Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Biochem Genet ; 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38965134

RESUMO

Laryngeal cancer is a common malignancy of the larynx with a generally poor prognosis. This study systematically assessed the functional role of lncRNA BBOX1-AS1 in laryngeal carcinoma progression and associated molecular regulatory mechanisms. The proliferation, migration, and invasion of laryngeal carcinoma cells were detected by Cell Counting Kit-8, wound healing, clonal formation, and transwell assays. In addition, the interaction between BBOX1-AS1, Serine/Arginine Splicing Factor 1 (SRSF1), and Ephrin-B2 (EFNB2) mRNA was examined employing RNA immunoprecipitation and RNA pull-down experiments. Furthermore, western blotting, and RT-qPCR assays were adopted to detect the expression levels of BBOX1-AS1, SRSF1, and EFNB2. The impact of BBOX1-AS1 and SRSF1 on EFNB2 mRNA stability was examined using the RNA stability assay. BBOX1-AS1 was highly expressed in human laryngeal carcinoma tissues and cell lines. BBOX1-AS1 knockdown suppressed the growth, proliferation, migration, and invasion of laryngeal carcinoma cells. BBOX1-AS1 maintained the stability of EFNB2 mRNA in laryngeal carcinoma cells by recruiting SRSF1. EFNB2 knockdown inhibited the growth and metastatic function of laryngeal carcinoma cells in vitro. EFNB2 overexpression reversed the influence of BBOX1-AS1 knockdown on laryngeal cancer tumorigenesis. BBOX1-AS1 maintained EFNB2 mRNA stability by recruiting SRSF1, thereby aggravating laryngeal carcinoma malignant phenotypes. BBOX1-AS1 might be a new theoretical target for the treatment of laryngeal carcinoma.

2.
Theranostics ; 14(8): 3385-3403, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38855175

RESUMO

Rationale: It has been emergingly recognized that apoptosis generates plenty of heterogeneous apoptotic vesicles (apoVs), which play a pivotal role in the maintenance of organ and tissue homeostasis. However, it is unknown whether apoVs influence postnatal ovarian folliculogenesis. Methods: Apoptotic pathway deficient mice including Fas mutant (Fasmut ) and Fas ligand mutant (FasLmut ) mice were used with apoV replenishment to evaluate the biological function of apoVs during ovarian folliculogenesis. Ovarian function was characterized by morphological analysis, biochemical examination and cellular assays. Mechanistical studies were assessed by combinations of transcriptomic and proteomic analysis as well as molecular assays. CYP17A1-Cre; Axin1fl /fl mice was established to verify the role of WNT signaling during ovarian folliculogenesis. Polycystic ovarian syndrome (PCOS) mice and 15-month-old mice were used with apoV replenishment to further validate the therapeutic effects of apoVs based on WNT signaling regulation. Results: We show that systemic administration of mesenchymal stem cell (MSC)-derived apoptotic vesicles (MSC-apoVs) can ameliorate impaired ovarian folliculogenesis, PCOS phenotype, and reduced birth rate in Fasmut and FasLmut mice. Mechanistically, transcriptome analysis results revealed that MSC-apoVs downregulated a number of aberrant gene expression in Fasmut mice, which were enriched by kyoto encyclopedia of genes and genomes (KEGG) pathway analysis in WNT signaling and sex hormone biosynthesis. Furthermore, we found that apoptotic deficiency resulted in aberrant WNT/ß-catenin activation in theca and mural granulosa cells, leading to responsive action of dickkopf1 (DKK1) in the cumulus cell and oocyte zone, which downregulated WNT/ß-catenin expression in oocytes and, therefore, impaired ovarian folliculogenesis via NPPC/cGMP/PDE3A/cAMP cascade. When WNT/ß-catenin was specially activated in theca cells of CYP17A1-Cre; Axin1fl /fl mice, the same ovarian impairment phenotypes observed in apoptosis-deficient mice were established, confirming that aberrant activation of WNT/ß-catenin in theca cells caused the impairment of ovarian folliculogenesis. We firstly revealed that apoVs delivered WNT membrane receptor inhibitor protein RNF43 to ovarian theca cells to balance follicle homeostasis through vesicle-cell membrane integration. Systemically infused RNF43-apoVs down-regulated aberrantly activated WNT/ß-catenin signaling in theca cells, contributing to ovarian functional maintenance. Since aging mice have down-regulated expression of WNT/ß-catenin in oocytes, we used MSC-apoVs to treat 15-month-old mice and found that MSC-apoVs effectively ameliorated the ovarian function and fertility capacity of these aging mice through rescuing WNT/ß-catenin expression in oocytes. Conclusion: Our studies reveal a previously unknown association between apoVs and ovarian folliculogenesis and suggest an apoV-based therapeutic approach to improve oocyte function and birth rates in PCOS and aging.


Assuntos
Apoptose , Células-Tronco Mesenquimais , Folículo Ovariano , Ovário , Síndrome do Ovário Policístico , Via de Sinalização Wnt , Animais , Feminino , Síndrome do Ovário Policístico/metabolismo , Camundongos , Células-Tronco Mesenquimais/metabolismo , Folículo Ovariano/metabolismo , Ovário/metabolismo , Modelos Animais de Doenças , Envelhecimento/fisiologia , Proteína Ligante Fas/metabolismo , Proteína Ligante Fas/genética
3.
Oncol Lett ; 27(6): 259, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38646492

RESUMO

Lung cancer is the most common cancer in the world due to its high incidence and recurrence. Genetic instability is one of the main factors leading to its occurrence, development and poor prognosis. Decreased xeroderma pigmentosum group C (XPC) expression notably enhances the stem cell properties of lung cancer cells and increases their proliferation and migration. Additionally, patients with lung cancer and low XPC expression had a poor prognosis. The purpose of the present study was to analyze the effect of XPC and IFN-γ on the clinical prognosis of patients with non-small cell lung cancer (NSCLC). Lung adenocarcinoma specimens were collected from a total of 140 patients with NSCLC. Additionally, from these 140 patients, 48 paracarcinoma tissue specimens were also collected, which were later used to construct tissue microarrays. The expression of XPC and IFN-γ in cancer tissues and in paraneoplastic tissues was detected using immunohistochemistry. The prognosis and overall survival of patients were determined through telephone follow-up. The results showed a positive correlation between expression of XPC and IFN-γ in NSCLC. Additionally, high expression of both markers was associated with a favorable prognosis in patients with NSCLC. The aforementioned findings suggest that the expression of XPC and IFN-γ has prognostic value in clinical practice and is expected to become a marker for clinical application.

4.
J Cell Commun Signal ; 18(1): e12016, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38545258

RESUMO

Long noncoding RNAs (lncRNAs) are involved in regulatory processes in laryngeal squamous cell carcinoma (LSCC) at posttranscriptional epigenetic modification level. Yet, the function and underlying mechanism behind lncRNA AC004943.2 in LSCC is still obscure. Therefore, the potential role of AC004943.2 in LSCC progression was investigated. The expression of gene or protein was tested by real-time quantitative polymerase chain reaction and western blot. MTT, colony formation, wound healing, and transwell experiments were applied to detect LSCC cell viability, proliferation, migration and invasion, respectively. The interaction among AC004943.2, miR-135a-5p, and protein tyrosine kinase 2 (PTK2) were analyzed by bioinformatics prediction and luciferase assay. AC004943.2 was highly expressed in LSCC cells compared with normal human bronchial epithelial cells, while miR-135a-5p was lowly expressed. AC004943.2 knockdown or miR-135a-5p overexpression inhibited LSCC cell viability, proliferation, migration and invasion. Mechanistically, AC004943.2 increased PTK2 expression in LSCC cells by sponging miR-135a-5p. Furthermore, miR-135a-5p knockdown inverted the inhibitory effect of AC004943.2 silencing on LSCC cell malignant behaviors. MiR-135a-5p upregulation attenuated the PTK2/PI3K pathway to inhibit progression of LSCC. AC004943.2 facilitated the cancerous phenotypes of LSCC cells by activating the PTK2/PI3K pathway through targeting miR-135a-5p, which furnished a therapeutic candidate for LSCC treatment.

5.
Theranostics ; 13(10): 3131-3148, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37351158

RESUMO

Background: Aged women and premature ovarian insufficiency (POI) patients have residual dormant primordial follicles that are hard to be activated through a physiological process. However, there are no effective and safe drugs to help them. Methods: We used the in vitro culture model of newborn mouse ovaries to identify the drugs that promote primordial follicle activation and study its mechanisms. It was verified by in vivo injection model of newborn mice and in vitro culture model of human ovarian tissue. In addition, we used the aged mice as a low infertility model to verify the effects of primordial follicle activation, and fertility by drugs. Results: Eleven metallic compounds activated mouse primordial follicles, and the five most effective compounds were selected for further study. Thapsigargin (TG), CrCl3, MnCl2, FeCl3 and ZnSO4 increased the levels of the glycolysis-related proteins (glucose transporter type 4, GLUT4; hexokinase 1, HK1; pyruvate kinase M2, PKM2; phosphofructokinase, liver type, PFKL), phosphorylated mammalian target of rapamycin (p-mTOR) in cultured mouse ovaries. The compound-promoted p-mTOR levels could be completely blocked by 2-DG (the inhibitor of glycolysis). The compounds also increased the levels of phosphorylated protein kinase B (p-Akt). TG-, CrCl3- and FeCl3-promoted p-Akt levels, but not MnCl2- and ZnSO4- promoted p-Akt levels, could be completely blocked by ISCK03 (the inhibitor of proto-oncogenic receptor tyrosine kinase, KIT). The injection of newborn mice with the compounds also activated primordial follicles and increased the levels of the glycolysis-related proteins, p-mTOR, and p-Akt. The oral administration of the compounds in adolescent and aged mice promoted primordial follicle activation, and had no obvious side effect. Importantly, ZnSO4 also increased ovulated oocytes, oocyte quality and offspring in aged mice. Furthermore, the compounds promoted human primordial follicle activation and increased the levels of the glycolysis-related proteins, p-mTOR, and p-Akt. Conclusion: The metallic compounds activate primordial follicles through the glycolysis-dependent mTOR pathway and/or the PI3K/Akt pathway, and the oral administration of ZnSO4 enhances fertility in aged mice. We suggest that these metallic compounds may be oral drugs to ameliorate fertility deficits in aged women and POI patients.


Assuntos
Insuficiência Ovariana Primária , Proteínas Proto-Oncogênicas c-akt , Animais , Humanos , Feminino , Camundongos , Adolescente , Idoso , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fosfatidilinositol 3-Quinases/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Insuficiência Ovariana Primária/tratamento farmacológico , Fertilidade , Mamíferos/metabolismo
6.
Reprod Biol Endocrinol ; 21(1): 36, 2023 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-37041518

RESUMO

BACKGROUND: Retinoic acid (RA) plays important role in the maintenance and differentiation of the Müllerian ducts during the embryonic stage via RA receptors (RARs). However, the function and mechanism of RA-RAR signaling in the vaginal opening are unknown. METHOD: We used the Rarα knockout mouse model and the wild-type ovariectomized mouse models with subcutaneous injection of RA (2.5 mg/kg) or E2 (0.1 µg/kg) to study the role and mechanism of RA-RAR signaling on the vaginal opening. The effects of Rarα deletion on Ctnnb1 mRNA levels and cell apoptosis in the vaginas were analyzed by real-time PCR and immunofluorescence, respectively. The effects of RA on the expression of ß-catenin and apoptosis in the vaginas were analyzed by real-time PCR and western blotting. The effects of E2 on RA signaling molecules were analyzed by real-time PCR and western blotting. RESULTS: RA signaling molecules were expressed in vaginal epithelial cells, and the mRNA and/or protein levels of RALDH2, RALDH3, RARα and RARγ reached a peak at the time of vaginal opening. The deletion of Rarα resulted in 25.0% of females infertility due to vaginal closure, in which the mRNA (Ctnnb1, Bak and Bax) and protein (Cleaved Caspase-3) levels were significantly decreased, and Bcl2 mRNA levels were significantly increased in the vaginas. The percentage of vaginal epithelium with TUNEL- and Cleaved Caspase-3-positive signals were also significantly decreased in Rarα-/- females with vaginal closure. Furthermore, RA supplementation of ovariectomized wild-type (WT) females significantly increased the expression of ß-catenin, active ß-catenin, BAK and BAX, and significantly decreased BCL2 expression in the vaginas. Thus, the deletion of Rarα prevents vaginal opening by reducing the vaginal ß-catenin expression and epithelial cell apoptosis. The deletion of Rarα also resulted in significant decreases in serum estradiol (E2) and vagina Raldh2/3 mRNA levels. E2 supplementation of ovariectomized WT females significantly increased the expression of RA signaling molecules in the vaginas, suggesting that the up-regulation of RA signaling molecules in the vaginas is dependent on E2 stimulation. CONCLUSION: Taken together, we propose that RA-RAR signaling in the vaginas promotes vaginal opening through increasing ß-catenin expression and vaginal epithelial cell apoptosis.


Assuntos
Tretinoína , beta Catenina , Feminino , Camundongos , Animais , Tretinoína/farmacologia , Caspase 3/metabolismo , beta Catenina/metabolismo , Proteína X Associada a bcl-2 , Receptor alfa de Ácido Retinoico/metabolismo , Células Epiteliais/metabolismo , Vagina , RNA Mensageiro/metabolismo , Apoptose , Aldeído Oxirredutases/metabolismo
7.
Kaohsiung J Med Sci ; 39(4): 354-363, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36919585

RESUMO

Laryngeal cancer is a usual malignant tumor of the head and neck. The role and mechanism of deubiquitinase USP21 in laryngeal cancer are still unclear. We aimed to explore whether USP21 affected laryngeal cancer progress through deubiquitinating AURKA. USP21 and AURKA levels were evaluated by qRT-PCR and Western blot. Kaplan-Meier analysis was conducted by survival package. MTT was performed to detect cell proliferation. The wound healing assay was applied to evaluate cell migration. Transwell was used to measure cell invasion. Co-IP and GST-pull down determined the interaction between USP21 and AURKA. In addition, AURKA ubiquitination levels were analyzed. USP21 was signally elevated in laryngeal cancer tissues and cells. USP21 level in clinical stages III-IV was higher than that in clinical stages I-II, and high levels of USP21 were highly correlated with poor prognosis in laryngeal cancer. USP21 inhibition suppressed AMC-HN-8 and TU686 cell proliferation, migration and invasion. Co-IP and GST-pull down confirmed the interaction between USP21 and AURKA. Knockdown of USP21 markedly increased the ubiquitination level of AURKA, and USP21 restored AURKA activity through deubiquitination. In addition, overexpression of AURKA reversed the effects of USP21 knockdown on cell growth, migration, and invasion. USP21 stabilized AURKA through deubiquitination to promote laryngeal cancer progression.


Assuntos
Neoplasias Laríngeas , Humanos , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patologia , Aurora Quinase A/genética , Proliferação de Células/genética , Linhagem Celular Tumoral , Ubiquitinação , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo
8.
Pathol Int ; 73(3): 109-119, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36285444

RESUMO

Laryngeal cancer (LC) is a rare and challenging clinical problem. Our aim was to investigate the mechanism of salt-like transcription factor 4 (SALL4) in LC. LC tissue and paracancerous tissue were collected. Relative mRNA or protein levels were measured by quantitative real-time polymerase chain reaction or Western blot. MTT, wound healing, and transwell assay were performed to evaluate cell proliferation, migration and invasion. The binding relationship between SALL4 and USP21 promoter was verified by dual-luciferase assay and ChIP. Co-IP and glutathione-S-transferase (GST)-pull down were performed to measure the protein interaction between USP21 and YY1. Additionally, YY1 ubiquitination level was analyzed. It was found that SALL4 mRNA and SALL4 protein levels were elevated in LC clinical tissues and various LC cells. Knockdown of SALL4 inhibited epithelial-mesenchymal transition (EMT) of LC cells. USP21 was transcriptionally activated by SALL4. Co-IP and GST-pull down confirmed USP21 interacted with YY1. USP21 protected YY1 from degradation through deubiquitination. Furthermore, overexpression of USP21 reversed the effect of knockdown of SALL4 on YY1 and EMT in LC cells. In general, SALL4 facilitated EMT of LC cells through modulating USP21/YY1 axis.


Assuntos
Neoplasias Laríngeas , Fatores de Transcrição , Humanos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Laríngeas/genética , RNA Mensageiro , Fator de Transcrição 4/genética , Fator de Transcrição 4/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Ubiquitina Tiolesterase/genética , Proteases Específicas de Ubiquitina/genética , Proteases Específicas de Ubiquitina/metabolismo , Yin-Yang
9.
Cell Death Dis ; 13(11): 963, 2022 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-36396932

RESUMO

Germ cell division and differentiation require intimate contact and interaction with the surrounding somatic cells. Luteinizing hormone (LH) triggers epidermal growth factor (EGF)-like growth factors to promote oocyte maturation and developmental competence by activating EGF receptor (EGFR) in somatic cells. Here, we showed that LH-EGFR signaling-activated sphingosine kinases (SphK) in somatic cells. The activation of EGFR by EGF increased S1P and calcium levels in cumulus-oocyte complexes (COCs), and decreased the binding affinity of natriuretic peptide receptor 2 (NPR2) for natriuretic peptide type C (NPPC) to release the cGMP-mediated meiotic arrest. These functions of EGF were blocked by the SphK inhibitor SKI-II, which could be reversed by the addition of S1P. S1P also activated the Akt/mTOR cascade reaction in oocytes and promoted targeting protein for Xklp2 (TPX2) accumulation and oocyte developmental competence. Specifically depleting Sphk1/2 in somatic cells reduced S1P levels and impaired oocyte meiotic maturation and developmental competence, resulting in complete female infertility. Collectively, SphK-produced S1P in somatic cells serves as a functional transmitter of LH-EGFR signaling from somatic cells to oocytes: acting on somatic cells to induce oocyte meiotic maturation, and acting on oocytes to improve oocyte developmental competence.


Assuntos
Fator de Crescimento Epidérmico , Oogênese , Animais , Feminino , Camundongos , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Peptídeos Natriuréticos/metabolismo , Oócitos/metabolismo , Hormônio Luteinizante/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)
10.
Mol Reprod Dev ; 89(11): 554-562, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36128893

RESUMO

Cumulus expansion is necessary for the release of a fertilizable oocyte from the ovary, which is critical for the normal fertilization of mammals. Cumulus expansion requires cooperation between epidermal growth factor (EGF)-like growth factors and oocyte paracrine factors. Growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) are well-known paracrine factors secreted by oocytes. In addition, transforming growth factor-ß2 (TGFB2) was primarily expressed in oocytes and its membrane receptors type 1 receptor (TGFBR1) and type 2 receptor (TGFBR2) were located in cumulus cells. In our present study, TGFB2 induced expansion of oocytectomized (OOX) complexes and increased the expression of expansion-related genes in the presence of EGF, suggesting that TGFB2 enables cumulus expansion. Inhibition of TGF-ß signaling with SD208 blocked TGFB2-promoted cumulus expansion. Furthermore, in the culture of OOX complexes from mice of Tgfbr2-specific depletion in granulosa cells, TGFB2-promoted cumulus expansion and the expression of expansion-related genes were impaired. These results suggest that TGFB2 could induce cumulus expansion through TGFBR-SMAD2/3 signaling. Tgfb2-specific depletion in oocytes using Zp3-Cre mice had no effect on cumulus expansion in vivo, possibly due to the compensatory effect of other cumulus expansion-enabling factors. Taken together, TGFB2 is involved in expansion-related gene expression and consequent cumulus expansion.


Assuntos
Células do Cúmulo , Fator de Crescimento Epidérmico , Fator de Crescimento Transformador beta2 , Animais , Feminino , Camundongos , Proteína Morfogenética Óssea 15/metabolismo , Células do Cúmulo/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Hormônio Foliculoestimulante/farmacologia , Fator 9 de Diferenciação de Crescimento/genética , Fator 9 de Diferenciação de Crescimento/metabolismo , Oócitos/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Fator de Crescimento Transformador beta2/metabolismo
11.
Mol Med Rep ; 26(3)2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35795988

RESUMO

Acidic nuclear phosphoprotein 32 family member E (ANP32E) is a histone chaperone that removes H2A.Z from chromatin. ANP32E is implicated in numerous cellular processes, including cell proliferation, apoptosis and cell differentiation. Increasing evidence suggests that dysregulation of ANP32E expression is strongly associated with carcinogenesis. However, the relationship between ANP32E in the development of gastric cancer (GC) is unknown. The present study aimed to explore the potential role of ANP32E in the development of GC using gain­of­function, loss­of­function, CCK­8, colony formation, apoptosis, reverse transcription­quantitative PCR, immunoblotting and luciferase reporter assay. The results of the present study demonstrated that ANP32E expression levels were significantly increased in GC tissues. ANP32E knockdown markedly inhibited GC cell proliferation and colony formation and significantly induced GC cell apoptosis, whereas overexpression of ANP32E significantly induced GC cell malignancy. Furthermore, the results demonstrated that there was a positive association between ANP32E and NUF2 component of NDC80 kinetochore complex (NUF2) expression levels. By assessing NUF2 expression levels, it was demonstrated that ANP32E promoted tumor cell proliferation and inhibited cell apoptosis by increasing NUF2 expression levels in GC cell lines. In conclusion, the present study indicated that ANP32E may function as an efficient oncogene, which promotes tumorigenesis of GC cells by inducing NUF2 expression.


Assuntos
Proteínas de Ciclo Celular , Chaperonas Moleculares , Neoplasias Gástricas , Carcinogênese/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/genética , Histonas/genética , Histonas/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Ativação Transcricional , Regulação para Cima
12.
Biol Reprod ; 107(4): 1059-1071, 2022 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-35871551

RESUMO

In mammals, dormant primordial follicles represent the ovarian reserve throughout reproductive life. In vitro activation of dormant primordial follicles has been used to treat patients with premature ovarian insufficiency (POI). However, there remains a lack of effective strategies to stimulate follicle activation in vivo. In this study, we used an in vitro ovarian culture system and intraperitoneal injection to study the effect of lithium treatment on primordial follicle activation. Lithium increased the number of growing follicles in cultured mouse ovaries and promoted pre-granulosa cell proliferation. Furthermore, lithium significantly increased the levels of phosphorylated protein kinase B (Akt) and the number of oocytes with forkhead Box O3a (FOXO3a) nuclear export. Inhibition of the phosphatidylinositol 3 kinase (PI3K)/Akt pathway by LY294002 reversed lithium-promoted mouse primordial follicle activation. These results suggest that lithium promotes mouse primordial follicle activation by the PI3K/Akt signaling. Lithium also promoted primordial follicle activation and increased the levels of p-Akt in mouse ovaries in vivo and in human ovarian tissue cultured in vitro. Taken together, lithium promotes primordial follicle activation in mice and humans by the PI3K/Akt signaling. Lithium might be a potential oral drug for treating infertility in POI patients with residual dormant primordial follicles.


Assuntos
Insuficiência Ovariana Primária , Proteínas Proto-Oncogênicas c-akt , Animais , Feminino , Humanos , Lítio/metabolismo , Lítio/farmacologia , Compostos de Lítio/metabolismo , Compostos de Lítio/farmacologia , Mamíferos/metabolismo , Camundongos , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
13.
Cell Death Dis ; 13(1): 87, 2022 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-35087042

RESUMO

In mammals, nonrenewable primordial follicles are activated in an orderly manner to maintain the longevity of reproductive life. Mammalian target of rapamycin (mTOR)-KIT ligand (KITL) signaling in pre-granulosa cells and phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt)-forkhead Box O3a (FOXO3a) signaling in oocytes are important for primordial follicle activation. The activation process is accompanied by the enhancement of energy metabolism, but the causal relationship is unclear. In the present study, the levels of glycolysis-related proteins GLUT4, HK1, PFKL, and PKM2 were significantly increased in granulosa cells but were decreased in oocytes during the mouse primordial-to-primary follicle transition. Both short-term pyruvate deprivation in vitro and acute fasting in vivo increased the glycolysis-related gene and protein levels, decreased AMPK activity, and increased mTOR activity in mouse ovaries. The downstream pathways Akt and FOXO3a were phosphorylated, resulting in mouse primordial follicle activation. The blockade of glycolysis by 2-deoxyglucose (2-DG), but not the blockade of the communication network between pre-granulosa cells and oocyte by KIT inhibitor ISCK03, decreased short-term pyruvate deprivation-promoted mTOR activity. Glycolysis was also increased in human granulosa cells during the primordial-to-primary follicle transition, and short-term pyruvate deprivation promoted the activation of human primordial follicles by increasing the glycolysis-related protein levels and mTOR activity in ovarian tissues. Taken together, the enhanced glycolysis in granulosa cells promotes the activation of primordial follicles through mTOR signaling. These findings provide new insight into the relationship between glycolytic disorders and POI/PCOS.


Assuntos
Células da Granulosa , Serina-Treonina Quinases TOR , Animais , Feminino , Glicólise , Células da Granulosa/metabolismo , Mamíferos , Camundongos , Oócitos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácido Pirúvico/metabolismo , Serina-Treonina Quinases TOR/metabolismo
14.
Chemosphere ; 288(Pt 1): 132490, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34624347

RESUMO

It is of great significance to develop novel membranes with dual-function of simultaneously separating oil/water emulsion and degrading the contained water-miscible toxic organic components. To meet this requirement, a dual-functional Ni nanoparticles (NPs)@Ag/C-carbon nanotubes (CNTs) composite membrane was fabricated via electroless nickel plating strategy in this study. The as-prepared composite membrane possessed superhydrophilicity with water contact angle of 0° and splendid underwater oleophobic property with oil contact angle of 142°. When the membrane was applied for separation of surfactant stabilized oil-in-water emulsion, high permeate flux (about 97 L m-2·h-1 under gravity), oil rejection (about 98.8%) and antifouling property were achieved. Benefitting from the NiNPs@Ag/C-CNTs layer on membrane surface, the composite membrane exhibited high catalytic degradation activity for water-miscible toxic organic pollutant (4-nitrophenol) with addition of NaBH4 in a flow-through mode. Meanwhile, the NiNPs@Ag/C-CNTs composite membrane possessed excellent durability, which was verified by the good structural integrity even under ultrasonic treatment. The cost-efficiency, high separation and degradation performance of the prepared membrane suggested its great potential for treatment of oily wastewater.


Assuntos
Nanotubos de Carbono , Purificação da Água , Emulsões , Membranas Artificiais , Águas Residuárias
15.
Obes Surg ; 30(1): 279-289, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31605365

RESUMO

BACKGROUND: Duodenal-jejunal bypass (DJB) can dramatically improve type 2 diabetes independent of weight loss and food restriction. Increasing evidence has demonstrated that brain insulin signaling plays an important role in the pathophysiology of type 2 diabetes. This study explores whether the antidiabetic effect of DJB is involved in brain insulin signaling activation and brain glucose utilization. METHODS: A diabetic rat model was established by high-fat and high-glucose diet. DJB or sham surgery was performed in diabetic rats. 18F-FDG PET scanning was used to detect glucose uptake in different organs, particularly in the brain. The levels of glucose transporters, glucose utilization-related proteins (HK1 and PFK2), insulin, and insulin signaling pathway-related proteins (InsR, IRS1/2, PI3K, and p-Akt) in the brain tissues were evaluated and analyzed. RESULTS: The results showed that DJB significantly improved basal glycemic parameters and reversed the decreasing glucose uptake in the brains of type 2 diabetic rats. DJB significantly increased not only the expression levels of brain insulin, IRS1/2, PI3K, and p-Akt but also the levels of the glucose utilization enzymes HK1 and PFK2 in the brain. CONCLUSION: These results indicate that enhanced brain insulin signaling transduction and brain glucose utilization play important roles in the antidiabetic effect of DJB.


Assuntos
Encéfalo/metabolismo , Diabetes Mellitus Tipo 2/cirurgia , Duodeno/cirurgia , Derivação Gástrica/métodos , Glucose/metabolismo , Insulina/metabolismo , Jejuno/cirurgia , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Duodeno/patologia , Resistência à Insulina/fisiologia , Jejuno/patologia , Fígado/metabolismo , Masculino , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Resultado do Tratamento , Redução de Peso
16.
Mol Reprod Dev ; 86(12): 1887-1900, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31638299

RESUMO

The release of a fertilizable oocyte from the ovary is dependent upon the expansion of the cumulus cells. The expansion requires cooperation between epidermal growth factor (EGF) family peptide-activated mitogen-activated protein kinase (MAPK)3/1 and oocyte paracrine factor-activated-Sma- and Mad-related protein (SMAD)2/3 signaling in cumulus cells. However, the mechanism underlying (MAPK)3/1 signaling is unclear. In the present study, the EGF-activation of EGF receptor (EGFR) induced cyclic adenosine 3',5'-monophosphate (cAMP) response element-binding protein (CREB) phosphorylation in cumulus cells, and the interruption of CREB functional complex formation by naphthol AS-E phosphate (KG-501) completely blocked the EGF-stimulated expansion-related gene expression. EGF-stimulated phosphorylation of CREB was completely inhibited by MAPK3/1 inhibitor U0126, suggesting that EGF-activated MAPK3/1 results in the activation of CREB for cumulus expansion. Also, the role of EGF-stimulated calcium signaling was studied. Calcium-elevating reagents ionomycin and sphingosine-1-phosphate mimicked, but calcium chelators bis-(o'aminophenoxy)-ethane-N,N,N,N-tetraacetic acid, tetra(acetoxymethyl)-ester, and 8-(N,N-diethylamino)-octyl-3,4,5-trimethoxybenzoate abolished the activity of EGF on CREB phosphorylation, cumulus expansion, and expansion-related gene expression. Furthermore, EGF-induced cumulus expansion was inhibited by calmodulin (CaM)-dependent protein kinase II (CaMKII) inhibitors, KN-93 and autocamtide-2-related inhibitory peptide. However, the inhibition of SMAD2/3 activity by removal of oocyte from cumulus-oocyte complexes did not affect the EGF-induced CREB phosphorylation, indicating that EGF-activated CREB is independent of oocyte-activated SMAD2/3 signaling. Therefore, EGF-induced CREB activity by MAPK3/1 and Ca2+ /CaMKII signaling pathways promotes the expansion-related gene expression and consequent cumulus expansion.


Assuntos
Células do Cúmulo/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Sistema de Sinalização das MAP Quinases , Animais , Células do Cúmulo/citologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo
17.
Medicine (Baltimore) ; 97(28): e11370, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29995774

RESUMO

This study retrospectively investigated the effect of neuromuscular electrical stimulation (NMES) for fatigue management in patients with advanced laryngeal cancer (ALC) receiving chemoradiotherapy.A total of 60 eligible patients with ALC receiving chemoradiotherapy were included. These patients were assigned equally to a treatment group and a control group. Patients in the treatment group received NMES therapy and were treated for a total of 8 weeks, while the patients in the control group did not receive NMES therapy. The primary outcome was fatigue, measured by the multidimensional fatigue inventory (MFI). The secondary outcomes included anxiety and depression, measured by the Hospital Anxiety and Depression Scale (HADS), and sleep quality, measured by the Pittsburgh Sleep Quality Index (PSQI). All outcomes were evaluated before and after 8-week NMES treatmentAfter 8-week NMES treatment, the patients in the treatment group did not exert better effect than patients in the control group in fatigue relief, measured by the MFI score, anxiety and depression decrease, assessed by HADS, and sleep quality improvement, evaluated by PSQI.The results of this study demonstrate that NMES may not benefit for fatigue relief in patients with ALC receiving chemoradiotherapy. Future studies should still focus on this topic and warrant these results.


Assuntos
Carcinoma de Células Escamosas/terapia , Quimiorradioterapia/efeitos adversos , Terapia por Estimulação Elétrica/métodos , Fadiga/terapia , Neoplasias Laríngeas/terapia , Ansiedade/etiologia , Ansiedade/prevenção & controle , Carcinoma de Células Escamosas/complicações , Carcinoma de Células Escamosas/psicologia , Depressão/etiologia , Depressão/prevenção & controle , Fadiga/etiologia , Feminino , Humanos , Neoplasias Laríngeas/complicações , Neoplasias Laríngeas/psicologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Transtornos do Sono-Vigília/etiologia , Transtornos do Sono-Vigília/prevenção & controle
18.
Medicine (Baltimore) ; 97(29): e11545, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30024547

RESUMO

This retrospective study explored the quality of life (QoL) in Chinese patients with laryngeal cancer (LC) after radiotherapy.Fifty-nine eligible patients with Tis-T4 LC were included in this retrospective study. All patients received radiotherapy. Outcomes were measured by the core measure Questionnaire-C30 (QLQ-C30), and the disease-specific Head & Neck cancer module (QLQ-H&N35). All outcomes were assessed before and 3 months after the radiotherapy.Three months after the radiotherapy, all items of QLQ-C30 and QLQ-H&N35 scales changed significantly (P < .05), except the social functioning (P = .09), role activities (P = .81), and global (P = .12) in QLQ-C30 scale and social contacts (P = 1.00), teeth problems (P = .21), trismus (P = 1.00), and feeling ill (P = .07) in QLQ-H&N35 scale, compared with these items before the radiotherapy.The results of this study showed that most items of QoL changed significantly after 3 months of radiotherapy in Chinese patients with LC.


Assuntos
Neoplasias Laríngeas/radioterapia , Qualidade de Vida , Idoso , Povo Asiático , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Inquéritos e Questionários , Resultado do Tratamento
19.
Medicine (Baltimore) ; 97(26): e11268, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29953001

RESUMO

This retrospective study examined the effect of voice rehabilitation training (VRT) for patients with laryngeal cancer (LC) after radiotherapy.Eighty-three eligible patients with LC were included. Forty-three patients were assigned to a treatment group, and underwent VRT, while the other 40 subjects were assigned to a control group, and were at waiting list. Primary outcome was measured by the Grade, Roughness, Breathiness, Asthenia, and Strain (GRBAS) scale. Secondary outcome was measured by Patient Perception Measures. All outcomes were measured before and 3 months after VRT intervention.Patients in the treatment group did not show better outcomes, measured by GRBAS scale (Grade, P = .78; Roughness, P = .61; Breathiness, P = .83; Ashenia, P = .89; and Strain, P = .41), and Patient Perception Measures (Vocal quality, P = .17; Acceptability, P = .35; Hoarseness, P = .23; Vocal fatigue, P = .39; and Ashamed, P = .51), compared with patients in the control group.The results of this study did not exert better outcomes in patients received VRT than those at waiting list.


Assuntos
Neoplasias Laríngeas/complicações , Distúrbios da Voz/etiologia , Distúrbios da Voz/reabilitação , Idoso , Feminino , Rouquidão , Humanos , Neoplasias Laríngeas/radioterapia , Masculino , Pessoa de Meia-Idade , Percepção , Qualidade de Vida , Estudos Retrospectivos , Qualidade da Voz
20.
J Cell Physiol ; 233(1): 226-237, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28218391

RESUMO

The majority of ovarian primordial follicles are preserved in a dormant state to maintain the female reproductive lifespan, and only a few primordial follicles are activated to enter the growing follicle pool in each wave. Recent studies have shown that primordial follicular activation depends on mammalian target of rapamycin complex 1 (mTORC1)-KIT ligand (KITL) signaling in pre-granulosa cells and its receptor (KIT)-phosphoinositol 3 kinase (PI3K) signaling in oocytes. However, the upstream regulator of mTORC1 signaling is unclear. The results of the present study showed that the phosphorylated mitogen-activated protein kinase3/1 (MAPK3/1) protein is expressed in some primordial follicles and all growing follicles. Culture of 3 days post-parturition (dpp) ovaries with the MAPK3/1 signaling inhibitor U0126 significantly reduced the number of activated follicles and was accompanied by dramatically reduced granulosa cell proliferation and increased oocyte apoptosis. Western blot and immunofluorescence analyses showed that U0126 significantly decreased the phosphorylation levels of Tsc2, S6K1, and rpS6 and the expression of KITL, indicating that U0126 inhibits mTORC1-KITL signaling. Furthermore, U0126 decreased the phosphorylation levels of Akt, resulting in a decreased number of oocytes with Foxo3 nuclear export. To further investigate MAPK3/1 signaling in primordial follicle activation, we used phosphatase and tensin homolog deleted on chromosome 10 (PTEN) inhibitor bpV(HOpic) to promote primordial follicle activation. In this model, U0126 also inhibited the activation of primordial follicles and mTORC1 signaling. Thus, these results suggest that MAPK3/1 participates in primordial follicle activation through mTORC1-KITL signaling.


Assuntos
Células da Granulosa/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Complexos Multiproteicos/metabolismo , Oócitos/enzimologia , Transdução de Sinais , Fator de Células-Tronco/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Proliferação de Células , Ativação Enzimática , Feminino , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos ICR , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Oócitos/efeitos dos fármacos , Oócitos/patologia , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA