Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 838
Filtrar
1.
Biomolecules ; 14(4)2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38672485

RESUMO

Restoring peripheral immune tolerance is crucial for addressing autoimmune diseases. An ancient mechanism in maintaining the balance between inflammation and tolerance is the ratio of extracellular ATP (exATP) and adenosine. Our previous research demonstrated the effectiveness of small spleen peptides (SSPs) in inhibiting psoriatic arthritis progression, even in the presence of the pro-inflammatory cytokine TNFα, by transforming dendritic cells (DCs) into tolerogenic cells and fostering regulatory Foxp3+ Treg cells. Here, we identified thymosins as the primary constituents of SSPs, but recombinant thymosin peptides were less efficient in inhibiting arthritis than SSPs. Since Tß4 is an ecto-ATPase-binding protein, we hypothesized that SSPs regulate exATP profiles. Real-time investigation of exATP levels in DCs revealed that tolerogenic stimulation led to robust de novo exATP synthesis followed by significant degradation, while immunogenic stimulation resulted in a less pronounced increase in exATP and less effective degradation. These contrasting exATP profiles were crucial in determining whether DCs entered an inflammatory or tolerogenic state, highlighting the significance of SSPs as natural regulators of peripheral immunological tolerance, with potential therapeutic benefits for autoimmune diseases. Finally, we demonstrated that the tolerogenic phenotype of SSPs is mainly influenced by adenosine receptors, and in vivo administration of SSPs inhibits psoriatic skin inflammation.


Assuntos
Trifosfato de Adenosina , Diferenciação Celular , Células Dendríticas , Baço , Células Dendríticas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Diferenciação Celular/efeitos dos fármacos , Baço/citologia , Baço/metabolismo , Baço/efeitos dos fármacos , Baço/imunologia , Camundongos , Timosina/farmacologia , Timosina/metabolismo , Peptídeos/farmacologia , Artrite Psoriásica/tratamento farmacológico , Artrite Psoriásica/metabolismo , Artrite Psoriásica/imunologia , Humanos , Camundongos Endogâmicos C57BL , Tolerância Imunológica/efeitos dos fármacos
2.
Int J Biol Macromol ; 267(Pt 1): 131562, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38626832

RESUMO

Angiogenesis is pivotal for osteogenesis during bone regeneration. A hydrogel that promotes both angiogenesis and osteogenesis is essential in bone tissue engineering. However, creating scaffolds with the ideal balance of biodegradability, osteogenic, and angiogenic properties poses a challenge. Thymosin beta 10 (TMSB10), known for its dual role in angiogenesis and osteogenesis differentiation, faces limitations due to protein activity preservation. To tackle this issue, ZIF-8 was engineered as a carrier for TMSB10 (TMSB10@ZIF-8), and subsequently integrated into the self-assembled sericin hydrogel. The efficacy of the composite hydrogel in bone repair was assessed using a rat cranial defect model. Characterization of the nanocomposites confirmed the successful synthesis of TMSB10@ZIF-8, with a TMSB10 encapsulation efficiency of 88.21 %. The sustained release of TMSB10 from TMSB10@ZIF-8 has significantly enhanced tube formation in human umbilical vein endothelial cells (HUVECs) in vitro and promoted angiogenesis in the chicken chorioallantoic membrane (CAM) model in vivo. It has markedly improved the osteogenic differentiation ability of MC 3 T3-E1 cells in vitro. 8 weeks post-implantation, the TMSB10@ZIF-8/ Sericin hydrogel group exhibited significant bone healing (86.77 ± 8.91 %), outperforming controls. Thus, the TMSB10@ZIF-8/Sericin hydrogel, leveraging ZIF-8 for TMSB10 delivery, emerges as a promising bone regeneration scaffold with substantial clinical application potential.


Assuntos
Regeneração Óssea , Células Endoteliais da Veia Umbilical Humana , Hidrogéis , Neovascularização Fisiológica , Osteogênese , Sericinas , Timosina , Regeneração Óssea/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Animais , Hidrogéis/química , Hidrogéis/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Humanos , Ratos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Timosina/farmacologia , Timosina/química , Sericinas/química , Sericinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Camundongos , Ratos Sprague-Dawley , Masculino , Angiogênese
3.
Int J Mol Sci ; 25(4)2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38396631

RESUMO

Resistance and toxicity associated with current treatments for human cytomegalovirus (HCMV) infection highlight the need for alternatives and immunotherapy has emerged as a promising strategy. This study examined the in vitro immunological effects of co-administration of Thymosin-alpha-1 (Tα1) and polyanionic carbosilane dendrimers (PCDs) on peripheral blood mononuclear cells (PBMCs) during HCMV infection. The biocompatibility of PCDs was assessed via MTT and LDH assays. PBMCs were pre-treated with the co-administered compounds and then exposed to HCMV for 48 h. Morphological alterations in PBMCs were observed using optical microscopy and total dendritic cells (tDCs), myeloid dendritic cells (mDCs), and plasmacytoid dendritic cells (pDCs), along with CD4+/CD8+ T cells and regulatory T cells (Treg), and were characterized using multiparametric flow cytometry. The findings revealed that Tα1 + PCDs treatments increased DC activation and maturation. Furthermore, increased co-receptor expression, intracellular IFNγ production in T cells and elevated Treg functionality and reduced senescence were evident with Tα1 + G2-S24P treatment. Conversely, reduced co-receptor expression, intracellular cytokine production in T cells, lower functionality and higher senescence in Treg were observed with Tα1 + G2S16 treatment. In summary, Tα1 + PCDs treatments demonstrate synergistic effects during early HCMV infection, suggesting their use as an alternative therapeutic for preventing virus infection.


Assuntos
Dendrímeros , Polieletrólitos , Silanos , Timosina , Humanos , Timalfasina/farmacologia , Dendrímeros/farmacologia , Timosina/farmacologia , Leucócitos Mononucleares/metabolismo
4.
Mol Med Rep ; 29(4)2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38391118

RESUMO

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.


Assuntos
Traumatismo por Reperfusão , Timosina , Timosina/análogos & derivados , Camundongos , Animais , Camundongos Transgênicos , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Regulação para Cima , Timosina/genética , Timosina/farmacologia , Timosina/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico
5.
Aesthetic Plast Surg ; 48(11): 2179-2189, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38409346

RESUMO

BACKGROUND: Autologous fat grafting (AFG) has emerged as a highly sought-after plastic surgery procedure, although its success has been hampered by the uncertain fat survival rate. Current evidence suggests that adipose-derived stem cells (ADSCs) may contribute to fat retention in AFG. In previous studies, it was confirmed that thymosin beta 4 (Tß4) could enhance fat survival in vivo, although the precise mechanism remains unclear. METHODS: ADSCs were isolated from patients undergoing liposuction and their proliferation, apoptosis, anti-apoptosis, and migration were analyzed under Tß4 stimulation using cell counting kit-8, flow cytometry, wound healing assay, and real-time quantitative PCR. The mRNA levels of genes relating to angiogenesis and Hippo signaling were also determined. RESULTS: Tß4 at 100 ng/mL (p-value = 0.0171) and 1000 ng/mL (p-value = 0.0054) significantly increased ADSC proliferation from day 1 compared to the control group (0 ng/mL). In addition, the mRNA levels of proliferation-associated genes were elevated in the Tß4 group. Furthermore, Tß4 enhanced the anti-apoptotic ability of ADSCs when stimulated with Tß4 and an apoptotic induction reagent (0 ng/mL vs. 1000 ng/mL, p-value = 0.011). Crucially, the mRNA expression levels of angiogenesis-related genes and critical genes in the Hippo pathway were affected by Tß4 in ADSCs. CONCLUSIONS: Tß4 enhances adipose viability in AFG via facilitating ADSC proliferation and reducing apoptosis, and acts as a crucial positive regulator of ADSC-associated angiogenesis. Additionally, Tß4 could be accountable for the phenotypic adjustment of ADSCs by regulating the Hippo pathway. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Assuntos
Tecido Adiposo , Timosina , Adulto , Feminino , Humanos , Adipócitos , Tecido Adiposo/citologia , Tecido Adiposo/transplante , Apoptose/efeitos dos fármacos , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Sobrevivência de Enxerto , Técnicas In Vitro , Células-Tronco , Timosina/genética , Timosina/farmacologia , Transplante Autólogo
6.
Phytomedicine ; 123: 155216, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38061285

RESUMO

BACKGROUND: Thymus is the most crucial organ connecting immunity and aging. The progressive senescence of thymic epithelial cells (TECs) leads to the involution of thymus under aging, chronic stress and other factors. Ligustilide (LIG) is a major active component of the anti-aging Chinese herbal medicine Angelica sinensis (Oliv.) Diels, but its role in preventing TEC-based thymic aging remains elusive. PURPOSE: This study explored the protective role of Ligustilide in alleviating ADM (adriamycin) -induced thymic immune senescence and its underlying molecular mechanisms. METHOD: The protective effect of Ligustilide on ADM-induced thymic atrophy was examined by mouse and organotypic models, and conformed by SA-ß-gal staining in TECs. The abnormal spatial distribution of TECs in the senescent thymus was analyzed using H&E, immunofluorescence and flow cytometry. The possible mechanisms of Ligustilide in ADM-induced thymic aging were elucidated by qPCR, fluorescence labeling and Western blot. The mechanism of Ligustilide was subsequently validated through actin polymerization inhibitor, genetic engineering to regulate Thymosin ß15 (Tß15) and Tß4 expression, molecular docking and ß Thymosin-G-actin cross-linking assay. RESULTS: At a 5 mg/kg dose, Ligustilide markedly ameliorated ADM-induced weight loss and limb grip weakness in mice. It also reversed thymic damage and restored positive selection impaired by ADM. In vitro, ADM disrupted thymic structure, reduced TECs number and hindered double negative (DN) T cell differentiation. Ligustilide counteracted these effects, promoted TEC proliferation and reticular differentiation, leading to an increase in CD4+ single positive (CD4SP) T cell proportion. Mechanistically, ADM diminished the microfilament quantity in immortalized TECs (iTECs), and lowered the expression of cytoskeletal marker proteins. Molecular docking and cross-linking assay revealed that Ligustilide inhibited the protein binding between G-actin and Tß15 by inhibiting the formation of the Tß15-G-actin complex, thus enhancing the microfilament assembly capacity in TECs. CONCLUSION: This study, for the first time, reveals that Ligustilide can attenuate actin depolymerization, protects TECs from ADM-induced acute aging by inhibiting the binding of Tß15 to G-actin, thereby improving thymic immune function. Moreover, it underscores the interesting role of Ligustilide in maintaining cytoskeletal assembly and network structure of TECs, offering a novel perspective for deeper understanding of anti thymic aging.


Assuntos
4-Butirolactona/análogos & derivados , Actinas , Timosina , Camundongos , Animais , Actinas/metabolismo , Timosina/farmacologia , Timosina/metabolismo , Simulação de Acoplamento Molecular , Células Epiteliais
7.
Mol Biol (Mosk) ; 57(6): 1006-1016, 2023.
Artigo em Russo | MEDLINE | ID: mdl-38062956

RESUMO

The aim of this work was to study the effects of thymosin-1 alpha (Tα1) on the anti-inflammatory response of RAW 264.7 macrophages cultured in the presence of lipopolysaccharide (LPS) from the walls of gram-negative bacteria. As well, we evaluated production of pro-inflammatory cytokines and the activity of the NF-κB and SAPK/JNK signaling pathways. In addition, the level of expression of a number of genes that regulate cell apoptosis, as well as the activity of receptors involved in the pro-inflammatory response, was determined. First, the addition of Tα1 normalized the level of cytokine production to varying degrees, with a particularly noticeable effect on IL-1ß and IL-6. Second, the addition of Tα1 normalized the activity of the NF-κB and SAPK/JNK signaling cascades and the expression of the Tlr4 gene. Third, Tα1 significantly reduced p53 and the activity of the P53 gene, which is a marker of cell apoptosis. Fourth, it was shown that the increase in Ar-1 gene expression under the influence of LPS was significantly reduced using Tα1. Thus, it was found that the presence of Tα1 in the RAW 264.7 cell culture medium significantly reduced the level of the pro-inflammatory response of cells.


Assuntos
NF-kappa B , Timosina , Animais , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Células RAW 264.7 , Endotoxinas , Lipopolissacarídeos/farmacologia , Timosina/genética , Timosina/farmacologia , Citocinas/metabolismo
8.
Expert Opin Ther Pat ; 33(12): 865-873, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38131310

RESUMO

INTRODUCTION: Thymosins are small proteins found mainly in the thymus. They are involved in several biological processes, including immunoregulation, angiogenesis, and anti-inflammatory activity. Due to these multiple activities, thymosins are widely used as therapeutics. In fact, these peptides have shown interesting results in the treatment of eye disorders, anticancer therapy, and dysregulated immune disorders. AREA COVERED: We analyzed the thymosins therapeutic patent landscape describing the most significant patents published after 2018 and originally written in English, classified according to the different type of functions and diseases. We searched 'Thymosin' on Patentscope and Espacenet. EXPERT OPINION: Thymalfasin (Zadaxin) is the only FDA-approved thymosine-based drug used to treat chronic hepatitis B and C and as a chemotherapy inducer. This outcome demonstrates how thymosins can be exploited as therapeutics, especially in immunological and anti-cancer therapies. However, the development of modified thymosins could expand their therapeutic interest and application in different diseases. In fact, by chemical modifications, it is possible to increase proteolytic stability in the biological environment, enhance cell permeability, and stabilize the secondary structure of the peptide. Finally, the development of shorter sequences could reduce the cost and production time of these thymosin-based drugs.


Assuntos
Timosina , Timo , Humanos , Patentes como Assunto , Timosina/farmacologia , Timosina/química , Timosina/metabolismo
9.
Cells ; 12(12)2023 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-37371128

RESUMO

Liver fibrosis is the most common feature of liver disease, and activated hepatic stellate cells (HSCs) are the main contributors to liver fibrosis. Thus, finding key targets that modulate HSC activation is important to prevent liver fibrosis. Previously, we showed that thymosin ß4 (Tß4) influenced HSC activation by interacting with the Hedgehog pathway in vitro. Herein, we generated Tß4 conditional knockout (Tß4-flox) mice to investigate in vivo functions of Tß4 in liver fibrosis. To selectively delete Tß4 in activated HSCs, double-transgenic (DTG) mice were generated by mating Tß4-flox mice with α-smooth muscle actin (α-Sma)-Cre-ERT2 mice, and these mice were administered carbon tetrachloride (CCl4) or underwent bile duct ligation to induce liver fibrosis. Tß4 was selectively suppressed in the activated HSCs of DTG mouse liver, and this reduction attenuated liver injury, including fibrosis, in both fibrotic models by repressing Hedgehog (Hh) signaling. In addition, the re-expression of Tß4 by an adeno-associated virus reversed the effect of HSC-specific Tß4 deletion and led to liver fibrosis with Hh activation in CCl4-exposed mice treated with tamoxifen. In conclusion, our results demonstrate that Tß4 is a crucial regulator of HSC activation, suggesting it as a novel therapeutic target for curing liver fibrosis.


Assuntos
Células Estreladas do Fígado , Cirrose Hepática , Timosina , Animais , Camundongos , Modelos Animais de Doenças , Proteínas Hedgehog/metabolismo , Células Estreladas do Fígado/metabolismo , Cirrose Hepática/metabolismo , Camundongos Transgênicos , Timosina/farmacologia , Timosina/metabolismo
10.
Molecules ; 28(9)2023 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-37175330

RESUMO

Prion protein peptide (PrP) has demonstrated neurotoxicity in brain cells, resulting in the progression of prion diseases with spongiform degenerative, amyloidogenic, and aggregative properties. Thymosin beta 4 (Tß4) plays a role in the nervous system and may be related to motility, axonal enlargement, differentiation, neurite outgrowth, and proliferation. However, no studies about the effects of Tß4 on prion disease have been performed yet. In the present study, we investigated the protective effect of Tß4 against synthetic PrP (106-126) and considered possible mechanisms. Hippocampal neuronal HT22 cells were treated with Tß4 and PrP (106-126) for 24 h. Tß4 significantly reversed cell viability and reactive oxidative species (ROS) affected by PrP (106-126). Apoptotic proteins induced by PrP (106-126) were reduced by Tß4. Interestingly, a balance of neurotrophic factors (nerve growth factor and brain-derived neurotrophic factor) and receptors (nerve growth factor receptor p75, tropomyosin related kinase A and B) were competitively maintained by Tß4 through receptors reacting to PrP (106-126). Our results demonstrate that Tß4 protects neuronal cells against PrP (106-126) neurotoxicity via the interaction of neurotrophic factors/receptors.


Assuntos
Doenças Priônicas , Timosina , Humanos , Neurônios/metabolismo , Transdução de Sinais , Fatores de Crescimento Neural/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Timosina/farmacologia , Hipocampo/metabolismo
11.
Biomed Pharmacother ; 162: 114633, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37018994

RESUMO

Vinorelbine, the standard chemotherapy drug on advanced lung cancer, causes adverse events such as immunosuppression and bone marrow suppression. Thus, it is necessary to find drugs that could improve immune function and synergistically enhance the anti-tumor effect of vinorelbine. Thymosin is reported to inhibit tumor growth as an immunomodulator. Herein, to study the synergistic anti-cancer and attenuation effects of thymosin on vinorelbine, human lung cancer A549 cells that were labeled with CM-DiI were transplanted into zebrafish to establish the lung cancer xenotransplanted model. After treatment of vinorelbine and different concentrations of thymosin, the fluorescence intensity of CM-DiI-labeled A549 cells and the number of apoptotic muscle cells in the tumor-bearing zebrafish were detected. Besides, effects of thymosin on vinorelbine-reduced macrophages and T cells were identified in the transgenic zebrafish (Tg:zlyz-EGFP and Tg:rag2-DsRed). Then, the qRT-PCR was used to determine the alterations of the immune-related factors at the transcription level. Thymosin showed a marked synergistic anti-cancer effect with vinorelbine for the xenograft human lung cancer A549 cells, and the synergistic effect enhanced in a dose-dependent manner. Moreover, thymosin alleviated vinorelbine-induced muscle cell apoptosis, macrophage reduction, and T cell suppression. Compared with the vinorelbine group, co-administration with thymosin raised the mRNA levels of TNF-α, TNF-ß, INF-γ, and GM-CSF. Thus, thymosin possesses synergistic anti-cancer effect on vinorelbine, and has protective effect on vinorelbine-induced immunosuppression. Thymosin, as an adjuvant immunomodulatory therapy, has great potential in enhancing the clinical application of vinorelbine.


Assuntos
Neoplasias Pulmonares , Timosina , Animais , Humanos , Vinorelbina , Peixe-Zebra , Timosina/farmacologia , Timosina/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Pulmonares/patologia
12.
Int Immunopharmacol ; 117: 109996, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36933449

RESUMO

The peculiar property of Thymosin alpha 1 (Tα1) to act as master regulator of immune homeostasis has been successfully defined in different physiological and pathological contexts ranging from cancer to infection. Interestingly, recent papers also demonstrated its mitigating effect on the "cytokine storm" as well as on the T-cell exhaustion/activation in SARS-CoV-2 infected individuals. Nevertheless, in spite of the increasing knowledge on Tα1-induced effects on T cell response confirming the distinctive features of this multifaceted peptide, little is known on its effects on innate immunity during SARS-CoV-2 infection. Here, we interrogated peripheral blood mononuclear cell (PBMC) cultures stimulated with SARS-CoV-2 to disclose Tα1 properties on the main cell players of early response to infection, namely monocytes and myeloid dendritic cells (mDC). Moving from ex vivo data showing an enhancement in the frequency of inflammatory monocytes and activated mDC in COVID-19 patients, a PBMC-based experimental setting reproduced in vitro a similar profile with an increased percentage of CD16+ inflammatory monocytes and mDC expressing CD86 and HLA-DR activation markers in response to SARS-CoV-2 stimulation. Interestingly, the treatment of SARS-CoV-2-stimulated PBMC with Tα1 dampened the inflammatory/activation status of both monocytes and mDC by reducing the release of pro-inflammatory mediators, including TNF-α, IL-6 and IL-8, while promoting the production of the anti-inflammatory cytokine IL-10. This study further clarifies the working hypothesis on Tα1 mitigating action on COVID-19 inflammatory condition. Moreover, these evidence shed light on inflammatory pathways and cell types involved in acute SARS-CoV-2 infection and likely targetable by newly immune-regulating therapeutic approaches.


Assuntos
COVID-19 , Timosina , Humanos , Timalfasina/uso terapêutico , Leucócitos Mononucleares/metabolismo , SARS-CoV-2/metabolismo , Citocinas/metabolismo , Inflamação/tratamento farmacológico , Timosina/farmacologia , Timosina/uso terapêutico
13.
Genes Genomics ; 45(4): 413-427, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36445571

RESUMO

BACKGROUND: The retention rate in autologous fat grafting is an increasing concern for surgeons and patients. Our previous research verified that thymosin beta 4 (Tß4) positively affected fat survival, while the mechanism was unknown. The endothelial cells (ECs) and exosomes derived from adipose-derived stem cells (ADSCs) were regarded to play a critical role in fat transplantation. OBJECTIVE: This study aimed to evaluate the effect of exosomes derived from Tß4-treated ADSCs on EC proliferation and to identify the exosomal microRNA (miRNA) profile compared with the Tß4-untreated group. Additionally, this research intended to recognize the related molecules and signaling pathways in the Tß4-treated group with potential roles in fat transplants. METHODS: ADSCs were collected from patients who underwent liposuction surgery. Depending on whether the medium was supplemented with exogenous Tß4 or not, exosomes derived from cultured ADSCs were divided into the Tß4-Exos group and Con-Exos group. Exosome uptake and cell counting kit-8 (CCK-8) assays assessed the influence of Tß4-Exos on EC proliferation. The exosomal miRNAs of the two groups were analyzed by next-generation sequencing. With the criteria at the |log2 (fold change)| ≥ 1 and p-value < 0.05, up-regulated and down-regulated differentially expressed miRNAs (DEMs) were obtained. Prediction databases were used to predict the downstream mRNAs for DEMs. And then, overlapping genes for the up-regulated DEMs and the down-regulated were screened out, followed by enrichment analysis, protein-protein interaction network construction, and the gene cluster and hub gene identification. RESULTS: ADSCs were obtained from four female patients. The exosome uptake and CCK-8 assays showed that the Tß4-Exos could increase cell growth rate compared with the control group (DMEM-H + PBS). In Tß4-Exos and Con-Exos groups, 2651 exosomal miRNAs were recognized, with 80 up-regulated and 99 down-regulated DEMs according to the criteria. After the prediction, 621 overlapping genes for the up-regulated and 572 for the down-regulated DEMs were screened. The subsequent bioinformatics analysis found specific molecules and pathways related to the positive effect on fat survival. CONCLUSIONS: The exosomes derived from Tß4-treated ADSCs probably positively affect EC proliferation. Compared with the Con-Exos group, several exosomal DEMs, genes, and pathways were distinguished. These findings of this exploratory study provide the potential direction for future in-depth research on fat grafting.


Assuntos
Tecido Adiposo , Exossomos , Células-Tronco , Timosina , Transplante Autólogo , Adulto , Feminino , Humanos , Adulto Jovem , Tecido Adiposo/citologia , Tecido Adiposo/transplante , Proliferação de Células , Análise por Conglomerados , Exossomos/química , Exossomos/genética , Exossomos/metabolismo , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/citologia , MicroRNAs/análise , MicroRNAs/genética , Ligação Proteica , Mapas de Interação de Proteínas , Análise de Sequência de RNA , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Timosina/farmacologia
14.
Curr Protein Pept Sci ; 24(1): 78-88, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36464872

RESUMO

Thymosin ß4 (Tß4) is the ß-thymosin (Tßs) with the highest expression level in human cells; it makes up roughly 70-80% of all Tßs in the human body. Combining the mechanism and activity studies of Tß4 in recent years, we provide an overview of the subtle molecular mechanism, pharmacological action, and clinical applications of Tß4. As a G-actin isolator, Tß4 inhibits the polymerization of G-actin by binding to the matching site of G-actin in a 1:1 ratio through conformational and spatial effects. Tß4 can control the threshold concentration of G-actin in the cytoplasm, influence the balance of depolymerization and polymerization of F-actin (also called Tread Milling of F-actin), and subsequently affect cell's various physiological activities, especially motility, development and differentiation. Based on this, Tß4 is known to have a wide range of effects, including regulation of inflammation and tumor metastasis, promotion of angiogenesis, wound healing, regeneration of hair follicles, promotion of the development of the nervous system, and improving bone formation and tooth growth. Tß4 therefore has extensive medicinal applications in many fields, and serves to preserve the kidney, liver, heart, brain, intestine, and other organs, as well as hair loss, skin trauma, cornea repairing, and other conditions. In this review, we focus on the mechanism of action and clinical application of Tß4 for its main biological functions.


Assuntos
Actinas , Timosina , Humanos , Actinas/genética , Actinas/metabolismo , Citoesqueleto de Actina/metabolismo , Timosina/farmacologia , Timosina/química , Timosina/metabolismo , Cicatrização
15.
Int Immunopharmacol ; 115: 109628, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36580759

RESUMO

Wound healing involves a rapid response to the injury by circulating cells, followed by inflammation with an influx of inflammatory cells that release various factors. Soon after, cellular proliferation begins to replace the damaged cells and extracellular matrix, and then tissue remodeling restores normal tissue function. Various factors can lead to pathological wound healing when excessive and irreversible connective tissue/extracellular matrix deposition occurs, resulting in fibrosis. The process is initiated when immune cells, such as macrophages, release soluble factors that stimulate fibroblasts. TGFß is the most well-characterized macrophage derived pro-fibrotic mediator. Other soluble mediators of fibrosis include connective tissue growth factor (CTGF), platelet-derived growth factor (PDGF), and interleukin 10 (IL-10). Thymosin ß4 (Tß4) has shown therapeutic benefit in preventing fibrosis/scarring in various animal models of fibrosis/scarring. The mechanism of action of Tß4 appears related, in part, to a reduction in the inflammatory response, including a reduction in macrophage infiltration, decreased levels of TGFß and IL-10, and reduced CTGF activation, resulting in both prevention of fibroblast conversion to myofibroblasts and production of normally aligned collagen fibers. The amino N-terminal end of Tß4, SDKP (serine-aspartate-lysine-proline), appears to contain the majority of anti-fibrotic activity and has shown excellent efficacy in many animal models of fibrosis, including liver, lung, heart, and kidney fibrosis. Ac-SDKP not only prevents fibrosis but can reverse fibrosis. Unanswered questions and future directions will be presented with regard to therapeutic uses alone and in combination with already approved drugs for fibrosis.


Assuntos
Interleucina-10 , Timosina , Animais , Cicatriz/tratamento farmacológico , Fibrose , Timosina/farmacologia , Timosina/uso terapêutico , Timosina/metabolismo , Fator de Crescimento Transformador beta
16.
Cells ; 11(9)2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35563721

RESUMO

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.


Assuntos
Morte Celular Imunogênica , Timosina , Alarminas/metabolismo , Biomarcadores , Humanos , Imunidade , Peptídeos , Precursores de Proteínas , Timosina/análogos & derivados , Timosina/farmacologia
17.
Zhonghua Shao Shang Za Zhi ; 38(4): 378-384, 2022 Apr 20.
Artigo em Chinês | MEDLINE | ID: mdl-35462518

RESUMO

With the aging of population and the development of social economy, the incidence of chronic wounds is increasing day by day, while the incidence of burns and trauma remains at a high level, making wound repair an increasingly concerned area in clinical practice. Thymosin ß4 is a naturally occurring small molecule protein in vivo, which is widely distributed in a variety of body fluids and cells, especially in platelets. Thymosin ß4 has biological activities of promoting angiogenesis, anti-inflammation, anti-apoptosis, and anti-fibrosis, and has many important functions in wound repair. Thymosin ß4 has been observed to promote the healing of various wounds, such as burns, diabetic ulcers, pressure ulcers. This paper will review the molecular structure, mechanism of wound healing promotion, pharmacokinetics, and clinical application of thymosin ß4, aiming to introduce its potential in wound treatment and the shortcomings of current researches.


Assuntos
Timosina , Cicatrização , Queimaduras/tratamento farmacológico , Humanos , Úlcera por Pressão , Timosina/química , Timosina/metabolismo , Timosina/farmacologia , Timosina/uso terapêutico , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologia
18.
Ann Clin Lab Sci ; 52(2): 230-239, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35414502

RESUMO

OBJECTIVE: Thymosin b10 (TMSB10), a member of the thymosin family, is mainly located in cells and participates in the assembly and occurrence of cytoskeleton. We aimed to investigate the regulatory mechanism of TMSB10 in ccRCC. METHODS: In this study, Xiantao Academic Tools were taken to perform the pan-cancer expression and immune infiltration analysis of TMSB10. Furthermore, it is found that there is a binding site for JUN in the promoter region of TMSB10 through the JASPAR database predictive analysis. The CHIP experiment is used to confirm that JUN regulates the expression of TMSB10 through transcription, and to further detect the mRNA expression level of TMSB10 and JUN in ccRCC cell lines by qRT-PCR. Proliferation and apoptosis function analysis was also carried out to determine the functional changes of ccRCC cell lines after the expression of TMSB10 was regulated by JUN transcription. RESULTS: The results show that TMSB10 is significantly up-regulated in a variety of cancers. Moreover, JUN regulates the high expression of TMSB10 through transcription and further promotes the proliferation of ccRCC cells and inhibits their apoptosis. CONCLUSIONS: In conclusion, this study shows that JUN transcription regulates the high expression of TMSB10 and promotes the progress of ccRCC.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Timosina , Apoptose/genética , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/genética , Prognóstico , Timosina/genética , Timosina/metabolismo , Timosina/farmacologia
19.
Int J Mol Sci ; 23(1)2022 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-35008976

RESUMO

Thymosin ß4 (Tß4) was extracted forty years agofrom calf thymus. Since then, it has been identified as a G-actin binding protein involved in blood clotting, tissue regeneration, angiogenesis, and anti-inflammatory processes. Tß4 has also been implicated in tumor metastasis and neurodegeneration. However, the precise roles and mechanism(s) of action of Tß4 in these processes remain largely unknown, with the binding of the G-actin protein being insufficient to explain these multi-actions. Here we identify for the first time the important role of Tß4 mechanism in ferroptosis, an iron-dependent form of cell death, which leads to neurodegeneration and somehow protects cancer cells against cell death. Specifically, we demonstrate four iron2+ and iron3+ binding regions along the peptide and show that the presence of Tß4 in cell growing medium inhibits erastin and glutamate-induced ferroptosis in the macrophage cell line. Moreover, Tß4 increases the expression of oxidative stress-related genes, namely BAX, hem oxygenase-1, heat shock protein 70 and thioredoxin reductase 1, which are downregulated during ferroptosis. We state the hypothesis that Tß4 is an endogenous iron chelator and take part in iron homeostasis in the ferroptosis process. We discuss the literature data of parallel involvement of Tß4 and ferroptosis in different human pathologies, mainly cancer and neurodegeneration. Our findings confronted with literature data show that controlled Tß4 release could command on/off switching of ferroptosis and may provide novel therapeutic opportunities in cancer and tissue degeneration pathologies.


Assuntos
Ferroptose/efeitos dos fármacos , Quelantes de Ferro/química , Quelantes de Ferro/farmacologia , Timosina/química , Timosina/farmacologia , Sequência de Aminoácidos , Ferroptose/genética , Expressão Gênica , Humanos , Ligação de Hidrogênio , Modelos Biológicos , Modelos Moleculares , Conformação Proteica , Análise Espectral , Relação Estrutura-Atividade , Timosina/genética
20.
Stem Cell Res Ther ; 13(1): 13, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35012642

RESUMO

BACKGROUND: Prior studies show that signature phenotypes of diabetic human induced pluripotent stem cells derived endothelial cells (dia-hiPSC-ECs) are disrupted glycine homeostasis, increased senescence, impaired mitochondrial function and angiogenic potential as compared with healthy hiPSC-ECs. In the current study, we aimed to assess the role of thymosin ß-4 (Tb-4) on endothelial function using dia-hiPSC-ECs as disease model of endothelial dysfunction. METHODS AND RESULTS: Using dia-hiPSC-ECs as models of endothelial dysfunction, we determined the effect of Tb-4 on cell proliferation, senescence, cyto-protection, protein expression of intercellular adhesion molecule-1 (ICAM-1), secretion of endothelin-1 and MMP-1, mitochondrial membrane potential, and cyto-protection in vitro and angiogenic potential for treatment of ischemic limb disease in a mouse model of type 2 diabetes mellitus (T2DM) in vivo. We found that 600 ng/mL Tb4 significantly up-regulated AKT activity and Bcl-XL protein expression, enhanced dia-hiPSC-EC viability and proliferation, limited senescence, reduced endothelin-1 and MMP-1 secretion, and improved reparative potency of dia-hiPSC-ECs for treatment of ischemic limb disease in mice with T2DM. However, Tb4 had no effect on improving mitochondrial membrane potential and glycine homeostasis and reducing intercellular adhesion molecule-1 protein expression in dia-hiPSC-ECs. CONCLUSIONS: Tb-4 improves endothelial dysfunction through enhancing hiPSC-EC viability, reducing senescence and endothelin-1 production, and improves angiogenic potency in diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Células-Tronco Pluripotentes Induzidas , Timosina , Animais , Diferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Tipo 2/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Timosina/genética , Timosina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA