diabetes, Gestational/genetics , Female , Fibrosis , Gene Expression Regulation , Humans , Hyperglycemia/complications , Inflammation/pathology , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Pregnancy , Reactive Oxygen Species/metabolism , Signal Transduction , Thymosin/metabolism , Trophoblasts/metabolism
15.
Food Chem ; 327: 127002, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32438262

ABSTRACT

This study was designed to investigate proteome changes in Japanese puffer fish (Takifugu rubripes) during short- and long-term frozen storage. In total, 1484 proteins were quantified, and 164 proteins were identified as differential abundance proteins (DAPs) in Japanese puffer fish from two frozen storage treatment groups (14 days and 60 days) compared with the fresh control group. Correlation analysis between the DAPs and quality traits of the puffer fish muscle showed that 106 proteins were correlated closely with colour and texture (hardness, elasticity, and chewiness). Bioinformatics analysis revealed and Western blot analysis verified that Putative prothymosin alpha species, Bridging integrator 3, NADH: the ubiquinone oxidoreductase subunit and Mx species are candidate biomarkers for puffer fish properties. This study offers valuable evidence to improve the quality control and monitoring of Japanese puffer fish during transportation and storage.


Subject(s)
Biomarkers/analysis , Fish Products/analysis , Fish Proteins/analysis , Takifugu , Animals , Color , Computational Biology/methods , Food Analysis/methods , Food Quality , Food Storage , Freezing , Muscle, Skeletal/chemistry , Protein Precursors/analysis , Thymosin/analogs & derivatives , Thymosin/analysis
16.
Mol Cell Biochem ; 469(1-2): 133-142, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32304006

ABSTRACT

Pro-inflammatory cytokines prevent bone regeneration in vivo and activation of nuclear factor-κB (NF-κB) signaling has been proposed to lead to suppression of bone morphogenetic protein (BMP)-induced osteogenesis via direct binding of p65 to Smad4 in vitro. Application of a small nuclear acidic protein (MTI-II) and its delivered peptide, MPAID (MTI-II peptide anti-inflammatory drug) has been described to elicit therapeutic potential via strong anti-inflammatory action following the physical association of MTI-II and MPAID with p65. However, it is unclear whether MTI-II attenuates tumor necrosis factor (TNF)-α inhibition of BMP-induced osteogenesis. Herein, we found that TNF-α-mediated suppression of responses associated with BMP4-induced osteogenesis, including expression of the osteocalcin encoding gene Ocn, Smad binding element (SBE)-dependent luciferase activity, alkaline phosphatase activity, and alizarin red S staining were largely restored by MTI-II and MPAID in MC3T3-E1 cells. Mechanistically, MTI-II and MPAID did not inhibit nuclear translocation of p65 or disassociate Smad4 from p65. Further, results from chromatin immunoprecipitation (ChIP) analyses revealed that Smad4 enrichment in cells over-expressing MTI-II and treated with TNF-α was equivalent to that in cells without TNF-α treatment. Alternatively, Smad4 enrichment was considerably decreased following TNF-α treatment in control cells. Moreover, p65 enrichment in the Id-1 promoter SBE was detected only when cells over-expressing MTI-II were stimulated with TNF-α. Overall, our study concludes that MTI-II restored TNF-α-inhibited suppression of BMP-Smad-induced osteogenic differentiation by enhancing accessibility of the Smad4-p65 complex to the SBE rather than by liberating Smad4 from p65.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bone Morphogenetic Proteins/pharmacology , Osteogenesis/drug effects , Smad4 Protein/metabolism , Thymosin/analogs & derivatives , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Alkaline Phosphatase/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Chromatin Immunoprecipitation , Mice , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Signal Transduction/drug effects , Thymosin/pharmacology
17.
J Pharmacol Sci ; 143(2): 127-131, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32156464

ABSTRACT

The inhibition of retinal ischemia-induced damage by post-ischemic prothymosin alpha (ProTα) was not affected in toll-like receptor 4 knockout (TLR4-/-) mice but blocked by the pretreatment with antibody against F0/F1 ATPase α- or ß-subunit, novel candidate for ProTα-receptor. In addition to the previous observation of ProTα-induced ATP release from cells, the present study showed a ProTα-induced enhancement of ATP hydrolysis activity of recombinant ATP5A1/5B complex. As the protection of retinal function by post-ischemic ProTα was abolished by anti-P2Y12 antibody, the activation of F0/F1 ATPase and subsequent P2Y12 receptor system may play roles in beneficial actions by post-ischemic ProTα.


Subject(s)
Ischemia/metabolism , Ischemia/prevention & control , Protein Precursors/administration & dosage , Protein Precursors/pharmacology , Proton-Translocating ATPases/metabolism , Receptors, Purinergic P2Y12/metabolism , Retina , Thymosin/analogs & derivatives , Animals , Hydrolysis/drug effects , Male , Mice, Inbred C57BL , Mitochondrial Proton-Translocating ATPases/metabolism , Recombinant Proteins/metabolism , Thymosin/administration & dosage , Thymosin/pharmacology
18.
Peptides ; 126: 170265, 2020 04.
Article in English | MEDLINE | ID: mdl-31982448

ABSTRACT

Prothymosin alpha (ProTα)-mimetic hexapeptide (amino acid: NEVDQE, P6Q) inhibits cerebral or retinal ischemia-induced behavioral, electrophysiological and histological damage. P6Q also abolishes cerebral hemorrhage induced by ischemia with tissue plasminogen activator (tPA). In the present study we examined the beneficial effects of P6Q on other post-stroke prognostic psychology-related symptoms, which obstruct the motivation toward physical therapy. Intravenous (i.v.) administration with tPA (10 mg/kg) at 6 h after photochemically induced thrombosis (PIT) in mice resulted in bilateral central post-stroke pain in thermal and mechanical nociception tests and loss of social activity in the nest building test, both of which were significantly blocked by P6Q (30 mg/kg, i.v.) given at 5 h after PIT. P6Q (30 mg/kg, i.v.) also improved the memory-learning deficit in the step-through test and depression-like behavior in the tail suspension test when it was given 1 day after bilateral common carotid arteries occlusion (BCCAO) in mice. Thus, these studies suggest that P6Q could be a promising candidate to prevent negative prognostic psychological symptoms following focal and global ischemia.


Subject(s)
Brain Ischemia/drug therapy , Depression/drug therapy , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Pain/drug therapy , Protein Precursors/pharmacology , Stroke/drug therapy , Thymosin/analogs & derivatives , Animals , Brain Ischemia/chemically induced , Brain Ischemia/pathology , Brain Ischemia/psychology , Depression/etiology , Depression/pathology , Learning , Male , Memory Disorders/etiology , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Pain/etiology , Pain/pathology , Peptide Fragments/pharmacology , Stroke/chemically induced , Stroke/pathology , Stroke/psychology , Thymosin/pharmacology , Tissue Plasminogen Activator/toxicity
19.
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
20.
J Neurochem ; 153(6): 772-789, 2020 06.
Article in English | MEDLINE | ID: mdl-31454420

ABSTRACT

Tissue plasminogen activator (tPA) administration beyond 4.5 h of stroke symptoms is beneficial for patients but has an increased risk of cerebral hemorrhage. Thus, increasing the therapeutic window of tPA is important for stroke recovery. We previously showed that prothymosin alpha (ProTα) or its mimetic hexapeptide (P6Q) has anti-ischemic activity. Here, we examined the beneficial effects of ProTα or P6Q against delayed tPA-induced brain damage following middle cerebral artery occlusion (MCAO) or photochemically induced thrombosis in mice. Brain hemorrhage was observed by tPA administration during reperfusion at 4.5 and 6 h after MCAO. Co-administration of ProTα with tPA at 4.5 h inhibited hemorrhage and motor dysfunction 2-4 days, but not 7 days after MCAO. ProTα administration at 2 and 4.5 h after MCAO significantly inhibited tPA (4.5 h)-induced motor dysfunction and death more than 7 days. Administration of tPA caused the loss of tight junction proteins, zona occulden-1 and occludin, and up-regulation of matrix metalloproteinase-2/9, in a ProTα-reversible manner. P6Q administration abolished tPA (4.5 h)-induced hemorrhage and reversed tPA (6 h)-induced vascular damage and matrix metalloproteinase-2 and 9 up-regulation. Twice administrations of P6Q at 2 h alone and 6 h with tPA significantly improved motor dysfunction more than 7 days. In photochemically induced thrombosis ischemia, similar vascular leakage and neuronal damage (infarction and motor dysfunction) by late tPA (4.5 or 6 h) were also inhibited by P6Q. Thus, these studies suggest that co-administration with ProTα or P6Q would be beneficial to inhibit delayed tPA-induced hemorrhagic mechanisms in acute ischemic stroke.


Subject(s)
Biomimetic Materials/therapeutic use , Brain Injuries/drug therapy , Brain Ischemia/drug therapy , Protein Precursors/therapeutic use , Thymosin/analogs & derivatives , Tissue Plasminogen Activator/toxicity , Animals , Brain Injuries/chemically induced , Brain Injuries/pathology , Brain Ischemia/chemically induced , Brain Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Thymosin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...