Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Adv Mater ; : e2401145, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38692574

RESUMO

Photopyroptosis is an emerging research branch of photodynamic therapy (PDT), whereas there remains a lack of molecular structural principles to fabricate photosensitizers for triggering a highly efficient pyroptosis. Herein, a general and rational structural design principle to implement this hypothesis, is proposed. The principle relies on the clamping of cationic moieties (e.g., pyridinium, imidazolium) onto one photosensitive core to facilitate a considerable mitochondrial targeting (both of the inner and the outer membranes) of the molecules, thus maximizing the photogenerated reactive oxygen species (ROS) at the specific site to trigger the gasdermin E-mediated pyroptosis. Through this design, the pyroptotic trigger can be achieved in a minimum of 10 s of irradiation with a substantially low light dosage (0.4 J cm⁻2), compared to relevant work reported (up to 60 J cm⁻2). Moreover, immunotherapy with high tumor inhibition efficiency is realized by applying the synthetic molecules alone. This structural paradigm is valuable for deepening the understanding of PDT (especially the mitochondrial-targeted PDT) from the perspective of pyroptosis, toward the future development of the state-of-the-art form of PDT.

2.
Aging Dis ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38502587

RESUMO

UDP-GalNAc polypeptide N-acetylgalactosaminyltransferases (GalNAc-Ts) catalyze mucin-type O-glycosylation by transferring α-N-acetylgalactosamine (GalNAc) from UDP-GalNAc to Ser or Thr residues of target proteins. This post-translational modification is common in eukaryotes, yet its biological functions remain unclear. Recent studies have identified specific receptors in the heart and vascular wall cells that can be mucin-type O-glycosylated, and there is now substantial evidence confirming that patients with various cardiovascular diseases (CVDs), such as heart failure, coronary artery disease, myocardial hypertrophy, and vascular calcification, exhibit abnormal changes in GalNAc-Ts. This review aims to highlight recent advances in GalNAc-Ts and their roles in the cardiovascular system, intending to provide evidence for clinical treatment and prevention of CVDs.

3.
Mol Cell Biochem ; 479(3): 653-664, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37155089

RESUMO

Pleckstrin homeolike domain, family A, member 1 (PHLDA1) is a multifunctional protein that plays diverse roles in A variety of biological processes, including cell death, and hence its altered expression has been found in different types of cancer. Although studies have shown a regulatory relationship between p53 and PHLDA1, the molecular mechanism is still unclear. Especially, the role of PHLDA1 in the process of apoptosis is still controversial. In this study, we found that the expression of PHLDA1 in human cervical cancer cell lines was correlated with the up-expression of p53 after treatment with apoptosis-inducing factors. Subsequently, the binding site and the binding effect of p53 on the promoter region of PHLDA1 were verified by our bioinformatics data analysis and luciferase reporter assay. Indeed, we used CRISPR-Cas9 to knockout the p53 gene in HeLa cells and further confirmed that p53 can bind to the promoter region of PHLDA1 gene, and then directly regulate the expression of PHLDA1 by recruiting P300 and CBP to change the acetylation and methylation levels in the promoter region. Finally, a series of gain-of-function experiments further confirmed that p53 re-expression in HeLap53-/- cell can up-regulate the reduction of PHLDA1 caused by p53 knockout, and affect cell apoptosis and proliferation. Our study is the first to explore the regulatory mechanism of p53 on PHLDA1 by using the p53 gene knockout cell model, which further proves that PHLDA1 is a target-gene in p53-mediated apoptosis, and reveals the important role of PHLDA1 in cell fate determination.


Assuntos
Fatores de Transcrição , Proteína Supressora de Tumor p53 , Humanos , Apoptose , Células HeLa , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/genética
5.
J Control Release ; 354: 701-712, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36690036

RESUMO

The therapeutic application of chemodynamic therapy (CDT) is severely limited by the insufficient intracellular H2O2 and acidity in tumor. Herein, an acid-sensitive nanoplatform (ZIF67-ICG/TAM@GOx) to promote H2O2 and acidity enhancement through intracellular cyclic amplification for enhanced CDT is rationally designed. Notably, the acidic conditions of the tumor microenvironment (TME) can turn on the switch of the nanoplatform, setting free the loaded tamoxifen (TAM) and indocyanine green (ICG). The mitochondrial respiration inhibitor TAM and the superoxide dismutase-mimicking ZIF67 synergistically lead to an increase in the content of O2 and H2O2, accelerating the depletion of ß-d-glucose by GOx to generate gluconate and H2O2. The gluconate in turn boosts the acidity to facilitate the collapse of nanoparticles, further significantly promoting the accumulation of intracellular H2O2 through a positive circulation. Consequently, the amplificated endogenous H2O2 is catalyzed by Co2+ to liberate hydroxyl radicals (•OH). Besides, ICG-mediated photothermal therapy (PTT) and GOx-induced starvation therapy along with CDT realize the synergistic cancer treatment. Importantly, in vitro and in vivo experiments verified that the nanoplatform performed superior specificity and excellent therapeutic responses. The smart nanoplatform overcomes H2O2 and acidity deficiency simultaneously for intensive CDT, providing new prospects for the development of biocompatible cancer synergistic therapy strategies.


Assuntos
Nanopartículas , Neoplasias , Humanos , Peróxido de Hidrogênio , Terapia Fototérmica , Gluconatos , Glucose , Verde de Indocianina , Tamoxifeno , Microambiente Tumoral , Linhagem Celular Tumoral
6.
J Am Chem Soc ; 145(4): 2252-2263, 2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36657461

RESUMO

It is highly desirable to fabricate an accessible catalyst surface that can efficiently activate reactants and desorb products to promote the local surface reaction equilibrium in heterogeneous catalysis. Herein, rare-earth oxycarbonates (Ln2O2CO3, where Ln = La and Sm), which have molecular-exchangeable (H2O and CO2) surface structures according to the ordered layered arrangement of Ln2O22+ and CO32- ions, are unearthed. On this basis, a series of Ln2O2CO3-supported Cu catalysts are prepared through the deposition precipitation method, which provides excellent catalytic activity and stability for the water-gas shift (WGS) reaction. Density functional theory calculations combined with systematic experimental characterizations verify that H2O spontaneously dissociates on the surface of Ln2O2CO3 to form hydroxyl by eliminating the carbonate through the release of CO2. This interchange efficiently promotes the WGS reaction equilibrium shift on the local surface and prevents the carbonate accumulation from hindering the active sites. The discovery of the unique layered structure provides a so-called "self-cleaning" active surface for the WGS reaction and opens new perspectives about the application of rare-earth oxycarbonate nanomaterials in C1 chemistry.

7.
Eur J Pharmacol ; 939: 175447, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36473594

RESUMO

Vascular calcification (VC) is the pathological deposition of calcium and phosphate minerals in blood vessels, which is a common complication of atherosclerosis. Polypeptide N-acetylgalactosamine transferase 3 (GALNT3) initiates O-glycosylation of proteins through addition of GalNAc to specific serine or threonine residues. Our previous studies revealed the potent role of GALNT3 in atherosclerosis, whereas the precise mechanisms remain obscure. This study investigated the regulatory effect and mechanism of GALNT3 on VC. Firstly, GALNT3 was overexpressed and knocked down by adenovirus in high-phosphate induced calcified HASMCs and overexpressed by adeno-associated virus in vitamin D3-induced arterial calcification mice. We showed that the calcium deposition and mRNA expression of osteogenic markers MSX2, ALPL, and Runx2 were all significantly reduced with GALNT3 overexpression. Moreover, overexpression of GALNT3 significantly down-regulated the expression of the oxidative stress markers Nox2 and Nox4, up-regulated total antioxidant capacity, decreased the expression of pro-inflammatory factors IL-1ß, TNF-α and IL-8, matrix metalloproteinases MMP2 and MMP9, as well as reduced the apoptosis of cells in phosphate induced HASMCs. Furthermore, Vicia Villosa Lectin (VVL) pull down and TNFR1 immunoprecipitation assays showed that GALNT3 overexpression increased O-GalNAcylation of TNFR1 and blocked the activation of NF-κB signaling pathway. In addition, GALNT3 attenuates vitamin D3-induced aortic calcification in mice by alleviating oxidative stress and apoptosis of smooth muscle cells. In conclusion, this study indicates that GALNT3 protects against VC by reducing oxidative stress, vascular inflammation, and apoptosis of smooth muscle cells through the TNFR1/NF-κB signaling pathway. Thus, GALNT3 may be a potential therapeutic target for VC.


Assuntos
Aterosclerose , Calcificação Vascular , Camundongos , Animais , Receptores Tipo I de Fatores de Necrose Tumoral/genética , NF-kappa B/metabolismo , Cálcio/metabolismo , Calcificação Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Estresse Oxidativo , Apoptose , Fosfatos/metabolismo , Aterosclerose/patologia , Células Cultivadas
8.
Cell Signal ; 100: 110477, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36162588

RESUMO

Vascular calcification (VC) acts as a notable risk factor in the cardiovascular system. Disorder of phosphorus (Pi) metabolism promotes VC. Recent findings show that polypeptide N-acetylgalactosaminyltransferase 3(GALNT3) is Pi responsive and with potent effects on Pi homeostasis. However, whether GALNT3 is involved in high Pi-induced VC remains unclear. The present study investigated the potential role of GALNT3 as a novel regulator of VC. In vitro, human aortic smooth muscle cells (HASMCs) calcification was induced by inorganic Pi, while in vivo, C57BL/6 J mice were used to determine the effects of GALNT3 on Vitamin D3-induced medial arterial calcification. Alizarin red staining, Von Kossa staining, calcium and alkaline phosphatase (ALP) activity were performed to test VC. We showed that expression of GALNT3 was increased in the calcified HASMCs and aortas of the calcified mice.In vitro, overexpression of GALNT3 increased the levels of active full-length FGF23, accompanied by suppression of the osteoblast-related factors (Runx2 and BMP2), and further inhibited the formation of calcified nodules. Moreover, the protein levels of Wnt3a and active ß-catenin were determined and it was found that GALNT3 significantly inhibited their expression. LiCl, a Wnt/ß-catenin signaling activator, was observed to reverse the protective effect of GALNT3 overexpression. The opposite results were observed in the GALNT3 knockdown cells. In vivo, overexpression of GALNT3 by adeno-associated virus decreased the serum Pi and slowed the formation of aortic calcification in the calcified mice. In conclusion, our results indicate that GALNT3 counteracts high Pi-induced osteoblastic differentiation of VSMCs and protects against the initiation and progression of VC by inhibiting the Wnt/ß-catenin signaling pathway.


Assuntos
Músculo Liso Vascular , Miócitos de Músculo Liso , N-Acetilgalactosaminiltransferases , Calcificação Vascular , Animais , Humanos , Camundongos , beta Catenina/metabolismo , Células Cultivadas , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fosfatos/efeitos adversos , Fosfatos/farmacologia , Calcificação Vascular/induzido quimicamente , Calcificação Vascular/genética , Calcificação Vascular/metabolismo , Calcificação Vascular/prevenção & controle , Via de Sinalização Wnt , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Colecalciferol/efeitos adversos , Colecalciferol/farmacologia , Polipeptídeo N-Acetilgalactosaminiltransferase
9.
J Agric Food Chem ; 70(35): 10899-10906, 2022 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-35998392

RESUMO

Despite their status of being widely used as food additives, bisulfite (HSO3-)/sulfite (SO32-) can pose serious health risks when they are excessively added. Therefore, it is vital to develop a new method for detecting HSO3-/SO32- in foodstuff. In this paper, a benzopyran-benzothiazole derivative (probe DCA-Btl) with near-infrared emission was designed and synthesized by constructing a "push-pull" electronic system. DCA-Btl can selectively recognize HSO3-/SO32- via a colorimetric and fluorescence dual channel in DMF/PBS (1:1, v/v, pH = 8.4), and the emission wavelength of DCA-Btl can reach 710 nm. The fluorescence quenching of DCA-Btl after recognition of HSO3- is attributed to the photoinduced electron transfer (PET) process of the adduct DCA-Btl-HSO3- as evaluated by the DFT/TD-DFT method. In addition, DCA-Btl has many advantages, including a large Stokes shift (95 nm), good anti-interference ability, and little cytotoxicity. What's more, DCA-Btl has been successfully applied for the detection of HSO3-/SO32- in actual water samples and food samples such as sugar, red wine, and biscuits with satisfying results, as well as for fluorescent imaging of HSO3- in living MCF-7 cells.


Assuntos
Colorimetria , Corantes Fluorescentes , Colorimetria/métodos , Humanos , Tomografia por Emissão de Pósitrons , Sulfitos , Água
10.
J Control Release ; 348: 660-671, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35716884

RESUMO

Since there are several limitations in cancer treatment for traditional chemotherapy, such as side effects, poor prognosis and drug resistance, developing new combined therapy is urgently needed. In this work, a biocompatible, simple and tumor microenvironment-responsive nanotheranostics (PCN-Oxpt/PEG) was built to favor the chemotherapy/ferroptosis/immunomodulation synergism in cancer. This nanotheranostics is constructed by modifying oxaliplatin prodrug and PEG on Fe(III) - porphyrin metal-organic frameworks (PCN(Fe) MOFs). After intravenous injection, the cloak of PEG leads to long circulation, and the Fe(III)-porphyrin MOFs enables dual-model guidance with fluorescence (FL) and magnetic resonance imaging (MRI). Inside the tumor, the intracellular H2O2 would be transferred into hydroxyl radicals (•OH) by iron ions released from MOFs, which could trigger the lethal ferroptosis to cancer cells. Meanwhile, oxaliplatin(II) transformed from the loaded oxaliplatin prodrug would result in the chemotherapy, as well as immunogenic cell death (ICD), and the prodrug strategy could also avoid the occurring of liver damage by the direct administration of oxaliplatin(II). It was noticed that the ferroptosis effect was enhanced by triple-assistance during the combined therapy, as followed: (1) glutathione (GSH) would be consumed in the process of oxaliplatin(II) generation from oxaliplatin prodrug; (2) the increased CD8+ T cells induced by ICD were able to produce interferon-γ (IFN-γ), which could inhibit the transport of cystine by tumor cells, and impair the activation of glutathione peroxidase 4 (GPX4); (3) the amount of H2O2 could be increased by the internalized oxaliplatin and thus further promote the Fenton reaction and ferroptosis. Both in vivo and in vitro results revealed that tumor growing was significantly inhibited by PCN-Oxpt/PEG, taken together, the concomitant of oxaliplatin-mediated chemotherapy and ICD with triple-enhanced ferroptosis offer great prospect in the clinical treatment of cancer.


Assuntos
Ferroptose , Neoplasias , Porfirinas , Pró-Fármacos , Humanos , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Compostos Férricos/uso terapêutico , Glutationa/metabolismo , Peróxido de Hidrogênio , Nanoestruturas , Neoplasias/patologia , Oxaliplatina , Porfirinas/uso terapêutico , Pró-Fármacos/uso terapêutico , Microambiente Tumoral
11.
Polymers (Basel) ; 14(12)2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35745990

RESUMO

Room-temperature afterglow (RTA) materials have a wide range of applications in imaging, lighting, and therapy, due to their long lifetime and persistent luminescence after the light source is removed. Additionally, near-infrared light with low energy and a high penetration rate ensures its irreplaceable importance in imaging and therapy. Thus, it is vital to design RTA materials excited by NIR. In the present study, we select up-conversion nanoparticles (UCNPs) as the donor and add them into hybrids, obtained by dispersing coronene tetra-carboxylate salt (CS) into a polyvinyl alcohol (PVA)-substrate through a series of mixing methods. Through radiation energy transfer between the donor UCNPs and the acceptor CS, a kind of RTA film with a photoluminescence lifetime of more than 2 s under NIR excitation was successfully achieved, and these films could maintain persistent naked-eye-distinguishable luminescence after withdrawing the excitation light source. Furthermore, the films obtained from UCNP doping into CS/PVA hybrids were found to exhibit better RTA performance than those from smearing. This idea of up-conversion afterglow broadens the tuning and application scope for polymer-based luminescent materials.

12.
Int Immunopharmacol ; 107: 108685, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35306282

RESUMO

BACKGROUND: Anti-TNF treatment has played a vital role in treating Inflammatory Bowel Disease (IBD). However, T helper 17 (Th17) cells, which facilitate the development of IBD, are not reduced and even increased during anti-TNF clinical studies. Therefore, inhibition of Th17 cells is of great significance for improving anti-TNF treatment. METHOD: DSS-induced colitis mice were treated with the anti-TNF nanobody V7 and ANX1, and the variation in intestinal Th17 cells and DCs was estimated by flow cytometry. RAW264.7 cells were used to study the differentiation mechanism of Th17 cells from colon and mesenteric lymphocyte nodes (mLN). RESULTS: Intestinal Th17 cells increased after DSS-induced colitis was well treated with V7 (10 mg/kg). Th17 cell differentiation induced by V7 was accounted for by the accumulation of the V7-TNF complex, which was phagocytized by lamina propria (LP) DCs and induced the upregulation of MHCII. V7-TNF complex phagocytized RAW264.7 (CPR) was constructed and directly induced Th17 cell differentiation in colon LPLs and mLN in vitro. After knocking down CIITA in RAW264.7 cells, the induction was inhibited. Furthermore, Annexin 1 (ANX1) was upregulated and activated the FPR2-STAT3 pathway to inhibit the differentiation of Th17 cells. Then, animal assay demonstrated that ANX1 (500 µg/kg) enhanced the therapeutic effect of V7 (10 mg/kg) on DSS-induced colitis accompanied by a decrease in Th17 cells in mLN and colon. CONCLUSION: The differentiation of Th17 cells induced by V7 was mediated by phagocytosis of V7-TNF complexes by DCs and regulated by exogenous ANX1 via activation of the FPR2-STAT3 pathway. The combination of V7 and ANX1 presented a better therapeutic effect than monotherapy on DSS-induced colitis.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Animais , Anexinas/metabolismo , Anexinas/farmacologia , Anexinas/uso terapêutico , Diferenciação Celular , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Colo , Sulfato de Dextrana/farmacologia , Modelos Animais de Doenças , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Th17 , Inibidores do Fator de Necrose Tumoral
13.
Acta Biomater ; 138: 463-477, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34718179

RESUMO

Photodynamic therapy (PDT) is an effective noninvasive therapeutic strategy that can convert oxygen to highly cytotoxic singlet oxygen (1O2) through the co-localization of excitation light and photosensitizers. However, compromised by the hypoxic tumor microenvironment, the therapeutic efficacy of PDT is reduced seriously. Herein, to overcome tumor-associated hypoxia, and further achieve tumor-targeted synergistic chemotherapy/PDT/photothermal therapy (PTT), we have constructed a biodegradable oxygen-producing nanoplatform (named Ini@PM-HP), which was composed of the porous metal-organic framework (PCN-224(Mn)), the poly (ADP-ribose) polymerase (PARP) inhibitor (Iniparib), and the polydopamine-modified hyaluronic acid (HA-PDA). Since HA can specifically bind to the overexpressed HA receptors (cluster determinant 44, CD44) on tumor cell, Ini@PM-HP prefers to accumulate at the tumor site once injected intravenously. Then iniparib can be released in tumor environment (TME), thereby dysfunctioning DNA damage repair and promoting cell apoptosis. At the same time, the chelating of Mn and tetrakis(4-carboxyphenyl) porphyrin (Mn-TCPP) can generate O2 in situ by reacting with endogenous H2O2, relieving the hypoxic TME and achieving enhanced PDT. Moreover, owing to the high photothermal conversion efficiency of PDA, PTT can be driven by the 808 nm laser irradiation. As systematically demonstrated in vitro and in vivo, this nanotherapeutic approach enables the combined therapy with great inhibition on tumor. Overall, the as-prepared nanoplatform provide a promising strategy to overcome tumor-associated hypoxia, and shows great potential for combination tumor therapy. STATEMENT OF SIGNIFICANCE: A delicately designed biodegradable oxygen-producing nanoplatform Ini@PM-HP is constructed to achieve combination therapy of solid tumors. Taking advantage of the active-targeting, PTT, enhanced PDT and PARPi, this nanotherapeutic approach successfully enables the combined chemo/photothermal/photodynamic therapy with great inhibition of solid tumors.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Fotoquimioterapia , Linhagem Celular Tumoral , Peróxido de Hidrogênio , Manganês , Estruturas Metalorgânicas/farmacologia , Oxigênio , Fármacos Fotossensibilizantes/farmacologia , Terapia Fototérmica
14.
ACS Appl Mater Interfaces ; 14(1): 57-68, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-34935343

RESUMO

Integrating chemodynamic therapy (CDT) and photodynamic therapy (PDT) into one nanoplatform can produce much more reactive oxygen species (ROS) for tumor therapy. Nevertheless, it is still a great challenge to selectively generate sufficient ROS in tumor regions. Meanwhile, CDT and PDT are restricted by insufficient H2O2 content in the tumor as well as by the limited tumor tissue penetration of the light source. In this study, a smart pH/ROS-responsive nanoplatform, Fe2+@UCM-BBD, is rationally designed for tumor combination therapy. The acidic microenvironment can induce the pH-responsive release of doxorubicin (DOX), which can induce tumor apoptosis through DNA damage. Beyond that, DOX can promote the production of H2O2, providing sufficient materials for CDT. Of note, upconversion nanoparticles at the core can convert the 980 nm light to red and green light, which are used to activate Ce6 to produce singlet oxygen (1O2) and achieve upconversion luminescence imaging, respectively. Then, the ROS-responsive linker bis-(alkylthio)alkene is cleaved by 1O2, resulting in the release of Fenton reagent (Fe2+) to realize CDT. Taken together, Fe2+@UCM-BBD exhibits on-demand therapeutic reagent release capability, excellent biocompatibility, and remarkable tumor inhibition ability via synergistic chemo/photodynamic/chemodynamic combination therapy.


Assuntos
Antineoplásicos/uso terapêutico , Doxorrubicina/uso terapêutico , Portadores de Fármacos/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Fármacos Fotossensibilizantes/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Clorofilídeos/química , Clorofilídeos/efeitos da radiação , Clorofilídeos/uso terapêutico , Terapia Combinada , Doxorrubicina/química , Portadores de Fármacos/química , Portadores de Fármacos/efeitos da radiação , Liberação Controlada de Fármacos , Tratamento Farmacológico , Érbio/química , Érbio/efeitos da radiação , Érbio/uso terapêutico , Feminino , Fluoretos/química , Fluoretos/efeitos da radiação , Fluoretos/uso terapêutico , Humanos , Ferro/química , Ferro/efeitos da radiação , Ferro/uso terapêutico , Nanopartículas Metálicas/química , Nanopartículas Metálicas/efeitos da radiação , Camundongos Endogâmicos BALB C , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Itérbio/química , Itérbio/efeitos da radiação , Itérbio/uso terapêutico , Ítrio/química , Ítrio/efeitos da radiação , Ítrio/uso terapêutico
15.
Small ; 17(47): e2103919, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34623753

RESUMO

Given that traditional anticancer therapies fail to significantly improve the prognoses of triple negative breast cancer (TNBC), new modalities with high efficiency are urgently needed. Herein, by mixing the metal-phenolic network formed by tannic acid (TA), bleomycin (BLM), and Fe3+ with glutathione peroxidase 4 (GPX4) inhibitor (ML210) loaded hollow mesoporous Prussian blue (HMPB) nanocubes, the HMPB/ML210@TA-BLM-Fe3+ (HMTBF) nanocomplex is prepared to favor the ferroptosis/apoptosis synergism in TNBC. During the intracellular degradation, Fe3+ /Fe2+ conversion mediated by TA can initiate the Fenton reaction to drastically upregulate the reactive oxygen species level in cells, subsequently induce the accumulation of lipid peroxidation, and thereby cause ferroptotic cell death; meanwhile, the released ML210 efficiently represses the activity of GPX4 to activate ferroptosis pathway. Besides, the chelation of Fe2+ with BLM leads to in situ BLM toxification at tumor site, then triggers an effective apoptosis to synergize with ferroptosis for tumor therapy. As a result, the superior in vivo antitumor efficacy of HMTBF is corroborated in a 4T1 tumor-bearing mice model regarding tumor growth suppression, indicating that the nanoformulations can serve as efficient ferroptosis and apoptosis inducers for use in combinatorial TNBC therapy.


Assuntos
Ferroptose , Nanopartículas , Neoplasias de Mama Triplo Negativas , Animais , Apoptose , Bleomicina , Linhagem Celular Tumoral , Ferrocianetos , Humanos , Camundongos , Polifenóis , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
16.
J Nanobiotechnology ; 19(1): 261, 2021 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-34481495

RESUMO

BACKGROUND: Chemodynamic therapy (CDT), employing Fenton or Fenton-like catalysts to convert hydrogen peroxide (H2O2) into toxic hydroxyl radicals (·OH) to kill cancer cells, holds great promise in tumor therapy due to its high selectivity. However, the therapeutic effect is significantly limited by insufficient intracellular H2O2 level in tumor cells. Fortunately, ß-Lapachone (Lapa) that can exert H2O2-supplementing functionality under the catalysis of nicotinamide adenine dinucleotide (phosphate) NAD(P)H: quinone oxidoreductase-1 (NQO1) enzyme offers a new idea to solve this problem. However, extensive DNA damage caused by high levels of reactive oxygen species can trigger the "hyperactivation" of poly(ADP-ribose) polymerase (PARP), which results in the severe interruption of H2O2 supply and further the reduced efficacy of CDT. Herein, we report a self-amplified nanocatalytic system (ZIF67/Ola/Lapa) to co-deliver the PARP inhibitor Olaparib (Ola) and NQO1-bioactivatable drug Lapa for sustainable H2O2 production and augmented CDT ("1 + 1 + 1 > 3"). RESULTS: The effective inhibition of PARP by Ola can synergize Lapa to enhance H2O2 formation due to the continuous NQO1 redox cycling. In turn, the high levels of H2O2 further react with Co2+ to produce the highly toxic ·OH by Fenton-like reaction, dramatically improving CDT. Both in vitro and in vivo studies demonstrate the excellent antitumor activity of ZIF67/Ola/Lapa in NQO1 overexpressed MDA-MB-231 tumor cells. Importantly, the nanocomposite presents minimal systemic toxicity in normal tissues due to the low NQO1 expression. CONCLUSIONS: This design of nanocatalytic system offers a new paradigm for combing PARP inhibitor, NQO1-bioactivatable drug and Fenton-reagents to obtain sustained H2O2 generation for tumor-specific self-amplified CDT.


Assuntos
Antineoplásicos/farmacologia , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Apoptose , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/metabolismo , Camundongos , NAD(P)H Desidrogenase (Quinona) , Nanopartículas , Naftoquinonas , Poli(ADP-Ribose) Polimerase-1 , Espécies Reativas de Oxigênio/metabolismo
17.
Cell Death Discov ; 7(1): 198, 2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34326318

RESUMO

Increasing evidence suggests the pivotal role of hematopoietic pre-B-cell leukemia transcription factor (PBX)-interacting protein (HPIP/PBXIP1) in cancer development and progression, indicating that HPIP inhibition may be a promising target for cancer therapy. Here, we screened compounds inhibiting breast cancer cell proliferation with HPIP fused with green fluorescent protein as a reporter. A novel agent named TXX-1-10 derived from rimonabant, an antagonist of cannabinoid receptor 1 with anticancer effects, has been discovered to reduce HPIP expression and has greater inhibitory effects on breast cancer cell growth and metastasis in vitro and in vivo than rimonabant. TXX-1-10 regulates HPIP downstream targets, including several important kinases involved in cancer development and progression (e.g., AKT, ERK1/2, and FAK) as well as cell cycle-, apoptosis-, migration-, and epithelial-to-mesenchymal transition (EMT)-related genes. Consistent with the results of anticancer effects, genome-wide RNA sequencing indicated that TXX-1-10 has more significant effects on regulation of the expression of genes related to DNA replication, cell cycle, apoptosis, cell adhesion, cell migration, and invasion than rimonabant. In addition, TXX-1-10 significantly regulated genes associated with the cell growth and extracellular matrix organization, many of which were shown to be regulated by HPIP. Moreover, compared with rimonabant, TXX-1-10 greatly reduces blood-brain barrier penetrability to avoid adverse central depressive effects. These findings suggest that HPIP inhibition may be a useful strategy for cancer treatment and TXX-1-10 is a promising candidate drug for cancer therapy.

18.
Small ; 17(13): e2007051, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33599061

RESUMO

Immune checkpoint inhibitor (ICI) therapy is considered to be a revolutionary anti-tumor strategy that may surpass other traditional therapies. Breast cancer is particularly suitable for it theoretically due to upregulation of programmed cell death 1 (PD-1) / programmed cell death ligand 1 (PD-L1) immune checkpoint pathway which exhausts the adaptive immune response mediated by T lymphocytes. However, its blockades exhibit very little effect in breast cancer, owing to the lack of T lymphocytes pre-infiltration and co-existing of intricate immune negative microenvironment including the macrophage-suppressed "Don't eat me" CD47 signal overexpression. Herein, a stimuli-responsive multifunctional nanoplatform (ZIF-PQ-PDA-AUN) is built. Its photothermal therapy can promote the infiltration of T lymphocytes in addition to ablating tumor cells and AUNP-12 and PQ912 further boost both the innate and adaptive immune reactions by cutting off PD-L1 and CD47 signals, respectively. In contrast to earlier single immunotherapy, the nanocomposites exhibit a stronger anti-tumor immune effect without obvious autoimmune side effects, promoting infiltration of T lymphocyte into the tumor site and strengthening phagocytosis of macrophages, even more exciting, significantly reversing pro-tumor M2-like tumor-associated macrophages (TAMs) to anti-tumor M1-like TAMs. The research may provide a promising strategy to develop high-efficient and low-toxic immunotherapy based on nanotechnology.


Assuntos
Neoplasias da Mama , Inibidores de Checkpoint Imunológico , Neoplasias da Mama/terapia , Feminino , Humanos , Imunoterapia , Fagocitose , Terapia Fototérmica , Microambiente Tumoral
19.
Chem Commun (Camb) ; 56(86): 13241-13244, 2020 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-33030161

RESUMO

Relying on an inhibitor-based probe, we reveal the clustered distribution of membrane PSMA by dSTORM imaging and uncover its potential interaction with folate receptor. This inhibitor-based strategy realizes more accurate labeling than antibody labeling, which would make it a powerful tool in the field of dSTORM imaging.


Assuntos
Antígenos de Superfície/análise , Corantes Fluorescentes/química , Glutamato Carboxipeptidase II/análise , Imagem Óptica , Neoplasias da Próstata/diagnóstico por imagem , Membrana Celular/química , Humanos , Masculino , Estrutura Molecular
20.
Nanoscale ; 12(41): 21234-21247, 2020 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-33063070

RESUMO

A combination of chemotherapy and phototherapy has been proposed as a promising treatment for esophageal cancer (EC). Irinotecan as a first-line treatment option is widely prescribed for metastatic EC, however, its clinical application is extremely restricted by the low conversion rate to SN38, severe myelosuppression and diarrhea. As a more potent active metabolite of irinotecan, SN38 is a better substitution for irinotecan, but the poor water solubility and the difficulty of encapsulation hindered its medical application. Herein, a multifunctional SN38-conjugated nanosystem (FA-PDA@PZM/SN38@BSA-MnO2, denoted as FA-PPSM) is designed for overcoming the above-mentioned drawbacks and achieving collaborative chemotherapy, photodynamic therapy (PDT) and photothermal therapy (PTT). The tumor acidic microenvironment induces decomposition of BSA-MnO2 nanoparticles into O2 and Mn2+, thus enhancing oxygen-dependent PDT efficacy; meanwhile, Mn2+ can be employed as a magnetic resonance imaging (MRI) contrast agent. Under 650 and 808 nm laser irradiation, the FA-PPSM nanocomposites exhibit superior antitumor efficacy in Eca-109-tumor bearing mice. Notably, there is low gastrointestinal toxicity and myelosuppression in the FA-PPSM treated mice compared with those treated with irinotecan (alone). Taken together, this work highlights the great potential of the FA-PPSM nanocomposites for MRI-guided chemotherapy in combination with endoscopic light therapy for esophageal cancer.


Assuntos
Neoplasias Esofágicas , Nanopartículas , Animais , Linhagem Celular Tumoral , Diarreia , Neoplasias Esofágicas/tratamento farmacológico , Irinotecano , Compostos de Manganês , Camundongos , Óxidos , Fototerapia , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA