Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Eur J Nutr ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38743096

RESUMO

PURPOSE: Diet-related factors are of great significance in the regulation of hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-gonad (HPG) axes. In this study, we aimed to investigate the effects of chronic exposure to a high fat diet (HFD), fructose or sucralose on the endocrine functions. METHODS: Male, Sprague-Dawley rats received a normal chow diet, HFD, 10% fructose or 0.02% sucralose for 10 weeks. Behavioral changes were assessed by open field (OFT) and elevated plus-maze (EPM) tests at week 8. H&E staining was used to observe pathological changes in adrenal cortex, testis and perirenal adipose tissue. Serum hormone concentrations were quantified via enzyme-linked immunosorbent assay (ELISA). The mRNA expression levels of genes along the HPA and HPG axes were determined using real-time PCR. RESULTS: All types of dietary interventions increased body weight and disturbed metabolic homeostasis, with anxiogenic phenotype in behavioral tests and damage to cell morphology of adrenal cortex and testis being observed. Along the HPA axis, significantly increased corticotropin releasing hormone (CRH), adrenocorticotropic hormone (ACTH) and corticosterone (CORT) concentrations were observed in the HFD or 0.02% sucralose group. For HPG axis, gonadotropin-releasing hormone (GnRH) and estradiol (E2) concentrations were significantly increased in all dietary intervention groups, while decreased concentrations of follicle-stimulating hormone (FSH) and testosterone (T) were also detected. Moreover, transcriptional profiles of genes involved in the synthesis of hormones and corresponding hormone receptors were significantly altered. CONCLUSION: Long-term consumption of HFD, fructose or sucralose manifested deleterious effects on endocrine system and resulted in the dysregulation of HPA and HPG axes.

3.
Sci Rep ; 14(1): 8193, 2024 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-38589544

RESUMO

The study aimed to determine the specific relative biological effectiveness (RBE) of various cells in the hippocampus following proton irradiation. Sixty Sprague-Dawley rats were randomly allocated to 5 groups receiving 20 or 30 Gy of proton or photon irradiation. Pathomorphological neuronal damage in the hippocampus was assessed using Hematoxylin-eosin (HE) staining. The expression level of NeuN, Nestin, Caspase-3, Olig2, CD68 and CD45 were determined by immunohistochemistry (IHC). The RBE range established by comparing the effects of proton and photon irradiation at equivalent biological outcomes. Proton20Gy induced more severe damage to neurons than photon20Gy, but showed no difference compared to photon30Gy. The RBE of neuron was determined to be 1.65. Similarly, both proton20Gy and proton30Gy resulted in more inhibition of oligodendrocytes and activation of microglia in the hippocampal regions than photon20Gy and photon30Gy. However, the expression of Olig2 was higher and CD68 was lower in the proton20Gy group than in the photon30Gy group. The RBE of oligodendrocyte and microglia was estimated to be between 1.1 to 1.65. For neural stem cells (NSCs) and immune cells, there were no significant difference in the expression of Nestin and CD45 between proton and photon irradiation (both 20 and 30 Gy). Therefore, the RBE for NSCs and immune cell was determined to be 1.1. These findings highlight the varying RBE values of different cells in the hippocampus in vivo. Moreover, the actual RBE of the hippocampus may be higher than 1.1, suggesting that using as RBE value of 1.1 in clinical practice may underestimate the toxicities induced by proton radiation.


Assuntos
Terapia com Prótons , Prótons , Ratos , Animais , Terapia com Prótons/métodos , Nestina , Eficiência Biológica Relativa , Ratos Sprague-Dawley , Hipocampo
4.
Front Immunol ; 15: 1334772, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38571956

RESUMO

Background: Autoimmune thyroid disease (AITD) ranks among the most prevalent thyroid diseases, with inflammatory cytokines playing a decisive role in its pathophysiological process. However, the causal relationship between the inflammatory cytokines and AITD remains elusive. Methods: A two-sample Mendelian randomization (MR) analysis was performed to elucidate the causal connection between AITD and 41 inflammatory cytokines. Genetic variations associated with inflammatory cytokines were sourced from the FinnGen biobank, whereas a comprehensive meta-analysis of genome-wide association studies (GWASs) yielded data on Graves' disease (GD) and Hashimoto thyroiditis. Regarding the MR analysis, the inverse variance-weighted, MR-Egger, and weighted median methods were utilized. Additionally, sensitivity analysis was conducted using MR-Egger regression, MR-pleiotropy residual sum, and outliers. Results: Seven causal associations were identified between inflammatory cytokines and AITD. High levels of tumor necrosis factor-ß and low levels of stem cell growth factor-ß were indicative of a higher risk of GD. In contrast, high levels of interleukin-12p70 (IL-12p70), IL-13, and interferon-γ and low levels of monocyte chemotactic protein-1 (MCP-1) and TNF-α suggested a higher risk of HD. Moreover, 14 causal associations were detected between AITD and inflammatory cytokines. GD increases the levels of macrophage inflammatory protein-1ß, MCP-1, monokine induced by interferon-γ (MIG), interferon γ-induced protein 10 (IP-10), stromal cell-derived factor-1α, platelet-derived growth factor BB, ß-nerve growth factor, IL-2ra, IL-4, and IL-17 in blood, whereas HD increases the levels of MIG, IL-2ra, IP-10, and IL-16 levels. Conclusion: Our bidirectional MR analysis revealed a causal relationship between inflammatory cytokines and AITD. These findings offer valuable insights into the pathophysiological mechanisms underlying AITD.


Assuntos
Citocinas , Doença de Hashimoto , Humanos , Interferon gama , Análise da Randomização Mendeliana , Doença de Hashimoto/genética , Quimiocina CXCL10 , Estudo de Associação Genômica Ampla
5.
J Cell Mol Med ; 28(7): e18212, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38516826

RESUMO

SBFI26, an inhibitor of FABP5, has been shown to suppress the proliferation and metastasis of tumour cells. However, the underlying mechanism by which SBFI26 induces ferroptosis in breast cancer cells remains largely unknown. Three breast cancer cell lines were treated with SBFI26 and CCK-8 assessed cytotoxicity. Transcriptome was performed on the Illumina platform and verified by qPCR. Western blot evaluated protein levels. Malondialdehyde (MDA), total superoxide dismutase (T-SOD), Fe, glutathione (GSH) and oxidized glutathione (GSSG) were measured. SBFI26 induced cell death time- and dose-dependent, with a more significant inhibitory effect on MDA-MB-231 cells. Fer-1, GSH and Vitamin C attenuated the effects but not erastin. RNA-Seq analysis revealed that SBFI26 treatment significantly enriched differentially expressed genes related to ferroptosis. Furthermore, SBFI26 increased intracellular MDA, iron ion, and GSSG levels while decreasing T-SOD, total glutathione (T-GSH), and GSH levels.SBFI26 dose-dependently up-regulates the expression of HMOX1 and ALOX12 at both gene and protein levels, promoting ferroptosis. Similarly, it significantly increases the expression of SAT1, ALOX5, ALOX15, ALOXE3 and CHAC1 that, promoting ferroptosis while downregulating the NFE2L2 gene and protein that inhibit ferroptosis. SBFI26 leads to cellular accumulation of fatty acids, which triggers excess ferrous ions and subsequent lipid peroxidation for inducing ferroptosis.


Assuntos
Ferroptose , Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Dissulfeto de Glutationa , Ferroptose/genética , Peroxidação de Lipídeos , Glutationa , Ferro , Superóxido Dismutase/genética , Espécies Reativas de Oxigênio , Proteínas de Ligação a Ácido Graxo
6.
J Cardiothorac Surg ; 19(1): 109, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38443970

RESUMO

BACKGROUND: Pulmonary primitive neuroectodermal tumor (PNET), a member of the Ewing sarcoma family of tumors, is a rare malignancy that is associated with a grim prognosis. To date, fewer than 30 cases of pulmonary PNET have been reported. In this case report, we present the clinical details of a 12-year-old girl with pulmonary PNET who underwent surgical treatment. We also conducted an analysis and summary of other relevant studies and the surgical outcomes. CASE PRESENTATION: In May 2018, a 12-year-old girl was admitted with symptoms of cough and blood-tinged phlegm. A computed tomography scan revealed a large mass, measuring 12.9 cm × 8.1 cm, in the right middle and lower lungs. A percutaneous lung biopsy confirmed poorly differentiated tumor cells with a nested growth pattern. Immunohistochemical staining demonstrated positive expression of CD99, CD56, Vimentin, and Synaptophysin. The patient was diagnosed with pulmonary PNET. Following three cycles of neoadjuvant chemotherapy, a substantial reduction in tumor volume was observed. Subsequently, the patient underwent a surgical procedure involving pneumonectomy and partial resection of the left atrium with the assistance of cardiopulmonary bypass. The patient was discharged 37 days after surgery. During a three-year follow-up period, she exhibited no signs of tumor recurrence and has successfully returned to school. CONCLUSIONS: This case highlights the successful management of an advanced PNET with neoadjuvant chemotherapy, pneumonectomy, and partial resection of the left atrium employing cardiopulmonary bypass. The patient remained disease-free after three years. Our analysis of surgically treated cases indicates that neoadjuvant chemotherapy can contribute to improved prognoses for PNET patients. It is crucial to emphasize that complete surgical excision remains the cornerstone of treatment, underscoring the importance of surgeons considering radical surgical approaches whenever feasible for patients with pulmonary PNETs.


Assuntos
Recidiva Local de Neoplasia , Tumores Neuroectodérmicos Primitivos , Feminino , Humanos , Criança , Pneumonectomia , Terapia Neoadjuvante , Pulmão , Tumores Neuroectodérmicos Primitivos/diagnóstico , Tumores Neuroectodérmicos Primitivos/cirurgia
7.
Eur J Cardiothorac Surg ; 65(4)2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38530803

RESUMO

OBJECTIVES: Tracheal reconstruction post-extensive resection remains an unresolved challenge in thoracic surgery. This study evaluates the use of aortic allografts (AAs) for tracheal replacement and reconstruction in a rat model, aiming to elucidate the underlying mechanisms of tracheal regeneration. METHODS: AAs from female rats were employed for tracheal reconstruction in 36 male rats, with the replacement exceeding half of the tracheal length. To avert collapse, silicone stents were inserted into the AA lumens. No immunosuppressive therapy was administered. The rats were euthanized biweekly, and the AAs were examined for neovascularization, cartilage formation, respiratory epithelial ingrowth, submucosal gland regeneration and the presence of the Sex-determining region of Y-chromosome (SRY) gene. RESULTS: All procedures were successfully completed without severe complications. The AA segments were effectively integrated into the tracheal framework, with seamless distinction at suture lines. Histological analysis indicated an initial inflammatory response, followed by the development of squamous and mucociliary epithelia, new cartilage ring formation and gland regeneration. In situ hybridization identified the presence of the SRY gene in newly formed cartilage rings, confirming that regeneration was driven by recipient cells. CONCLUSIONS: This study demonstrates the feasibility of AAs transforming into functional tracheal conduits, replicating the main structural and functional characteristics of the native trachea. The findings indicate that this approach offers a novel pathway for tissue regeneration and holds potential for treating extensive tracheal injuries.


Assuntos
Aorta , Procedimentos de Cirurgia Plástica , Masculino , Feminino , Animais , Ratos , Estudos de Viabilidade , Aorta/cirurgia , Traqueia/cirurgia , Traqueia/fisiologia , Stents , Aloenxertos/cirurgia , Engenharia Tecidual
8.
STAR Protoc ; 5(1): 102881, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38386546

RESUMO

Here, we present a protocol for inducing fibrosis in human kidney-2 (HK2) cells followed by quantitative real-time PCR analysis of fibrosis-related genes. We describe steps for growing and expanding cells, inducing HK2 fibrosis, and collecting cells for downstream applications. Given the limited cell quantity in culture flasks and the challenges of cell collection, we utilized 10-cm Petri dishes for cell harvesting, with each experimental group comprising five replicate samples. For complete details on the use and execution of this protocol, please refer to Zhang et al.1.


Assuntos
Células Epiteliais , Rim , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Fibrose
9.
Angew Chem Int Ed Engl ; 63(15): e202318534, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38343199

RESUMO

Click chemistry is a powerful molecular assembly strategy for rapid functional discovery. The development of click reactions with new connecting linkage is of great importance for expanding the click chemistry toolbox. We report the first selenium-nitrogen exchange (SeNEx) click reaction between benzoselenazolones and terminal alkynes (Se-N to Se-C), which is inspired by the biochemical SeNEx between Ebselen and cysteine (Cys) residue (Se-N to Se-S). The formed selenoalkyne connection is readily elaborated, thus endowing this chemistry with multidimensional molecular diversity. Besides, this reaction is modular, predictable, and high-yielding, features fast kinetics (k2≥14.43 M-1 s-1), excellent functional group compatibility, and works well at miniaturization (nanomole-scale), opening up many interesting opportunities for organo-Se synthesis and bioconjugation, as exemplified by sequential click chemistry (coupled with ruthenium-catalyzed azide-alkyne cycloaddition (RuAAC) and sulfur-fluoride exchange (SuFEx)), selenomacrocycle synthesis, nanomole-scale synthesis of Se-containing natural product library and DNA-encoded library (DEL), late-stage peptide modification and ligation, and multiple functionalization of proteins. These results indicated that SeNEx is a useful strategy for new click chemistry developments, and the established SeNEx chemistry will serve as a transformative platform in multidisciplinary fields such as synthetic chemistry, material science, chemical biology, medical chemistry, and drug discovery.


Assuntos
Química Click , Selênio , Química Click/métodos , Química Farmacêutica/métodos , Proteínas/química , Alcinos/química , Azidas/química , Reação de Cicloadição
10.
J Ovarian Res ; 17(1): 6, 2024 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-38184624

RESUMO

BACKGROUND: Polycystic ovarian syndrome (PCOS) is recognized as the most prevalent endocrine disorder among women of reproductive age. While the utilization of assisted reproductive technology (ART) has resulted in favorable outcomes for infertility treatment in PCOS patients, the inherent pathophysiological features of the condition give rise to complications and consequences during pregnancy and delivery for both the mother and offspring. This study was to assess the correlation between maternal PCOS and various pregnancy complications and neonatal outcomes undergone ART. METHODS: A systematic search was conducted on PubMed, EmBase, and the Cochrane Library to identify observational studies that investigated the association between PCOS and the risk of various pregnancy complications and neonatal outcomes, including gestational diabetes mellitus (GDM), hypertension in pregnancy (PIH), preeclampsia (PE), preterm birth, abortion, congenital malformations (CA), small for gestational age (SGA), large for gestational age (LGA), low birth weight (LBW), macrosomia, neonatal intensive care unit (NICU) admission and birth weight. Eligible studies were selected based on predetermined inclusion and exclusion criteria. The meta-analysis was conducted using Review Manager and Stata software, with odds ratios (ORs) or mean difference (MD), confidence intervals (CIs), and heterogeneity (I2) being calculated. The search was conducted up to March 2023. RESULTS: A total of 33 studies with a combined sample size of 92,810 participants were identified. The findings indicate that PCOS is significantly associated with an increased risk of GDM (OR 1.51, 95% CI:1.17-1.94), PIH (OR 1.72, 95% CI:1.25-2.39), PE (OR 2.12, 95% CI:1.49-3.02), preterm birth (OR 1.29, 95% CI:1.21-1.39), and LBW (OR 1.29, 95% CI:1.14-1.47). In subgroup analyses, the risks of GDM (OR 1.80, 95% CI:1.23-2.62) and abortion (OR 1.41, 95% CI:1.08-1.84) were elevated in fresh embryo transferred (ET) subgroup, whereas elevated risk of PE (OR 1.82, 95% CI:1.17-2.83) and preterm birth (OR 1.31, 95% CI:1.21-1.42) was identified in frozen ET subgroup. Whatever with or without hyperandrogenism, patients with PCOS had a higher risk in preterm birth (OR 1.69, 95% CI: 1.31-2.18; OR 1.24, 95% CI:1.02-1.50) and abortion (OR 1.38, 95% CI:1.12-1.71; OR 1.23, 95% CI:1.06-1.43). CONCLUSION: Our findings suggest that individuals with PCOS undergone ART are at a notably elevated risk for experiencing pregnancy complications and unfavorable neonatal outcomes. Nevertheless, to establish a definitive association between PCOS and pregnancy-related outcomes, it is necessary to conduct extensive prospective, blinded cohort studies and effectively control for confounding variables.


Assuntos
Síndrome do Ovário Policístico , Complicações na Gravidez , Feminino , Humanos , Recém-Nascido , Gravidez , Transferência Embrionária , Síndrome do Ovário Policístico/complicações , Nascimento Prematuro/epidemiologia , Nascimento Prematuro/etiologia , Estudos Prospectivos , Técnicas de Reprodução Assistida/efeitos adversos , Complicações na Gravidez/etiologia
11.
Biomed Pharmacother ; 168: 115659, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37864896

RESUMO

The anti-tumoral effects of metformin have been widely studied in several types of cancer, including thyroid cancer; however, the underlying molecular mechanisms remain poorly understood. As an oral hypoglycemic drug, metformin facilitates glucose catabolism and disrupts metabolic homeostasis. Metabolic reprogramming, particularly cellular glucose metabolism, is an important characteristic of malignant tumors. This study aimed to explore the therapeutic effects of metformin in thyroid cancer and the underlying metabolic mechanism. In the present study, it was shown that metformin reduced cell viability, invasion, migration, and EMT, and induced apoptosis and cell cycle G1 phase arrest in thyroid cancer. Transcriptome analysis demonstrated that the differentially expressed genes induced by metformin were involved in several signaling pathways including apoptosis singling pathways, TGF-ß signaling, and cell cycle regulation in human thyroid cancer cell lines. In addition, the helicase activity of the CDC45-MCM2-7-GINS complex and DNA replication related genes such as RPA2, RAD51, and PCNA were downregulated in metformin-treated thyroid cancer cells. Moreover, metabolomics analysis showed that metformin-induced significant alterations in metabolic pathways such as glutathione metabolism and polyamine synthesis. Integrative analysis of transcriptomes and metabolomics revealed that metformin suppressed glycolysis by downregulating the key glycolytic enzymes LDHA and PKM2 and upregulating IDH1 expression in thyroid cancer. Furthermore, the anti-tumor role of metformin in thyroid cancer in vivo was shown. Together these results show that metformin plays an anti-tumor role by inhibiting glycolysis and restraining DNA replication in thyroid cancer.


Assuntos
Metformina , Neoplasias da Glândula Tireoide , Humanos , Metformina/farmacologia , Transcriptoma , Linhagem Celular Tumoral , Glicólise , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Perfilação da Expressão Gênica , Replicação do DNA , Proliferação de Células
12.
Cell Signal ; 109: 110792, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37406787

RESUMO

OBJECTIVES: miR-142-3P is a tumor suppressor in various malignant cancers. However, the function of miR-142-3P in papillary thyroid carcinoma (PTC) remains to be elucidated. The aim of this study was to explore the function and mechanism of miR-142-3P in PTC. METHODS: Real Time Quantitative PCR (RT-qPCR) was used to assess the expression of miR-142-3P and Fibronectin 1 (FN1) in PTC. The correlation between FN1 and miR-142-3P expression was analyzed by Spearman's correlation analysis. Cell Counting Kit 8 (CCK8), 5-ethynyl-2'-deoxyuridine (EDU) assay, cell migration and invasion assay and wound healing measures evaluated the effect of miR-142-3P and FN1 on cell proliferation, migration and invasion. Dural Luciferase reported gene assay evaluated the interaction between miR-142-3P and 3' untranslated region (UTR) of FN1. The Epithelial-Mesenchymal-Transition (EMT) and apoptosis related marker genes were measured using western blot analysis (WB). RESULTS: miR-142-3P was significantly decreased in both PTC specimens and relevant cell lines. Functionally, miR-142-3P inhibited cell proliferation, migration, invasion and EMT, and induced the cell apoptosis in PTC. In addition, miR-142-3P bound directly with 3' UTR of FN1 and negatively regulated the expression of FN1 in PTC. FN1 expression is elevated in PTC, and its aberrant high correlated with declines in recurrence-free survival (RFS). Moreover, FN1 promoted cell proliferation, migration, invasion and EMT, induced cell apoptosis in PTC cells. Depletion of FN1 rescues the effect of miR-142-3P inhibitor on cell proliferation, invasion, apoptosis and EMT via inactivating Focal Adhesion Kinase (FAK)/Extracellular Signal-Regulated Kinase (ERK) / Phosphoinostide 3-kinase (P13K) signaling. CONCLUSION: miR-142-3P suppressed cell proliferation, migration, invasion and EMT through modulating FN1/FAK/ERK/PI3K signaling in PTC, suggesting it as a potential therapeutic target for PTC.


Assuntos
MicroRNAs , Neoplasias da Glândula Tireoide , Humanos , Câncer Papilífero da Tireoide/genética , Câncer Papilífero da Tireoide/metabolismo , MicroRNAs/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias da Glândula Tireoide/patologia , Fibronectinas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica
14.
Protein Cell ; 14(3): 202-216, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-36929036

RESUMO

Although the mTOR-4E-BP1 signaling pathway is implicated in aging and aging-related disorders, the role of 4E-BP1 in regulating human stem cell homeostasis remains largely unknown. Here, we report that the expression of 4E-BP1 decreases along with the senescence of human mesenchymal stem cells (hMSCs). Genetic inactivation of 4E-BP1 in hMSCs compromises mitochondrial respiration, increases mitochondrial reactive oxygen species (ROS) production, and accelerates cellular senescence. Mechanistically, the absence of 4E-BP1 destabilizes proteins in mitochondrial respiration complexes, especially several key subunits of complex III including UQCRC2. Ectopic expression of 4E-BP1 attenuates mitochondrial abnormalities and alleviates cellular senescence in 4E-BP1-deficient hMSCs as well as in physiologically aged hMSCs. These f indings together demonstrate that 4E-BP1 functions as a geroprotector to mitigate human stem cell senescence and maintain mitochondrial homeostasis, particularly for the mitochondrial respiration complex III, thus providing a new potential target to counteract human stem cell senescence.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ciclo Celular , Senescência Celular , Complexo III da Cadeia de Transporte de Elétrons , Células-Tronco Mesenquimais , Mitocôndrias , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/fisiologia , Homeostase , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Mitocôndrias/metabolismo , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Humanos , Células Cultivadas
15.
J Exp Bot ; 74(6): 2047-2066, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36622787

RESUMO

Grapevine downy mildew, caused by the oomycete Plasmopara viticola, is one of the most significant production challenges for the grape and wine industry. P. viticola injects a plethora of effectors into its host cells to disrupt immune processes, but the mechanisms by which these effectors act at the molecular level have not been well characterized. Herein, we show that a candidate P. viticola avirulence homolog (Avh) RxLR effector gene, designated PvAvh77, was strongly up-regulated during the initial stages of P. viticola infection in Vitis vinifera. Further experiments demonstrated that PvAvh77 could trigger non-specific cell death when expressed in the wild grapevine Vitis riparia and in tobacco (Nicotiana benthamiana and Nicotiana tabacum). In addition, a truncated form of PvAvh77, designated PvAvh77-M2, was more active in inducing cell death in N. benthamiana and V. riparia than full-length PvAvh77. Ectopic expression of PvAvh77 in V. vinifera 'Thompson Seedless' leaves neutralized host immunity and enhanced colonization by P. viticola, and the immune-inhibiting activity of PvAvh77 on susceptible Eurasian grapevine depended on its nuclear localization. Using a yeast signal sequence trap approach, we showed that the signal peptide of PvAvh77 is functional in yeast. Moreover, PvAvh77 with a signal peptide stimulated plant immune responses in the apoplast. Notably, application of exogenous purified PvAvh77-M2 effectively initiated defence responses in grapevine extracellularly, as evidenced by increased accumulation of salicylic acid and H2O2, and reduced infection of inoculated P. viticola. In summary, we identified a novel effector, PvAvh77, from P. viticola, which has the potential to serve as an inducer of plant immunity.


Assuntos
Oomicetos , Phytophthora infestans , Vitis , Saccharomyces cerevisiae , Peróxido de Hidrogênio/metabolismo , Doenças das Plantas , Nicotiana/genética , Vitis/genética , Vitis/metabolismo , Morte Celular , Sinais Direcionadores de Proteínas , Resistência à Doença
16.
Acta Pharmacol Sin ; 44(5): 999-1013, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36347996

RESUMO

Non-healing diabetic wounds (DW) are a serious clinical problem that remained poorly understood. We recently found that topical application of growth differentiation factor 11 (GDF11) accelerated skin wound healing in both Type 1 DM (T1DM) and genetically engineered Type 2 diabetic db/db (T2DM) mice. In the present study, we elucidated the cellular and molecular mechanisms underlying the action of GDF11 on healing of small skin wound. Single round-shape full-thickness wound of 5-mm diameter with muscle and bone exposed was made on mouse dorsum using a sterile punch biopsy 7 days following the onset of DM. Recombinant human GDF11 (rGDF11, 50 ng/mL, 10 µL) was topically applied onto the wound area twice a day until epidermal closure (maximum 14 days). Digital images of wound were obtained once a day from D0 to D14 post-wounding. We showed that topical application of GDF11 accelerated the healing of full-thickness skin wounds in both type 1 and type 2 diabetic mice, even after GDF8 (a muscle growth factor) had been silenced. At the cellular level, GDF11 significantly facilitated neovascularization to enhance regeneration of skin tissues by stimulating mobilization, migration and homing of endothelial progenitor cells (EPCs) to the wounded area. At the molecular level, GDF11 greatly increased HIF-1ɑ expression to enhance the activities of VEGF and SDF-1ɑ, thereby neovascularization. We found that endogenous GDF11 level was robustly decreased in skin tissue of diabetic wounds. The specific antibody against GDF11 or silence of GDF11 by siRNA in healthy mice mimicked the non-healing property of diabetic wound. Thus, we demonstrate that GDF11 promotes diabetic wound healing via stimulating endothelial progenitor cells mobilization and neovascularization mediated by HIF-1ɑ-VEGF/SDF-1ɑ pathway. Our results support the potential of GDF11 as a therapeutic agent for non-healing DW.


Assuntos
Diabetes Mellitus Experimental , Células Progenitoras Endoteliais , Fatores de Diferenciação de Crescimento , Cicatrização , Animais , Humanos , Camundongos , Proteínas Morfogenéticas Ósseas/metabolismo , Quimiocina CXCL12/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Fatores de Diferenciação de Crescimento/uso terapêutico , Fatores de Diferenciação de Crescimento/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo
17.
Front Public Health ; 10: 969030, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36203704

RESUMO

Background: For patients with locally advanced breast cancer (LABC), conventional TNM staging is not accurate in predicting survival outcomes. The aim of this study was to develop two accurate survival prediction models to guide clinical decision making. Methods: A retrospective analysis of 22,842 LABC patients was performed from 2010 to 2015 using the Surveillance, Epidemiology and End Results (SEER) database. An additional cohort of 200 patients from the Binzhou Medical University Hospital (BMUH) was analyzed. The least absolute shrinkage and selection operator (LASSO) regression was used to screen for variables. The identified variables were used to build a survival prediction model. The performance of the nomogram models was assessed based on the concordance index (C-index), calibration plot, receiver operating characteristic (ROC) curve, and decision curve analysis (DCA). Results: The LASSO analysis identified 9 variables in patients with LABC, including age, marital status, Grade, histological type, T-stage, N-stage, surgery, radiotherapy, and chemotherapy. In the training cohort, the C-index of the nomogram in predicting the overall survival (OS) was 0.767 [95% confidence intervals (95% CI): 0.751-0.775], cancer specific survival (CSS) was 0.765 (95% CI: 0.756-0.774). In the external validation cohort, the C-index of the nomogram in predicting the OS was 0.858 (95% CI: 0.812-0.904), the CSS was 0.866 (95% CI: 0.817-0.915). In the training cohort, the area under the receiver operator characteristics curve (AUC) values of the nomogram in prediction of the 1, 3, and 5-year OS were 0.836 (95% CI: 0.821-0.851), 0.769 (95% CI: 0.759-0.780), and 0.750 (95% CI: 0.738-0.762), respectively. The AUC values for prediction of the 1, 3, and 5-year CSS were 0.829 (95% CI: 0.811-0.847), 0.769 (95% CI: 0.757-0.780), and 0.745 (95% CI: 0.732-0.758), respectively. Results of the C-index, ROC curve, and DCA demonstrated that the nomogram was more accurate in predicting the OS and CSS of patients compared with conventional TNM staging. Conclusion: Two prediction models were developed and validated in this study which provided more accurate prediction of the OS and CSS in LABC patients than the TNM staging. The constructed models can be used for predicting survival outcomes and guide treatment plans for LABC patients.


Assuntos
Neoplasias da Mama , Nomogramas , Feminino , Humanos , Prognóstico , Estudos Retrospectivos , Programa de SEER
18.
J Cancer Res Ther ; 18(5): 1261-1267, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36204871

RESUMO

Context: Previous studies show that dose escalation for gross tumor volume (GTV) improves local control of esophageal cancer (EC). However, optimal boosting remains uncertain. Recently, functional imaging guidance to achieve dose escalation in high-risk areas of tumors has been proposed. Aims: This study evaluated the feasibility of dose escalation in tumor regions with high fluorodeoxyglucose (FDG) uptake using intensity-modulated radiotherapy (IMRT) and intensity-modulated proton therapy (IMPT). Settings and Design: GTVPET was defined as a high FDG uptake region with 50% SUVmax threshold for dose escalation. IMRT and IMPT plans were generated for three boosting modes: plan 50.4 (50.4 Gy in clinical target volume, CTV), plan 63 (50.4 Gy in CTV, 63 Gy in GTV), plan 70 (50.4 Gy in CTV, 63 Gy in GTV, and 70 Gy in GTVPET). Methods and Material: Eleven patients with squamous cell carcinoma were evaluated. Dose parameters for heart, lung, and spinal cord were compared based on the dose-volume histogram (DVH). Statistical Analysis Used: Paired t-test was performed on the doses to organs-at-risk (OARs) among plan 50.4, plan 63, and plan 70 for IMRT and IMPT. Results: Dosimetric parameters for IMRT for heart, lung, and spinal cord increased significantly for plan 63 and some parameters even exceeded dose limits for OARs. Further dose escalation in GTV-PET did not increase dosimetric parameters significantly. Most dosimetric parameters of OARs in IMPT exhibited no statistical change compared with plan 50.4, and doses to OARs were far less than dose constraints. Conclusions: Dose escalation by IMRT may lead to increased risk of radiation-related injury. Further dose escalation in high FDG uptake regions did not increase doses to OARs. This dose escalation is ideal for achieving better outcomes for EC treatment.


Assuntos
Neoplasias Esofágicas , Terapia com Prótons , Lesões por Radiação , Radioterapia de Intensidade Modulada , Neoplasias Esofágicas/diagnóstico por imagem , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/radioterapia , Estudos de Viabilidade , Fluordesoxiglucose F18 , Humanos , Órgãos em Risco/patologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador/métodos , Radioterapia de Intensidade Modulada/métodos
20.
ACS Appl Mater Interfaces ; 14(31): 35366-35375, 2022 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-35914110

RESUMO

Gating systems have been extensively researched in energy harvesting, lab-on-chip applications, and so forth. However, the controlled drug delivery system with artificial hydrogel-based porous gating systems (HPGSs) is rarely reported. Herein, a biomimetic HPGS with a pH-responsive hydrogel as the valve and polydimethylsiloxane as the frame is fabricated by in situ femtosecond laser microdrilling and subsequent ultraviolet exposure. The proposed HPGS loaded with doxorubicin hydrochloride (DOX) is stable under physiological conditions, has a low drug leakage rate, and can achieve sustained drug release in a low pH environment. The experimental results show that the drug release is mainly controlled by non-Fickian diffusion, which renders the dynamic speed control of molecular transport possible. Moreover, the HPGS can also be prepared into an antitumor microcapsule. The results of in vitro cell experiments demonstrate that DOX@HPGS can release drugs and achieve terrific therapeutic efficacy in the elimination of HeLa cells in the acidic environments around tumor cells. This functional HPGS is envisioned to be an ideal pH-response carrier for sustained drug release treatment of digestive diseases such as inflammatory bowel disease and gastrointestinal cancer.


Assuntos
Doxorrubicina , Hidrogéis , Doxorrubicina/química , Doxorrubicina/farmacologia , Portadores de Fármacos , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Células HeLa , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Concentração de Íons de Hidrogênio , Porosidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA