Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
MedComm (2020) ; 5(9): e704, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39268354

RESUMO

The continuous production of mature blood cell lineages is maintained by hematopoietic stem cells but they are highly susceptible to damage by ionizing radiation (IR) that induces death. Thus, devising therapeutic strategies that can mitigate hematopoietic toxicity caused by IR would benefit acute radiation syndrome (ARS) victims and patients receiving radiotherapy. Herein, we describe the preparation of an injectable hydrogel formulation based on Arg-Gly-Asp-alginate (RGD-Alg) and Laponite using a simple mixing method that ensured a slow and sustained release of interleukin-12 (IL-12) (RGD-Alg/Laponite@IL-12). The local administration of RGD-Alg/Laponite@IL-12 increased survival rates and promoted the hematopoietic recovery of mice who had received sublethal-dose irradiation. Local intra-bone marrow (intra-BM) injection of RGD-Alg/Laponite@IL-12 hydrogel effectively stimulated IL12 receptor-phosphoinositide 3-kinase/protein kinase B (IL-12R-PI3K/AKT) signaling axis, which promoted proliferation and hematopoietic growth factors secretion of BM mesenchymal stem/stromal cells. This signaling axis facilitates the repair of the hematopoietic microenvironment and plays a pivotal role in hematopoietic reconstitution. In conclusion, we describe a biomaterial-sustained release of IL-12 for the treatment of irradiated hematopoietic injury and provide a new therapeutic strategy for hematopoietic ARS.

2.
Redox Biol ; 75: 103260, 2024 09.
Artigo em Inglês | MEDLINE | ID: mdl-38955114

RESUMO

Tumor metabolic reprogramming requires high levels of adenosine triphosphate (ATP) to maintain treatment resistance, which poses major challenges to chemotherapy and photothermal therapy. Especially, high levels of ATP promote copper ion efflux for limiting the curative effect of cuproptosis. Here, an H2S-responsive mesoporous Cu2Cl(OH)3-loading chemotherapeutic cisplatin (CDDP) was synthesized, and the final nanoparticle, CDDP@Cu2Cl(OH)3-CDs (CDCuCDs), was encapsulated by electrostatic action with carbon dots (CDs). CDCuCDs reacted with overproduction H2S in colon tumor to produce photothermic copper sulfide for photothermal therapy. CDDP was released by lysis to achieve chemotherapeutic effects. Importantly, CDDP elevated H2O2 levels in cells through a cascade reaction and continuously transforms H2O2 into highly cytotoxic •OH through chemodynamic therapy between H2O2 and Cu+, which enables nanoparticles to generate •OH and improve the chemotherapeutic efficacy. Highly toxic •OH disrupts mitochondrial homeostasis, prohibiting it from performing normal energy-supplying functions. Down-regulated ATP inhibits heat shock protein expression, which promotes the therapeutic effect of mild photothermal therapy and reduces the efflux of intracellular copper ions, thus improving the therapeutic effect of cuproptosis. Our research provides a potential therapeutic strategy using overproduction H2S responses in tumors, allowing tumor microenvironment-activated •OH nanogenerators to promote tumor energy remodeling for cancer treatment.


Assuntos
Cobre , Estresse Oxidativo , Terapia Fototérmica , Microambiente Tumoral , Terapia Fototérmica/métodos , Microambiente Tumoral/efeitos dos fármacos , Cobre/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Humanos , Animais , Camundongos , Linhagem Celular Tumoral , Nanopartículas/química , Cisplatino/farmacologia , Peróxido de Hidrogênio/metabolismo
3.
World J Stem Cells ; 16(5): 538-550, 2024 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-38817334

RESUMO

BACKGROUND: Thrombocytopenia 2, an autosomal dominant inherited disease characterized by moderate thrombocytopenia, predisposition to myeloid malignancies and normal platelet size and function, can be caused by 5'-untranslated region (UTR) point mutations in ankyrin repeat domain containing 26 (ANKRD26). Runt related transcription factor 1 (RUNX1) and friend leukemia integration 1 (FLI1) have been identified as negative regulators of ANKRD26. However, the positive regulators of ANKRD26 are still unknown. AIM: To prove the positive regulatory effect of GATA binding protein 2 (GATA2) on ANKRD26 transcription. METHODS: Human induced pluripotent stem cells derived from bone marrow (hiPSC-BM) and urothelium (hiPSC-U) were used to examine the ANKRD26 expression pattern in the early stage of differentiation. Then, transcriptome sequencing of these iPSCs and three public transcription factor (TF) databases (Cistrome DB, animal TFDB and ENCODE) were used to identify potential TF candidates for ANKRD26. Furthermore, overexpression and dual-luciferase reporter experiments were used to verify the regulatory effect of the candidate TFs on ANKRD26. Moreover, using the GENT2 platform, we analyzed the relationship between ANKRD26 expression and overall survival in cancer patients. RESULTS: In hiPSC-BMs and hiPSC-Us, we found that the transcription levels of ANKRD26 varied in the absence of RUNX1 and FLI1. We sequenced hiPSC-BM and hiPSC-U and identified 68 candidate TFs for ANKRD26. Together with three public TF databases, we found that GATA2 was the only candidate gene that could positively regulate ANKRD26. Using dual-luciferase reporter experiments, we showed that GATA2 directly binds to the 5'-UTR of ANKRD26 and promotes its transcription. There are two identified binding sites of GATA2 that are located 2 kb upstream of the TSS of ANKRD26. In addition, we discovered that high ANKRD26 expression is always related to a more favorable prognosis in breast and lung cancer patients. CONCLUSION: We first discovered that the transcription factor GATA2 plays a positive role in ANKRD26 transcription and identified its precise binding sites at the promoter region, and we revealed the importance of ANKRD26 in many tissue-derived cancers.

4.
Br J Cancer ; 130(10): 1621-1634, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38575732

RESUMO

BACKGROUND: DNA double-strand break (DSB) induction and repair are important events for determining cell survival and the outcome of cancer radiotherapy. The DNA-dependent protein kinase (DNA-PK) complex functions at the apex of DSBs repair, and its assembly and activity are strictly regulated by post-translation modifications (PTMs)-associated interactions. However, the PTMs of the catalytic subunit DNA-PKcs and how they affect DNA-PKcs's functions are not fully understood. METHODS: Mass spectrometry analyses were performed to identify the crotonylation sites of DNA-PKcs in response to γ-ray irradiation. Co-immunoprecipitation (Co-IP), western blotting, in vitro crotonylation assays, laser microirradiation assays, in vitro DNA binding assays, in vitro DNA-PK assembly assays and IF assays were employed to confirm the crotonylation, identify the crotonylase and decrotonylase, and elucidate how crotonylation regulates the activity and function of DNA-PKcs. Subcutaneous xenografts of human HeLa GCN5 WT or HeLa GCN5 siRNA cells in BALB/c nude mice were generated and utilized to assess tumor proliferation in vivo after radiotherapy. RESULTS: Here, we reveal that K525 is an important site of DNA-PKcs for crotonylation, and whose level is sharply increased by irradiation. The histone acetyltransferase GCN5 functions as the crotonylase for K525-Kcr, while HDAC3 serves as its dedicated decrotonylase. K525 crotonylation enhances DNA binding activity of DNA-PKcs, and facilitates assembly of the DNA-PK complex. Furthermore, GCN5-mediated K525 crotonylation is indispensable for DNA-PKcs autophosphorylation and the repair of double-strand breaks in the NHEJ pathway. GCN5 suppression significantly sensitizes xenograft tumors of mice to radiotherapy. CONCLUSIONS: Our study defines K525 crotonylation of DNA-PKcs is important for the DNA-PK complex assembly and DSBs repair activity via NHEJ pathway. Targeting GCN5-mediated K525 Kcr of DNA-PKcs may be a promising therapeutic strategy for improving the outcome of cancer radiotherapy.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Tolerância a Radiação , Fatores de Transcrição de p300-CBP , Animais , Feminino , Humanos , Camundongos , Proteína Quinase Ativada por DNA/metabolismo , Células HeLa , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/radioterapia , Neoplasias/metabolismo , Neoplasias/genética , Fatores de Transcrição de p300-CBP/metabolismo , Processamento de Proteína Pós-Traducional , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Nanobiotechnology ; 22(1): 205, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658965

RESUMO

The elevated level of hydrogen sulfide (H2S) in colon cancer hinders complete cure with a single therapy. However, excessive H2S also offers a treatment target. A multifunctional cascade bioreactor based on the H2S-responsive mesoporous Cu2Cl(OH)3-loaded hypoxic prodrug tirapazamine (TPZ), in which the outer layer was coated with hyaluronic acid (HA) to form TPZ@Cu2Cl(OH)3-HA (TCuH) nanoparticles (NPs), demonstrated a synergistic antitumor effect through combining the H2S-driven cuproptosis and mild photothermal therapy. The HA coating endowed the NPs with targeting delivery to enhance drug accumulation in the tumor tissue. The presence of both the high level of H2S and the near-infrared II (NIR II) irradiation achieved the in situ generation of photothermic agent copper sulfide (Cu9S8) from the TCuH, followed with the release of TPZ. The depletion of H2S stimulated consumption of oxygen, resulting in hypoxic state and mitochondrial reprogramming. The hypoxic state activated prodrug TPZ to activated TPZ (TPZ-ed) for chemotherapy in turn. Furthermore, the exacerbated hypoxia inhibited the synthesis of adenosine triphosphate, decreasing expression of heat shock proteins and subsequently improving the photothermal therapy. The enriched Cu2+ induced not only cuproptosis by promoting lipoacylated dihydrolipoamide S-acetyltransferase (DLAT) heteromerization but also performed chemodynamic therapy though catalyzing H2O2 to produce highly toxic hydroxyl radicals ·OH. Therefore, the nanoparticles TCuH offer a versatile platform to exert copper-related synergistic antitumor therapy.


Assuntos
Cobre , Ácido Hialurônico , Sulfeto de Hidrogênio , Mitocôndrias , Nanopartículas , Terapia Fototérmica , Pró-Fármacos , Tirapazamina , Terapia Fototérmica/métodos , Sulfeto de Hidrogênio/metabolismo , Sulfeto de Hidrogênio/farmacologia , Animais , Cobre/química , Cobre/farmacologia , Camundongos , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Pró-Fármacos/farmacologia , Pró-Fármacos/química , Tirapazamina/farmacologia , Tirapazamina/química , Nanopartículas/química , Ácido Hialurônico/química , Linhagem Celular Tumoral , Neoplasias do Colo/terapia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/tratamento farmacológico , Camundongos Endogâmicos BALB C , Antineoplásicos/farmacologia , Antineoplásicos/química , Camundongos Nus
6.
J Enzyme Inhib Med Chem ; 39(1): 2287990, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38062554

RESUMO

Doublecortin-like kinase 1 (DCLK) is a microtubule-associated serine/threonine kinase that is upregulated in a wide range of cancers and is believed to be related to tumour growth and development. Upregulated DCLK1 has been used to identify patients at high risk of cancer progression and tumours with chemotherapy-resistance. Moreover, DCLK1 has been identified as a cancer stem cell (CSC) biomarker in various cancers, which has received considerable attention recently. Herein, a series of DCLK1 inhibitors were prepared based on the previously reported XMD8-92 structure. Among all the synthesised compounds, D1, D2, D6, D7, D8, D12, D14, and D15 showed higher DCLK1 inhibitory activities (IC50 40-74 nM) than XMD8-92 (IC50 161 nM). Compounds D1 and D2 were selective DCLK1 inhibitors as they showed a rather weak inhibitory effect on LRRK2. The antiproliferative activities of these compounds were also preliminarily evaluated. The structure-activity relationship revealed by our compounds provides useful guidance for the further development of DCLK1 inhibitors.


Assuntos
Quinases Semelhantes a Duplacortina , Inibidores de Proteínas Quinases , Humanos , Quinases Semelhantes a Duplacortina/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Relação Estrutura-Atividade , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia
7.
Biol Pharm Bull ; 46(11): 1569-1575, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37914359

RESUMO

Ovarian cancer (OC) is one of the most common and high mortality type of cancer among women worldwide. The majority of patients with OC respond to chemotherapy initially; however, most of them become resistant to chemotherapy and results in a high level of treatment failure in OC. Therefore, novel agents for the treatment of OC are urgently required. Benzimidazole anthelmintics might have the promising efficacy for cancer therapy as their selectively binding activity to ß-tubulin. Recent study has shown that one of the benzimidazole anthelmintics oxfendazole inhibited cell growth of non-small cell lung cancer cells, revealing its anti-cancer activity; however, the pharmacological action and detailed mechanism underlying the effects of oxfendazole on OC cells remain unclear. Therefore, the present study investigated the cytotoxic effects of oxfendazole on OC cells. Our results demonstrated that oxfendazole significantly decreased the viability of OC cells. Oxfendazole inhibited the proliferation, induced G2/M phase arrest and apoptotic cell death in A2780 cells. The c-Jun N-terminal kinase (JNK)/mitogen-activated protein kinase (MAPK) pathway was activated and reactive oxygen species (ROS) generation was increased in OC cells treated with oxfendazole; oxfendazole-induced apoptosis was notably abrogated when co-treated with JNK inhibitor SP600125 and ROS scavenger N-acetyl-L-cysteine (NAC), indicating that JNK/MAPK pathway activation and ROS accumulation was associated with the oxfendazole-induced apoptosis of OC cells. Moreover, oxfendazole could also induce the proliferation inhibition and apoptosis of cisplatin resistant cells. Collectively, these results revealed that oxfendazole may serve as a potential therapeutic agent for the treatment of OC.


Assuntos
Anti-Helmínticos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Neoplasias Ovarianas , Humanos , Feminino , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Neoplasias Ovarianas/tratamento farmacológico , Apoptose , Benzimidazóis/farmacologia , Sistema de Sinalização das MAP Quinases , Anti-Helmínticos/farmacologia
8.
Int J Mol Sci ; 24(22)2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-38003480

RESUMO

Inhibition of the extracellular signal-regulated kinases 1/2 (ERK1/2) alone or in combination with other targets has emerged as a promising treatment strategy for a variety of human tumors. In addition to the development of inhibitors, the development of ERK1/2 degraders is an alternative approach to decrease its activity. We synthesized proteolysis-targeting chimeras (PROTACs) as effective ERK1/2 degraders, among which B1-10J showed high degradative activity, with DC50 of 102 nM and cytotoxic IC50 of 2.2 µM against HCT116 cells. Moreover, B1-10J dose-dependently inhibited tumor cell migration. Xenograft experiments in nude mice demonstrated that B1-10J inhibited HCT116 tumor cell growth and achieved significant regression of tumors at a daily dose of 25 mg/kg.


Assuntos
Antineoplásicos , Animais , Camundongos , Humanos , Proteólise , Proliferação de Células , Camundongos Nus , Antineoplásicos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular
9.
Radiat Res ; 200(5): 489-502, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37815199

RESUMO

Radiation exposure arising from radiotherapy may induce rapid bone loss and an increase in the extent of bone resorption. Reactive oxygen species (ROS) caused by radiation exposure play a crucial role during the process of osteoclastogenesis. However, the pathological mechanisms underlying radiation-induced osteoclastogenesis have yet to be fully elucidated. CR6-interacting factor-1 (Crif1) as a multifunctional protein is involved in regulating multiple biological functions in cells. Here, we investigated the role of Crif1 in radiation-induced osteoclastogenesis and found that radiation exposure induced an increase in the expression level of Crif1 and enhanced osteoclastogenesis in osteoclast progenitors. Crif1 and NF-κB p65 co-localized in the cytoplasm after radiation exposure. Crif1 knockdown did not affect the phosphorylation and total protein levels of extracellular signal-regulated kinases (ERK), c-Jun amino (N)-terminal kinases (JNK), p38, and IκB-α before and after irradiation. However, Crif1 knockdown did lead to the reduced phosphorylation and nuclear translocation of NF-κB p65 after irradiation and resulted in a reduced level of osteoclastogenesis in RAW264.7 cells after irradiation. In vivo studies involving Lyz2Cre;Crif1fl/fl mice possessing the myeloid-specific deletion of Crif1 demonstrated the alleviation of bone loss after irradiation when compared with Crif1fl/fl mice. Our findings demonstrate that Crif1 mediated the phosphorylation and nuclear translocation of NF-κB p65 and promoted osteoclastogenesis via the NF-κB signaling pathway after radiation exposure. Thus, our analysis revealed a specific role for Crif1 in the mediation of radiation-induced bone loss and may provide new insight into potential therapeutic strategies for radiation-induced bone loss.


Assuntos
Reabsorção Óssea , NF-kappa B , Camundongos , Animais , NF-kappa B/metabolismo , Osteogênese , Transdução de Sinais , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteoclastos/efeitos da radiação , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Diferenciação Celular , Proteínas de Ciclo Celular/metabolismo
10.
Platelets ; 34(1): 2267676, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37849076

RESUMO

Inherited thrombocytopenia (IT) is a group of hereditary disorders characterized by a reduced platelet count as the main clinical manifestation, and often with abnormal platelet function, which can subsequently lead to impaired hemostasis. In the past decades, humanized mouse models (HMMs), that are mice engrafted with human cells or genes, have been widely used in different research areas including immunology, oncology, and virology. With advances of the development of immunodeficient mice, the engraftment, and reconstitution of functional human platelets in HMM permit studies of occurrence and development of platelet disorders including IT and treatment strategies. This article mainly reviews the development of humanized mice models, the construction methods, research status, and problems of using humanized mice for the in vivo study of human thrombopoiesis.


Humanized mouse models (HMMs) refer to immunodeficient mice that have been used for the investigation of human hematopoiesis and immunity for years. With engrafted human hematopoietic stem cells (HSCs), the differentiation process of HSCs and re-construction of platelets can be monitored in the mice. Until now, several strains of HMMs have been used in the studies of inherited thrombocytopenia (IT), a genetic disorder associated with low platelet count in the blood. In this study, we reviewed the development of these HMMs in IT studies, compared the different sources of HSCs transplanted into HMMs and summarize the strategies of HSC transplantation in HMMs. The Kit−/− immunodeficient mice showed effectively long-term and stable implantation of human HSC without irradiation and higher implantation levels, and they also support multilinear differentiation of human HSC, such as platelets and red blood cells. The source and count of HSCs and the transplantation strategy may also impact the result. This study provides a basis information for HMMs used in IT and will help other investigators in this field choosing the right research plan.


Assuntos
Transtornos Plaquetários , Transplante de Células-Tronco Hematopoéticas , Trombocitopenia , Animais , Camundongos , Humanos , Modelos Animais de Doenças , Plaquetas , Trombopoese , Trombocitopenia/genética , Transplante de Células-Tronco Hematopoéticas/métodos
11.
Int J Biol Macromol ; 253(Pt 1): 126499, 2023 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-37659484

RESUMO

Although a high prevalence of benign prostate hyperplasia (BPH) has been documented, the risk factors are poorly understood. Metabolic syndrome increases the risk of BPH. Succinylation, a type of posttranslational modification, mostly targets metabolic processes. The level of succinylation was investigated in 4 BPH patients and 4 healthy controls. Additionally, 176 patients with BPH were analyzed by using pan-antisuccinyllysine antibody blotting. TMT-labeling proteomic and sc-RNAseq Cellchat analyses were employed to identify key signaling factors involved in the development of BPH. In vivo and in vitro experiments were used to confirm the role of integrin receptors. The global succinylation level in BPH was higher than that in the healthy prostate. Positive correlations of prostate volume with IHC score sand urodynamics testing were found in large clinical cohorts. The extracellular matrix (ECM), metabolic processes and immune signaling were involved in succinylation in BPH, as indicated by using TMT-labeling proteomic analysis, and this finding was also confirmed by sc-RNAseq CellChat analysis. The proteins upregulated in SIRT5 knockout WPMY-1 cells were also enriched in the extracellular matrix and metabolic processes. More importantly, integrin receptor inhibition in a mouse model of BPH significantly ameliorated prostate hyperplasia. High levels of succinylation modifications were found in BPH, and succinylated proteins influenced the activation of the ECM. Inhibition of ECM signaling further ameliorated prostate hyperplasia in mice.


Assuntos
Hiperplasia Prostática , Masculino , Humanos , Camundongos , Animais , Próstata/metabolismo , Hiperplasia/complicações , Hiperplasia/metabolismo , Hiperplasia/patologia , Proteômica , Matriz Extracelular/metabolismo
12.
Front Oncol ; 13: 1271492, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37692858

RESUMO

[This corrects the article DOI: 10.3389/fonc.2022.1009948.].

13.
Oncol Rep ; 50(5)2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37711054

RESUMO

As a newly identified circular RNA (circRNA), the role of circBLNK in cancer progression has not been probed. The objective of the present study was to functionally dissect the role of circBLNK in osteosarcoma (OS) tumorigenesis and progression. With regards of the experimental procedure, the levels of mRNAs and proteins were assessed using reverse transcription­quantitative PCR and western blot analysis, respectively. The subcellular location of circBLNK in OS cells was determined by cell cytosolic/nuclear fractionation assay. Cell ferroptosis ability was assessed through MTT assay. Cell proliferative abilities were assessed by clonogenic and Cell Counting Kit­8 assays, and cell apoptosis was measured using flow cytometry. The relationships among circBLNK, miR­188­3p, and glutathione peroxidase 4 (GPX4) were validated by luciferase reporter and RNA pull­down assays, as well as RNA immunoprecipitation. The stability of circBLNK and linear BLNK was confirmed using RNase R treatment assay. The association between circBLNK expression and overall survival rate was assessed by Kaplan­Meier plot. The correlation between the expression levels of circBLNK, miR­188­3p, and GPX4 in OS tissues was assessed by Pearson's χ2 test. The results revealed that CircBLNK and GPX4 were significantly upregulated in OS tissues, which predicted the poor prognosis. CircBLNK knockdown led to suppressed cell proliferation and enhanced cell apoptosis, an effect that could be reversed by the inhibition of miR­188­3p. In an in vivo circBLNK deficiency model, tumor growth was observed to be markedly suppressed. Moreover, circBLNK deficiency elevated levels of intracellular free iron (Fe2+), malondialdehyde, lipid reactive oxygen species and mitochondrial superoxide, while diminishing mitochondrial membrane potential in Erastin­treated OS cells, which were eliminated by overexpressing GPX4. Furthermore, mechanistic investigations revealed that circBLNK sponged miR­188­3p to regulate the expression of GPX4, thereby affecting OS progression. In conclusion, the present study delineated a new regulatory axis involving circBLNK/miR­188­3p/GPX4 in OS progression, adding to the growing evidence that circRNAs are critical gene regulators in cancer progression.


Assuntos
Neoplasias Ósseas , Ferroptose , MicroRNAs , Osteossarcoma , Humanos , RNA Circular/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Ferroptose/genética , Osteossarcoma/genética , Neoplasias Ósseas/genética , MicroRNAs/genética
14.
J Med Chem ; 66(19): 13568-13586, 2023 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-37751283

RESUMO

Extracellular signal-regulated kinase 5 (ERK5) is recognized as a key member of the mitogen-activated protein kinase family and is involved in tumor growth, migration, and angiogenesis. However, the results of ERK5 inhibition in multiple studies are controversial, and a highly specific ERK5-targeting agent is required to confirm physiological functions. Using proteolysis-targeting chimera technology, we designed the selective ERK5 degrader PPM-3 and examined its biological effect on cancer cells. Interestingly, the selective degradation of ERK5 with PPM-3 did not influence tumor cell growth directly. Based on proteomics analysis, the ERK5 deletion may be associated with tumor immunity. PPM-3 influences tumor development by affecting the differentiation of macrophages. Therefore, PPM-3 is an effective small-molecule tool for studying ERK5 and a promising immunotherapy drug candidate.

15.
Biomed Mater ; 18(5)2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37567189

RESUMO

Chemodynamic therapy (CDT) is a rising technology for cancer therapy by converting intracellular hydrogen peroxide (H2O2) into hydroxyl radical (•OH) via transition-metal-containing nanoparticles (NPs) catalysis reaction (i.e. Fenton reaction) to kill tumor cells. Highly efficient Fenton reaction and favorable delivery of the catalytic NPs 'nanoenzyme' are the key for successful treatment of cancer. In this work, we developed a novel nanoenzyme MnFe2O4@GFP forin vitroandin vivoantitumor therapy. A new MnFe2O4nanoparticle containing two transition-metal-element Fe and Mn was synthesized for enhanced Fenton reaction and used to co-deliver protein with high biocompatibility through post-modification with dopamine polymerization, green fluorescent protein adsorption, and PEG coating. The enrichment of H2O2and glutathione (GSH) in tumor tissue provided a favorable microenvironment forin situgeneration of toxic free radicals. Fe3+and GSH triggered a redox reaction to produce Fe2+, which in turn catalyzed H2O2into •OH, with the consumption of antioxidant GSH. By combining Fe3+with another catalyzer, the catalytic efficiency of the nanoenzyme were greatly improved. Consequently, the nanoenzyme showed efficient antitumor ability bothin vitroandin vivo. Thus, the multifunctional CDT nanoenzyme platform shows great promising for antitumor therapy through the combination of catalyzers Fe3+and Mn2+and codelivery of protein cargo.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Neoplasias , Humanos , Peróxido de Hidrogênio , Adsorção , Antioxidantes , Dopamina , Glutationa , Proteínas de Fluorescência Verde , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico , Microambiente Tumoral
16.
Acta Biomater ; 169: 434-450, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37516418

RESUMO

Radiotherapy is a mainstream modality for breast cancer treatment that employs ionizing radiation (IR) to damage tumor cell DNA and elevate ROS stress, which demonstrates multiple clinically-favorable advantages including localized treatment and low invasiveness. However, breast cancer cells may activate the p53-mediated cell cycle regulation in response to radiotherapy to repair IR-induced cellular damage and facilitate post-treatment survival. F-Box and WD Repeat Domain Containing 7 (FBXW7) is a promoter of p53 degradation and critical nexus of cell proliferation and survival events. Herein, we engineered a cooperative radio-ferroptosis-stimulatory nanomedicine through coordination-driven self-assembly between ferroptosis-inducing Fe2+ ions and FBXW7-inhibiting DNAzymes and further modification of tumor-targeting dopamine-modified hyaluronic acid (HA). The nanoassembly could be selectively internalized by breast cancer cells and disintegrated in lysosomes to release the therapeutic payload. DNAzyme readily abolishes FBXW7 expression and stabilizes phosphorylated p53 to cause irreversible G2 phase arrest for amplifying post-IR tumor cell apoptosis. Meanwhile, the p53 stabilization also inhibits the SLC7A11-cystine-GSH axis, which combines with the IR-upregulated ROS levels to amplify Fe2+-mediated ferroptotic damage. The DNAzyme-Fe-HA nanoassembly could thus systematically boost the tumor cell damaging effects of IR, presenting a simple and effective approach to augment the response of breast cancer to radiotherapy. STATEMENT OF SIGNIFICANCE: To overcome the intrinsic radioresistance in breast cancer, we prepared co-assembly of Fe2+ and FBXW7-targeted DNAzymes and modified surface with dopamine conjugated hyaluronic acid (HA), which enabled tumor-specific FBXW7-targeted gene therapy and ferroptosis therapy in IR-treated breast cancers. The nanoassembly could be activated in acidic condition to release the therapeutic contents. Specifically, the DNAzymes could selectively degrade FBXW7 mRNA in breast cancer cells to simultaneously induce accumulation of p53 and retardation of NHEJ repair, eventually inducing irreversible cell cycle arrest to promote apoptosis. The p53 stabilization would also inhibit the SLC7A11/GSH/GPX4 axis to enhance Fe2+ mediated ferroptosis. These merits could act in a cooperative manner to induce pronounced tumor inhibitory effect, offering new approaches for tumor radiosensitization in the clinics.


Assuntos
Neoplasias da Mama , DNA Catalítico , Proteínas F-Box , Ferroptose , Humanos , Feminino , Proteína 7 com Repetições F-Box-WD/genética , Proteína 7 com Repetições F-Box-WD/metabolismo , DNA Catalítico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias da Mama/genética , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína Supressora de Tumor p53/genética , Dopamina , Ácido Hialurônico , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Pontos de Checagem do Ciclo Celular
17.
J Colloid Interface Sci ; 646: 78-88, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37182261

RESUMO

The exploitation of high-performance electrode materials is significant to develop supercapacitors with satisfied energy and power output properties. In this study, a g-C3N4/Prussian-blue analogue (PBA)/Nickel foam (NF) with hierarchical micro/nano structures was developed by a simple salts-directed self-assembly approach. In this synthetic strategy, NF acted as both 3D macroporous conductive substrate and Ni source for PBA formation. Moreover, the incidental salt in molten salt-synthesized g-C3N4 nanosheets could regulate the combination mode between g-C3N4 and PBA to generate interactive networks of g-C3N4 nanosheets-covered PBA nano-protuberances on NF surfaces, which further expended the electrode/electrolyte interfaces. Based on the merits from this unique hierarchical structure and the synergy effect of PBA and g-C3N4, the optimized g-C3N4/PBA/NF electrode exhibited a maximum areal capacitance of 3366 mF cm-2 at current of 2 mA cm-2, as well as 2118 mF cm-2 even under large current of 20 mA cm-2. The solid-state asymmetric supercapacitor using g-C3N4/PBA/NF electrode possessed an extended working potential window of 1.8 V, prominent energy density of 0.195 mWh cm-2 and power density of 27.06 mW cm-2. Compared to the device with pure NiFe-PBA electrode, a better cyclic stability with capacitance retention rate of 80% after 5000 cycles was also achieved due to the protective effect of g-C3N4 shells on the etching of PBA nano-protuberances in electrolyte. This work not only builds a promising electrode material for supercapacitors, but also provide an effective way to apply molten salt-synthesized g-C3N4 nanosheet without purification.

18.
ACS Appl Mater Interfaces ; 15(12): 15893-15906, 2023 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-36940438

RESUMO

The hypopermeability and hypoxia in the tumor milieu are important factors that limit multiple treatments. Herein, the reactive oxygen species (ROS)-triggered self-assembled nanoparticles (RP-NPs) was constructed. The natural small molecule Rhein (Rh) was encapsulated into RP-NPs as a sonosensitizer highly accumulated at the tumor site. Then highly tissue-permeable ultrasound (US) irradiation induced apoptosis of tumor cells through the excitation of Rh and acoustic cavitation, which prompted the rapid production of large amounts of ROS in the hypoxic tumor microenvironment. In addition, the thioketal bond structures in the innovatively designed prodrug LA-GEM were triggered and broken by ROS to achieve rapid targeted release of the gemcitabine (GEM). Sonodynamic therapy (SDT) increased the tissue permeability of solid tumors and actively disrupted redox homeostasis via mitochondrial pathways to kill hypoxic tumor cells, and the triggered response mechanism to GEM synergistically amplified the effect of chemotherapy. The chemo-sonodynamic combinational treatment approach is highly effective and noninvasive, with promising applications for hypoxic tumor elimination, such as in cervical cancer (CCa) patients who want to maintain their reproductive function.


Assuntos
Nanopartículas , Neoplasias , Hipóxia Tumoral , Espécies Reativas de Oxigênio/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Espaço Intracelular , Microambiente Tumoral , Sistemas de Liberação de Medicamentos , Gencitabina/química , Gencitabina/farmacologia , Terapia Combinada , Humanos , Animais , Camundongos , Células HeLa
19.
Front Oncol ; 12: 1009948, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263222

RESUMO

Sustaining proliferative signaling and enabling replicative immortality are two important hallmarks of cancer. The complex of cyclin-dependent kinase (CDK) and its cyclin plays a decisive role in the transformation of the cell cycle and is also critical in the initiation and progression of cancer. CRIF1, a multifunctional factor, plays a pivotal role in a series of cell biological progresses such as cell cycle, cell proliferation, and energy metabolism. CRIF1 is best known as a negative regulator of the cell cycle, on account of directly binding to Gadd45 family proteins or CDK2. In addition, CRIF1 acts as a regulator of several transcription factors such as Nur77 and STAT3 and partly determines the proliferation of cancer cells. Many studies showed that the expression of CRIF1 is significantly altered in cancers and potentially regarded as a tumor suppressor. This suggests that targeting CRIF1 would enhance the selectivity and sensitivity of cancer treatment. Moreover, CRIF1 might be an indispensable part of mitoribosome and is involved in the regulation of OXPHOS capacity. Further, CRIF1 is thought to be a novel target for the underlying mechanism of diseases with mitochondrial dysfunctions. In summary, this review would conclude the latest aspects of studies about CRIF1 in cancers and mitochondria-related diseases, shed new light on targeted therapy, and provide a more comprehensive holistic view.

20.
Front Oncol ; 12: 983724, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36185270

RESUMO

Cancer is still a leading cause of death worldwide and liquid biopsy is a powerful tool that can be applied to different stages of cancer screening and treatment. However, as the second most abundant cell type in the bloodstream, platelets are isolated through well-established and fast methods in clinic but their value as a BioSource of cancer biomarkers is relatively recent. Many studies demonstrated the bidirectional interaction between cancer cells and platelets. Platelets transfer various proteins (e.g., growth factors, cytokine, chemokines) and RNAs (e.g., mRNA, lncRNA, miRNA, circRNA) into the tumor cells and microenvironment, leading the stimulation of tumor growth and metastasis. In turn, the platelet clinical characteristics (e.g., count and volume) and contents (e.g., RNA and protein) are altered by the interactions with cancer cells and this enables the early cancer detection using these features of platelets. In addition, platelet-derived microparticles also demonstrate the prediction power of being cancer biomarkers. In this review, we focus on the clinical applications of platelet detection using the platelet count, mean platelet volume, platelet RNA and protein profiles for human cancers and discuss the gap in bringing these implementations into the clinic.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA