Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
2.
J Colloid Interface Sci ; 663: 167-176, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38401438

RESUMO

Photothermal therapy (PTT), which utilizes nanomaterials to harvest laser energy and convert it into heat to ablate tumor cells, has been rapidly developed for lung tumor treatment, but most of the PTT-related nanomaterials are not degradable, and the immune response associated with PTT is unclear, which leads to unsatisfactory results of the actual PTT. Herein, we rationally designed and prepared a manganese ion-doped polydopamine nanomaterial (MnPDA) for immune-activated PTT with high efficiency. Firstly, MnPDA exhibited 57.2% photothermal conversion efficiency to accomplish high-efficiency PTT, and secondly, MnPDA can be stimulated by glutathione (GSH) to the release of Mn2+, and it can produce ·OH in a Fenton-like reaction with the overexpressed H2O2 and stimulate the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. These two synergistically can effectively remove lung tumor cells that have not been ablated by PTT, resulting in an 86.7% tumor suppression rate under laser irradiation of MnPDA in vivo, and further significantly activated the downstream immune response, as evidenced by an increased ratio of cytotoxic T cells to immunosuppressive Treg cells. Conclusively, the GSH degradable MnPDA nanoparticles can be used for photothermal therapy and cGAS-STING-activated immunotherapy of lung tumors, which provides a new idea and strategy for the future treatment of lung tumors.


Assuntos
Indóis , Neoplasias Pulmonares , Nanopartículas , Neoplasias , Polímeros , Humanos , Manganês , Peróxido de Hidrogênio , Terapia Fototérmica , Imunoterapia , Neoplasias Pulmonares/terapia , Glutationa
3.
J Control Release ; 363: 221-234, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37717657

RESUMO

Checkpoint blockade immunotherapy (CBI) have exhibited remarkable benefits for cancer therapy. However, the low responsivity of CBI hinders its application in treatment of bladder cancer. Ferroptosis shows potential for increasing the responsivity of CBI by inducing immunogenic cell death (ICD) process. Herein, we developed a mitochondrial-targeted liposome loaded with brequinar (BQR) (BQR@MLipo) for enhancing the mitochondrial-related ferroptosis in bladder cancer in situ. It could be found that BQR@MLipo could selectively accumulate into mitochondria and inactivate dihydroorotate dehydrogenase (DHODH), which induced extensive mitochondrial lipid peroxidation and ROS, finally triggering ferroptosis of bladder cancer cells to boost the release of intracellular damage-associated molecular patterns (DAMPs) such as calreticulin (CRT), adenosine triphosphate (ATP), high mobility group box 1 (HMGB1). In addition, BQR@MLipo further promoted the release of mtDNA into the cytoplasm to activate the cGAS-STING pathway for the secretion of IFN-ß, which would increase the cross-presentation of antigens by dendritic cells and macrophage phagocytosis. Furthermore, the in vivo studies revealed that BQR@MLipo could remarkably accumulate into the bladder tumor and successfully initiate the infiltration of CD8+ T cells into tumor microenvironment for enabling efficient CBI to inhibit bladder tumor growth. Therefore, BQR@MLipo may represent a clinically promising modality for enhancing CBI in bladder tumor.


Assuntos
Ferroptose , Neoplasias da Bexiga Urinária , Humanos , Linfócitos T CD8-Positivos , Inibidores de Checkpoint Imunológico , Lipossomos , Imunoterapia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Mitocôndrias , Linhagem Celular Tumoral , Microambiente Tumoral
4.
Clin Exp Pharmacol Physiol ; 50(11): 855-866, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37582493

RESUMO

One of the toxic side effects of methotrexate (MTX) is enteritis. Aucubin, an iridoid glycoside derived from traditional medicinal herbs, has been proven to have anti-inflammation, anti-apoptosis and anti-oxidation properties. This work explored the effect and mechanism of aucubin in treating MTX-induced enteritis in a rat model. Two doses of aucubin (5 and 10 mg/kg) were adopted for the assessment of its pharmacological activity. We observed that in rats with MTX-induced enteritis, the body weight and small intestinal weight decreased. The intestine barrier was injured, as reflected by pathological examinations and an increase in D-lactate and diamine oxidase concentration in serum. Intestinal inflammation was shown by the observation of macrophages in the intestine and the concentrations of inflammatory cytokines tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in serum. The NLR family pyrin domain containing 3 (NLRP3) inflammasome was shown to be activated by the enhancement of NLRP3, cleaved-caspase 1, IL-18 and IL-1ß. Moreover, autophagy was reflected by transmission electron microscopy as slightly induced, along with changes in autophagy-related markers microtubule-associated protein 1 light chain 3 (LC3) and Beclin1. Remarkably, aucubin treatment attenuated the MTX-induced disease activity index increase, intestinal damage, inflammatory response and NLRP3 inflammasome activation, but provoked autophagy. Rapamycin, an autophagy activator, showed similar therapeutic effects to aucubin on MTX-induced enteritis. However, 3-methyladenine, an autophagy inhibitor, reversed the protective effects of aucubin. These findings prompted the hypothesis that aucubin alleviates MTX-induced enteritis by aggravating autophagy. This study might provide evidence for further investigation on the therapeutic role of aucubin in MTX-resulted enteritis.


Assuntos
Enterite , Inflamassomos , Ratos , Animais , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Metotrexato/toxicidade , Autofagia , Enterite/induzido quimicamente , Enterite/tratamento farmacológico
5.
Nanoscale ; 15(32): 13272-13279, 2023 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-37526608

RESUMO

Metal fluoride conversion cathodes are promising for the production of cheap, sustainable, and high-energy lithium-ion batteries. Yet, such systems are plagued by active material dissolution that causes capacity fade and hinders commercialization. Here, a covalent netting strategy is proposed to overcome this hurdle. In a proof-of-concept design, polydopamine derived carbon-mediated covalent binding inhibited the dissolution, while the pyrolyzed bacterial cellulose netting structure furnished fast electronic and ionic transport pathways. We demonstrate high-capacity, high-rate and long-lasting stability attained at practical loading levels. Our investigations suggest that the covalent netting-enabled formation of a robust and efficient blocking layer, highly competent in suppressing the leaching, is key for a stable performance. The successful stabilization of metal difluorides in the absence of electrolyte engineering opens an avenue for their practical deployment in future higher-level but lower-cost batteries, and provides a solution to similar challenges encountered by other dissolving energy electrode materials.

6.
Adv Healthc Mater ; 12(6): e2202467, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36377480

RESUMO

Lacking blood vessels is one of the main characteristics of most solid tumors due to their rapid and unrestricted growth, which thus causes the inefficient delivery efficiency of nanomedicine and tumor hypoxia. Herein, a dual "unlocking" strategy to overcome these obstacles is proposed by combining engineered hybrid nanoparticles (named ZnPc@FOM-Pt) with dexamethasone (DXM). It is verified that pretreatment of tumors with DXM can increase intratumorally micro-vessel density (delivery "unlocking") to enhance the tumor delivery efficiency of ZnPc@FOM-Pt and decrease HIF-1α expression. Correspondingly, more Pt can catalyze tumor-overexpressed H2 O2 to produce oxygen to further cause hypoxia "unlocking," ultimately achieving boosted ZnPc-based photodynamic therapy in vivo (tumor inhibition rate: 99.1%). Moreover, the immunosuppressive tumor microenvironment is efficiently reversed and the therapeutic effect of anti-PD-L1-based immunotherapy is promoted by this newly designed nanomedicine. This dual "unlocking" strategy provides an innovative paradigm on simultaneously enhancing nanomedicine delivery efficacy and hypoxia relief for tumor therapy.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Nanomedicina , Hipóxia Tumoral , Neoplasias/tratamento farmacológico , Hipóxia/tratamento farmacológico , Imunoterapia , Linhagem Celular Tumoral , Microambiente Tumoral
7.
Adv Healthc Mater ; 12(2): e2201262, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36213949

RESUMO

Glucose oxidase (GOD)-mediated starvation therapy (ST) that causes intratumoral glucose depletion is a promising strategy for tumor treatment. However, the ultimate efficacy is inevitably limited by tumor hypoxia, as oxygen is a key component in the consumption of glucose by GOD. In this study, a kind of glutathione (GSH)-responsive organosilica hybrid micelles loaded with Mn3 O4 and GOD (denoted as Mn3 O4 @PDOMs-GOD) is ingeniously designed for enhanced ST and chemodynamic therapy (CDT). Specifically, the internalized Mn3 O4 @PDOMs-GOD in tumor cells consumes intracellular glucose and oxygen (O2 ) under the catalysis of GOD to generate hydrogen peroxide (H2 O2 ), which is subsequently decomposed by Mn3 O4 to liberate O2 . This cyclically regenerated O2 will form a virtuous cycle of O2 and H2 O2 compensation to enhance the ST outcome. Meanwhile, Mn3 O4 can oxidize and deplete the overexpressed GSH in the tumor microenvironment (TME) to release Mn2+ , which then catalyzes H2 O2 into highly toxic hydroxyl radicals (·OH) to accomplish chemodynamic therapy (CDT). Both in vitro and in vivo experiment results demonstrate the significant antitumor efficacy of Mn3 O4 @PDOMs-GOD by the cooperatively enhanced ST and CDT, suggesting the feasibility to develop promising therapeutic platforms with higher treatment efficacies.


Assuntos
Nanopartículas , Neoplasias , Humanos , Catálise , Glucose , Glucose Oxidase , Glutationa , Peróxido de Hidrogênio , Oxigênio , Microambiente Tumoral , Linhagem Celular Tumoral , Neoplasias/tratamento farmacológico
8.
Front Nutr ; 9: 934621, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35967807

RESUMO

Polydextrose is a nutrient supplement, which is widely applied in the food industry. The use of polydextrose in combination with prebiotics and probiotics has recently increased, whereas the fermentation properties of its blend have not yet been fully revealed. We evaluated the metabolic profile of polydextrose, inulin, and their blends by a batch in vitro fermentation of fifteen human fecal inocula. After 24 h of fermentation, polydextrose increased the production of gas, ammonia, and several short chain fatty acids, including propionate and butyrate, when compared to its blends, inulin, and fructo-oligosaccharides. Furthermore, polydextrose had the slowest degradation rate of all the carbohydrates tested, consistent with its partial fermentation in the distal colon. The 16S rRNA gene sequencing analysis of the gut microbiome exhibited significantly increased relative abundance of Clostridium_XVIII, Megamonas, Mitsuokella, and Erysipelotrichaceae_incertae_sedis in polydextrose compared to other carbohydrates. On the other hand, the blends of polydextrose and inulin (1:1 or 2:1) showed reduced gas production and similar bifidogenicity to inulin alone. The blends not only had similar alpha-diversity and PCoA to inulin but also had a similar abundance of beneficial bacteria, such as Faecalibacterium and Roseburia, suggesting potential health benefits. Also their low gas production was likely due to the abundance of Faecalibacterium and Anaerostipes, which were negatively correlated with gas production. Additionally, our in vitro fermentation model shows advantages in the large-scale assessment of fermentation performance.

9.
Chem Asian J ; 17(17): e202200570, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35785417

RESUMO

Glucose oxidase (GOD)-based starvation therapy (ST), which inhibits the growth and proliferation of cancer cells by consuming glucose, has attracted intensive attention as an emerging non-invasive method for fighting cancers. However, the enzyme activity of GOD is greatly limited in vivo because of its optimal catalytic activity in the temperature range of 43-60 °C. Herein, a photothermal-enhanced starvation strategy is developed based on our engineered organosilica hybrid micelles (TiO2-x @POMs-GOD), in which the fluoride-doped TiO2-x with photothermal properties is encapsulated in the cores of organosilica cross-linked micelles and GOD is immobilized on the carboxyl groups of PAA segments. With its internalization by cancer cells, the conjugated GOD can effectively deplete glucose to achieve the ST effect, which can be remarkably enhanced by the loaded fluoride-doped TiO2-x with NIR laser irradiation, thus cooperatively contributing to the efficient treatment of TiO2-x @POMs-GOD on various cancer cells. This suggests great potential for TiO2-x @POMs-GOD in photothermal-enhanced ST in vivo.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Fluoretos , Glucose , Glucose Oxidase , Humanos , Micelas , Neoplasias/tratamento farmacológico , Fototerapia/métodos
10.
Biomater Sci ; 10(15): 4208-4217, 2022 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-35734909

RESUMO

Reactive oxygen species (ROS) based nanoplatforms have been considered as attractive and feasible candidates for cancer therapy. However, the activated endogenous antioxidant defense of cancer cells in response to the ROS attack greatly hinders their therapeutic efficacy. Although cancer-specific ROS amplification strategies have been widely explored, most of them suffer from tedious synthesis procedures and complex components, which will bring about undesired side effects and unsatisfactory results. Herein, we design a cancer-specific oxidative stress amplification nanomedicine (CA-Cu-PDA), which is simply fabricated through integrating the glutathione (GSH) responsive/depleting nanocarrier of copper-polydopamine (Cu-PDA) nanoparticles with a ROS-generating drug cinnamaldehyde (CA) via a facile one-pot polymerization route. It is verified that GSH could trigger the breakage of CA-Cu-PDA networks and the subsequent release of both copper ions and CA in cancer cells. The released copper ions efficiently oxidize GSH, thereby weakening the antioxidant system of cancer cells and increasing the ROS levels. On the other hand, extra ROS are generated by the reduced copper ions through a Fenton reaction, so that a synergistic ROS therapy with CA is achieved. Consequently, oxidative stress is specifically increased within cancer cells, leading to efficient cancer cell apoptosis, significant tumor suppression and minimized side effects. Such an ingenious structure realizes the interlocking cooperation and full utilization of each component's function, presenting promising perspectives for nanomedicine design.


Assuntos
Nanopartículas , Neoplasias , Antioxidantes , Linhagem Celular Tumoral , Cobre/uso terapêutico , Glutationa , Humanos , Indóis , Íons , Nanopartículas/química , Neoplasias/tratamento farmacológico , Polímeros , Espécies Reativas de Oxigênio
11.
J Mater Chem B ; 10(26): 5035-5044, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35726686

RESUMO

Photothermal therapy combined with chemotherapy based on nanomedicine has been considered a promising strategy for improving therapeutic efficacy in a tumor. However, nanomedicine can be easily cleared by the immune system without specific surface engineering modifications, thus affecting the ultimate efficacy. Herein, multifunctional biomimetic nanoparticles (Bio-RBCm@PDA@MSN-DOX) with enhanced long circulation and targeting ability are constructed by coating large pore-sized mesoporous silica (MSN) with polydopamine (PDA) layers in a biotin modified red blood cell membrane (Bio-RBCm) for efficient chemo/photothermal synergistic therapy. It is demonstrated that Bio-RBCm@PDA@MSN-DOX presents high photothermal conversion efficiency (40.17%) and enhanced capability to accelerate the release of the anticancer drug (doxorubicin, DOX), thus showing a good synergistic therapeutic effect in cell experiments. More importantly, with the assistance of the biotin and RBC membrane, Bio-RBCm@PDA@MSN-DOX can successfully evade immune clearance and effectively target transport to HeLa tumor sites, finally accomplishing up to 98.95% tumor inhibition with negligible side effects to normal tissues. This multilayer structure presents a valuable model for future therapeutic applications with safe and effective tumor chemotherapy and photothermal therapy.


Assuntos
Nanopartículas , Neoplasias , Biomimética , Biotina , Doxorrubicina/química , Humanos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Fototerapia , Terapia Fototérmica
12.
J Hazard Mater ; 434: 128858, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35405607

RESUMO

Porous membranes with fascinating super-wettable surface and tunable porous architecture for oil-water separation have been developed rapidly, however, the serious secondary marine pollution caused by the non-degradable defectiveness of membranes themselves is still a thorny problem. Herein, we create an eco-friendly membrane with biomimetic cobweb-like nanostructure via assembling two-dimensional bacterial cellulose nanonets on the starch nanofibrous membrane on a large scale. The obtained novel composite membranes exhibit integrated properties of sub-micron pore size, ultrahigh porosity, superhydrophilicity, and underwater superoleophobicity, stemming from the synergistic effect of the hydrated nanonet-skin-layer and porous starch matrix. By virtue of the narrow-distributed sub-micron pores, ultrahigh porosity, and ultrathin thickness, the resulting membrane shows outstanding performance of excellent separation efficiency (up to 99.996%), high percolation flux (maximum of 15968 L m-2 h-1), well surpassing the conventional microfiltration membranes. More significantly, with the advantage of biodegradability and anti-oil-fouling property, the membrane could serve as the robust platform for long-term wastewater remediation.


Assuntos
Purificação da Água , Biomimética , Membranas Artificiais , Amido , Águas Residuárias , Purificação da Água/métodos
13.
Crit Rev Eukaryot Gene Expr ; 32(1): 59-65, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35377981

RESUMO

Previous studies have revealed the role of lncRNA FAM230B in promoting papillary thyroid cancer and gastric cancer. We predicted that FAM230B may interact with miR-203 and studied the crosstalk between FAM230B and miR-203 in osteosarcoma (OS). Paired OS and nontumor tissues donated by 60 patients with OS were subjected to RNA isolations and quantitative real-time PCR (RT-qPCR) to analyze the expression of both FAM230B and miR-203 (mature and premature levels). Subcellular location of FAM230B in OS cells was detected using subcellular fractionation assay. RNA pull-down assay was performed to investigate the direct interaction between FAM230B and miR-203. Overexpression assays followed by RT-qPCR were performed to analyze the role of FAM230B in miR-203 maturation. Cell proliferation was studied with 5-Bromo-2-deoxyUridine (BrdU) assay. FAM230B and premature miR-203 were upregulated in OS, whereas mature miR-203 was downregulated in OS cells. FAM230B was detected in the cytoplasm and directly interacted with premature miR-203. FAM230B overexpression in OS cells increased premature miR-203 level and decreased mature miR-203 level. FAM230B increased cell proliferation and suppressed the role of miR-203 in inhibiting cell proliferation. FAM230B in the cytoplasm may sponge premature miR-203, thereby inhibiting miR-203 maturation to increase OS cell proliferation.


Assuntos
Neoplasias Ósseas , MicroRNAs , Osteossarcoma , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia
14.
Cell Death Dis ; 13(2): 160, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35181676

RESUMO

Long non-coding RNAs (lncRNAs) have been manifested to manipulate diverse biological processes, including tumor-induced immune tolerance. Thus, we aimed in this study to identify the expression pattern of lncRNA homeobox A cluster antisense RNA 2 (HOXA-AS2) in glioma and decipher its role in immune tolerance and glioma progression. We found aberrant upregulation of lncRNA HOXA-AS2, lysine demethylase 2A (KDM2A), and jagged 1 (JAG1) and a downregulation of microRNA-302a (miR-302a) in glioma specimens. Next, RNA immunoprecipitation, chromatin immunoprecipitation, and dual-luciferase reporter gene assay demonstrated that lncRNA HOXA-AS2 upregulated KDM2A expression by preventing miR-302a from binding to its 3'untranslated region. The functional experiments suggested that lncRNA HOXA-AS2 could promote regulatory T (Treg) cell proliferation and immune tolerance, which might be achieved through inhibition of miR-302a and activation of KDM2A/JAG1 axis. These findings were validated in a tumor xenograft mouse model. To conclude, lncRNA HOXA-AS2 facilitates KDM2A/JAG1 expression to promote Treg cell proliferation and immune tolerance in glioma by binding to miR-302a. These findings may aid in the development of novel antitumor targets.


Assuntos
Proteínas F-Box , Glioma , RNA Antissenso , RNA Longo não Codificante , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteínas F-Box/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Glioma/genética , Humanos , Tolerância Imunológica , Proteína Jagged-1/genética , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , MicroRNAs/metabolismo , RNA Antissenso/metabolismo , RNA Longo não Codificante/metabolismo
15.
Adv Sci (Weinh) ; 9(8): e2104671, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35038243

RESUMO

Starvation-dependent differential stress sensitization effect between normal and tumor cells provides a potentially promising strategy to amplify chemotherapy effects and reduce side effects. However, the conventional starvation approaches such as glucose oxidase (Gox)-induced glucose depletion and nanomedicine-enabled vascular embolism usually suffer from aggravated tumor hypoxia, systemic toxicity, and unpredictable metabolic syndrome. Herein, a novel "valve-closing" starvation strategy is developed to amplify the chemotherapy effects via closing the "valve" of glucose transported into tumor cells, which is accomplished by a glucose transporters 1 (GLUT1, valve of glucose uptake) inhibitor (Genistein, Gen) and chemotherapeutic agent (Curcumin, Cur) coloaded hybrid organosilica-micelles nanomedicine (designated as (Gen + Cur)@FOS) with controllable stability. In vitro and in vivo results demonstrate that (Gen + Cur)@FOS can effectively reduce glucose/adenosine triphosphate levels in tumor cells by inhibiting GLUT1 expression (i.e., "valve-closing") to induce the starvation of tumor cells, thus weakening the resistance of tumor cells to apoptosis caused by chemotherapy, and consequently contributing to the remarkably improved antitumor efficiency and minimized side effects based on the stress sensitization effect mediated by GLUT1 inhibition-induced starvation. This "valve-closing" starvation strategy provides a promising paradigm for the development of novel nanotherapeutics with amplified chemotherapy effect.


Assuntos
Antineoplásicos , Neoplasias , Antineoplásicos/uso terapêutico , Glucose Oxidase/uso terapêutico , Humanos , Micelas , Nanomedicina , Neoplasias/tratamento farmacológico
16.
Neural Regen Res ; 17(5): 1096-1105, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34558538

RESUMO

Long noncoding RNAs (lncRNAs) participate in many pathophysiological processes after traumatic brain injury by mediating neuroinflammation and apoptosis. Homeobox A11 antisense RNA (HOXA11-AS) is a member of the lncRNA family that has been reported to participate in many inflammatory reactions; however, its role in traumatic brain injury remains unclear. In this study, we established rat models of traumatic brain injury using a weight-drop hitting device and injected LV-HOXA11-AS into the right lateral ventricle 2 weeks before modeling. The results revealed that overexpression of HOXA11-AS aggravated neurological deficits in traumatic brain injury rats, increased brain edema and apoptosis, promoted the secretion of proinflammatory factors interleukin-1ß, interleukin-6, and tumor necrosis factor α, and promoted the activation of astrocytes and microglia. Microglia were treated with 100 ng/mL lipopolysaccharide for 24 hours to establish in vitro cell models, and then transfected with pcDNA-HOXA11-AS, miR-124-3p mimic, or sh-MDK. The results revealed that HOXA11-AS inhibited miR-124-3p expression and boosted MDK expression and TLR4-nuclear factor-κB pathway activation. Furthermore, lipopolysaccharide enhanced potent microglia-induced inflammatory responses in astrocytes. Forced overexpression of miR-124-3p or downregulating MDK repressed microglial activation and the inflammatory response of astrocytes. However, the miR-124-3p-mediated anti-inflammatory effects were reversed by HOXA11-AS. These findings suggest that HOXA11-AS can aggravate neuroinflammation after traumatic brain injury by modulating the miR-124-3p-MDK axis. This study was approved by the Animal Protection and Use Committee of Southwest Medical University (approval No. SMU-2019-042) on February 4, 2019.

17.
Oncol Res ; 28(9): 929-944, 2022 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-34544526

RESUMO

This phase II randomized clinical trial aimed to assess the efficacy and toxicity of Endostar, an antiangiogenesis inhibitor, combined with concurrent chemoradiotherapy (CCRT) for locally advanced cervical cancer (LACC). Patients with LACC were randomly assigned to either CCRT plus Endostar (CCRT+E arm) or CCRT alone (CCRT arm). All patients received pelvic intensity-modulated radiation therapy (IMRT) and brachytherapy. Weekly cisplatin was administered concurrently with IMRT. Patients in the CCRT+E arm also received concurrent Endostar every 3 weeks for two cycles. The primary endpoint was progression-free survival (PFS) and acute toxicities. The exploratory endpoint was the impact of vascular endothelial growth factor receptor-2 (VEGFR2) expression on long-term survival. A total of 116 patients were enrolled. Patients in the CCRT+E arm and in the CCRT arm had similar acute and late toxicity profile. The 1- and 2-year PFS were 91.4% versus 82.1% and 80.8% versus 63.5% (p=0.091), respectively. The 1- and 2-year distance metastasis-free survival (DMFS) were 92.7% versus 81.1% and 86.0% versus 65.1% (p=0.031), respectively. Patients with positive VEGFR2 expression had significant longer PFS and overall survival (OS) compared with those with negative VEGFR2 expression. Patients in the CCRT+E arm had significantly longer PFS, OS, and DMFS than those in the CCRT arm when VEGFR2 expression was positive. In conclusion, CCRT plus Endostar significantly improved DMFS but not PFS over CCRT alone. The addition of Endostar could significantly improve survival for patients with positive VEGFR2 expression.


Assuntos
Neoplasias Nasofaríngeas , Neoplasias do Colo do Útero , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Quimiorradioterapia , Cisplatino/uso terapêutico , Endostatinas/uso terapêutico , Feminino , Humanos , Neoplasias Nasofaríngeas/tratamento farmacológico , Proteínas Recombinantes , Resultado do Tratamento , Neoplasias do Colo do Útero/terapia , Fator A de Crescimento do Endotélio Vascular
18.
Adv Healthc Mater ; 11(2): e2101634, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34706163

RESUMO

In eukaryotes and prokaryotes, some copper transportations driven by gradient copper-binding affinities exhibit typical glutathione (GSH)-responsive features. Inspired by these delicate endogenous processes, a biomimic copper-ion mediated GSH-responsive nanomedicine is designed based on the gradient copper-binding strengths between polydopamine (PDA) species and GSH. The nanomedicine is constructed as core-shell nanoparticles with copper-polydopamine (Cu-PDA) coordinated shell and micellar core encapsulating chemotherapeutic drug of ß-lapachone (ß-lapa). In tumor cells, the excess intracellular GSH will reduce and extract the Cu(II) from the Cu-PDA network, triggered by the binding affinity gradients between Cu-PDA and Cu-GSH, resulting in the breaking of the shell and the releasing of ß-lapa and Fenton agent copper. The additional Fenton reaction of copper ions induces excess oxidative damage of tumor cells assisted by the abundant H2 O2 amplified by ß-lapa, achieving cascade anticancer effects combining chemodynamic therapy with chemotherapy. This multilevel anticancer system exhibits an efficient tumor inhibitory rate and a negligible systematic toxicity for normal organs in vivo, presenting a new bioinspired GSH-responsive strategie to develop stimuli-responsive structures.


Assuntos
Glutationa , Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Cobre/química , Glutationa/química , Humanos , Peróxido de Hidrogênio/química , Ferro/química , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Estresse Oxidativo
19.
Clin Lab ; 67(11)2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34758231

RESUMO

BACKGROUND: Immunoglobulin D multiple myeloma (IgD-MM) is a rare but aggressive disease. The safety and effectiveness of anti-CD38 monoclonal antibody (daratumumab) have not been known in either IgD-MM or MM complicated with secondary neoplasm. METHODS: A fragile IgD-MM patient had an aggressively relapsed disease concurrent with lung cancer and severe thrombocytopenia, which led to a dilemma for management. After a failure of ixazomib-based chemotherapy, a salvage therapy with daratumumab unexpectedly induced complete remission and platelet recovery, and the patient successfully proceeded to lung cancer surgery. CONCLUSIONS: Our case indicates daratumumab is both safe and effective for refractory IgD-MM with severe complications.


Assuntos
Neoplasias Pulmonares , Mieloma Múltiplo , Trombocitopenia , Anticorpos Monoclonais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Humanos , Imunoglobulina D , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/tratamento farmacológico , Mieloma Múltiplo/complicações , Mieloma Múltiplo/tratamento farmacológico , Trombocitopenia/induzido quimicamente , Trombocitopenia/tratamento farmacológico
20.
Oxid Med Cell Longev ; 2021: 3823122, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790286

RESUMO

BACKGROUND: Circular RNA phosphorylase kinase regulatory subunit alpha 2 (circPHKA2; hsa_circ_0090002) has a significantly, specifically different expression in acute ischemic stroke (AIS) patients' blood. Here, we intended to investigate the role and mechanism of circPHKA2 in oxygen-glucose deprivation- (OGD-) induced stoke model in human brain microvascular endothelial cells (HBMEC). METHODS: Expression of circPHKA2, microRNA- (miR-) 574-5p, and superoxide dismutase-2 (SOD2) was detected by quantitative PCR and western blotting. Cell injury was measured by detecting cell proliferation (EdU assay and CCK-8 assay), migration (transwell assay), neovascularization (tube formation assay), apoptosis (flow cytometry and western blotting), endoplasmic reticulum stress (western blotting), and oxidative stress (assay kits). Direct intermolecular interaction was determined by bioinformatics algorithms, dual-luciferase reporter assay, biotin-labelled miRNA capture, and argonaute 2 RNA immunoprecipitation. RESULTS: circPHKA2 was downregulated in AIS patients' blood in SOD2-correlated manner. Reexpressing circPHKA2 rescued EdU incorporation, cell viability and migration, tube formation, B cell lymphoma-2 (Bcl-2) expression, and SOD activity of OGD-induced HBMEC and alleviate apoptotic rate and levels of Bcl-2-associated protein (Bax), glucose-regulated protein 78 kD (GRP78), C/EBP-homologous protein (CHOP), caspase-12, reactive oxygen species (ROS), and malondialdehyde (MDA). Additionally, blocking SOD2 partially attenuated these roles of circPHKA2 overexpression. Molecularly, circPHKA2 upregulated SOD2 expression via interacting with miR-574-5p, and miR-574-5p could target SOD2. Similarly, allied to neurovascular protection of circPHKA2 was the downregulation of miR-574-5p. CONCLUSION: circPHKA2 could protect HBMEC against OGD-induced cerebral stroke model via the miR-574-5p/SOD2 axis, suggesting circPHKA2 as a novel and promising candidate in ischemic brain injury.


Assuntos
Endotélio Vascular/metabolismo , Glucose/deficiência , Hipóxia/complicações , AVC Isquêmico/terapia , MicroRNAs/genética , Fosforilase Quinase/genética , RNA Circular/administração & dosagem , Superóxido Dismutase/metabolismo , Estudos de Casos e Controles , Endotélio Vascular/patologia , Regulação da Expressão Gênica , Humanos , AVC Isquêmico/etiologia , AVC Isquêmico/metabolismo , AVC Isquêmico/patologia , RNA Circular/genética , Superóxido Dismutase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA