Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Phytomedicine ; 132: 155890, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39033726

RESUMO

BACKGROUND: Reactive Oxygen Species (ROS) is a key factor in the pathogenesis of osteoporosis (OP) primarily characterized by excessive osteoclast activity. Active fraction of Polyrhachis vicina Rogers (AFPR) exerts antioxidant effects and possesses extensive promising therapeutic effects in various conditions, however, its function in osteoclastogenesis and OP is unknown. PURPOSE: The aim of this study is to elucidate the cellular and molecular mechanisms of AFPR in OP. STUDY DESIGN AND METHODS: CCK8 assay was used to evaluate the cell viability under AFPR treatment. TRAcP staining, podosome belts staining and bone resorption were used to test the effect of AFPR on osteoclastogenesis. Immunofluorescence staining was used to observe the effect of AFPR on ROS production. si-RNA transfection, coimmunoprecipitation and Western-blot were used to clarify the underlying mechanisms. Further, an ovariectomy (OVX) -induced OP mice model was used to identify the effect of AFPR on bone loss using Micro-CT scanning and histological examination. RESULTS: In the present study, AFPR inhibited osteoclast differentiation and bone resorption induced by nuclear factor-κB receptor activator (NF-κB) ligand (RANKL) in dose-/ time-dependent with no cytotoxicity. Meanwhile, AFPR decreased RANKL-mediated ROS levels and enhanced ROS scavenging enzymes. Mechanistically, AFPR promoted proteasomal degradation of TRAF6 by significantly upregulating its K48-linked ubiquitination, subsequently inhibiting NFATc1 activity. We further observed that tripartite motif protein 38 (TRIM38) could mediate the ubiquitination of TRAF6 in response to RANKL. Moreover, TRIM38 could negatively regulate the RANKL pathway by binding to TRAF6 and promoting K48-linked polyubiquitination. In addition, TRIM38 deficiency rescued the inhibition of AFPR on ROS and NFATc1 activity and osteoclastogenesis. In line with these results, AFPR reduced OP caused by OVX through ameliorating osteoclastogenesis. CONCLUSION: AFPR alleviates ovariectomized-induced bone loss via suppressing ROS and NFATc1 by targeting Trim38 mediated proteasomal degradation of TRAF6. The research offers innovative perspectives on AFPR's suppressive impact in vivo OVX mouse model and in vitro, and clarifies the fundamental mechanism.


Assuntos
Osteoclastos , Osteogênese , Osteoporose , Extratos Vegetais , Espécies Reativas de Oxigênio , Fator 6 Associado a Receptor de TNF , Animais , Fator 6 Associado a Receptor de TNF/metabolismo , Camundongos , Osteogênese/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Feminino , Osteoporose/tratamento farmacológico , Extratos Vegetais/farmacologia , Proteínas com Motivo Tripartido/metabolismo , Ligante RANK/metabolismo , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo , Ovariectomia , Reabsorção Óssea/tratamento farmacológico , Células RAW 264.7 , Diferenciação Celular/efeitos dos fármacos , Fatores de Transcrição NFATC/metabolismo , Humanos
2.
Biomed Pharmacother ; 177: 116933, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38901204

RESUMO

Reactive oxidative species (ROS) generation triggers pyroptosis and induces development of inflammatory osteolysis. Hecogenin (HG) has anti-inflammatory and antioxidative property, but its effects on inflammatory osteolysis remains unclear. In our study, we investigated the mechanism of HG on pyroptosis and its effect on inflammatory osteolysis in vitro and in vivo. The impact of HG on osteoclastogenesis was evaluated using cytotoxicity, TRAcP staining and bone resorption assays. The RNA-sequencing was employed to identify potential signaling pathways, and then RT-qPCR, western blot, immunofluorescence, and ELISA were used to verify. To determine the protective effect of HG in vivo, Lipopolysaccharide (LPS)-induced animal models were utilized, along with micro-CT and histological examination. HG suppressed RANKL-induced osteoclast differentiation, bone resorption, NFATc1 activity and downstream factors. RNA-sequencing results showed that HG inhibited osteoclastogenesis by modulating the inflammatory response and macrophage polarization. Furthermore, HG inhibited the NF-κB pathway, and deactivated the NLRP3 inflammasome. HG activated the expression of nuclear factor E2-related factor 2 (Nrf2) to eliminate ROS generation. Importantly, the inhibitory effect of HG on NLRP3 inflammasome could be reversed by treatment with the Nrf2 inhibitor ML385. In vivo, HG prevented the mice against LPS-induced osteolysis by suppressing osteoclastogenesis and inflammatory factors. In conclusion, HG could activate Nrf2 to eliminate ROS generation, inactivate NLRP3 inflammasome and inhibit pyroptosis, thereby suppressing osteoclastogenesis in vitro and alleviating inflammatory osteolysis in vivo, which indicating that HG might be a promising candidate to treat inflammatory osteolysis.


Assuntos
Lipopolissacarídeos , Fator 2 Relacionado a NF-E2 , Proteína 3 que Contém Domínio de Pirina da Família NLR , Osteoclastos , Osteólise , Piroptose , Espécies Reativas de Oxigênio , Animais , Masculino , Camundongos , Anti-Inflamatórios/farmacologia , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese/efeitos dos fármacos , Osteólise/induzido quimicamente , Osteólise/tratamento farmacológico , Osteólise/metabolismo , Osteólise/patologia , Piroptose/efeitos dos fármacos , Ligante RANK/metabolismo , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
Adv Healthc Mater ; 13(22): e2400797, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38726796

RESUMO

The Notch signaling is a key molecular pathway that regulates cell fate and development. Aberrant Notch signaling can lead to carcinogenesis and progression of malignant tumors. However, current therapies targeting Notch pathway lack specificity and induce high toxicity. In this report, a tumor microenvironment-responsive and injectable hydrogel is designed to load plasmid DNA complexes as a cascade gene delivery system to achieve precise Notch-targeted gene therapy of colorectal cancer (CRC). The hydrogels are prepared through cross-linking between phenylboric acid groups containing poly(oligo(ethylene glycol)methacrylate) (POEGMA) and epigallocatechin gallate (EGCG), used to load the complexes between plasmid DNA encoding short hairpin RNAs of Notch1 (shNotch1) and fluorinated polyamidoamine (PAMAM-F) (PAMAM-F/shNotch1). In response to low pH and H2O2 in tumor microenvironment, the hydrogel can be dissociated and release the complexes for precise delivery of shNotch1 into tumor cells and inhibit Notch1 activity to suppress malignant biological behaviors of CRC. In the subcutaneous tumor model of CRC, PAMAM-F/shNotch1-loaded hydrogels can accurately attenuate Notch1 activity and significantly inhibit tumor growth without affecting Notch signal in adjacent normal tissues. Therefore, this therapeutic system can precisely inhibit Notch1 signal in CRC with high responsiveness and low toxicity, providing a promising Notch-targeted gene therapeutic for human malignancy.


Assuntos
Neoplasias Colorretais , Hidrogéis , Receptor Notch1 , Microambiente Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/terapia , Receptor Notch1/metabolismo , Humanos , Microambiente Tumoral/efeitos dos fármacos , Animais , Hidrogéis/química , Camundongos , Linhagem Celular Tumoral , DNA/química , Terapia Genética/métodos , Catequina/análogos & derivados , Catequina/farmacologia , Catequina/química , Técnicas de Transferência de Genes , Camundongos Nus , Camundongos Endogâmicos BALB C
4.
Phytother Res ; 38(4): 1971-1989, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38358727

RESUMO

BACKGROUND AND AIM: Osteoporosis, a systemic metabolic bone disease, is characterized by the decline of bone mass and quality due to excessive osteoclast activity. Currently, drug-targeting osteoclasts show promising therapy for osteoporosis. In this study, we investigated the effect of cichoric acid (CA) on receptor activator of nuclear kappa-B ligand (RANKL)-induced osteoclastogenesis and the bone loss induced by ovariectomy in mice. EXPERIMENTAL PROCEDURE: Molecular docking technologies were employed to examine the interaction between CA and RANKL. CCK8 assay was used to evaluate the cell viability under CA treatment. TRAcP staining, podosome belt staining, and bone resorption assays were used to test the effect of CA on osteoclastogenesis and osteoclast function. Further, an OVX-induced osteoporosis mice model was employed to identify the effect of CA on bone loss using micro-CT scanning and histological examination. To investigate underlying mechanisms, network pharmacology was applied to predict the downstream signaling pathways, which were verified by Western blot and immunofluorescence staining. KEY RESULTS: The molecular docking analysis revealed that CA exhibited a specific binding affinity to RANKL, engaging multiple binding sites. CA inhibited RANKL-induced osteoclastogenesis and bone resorption without cytotoxic effects. Mechanistically, CA suppressed RANKL-induced intracellular reactive oxygen species, nuclear factor-kappa B, and mitogen-activated protein kinase pathways, followed by abrogated nuclear factor activated T-cells 1 activity. Consistent with this finding, CA attenuated post-ovariectomy-induced osteoporosis by ameliorating osteoclastogenesis. CONCLUSIONS AND IMPLICATIONS: CA inhibited osteoclast activity and bone loss by targeting RANKL. CA might represent a promising candidate for treating osteoclast-related diseases, such as osteoporosis.


Assuntos
Reabsorção Óssea , Ácidos Cafeicos , Osteoporose , Succinatos , Animais , Feminino , Humanos , Camundongos , Reabsorção Óssea/prevenção & controle , Diferenciação Celular , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular , NF-kappa B/metabolismo , Osteoclastos , Osteogênese , Osteoporose/patologia , Ovariectomia/efeitos adversos , Ligante RANK/metabolismo
5.
Biochem Pharmacol ; 218: 115895, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-38084677

RESUMO

Reactive Oxygen Species (ROS) play an essential role in the pathogenesis of osteoporosis mainly characterized by excessive osteoclasts (OCs) activity. OCs are rich in mitochondria for energy support, which is a major source of total ROS. Tussilagone (TSG), a natural Sesquiterpenes from the flower of Tussilago farfara, has plentiful beneficial pharmacological characteristics with anti-inflammatory and anti-oxidative activity, but its effects and mechanism in osteopathology are still unclear. In our study, we investigated the regulation of ROS generated from the mitochondria in OCs. We found that TSG inhibited OCs differentiation and bone resorption without any cytotoxicity. Mechanistically, TSG reduced RANKL-mediated total ROS level by down-regulating intracellular ROS production and mitochondrial function, leading to the suppression of NFATc1 transcription. We also found that nuclear factor erythroid 2-related factor 2 (Nrf2) could enhance ROS scavenging enzymes in response to RANKL-induced oxidative stress. Furthermore, TSG up-regulated the expression of Nrf2 by inhibiting its proteosomal degradation. Interestingly, Nrf2 deficiency reversed the suppressive effect of TSG on mitochondrial activity and ROS signaling in OCs. Consistent with this finding, TSG attenuated post-ovariectomy (OVX)- and lipopolysaccharide (LPS) induced bone loss by ameliorating osteoclastogenesis. Taken together, TSG has an anti-bone resorptive effect by modulating mitochondrial function and ROS production involved Nrf2 activation.


Assuntos
Reabsorção Óssea , Sesquiterpenos , Feminino , Humanos , Osteogênese , Espécies Reativas de Oxigênio/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Osteoclastos , Sesquiterpenos/farmacologia , Sesquiterpenos/metabolismo , Reabsorção Óssea/metabolismo , Ligante RANK/farmacologia , Diferenciação Celular , NF-kappa B/metabolismo
6.
Molecules ; 28(13)2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37446806

RESUMO

Cancer continues to pose a severe threat to global health, making pursuing effective treatments more critical than ever. Traditional therapies, although pivotal in managing cancer, encounter considerable challenges, including drug resistance, poor drug solubility, and difficulties targeting tumors, specifically limiting their overall efficacy. Nanomedicine's application in cancer therapy signals a new epoch, distinguished by the improvement of the specificity, efficacy, and tolerability of cancer treatments. This review explores the mechanisms and advantages of nanoparticle-mediated drug delivery, highlighting passive and active targeting strategies. Furthermore, it explores the transformative potential of nanomedicine in tumor therapeutics, delving into its applications across various treatment modalities, including surgery, chemotherapy, immunotherapy, radiotherapy, photodynamic and photothermal therapy, gene therapy, as well as tumor diagnosis and imaging. Meanwhile, the outlook of nanomedicine in tumor therapeutics is discussed, emphasizing the need for addressing toxicity concerns, improving drug delivery strategies, enhancing carrier stability and controlled release, simplifying nano-design, and exploring novel manufacturing technologies. Overall, integrating nanomedicine in cancer treatment holds immense potential for revolutionizing cancer therapeutics and improving patient outcomes.


Assuntos
Nanopartículas , Neoplasias , Humanos , Nanomedicina , Neoplasias/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Imunoterapia , Diagnóstico por Imagem , Nanopartículas/uso terapêutico
8.
Arch Biochem Biophys ; 741: 109596, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37030589

RESUMO

OBJECTIVE: Preeclampsia (PE) is a maternal multisystem disease with an unclear mechanism. Data showed that MiR-95-3p promoted cell migration, invasion and proliferation, leading to the occurrence and development of many cancers, and placental trophoblasts and tumor cells had similar migration, invasion and proliferation abilities. Meanwhile we found that MiR-95-3p was differentially expressed in PE and normal placenta. Therefore, this article aimed to explore the biological function and mechanism of miR-95-3p in PE. METHODS: The expression of miR-95-3p in PE and normal placental tissue was explored by high-throughput sequencing and qRT-PCR. The effects of miR-95-3p on trophoblast migration, invasion, proliferation, angiogenesis and apoptosis were investigated by Transwell migration and invasion assays, cell viability assay, tube formation assay and flow cytometry in two trophoblast cell lines (HTR-8/SVneo and JAR). The miR-95-3p target gene EPM2A was identified and verified by unique identifier mRNA next-generation sequencing and dual-luciferase reporter gene experiments. Rescue experiments were conducted to investigate whether miR-95-3p regulated EPM2A to participate in trophoblast migration and invasion. Finally, the effects of miR-95-3p and EPM2A on the expression of angiogenic factors and inflammation-related factors were investigated by ELISA. RESULTS: We found that miR-95-3p was expressed at low levels in the placental tissue of patients with PE and was negatively correlated with EPM2A expression. In vitro upregulation of miR-95-3p and downregulation of EPM2A promote trophoblast migration, invasion and proliferation. Furthermore, EPM2A was confirmed as a target mRNA of miR-95-3p. Upregulation of EPM2A mitigated miR-95-3p-mediated promotion of trophoblast migration and invasion and vice versa. Finally, both miR-95-3p and EPM2A regulate the expression of trophoblast angiogenesis-related factors and inflammation-related factors. CONCLUSION: Our findings demonstrated that miR-95-3p promoted the migration and invasion of trophoblast cells by targeting EPM2A to inhibit the occurrence and development of PE.


Assuntos
MicroRNAs , Pré-Eclâmpsia , Trofoblastos , Feminino , Humanos , Gravidez , Movimento Celular/genética , Proliferação de Células/genética , Metaloproteinase 2 da Matriz/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Proteínas Tirosina Fosfatases não Receptoras , RNA Mensageiro/metabolismo , Trofoblastos/metabolismo
9.
Gene ; 860: 147211, 2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-36708847

RESUMO

Sirt1 is a member of the Sirtuins family that regulates ovarian senescence, follicular development, and oocyte maturation in vertebrates. To understand its role in the ovary of Pelodiscus sinensis, we cloned the full-length cDNA of Ps-Sirt1 and characterized its potential function by intraperitoneally injecting agonist (resveratrol) and antagonist (EX527) in the female juvenile turtle. The full-length cDNA of Ps-Sirt1 was 2106 bp, comprising 203 bp 5'UTR, a 226 bp 3'UTR, and a 1677 bp ORF encoding 558 amino acids. The calculated molecular weight of predicted protein was 63 kDa, and the isoelectric point was 4.65. The predicted protein comprised a conserved Sir2 domain. Amino acid sequence alignment and phylogenetic analyses showed that Ps-Sirt1 was most closely related to turtles, and distantly related to fish. Expression pattern analysis showed Ps-Sirt1 was highest expressed in ovary, followed by testis, liver, heart, and brain. In the ovarian differentiation processes, Sirt1 showed significantly higher expression at embryonic stage 15 and 21. In the testis differentiation process, Sirt1 expression was downregulated at embryonic stages 15-19. Activated and inactivated Sirt1 decreased the number of primordial follicles in juvenile turtles. Bcl2, Bax, mTOR, and rpS6 expressions were up-regulated, whereas GnRH, Fshb, p50, and p65 were down-regulated after agonist treatment. The inaction of Sirt1 with antagonist up-regulated GnRH, Fshb, p65, p53, Foxo3a, Bcl2, Bax, mTOR, and rpS6, but down-regulated p50. In summary, Sirt1 might be involved in the ovarian follicle development of P. sinensis.


Assuntos
Tartarugas , Masculino , Animais , Feminino , Tartarugas/genética , Tartarugas/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Filogenia , DNA Complementar/metabolismo , Proteína X Associada a bcl-2/metabolismo , Clonagem Molecular , Folículo Ovariano , Serina-Treonina Quinases TOR/metabolismo , Hormônio Liberador de Gonadotropina/genética
10.
Dis Markers ; 2022: 9872243, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36340581

RESUMO

Purpose: Osteoporosis is a complication of type 2 diabetes, and it is characterized by reduced bone mass, augmented bone fragility, and increased risk of fracture, thus reducing patient quality of life, especially in the elderly. Ferroptosis has been implicated in the pathological process of type 2 diabetic osteoporosis (T2DOP), but the specific underlying mechanisms remain largely unknown. This study clarified the role of activating transcription factor 3 (ATF3) in T2DOP and explored its specific regulatory mechanism, providing a new treatment target for T2DOP. Methods: We cultured hFob1.19 cells in high glucose (HG, 35 mM) and knocked down ATF3 using short hairpin RNA (shRNA). We then measured cell viability, assessed morphology, quantified the expression of ATF3 and glutathione peroxidase 4 (GPX4), detected the levels of reactive oxygen species (ROS) and lipid peroxides, and determined the osteogenic function of osteoblasts. Cystine/glutamate antiporter (system Xc-) activity was evaluated by determining the expression of SLC7A11 and the levels of glutathione (GSH) and extracellular glutamate. We constructed a T2DOP rat model and observed the effect of ATF3 on ferroptosis and T2DOP by knocking down ATF3 using small interfering RNA (siRNA). Then, we evaluated the levels of iron metabolism, lipid peroxidation, and bone turnover in serum, detected the expression of ATF3, SLC7A11, and GPX4 in bone tissues, and assessed bone microstructure using microcomputed tomography. Results: ATF3 expression was increased in osteoblasts under HG condition and in T2DOP rats. Inhibiting the function of ATF3 increased GPX4 levels and reduced the accumulation of ROS and lipid peroxides. These changes inhibited the ferroptosis of osteoblasts and improved osteogenic function. In addition, HG induced ATF3 upregulation, resulting in decreased SLC7A11 expression and lower levels of intracellular GSH and extracellular glutamate. Conclusion: Osteoblast ferroptosis under HG conditions is induced by ATF3-mediated inhibition of system Xc- activity, and these events contribute to T2DOP pathogenesis.


Assuntos
Diabetes Mellitus Tipo 2 , Ferroptose , Osteoporose , Ratos , Animais , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Peróxidos Lipídicos , Diabetes Mellitus Tipo 2/complicações , Microtomografia por Raio-X , Qualidade de Vida , Osteoblastos/metabolismo , Osteoporose/genética , Glutamatos
11.
Hypertension ; 79(12): 2830-2842, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36252138

RESUMO

BACKGROUND: Preeclampsia is a pregnancy syndrome that may utilize multiple pathogenic mechanisms. Insufficient trophoblast invasion and impaired uterine spiral artery remodeling are believed to be the pathological basis; yet the underlying mechanisms remain largely unclear. METHODS: The placental BCAM (basal cell adhesion molecule) expression and important clinical indicators were detected and correlation analysis was performed. MiRNAs directly targeting BCAM were predicted and further verified by dual-luciferase reporter gene, and the downstream molecular mechanisms of BCAM were investigated in both HTR-8/SVneo and JAR cells. In addition, pregnant/nonpregnant rats were treated with adenoviruses containing BCAM shRNA genes (Ad-shBCAM) on gestational 9.5 days to detect the preeclamptic features. RESULTS: The BCAM is highly expressed on the trophoblast membrane and decreased in the preeclamptic placentae. In HTR-8/SVneo and JAR cells, BCAM knockdown inhibited trophoblast proliferation, migration, and invasion, and suppressed phosphorylation on Y705 of STAT3 dependent on the downregulation of PIK3R6. Moreover, miR-199a-5p mediated the degradation of BCAM and also inhibited trophoblast proliferation, migration, and invasion. In vivo, BCAM deficiency induced a preeclampsia-like phenotype included elevated systolic blood pressure, proteinuria, impaired morphology and function of multiple organs (placenta, liver, and kidney), and fetal growth restriction. The expression of placenta BCAM/PIK3R6/p-STAT3 signaling was also downregulated in this preeclampsia rat model. CONCLUSIONS: MiR-199a-5p mediated-BCAM deficiency contributes to the suppression of trophoblast proliferation, migration, and invasion by inhibiting PIK3R6/p-STAT3 signaling, which may lead to poor placentation and result in preeclampsia-like phenotypes. Our study provides a new academic perspective on the pathogenesis of preeclampsia.


Assuntos
MicroRNAs , Pré-Eclâmpsia , Animais , Feminino , Gravidez , Ratos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Movimento Celular , Sistema do Grupo Sanguíneo Lutheran/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Placenta/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Trofoblastos/metabolismo
12.
BMC Surg ; 21(1): 315, 2021 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-34301235

RESUMO

PURPOSE: Amyand's hernia is a rare hernia defined as an inguinal hernia that contains the appendix within the hernia sac. Current treatment of Amyand's hernia remains controversial. Our study retrospectively reviewed 6 cases of Amyand's hernia, aiming to provide a reference for the surgical treatment of Amyand's hernia. METHODS: Six patients diagnosed with Amyand's hernia from September 2010 to May 2020 were retrospectively enrolled in our study. We summarized clinical data of six patients including the chief complaint, physical examinations, laboratory examinations, imaging examinations, surgical methods, and postoperative treatments and outcomes. RESULTS: The diagnosis of six cases with Amyand's hernia was made during surgery. Two patients had normal appendixes whereas the remaining four patients had appendicitis. Two patients with normal appendix received tension-free mesh repair through the inguinal incision. Among those with inflamed or perforated appendixes, two received mesh repair and the other two did not. The discharge time after surgery of six patients was 9.8 ± 6.1 days. One patient suffered from a wound infection. No additional postoperative complications were detected. CONCLUSIONS: Computed tomography and ultrasonography are helpful but limited in the definite diagnosis of Amyand's hernia. The presence of a normal appendix does not require to be resected, but appendicectomy is necessary if the appendix is inflamed. The treatment of Amyand's hernia should be tailored based on the patient's condition and the type of Amyand's hernia.


Assuntos
Apendicite , Apêndice , Hérnia Inguinal , Apendicectomia , Apendicite/complicações , Apendicite/diagnóstico , Apendicite/cirurgia , Hérnia Inguinal/complicações , Hérnia Inguinal/diagnóstico , Hérnia Inguinal/cirurgia , Humanos , Estudos Retrospectivos
13.
Cell Rep Med ; 2(5): 100279, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34095881

RESUMO

Aberrant NOTCH3 signaling and overexpression is oncogenic, associated with cancer stem cells and drug resistance, yet therapeutic targeting remains elusive. Here, we develop NOTCH3-targeted antibody drug conjugates (NOTCH3-ADCs) by bioconjugation of an auristatin microtubule inhibitor through a protease cleavable linker to two antibodies with differential abilities to inhibit signaling. The signaling inhibitory antibody rapidly induces ligand-independent receptor clustering and internalization through both caveolin and clathrin-mediated pathways. The non-inhibitory antibody also efficiently endocytoses via clathrin without inducing receptor clustering but with slower lysosomal co-localization kinetics. In addition, DLL4 ligand binding to the NOTCH3 receptor mediates transendocytosis of NOTCH3-ADCs into ligand-expressing cells. NOTCH3-ADCs internalize into receptor and ligand cells independent of signaling and induce cell death in both cell types representing an atypical mechanism of ADC cytotoxicity. Treatment of xenografts with NOTCH3-ADCs leads to sustained tumor regressions, outperforms standard-of-care chemotherapy, and allows targeting of tumors that overexpress NOTCH3 independent of signaling inhibition.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Imunoconjugados/farmacologia , Receptor Notch3/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Humanos , Imunoconjugados/metabolismo , Oncogenes/efeitos dos fármacos , Receptor Notch3/imunologia , Receptores Notch/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Cancer Lett ; 328(2): 261-70, 2013 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-23041621

RESUMO

Recent evidence suggests that Notch signaling may play a role in regulation of cancer stem cell (CSC) self-renewal and differentiation hence presenting a promising target for development of novel therapies for aggressive cancers such as triple negative breast cancer (TNBC). We generated Notch1 monoclonal antibodies (mAbs) that specifically bind to the negative regulatory region of human Notch1. Notch1 inhibition in TNBC Sum149 and patient derived xenograft (PDX) 144580 models led to significant TGI particularly in combination with docetaxel. More interestingly, Notch1 mAbs caused a reduction in mammosphere formation and CD44+/CD24-/lo cell population. It also resulted in decreased tumor incidence upon re-implantation and delay in tumor recurrence. Our data demonstrated a potent antitumor efficacy of Notch1 mAbs, with a remarkable activity against CSCs. These findings suggest that anti-Notch1 mAbs may provide novel therapies to improve the efficacy of conventional therapies by directly targeting the CSC niche. They may also delay tumor recurrence and hence have a major impact on cancer patient survival.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Anticorpos Monoclonais/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Docetaxel , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Camundongos Nus , Recidiva , Esferoides Celulares/efeitos dos fármacos , Taxoides/farmacologia , Carga Tumoral , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Proc Natl Acad Sci U S A ; 103(19): 7378-83, 2006 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-16670198

RESUMO

Inactivation of the XRCC4 nonhomologous end-joining factor in the mouse germ line leads to embryonic lethality, in association with apoptosis of newly generated, postmitotic neurons. We now show that conditional inactivation of the XRCC4 in nestin-expressing neuronal progenitor cells, although leading to no obvious phenotype in a WT background, leads to early onset of neuronally differentiated medulloblastomas (MBs) in a p53-deficient background. A substantial proportion of the XRCC4/p53-deficient MBs have high-level N-myc gene amplification, often intrachromosomally in the context of complex translocations or other alterations of chromosome 12, on which N-myc resides, or extrachromosomally within double minutes. In addition, most XRCC4/p53-deficient MBs harbor clonal translocations of chromosome 13, which frequently involve chromosome 6 as a partner. One copy of the patched gene (Ptc), which lies on chromosome 13, was deleted in all tested XRCC4/p53-deficient MBs in the context of translocations or interstitial deletions. In addition, Cyclin D2, a chromosome 6 gene, was amplified in a subset of tumors. Notably, amplification of Myc-family or Cyclin D2 genes and deletion of Ptc also have been observed in human MBs. We therefore conclude that, in neuronal cells of mice, the nonhomologous end-joining pathway plays a critical role in suppressing genomic instability that, in a p53-deficient background, routinely contributes to genesis of MBs with recurrent chromosomal alterations.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Meduloblastoma/metabolismo , Translocação Genética/genética , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo , Alelos , Animais , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Regulação para Baixo/genética , Amplificação de Genes , Proteínas de Filamentos Intermediários/metabolismo , Meduloblastoma/genética , Meduloblastoma/patologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Nestina , Taxa de Sobrevida , Fatores de Tempo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
16.
Cell Signal ; 17(3): 365-75, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15567067

RESUMO

Growth differentiation factor-8 (GDF-8), a member of the TGF-beta superfamily, is a negative regulator of skeletal muscle growth, which functions through activation of the Smad proteins. We found that GDF-8 can activate the p38 mitogen-activated protein kinase (MAPK) through the TGF-beta-activated kinase 1 (TAK1), and this appeared to be independent of Smad signaling. GDF-8-induced transcriptional activation was inhibited by expression of dominant negative MKK6 or treatment with the p38 inhibitor SB203580, while overexpression of wild-type forms of either MKK6 or p38 augmented GDF-8-induced transcriptional activity. In addition, ATF-2, a known transcription factor target of p38, was found to be phosphorylated on GDF-8 stimulation and was detected in a complex with Smad3/Smad4 upon GDF-8 treatment. Furthermore, we found that the p38 MAPK played an important role in GDF-8-induced inhibition of proliferation and upregulation of the cyclin kinase inhibitor p21. Together, these results highlight a functional link between the p38 MAPK and GDF-8-activated Smad pathways, and identify a critical role for the p38 MAPK in GDF-8's function as a negative regulator of muscle growth.


Assuntos
Células Musculares/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Fator 2 Ativador da Transcrição , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Humanos , Imidazóis/farmacologia , MAP Quinase Quinase 6/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Miostatina , Piridinas/farmacologia , Transdução de Sinais , Proteínas Smad , Proteína Smad3 , Proteína Smad4 , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
17.
Cell ; 109(7): 811-21, 2002 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-12110179

RESUMO

Amplification of large genomic regions associated with complex translocations (complicons) is a basis for tumor progression and drug resistance. We show that pro-B lymphomas in mice deficient for both p53 and nonhomologous end-joining (NHEJ) contain complicons that coamplify c-myc (chromosome 15) and IgH (chromosome 12) sequences. While all carry a translocated (12;15) chromosome, coamplified sequences are located within a separate complicon that often involves a third chromosome. Complicon formation is initiated by recombination of RAG1/2-catalyzed IgH locus double-strand breaks with sequences downstream of c-myc, generating a dicentric (15;12) chromosome as an amplification intermediate. This recombination event employs a microhomology-based end-joining repair pathway, as opposed to classic NHEJ or homologous recombination. These findings suggest a general model for oncogenic complicon formation.


Assuntos
Dano ao DNA/genética , Reparo do DNA , Amplificação de Genes/genética , Genes myc/genética , Translocação Genética/genética , Proteína Supressora de Tumor p53/deficiência , Animais , Sequência de Bases , Cromossomos/genética , Regulação Neoplásica da Expressão Gênica , Genes RAG-1 , Hibridização in Situ Fluorescente , Linfoma de Células B/genética , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recombinação Genética/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA