Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Adv Sci (Weinh) ; 11(15): e2306031, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38342617

RESUMO

Overproduction of reactive oxygen species (ROS), metal ion accumulation, and tricarboxylic acid cycle collapse are crucial factors in mitochondria-mediated cell death. However, the highly adaptive nature and damage-repair capabilities of malignant tumors strongly limit the efficacy of treatments based on a single treatment mode. To address this challenge, a self-reinforced bimetallic Mito-Jammer is developed by incorporating doxorubicin (DOX) and calcium peroxide (CaO2) into hyaluronic acid (HA) -modified metal-organic frameworks (MOF). After cellular, Mito-Jammer dissociates into CaO2 and Cu2+ in the tumor microenvironment. The exposed CaO2 further yields hydrogen peroxide (H2O2) and Ca2+ in a weakly acidic environment to strengthen the Cu2+-based Fenton-like reaction. Furthermore, the combination of chemodynamic therapy and Ca2+ overload exacerbates ROS storms and mitochondrial damage, resulting in the downregulation of intracellular adenosine triphosphate (ATP) levels and blocking of Cu-ATPase to sensitize cuproptosis. This multilevel interaction strategy also activates robust immunogenic cell death and suppresses tumor metastasis simultaneously. This study presents a multivariate model for revolutionizing mitochondria damage, relying on the continuous retention of bimetallic ions to boost cuproptosis/immunotherapy in cancer.


Assuntos
Peróxido de Hidrogênio , Neoplasias , Humanos , Espécies Reativas de Oxigênio , Trifosfato de Adenosina , Morte Celular , Mitomicina , Microambiente Tumoral
2.
Biochem Biophys Res Commun ; 671: 192-199, 2023 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-37302294

RESUMO

The therapeutic effects and application of radiotherapy are restricted to some extent due to low radiosensitivity of tumor tissues and adverse effects by excess dosage. Current radiosensitizers are confronted with problems in clinical translation because of complicated manufacture technique and high cost. In this research, we have synthesized a radiosensitizer with advantages in low cost and mass production, which could be applied to CT imaging and enhanced radiotherapy in breast cancer, namely Bi-DTPA. It not only enhanced tumor CT imaging which resulted in better therapeutic accuracy, but also realized radiotherapy sensitization by producing massive ROS and inhibit tumor proliferation, providing a sound perspective in the clinical translation of the radiosensitizer.


Assuntos
Neoplasias , Radiossensibilizantes , Humanos , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico , Tolerância a Radiação , Neoplasias/tratamento farmacológico , Ácido Pentético/farmacologia , Ácido Pentético/uso terapêutico , Tomografia Computadorizada por Raios X/métodos
3.
J Nanobiotechnology ; 21(1): 165, 2023 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-37221521

RESUMO

BACKGROUND: Oxidative stress (OS) induced by an imbalance of oxidants and antioxidants is an important aspect in anticancer therapy, however, as an adaptive response, excessive glutathione (GSH) in the tumor microenvironment (TME) acts as an antioxidant against high reactive oxygen species (ROS) levels and prevents OS damage to maintain redox homoeostasis, suppressing the clinical efficacy of OS-induced anticancer therapies. RESULTS: A naturally occurring ROS-activating drug, galangin (GAL), is introduced into a Fenton-like catalyst (SiO2@MnO2) to form a TME stimulus-responsive hybrid nanopharmaceutical (SiO2-GAL@MnO2, denoted SG@M) for enhancing oxidative stress. Once exposed to TME, as MnO2 responds and consumes GSH, the released Mn2+ converts endogenous hydrogen peroxide (H2O2) into hydroxyl radicals (·OH), which together with the subsequent release of GAL from SiO2 increases ROS. The "overwhelming" ROS cause OS-mediated mitochondrial malfunction with a decrease in mitochondrial membrane potential (MMP), which releases cytochrome c from mitochondria, activates the Caspase 9/Caspase 3 apoptotic cascade pathway. Downregulation of JAK2 and STAT3 phosphorylation levels blocks the JAK2/STAT3 cell proliferation pathway, whereas downregulation of Cyclin B1 protein levels arrest the cell cycle in the G2/M phase. During 18 days of in vivo treatment observation, tumor growth inhibition was found to be 62.7%, inhibiting the progression of pancreatic cancer. Additionally, the O2 and Mn2+ released during this cascade catalytic effect improve ultrasound imaging (USI) and magnetic resonance imaging (MRI), respectively. CONCLUSION: This hybrid nanopharmaceutical based on oxidative stress amplification provides a strategy for multifunctional integrated therapy of malignant tumors and image-visualized pharmaceutical delivery.


Assuntos
Peróxido de Hidrogênio , Neoplasias Pancreáticas , Humanos , Espécies Reativas de Oxigênio , Compostos de Manganês , Dióxido de Silício , Óxidos , Estresse Oxidativo , Antioxidantes , Microambiente Tumoral , Neoplasias Pancreáticas
4.
Nutrients ; 15(6)2023 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-36986171

RESUMO

Gestational diabetes mellitus (GDM) is a common disease of pregnancy, but with very limited knowledge of its impact on human milk oligosaccharides (HMOs) in breast milk. This study aimed to explore the lactational changes in the concentration of HMOs in exclusively breastfeeding GDM mothers and the differences between GDM and healthy mothers. A total of 22 mothers (11 GDM mothers vs. 11 healthy mothers) and their offspring were enrolled in the study and the levels of 14 HMOs were measured in colostrum, transitional milk, and mature milk. Most of the HMOs showed a significant temporal trend with decreasing levels over lactation; however, there were some exceptions for 2'-Fucosyllactose (2'-FL), 3-Fucosyllactose (3-FL), Lacto-N-fucopentaose II (LNFP-II), and Lacto-N-fucopentaose III (LNFP-III). Lacto-N-neotetraose (LNnT) was significantly higher in GDM mothers in all time points and its concentrations in colostrum and transitional milk were correlated positively with the infant's weight-for-age Z-score at six months postnatal in the GDM group. Significant group differences were also found in LNFP-II, 3'-Sialyllactose (3'-SL), and Disialyllacto-N-tetraose (DSLNT) but not in all lactational periods. The role of differently expressed HMOs in GDM needs to be further explored by follow-up studies.


Assuntos
Diabetes Gestacional , Leite Humano , Lactente , Feminino , Gravidez , Humanos , Lactação , Aleitamento Materno , Mães , Oligossacarídeos
5.
J Nanobiotechnology ; 21(1): 30, 2023 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-36698190

RESUMO

BACKGROUND: Combined therapy based on the effects of cascade reactions of nanoplatforms to combat specific solid tumor microenvironments is considered a cancer treatment strategy with transformative clinical value. Unfortunately, an insufficient O2 supply and the lack of a visual indication hinder further applications of most nanoplatforms for solid tumor therapy. RESULTS: A visualizable nanoplatform of liposome nanoparticles loaded with GOD, H(Gd), and PFP and grafted with the peptide tLyP-1, named tLyP-1H(Gd)-GOD@PFP, was constructed. The double-domain peptide tLyP-1 was used to specifically target and penetrate the tumor cells; then, US imaging, starvation therapy and sonodynamic therapy (SDT) were then achieved by the ultrasound (US)-activated cavitation effect under the guidance of MR/PA imaging. GOD not only deprived the glucose for starvation therapy but also produced H2O2, which in coordination with 1O2 produced by H(Gd), enable the effects of SDT to achieve a synergistic therapeutic effect. Moreover, the synergistic therapy was enhanced by O2 from PFP and low-intensity focused ultrasound (LIFU)-accelerated redox effects of the GOD. The present study demonstrated that the nanoplatform could generate a 3.3-fold increase in ROS, produce a 1.5-fold increase in the maximum rate of redox reactions and a 2.3-fold increase in the O2 supply in vitro, and achieve significant tumor inhibition in vivo. CONCLUSION: We present a visualizable nanoplatform with tumor-penetrating ability that can be unlocked by US to overcome the current treatment problems by improving the controllability of the O2 supply, which ultimately synergistically enhanced cascade therapy.


Assuntos
Retroalimentação Sensorial , Nanopartículas , Humanos , Peróxido de Hidrogênio , Linhagem Celular Tumoral , Nanopartículas/química , Peptídeos , Hipóxia
6.
Small ; 19(14): e2206174, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36651135

RESUMO

Multidrug resistance (MDR) and metastasis in cancer have become increasingly serious problems since antitumor efficiency is greatly restricted by a single therapeutic modality and the insensitive tumor microenvironment (TME). Herein, metal-phenolic network-functionalized nanoparticles (t-P@TFP NPs) are designed to realize multiple therapeutic modalities and reshape the TME from insensitive to sensitive under multimodal imaging monitoring. After a single irradiation, a near-infrared laser-activated multistage reaction occurs. t-P@TFP NPs trigger the phase transition of perfluoropentane (PFP) to release tannic acid (TA)/ferric ion (Fe3+ )-coated paclitaxel (PTX) and cause hyperthermia in the tumor region to efficiently kill cancer cells. Additionally, PTX is released after the disassembly of the TA-Fe3+ film by the abundant adenosine triphosphate (ATP) in the malignant tumor, which concurrently inhibits ATP-dependent drug efflux to improve sensitivity to chemotherapeutic agents. Furthermore, hyperthermia-induced immunogenic cell death (ICD) transforms "cold" tumors into "hot" tumors with the assistance of PD-1/PD-L1 blockade to evoke antitumor immunogenicity. This work carefully reveals the mechanisms underlying the abilities of these multifunctional NPs, providing new insights into combating the proliferation and metastasis of multidrug-resistant tumors.


Assuntos
Nanopartículas , Neoplasias , Humanos , Fototerapia/métodos , Paclitaxel/farmacologia , Neoplasias/terapia , Sistemas de Liberação de Medicamentos/métodos , Resistência a Múltiplos Medicamentos , Metais , Linhagem Celular Tumoral , Microambiente Tumoral
7.
ACS Appl Mater Interfaces ; 15(1): 309-326, 2023 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-36576435

RESUMO

Cancer multidrug resistance (MDR) is an important reason that results in chemotherapy failure. As a main mechanism of MDR, overexpressed P-glycoprotein (P-gp) utilizes adenosine triphosphate (ATP) to actively pump chemotherapy drugs out of cells. In addition, metabolic reprogramming of drug-resistant tumor cells (DRTCs) exacerbates the specific hypoxic microenvironment and promotes tumor metastasis and recurrence. Therefore, we propose a novel sonodynamic therapy (SDT) paradigm to induce energy metabolism disorder and drug resistance change of DRTCs. A US-controlled "Nanoenabled Energy Metabolism Jammer" (TL@HPN) is designed using perfluoropentane (PFP) adsorbing oxygen in the core, and a targeting peptide (CGNKRTR) is attached to the liposome as the delivery carrier shell to incorporate hematoporphyrin monomethyl ether (HMME) and paclitaxel (PTX). The TL@HPN with ultrasonic/photoacoustic imaging (PAI/USI) precisely controlled the release of drugs and oxygen after being triggered by ultrasound (US), which attenuated the hypoxic microenvironment. SDT boosted the reactive oxygen species (ROS) content in tumor tissues, preferentially inducing mitochondrial apoptosis and maximizing immunogenic cell death (ICD). Persistently elevated oxidative stress levels inhibited ATP production and downregulated P-gp expression by disrupting the redox balance and electron transfer of the respiratory chain. We varied the effect of TL@HPN combined with PD-1/PD-L1 to activate autoimmunity and inhibit tumor metastasis, providing a practical strategy for expanding the use of SDT-mediated tumor energy metabolism.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Resistência a Múltiplos Medicamentos , Espécies Reativas de Oxigênio/metabolismo , Oxigênio , Metabolismo Energético , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Microambiente Tumoral
8.
Adv Sci (Weinh) ; 10(4): e2204989, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36494092

RESUMO

The emergence of aggregation-induced emission luminogens (AIEgens) has attracted substantial scientific attention. However, their antitumor efficacy in photodynamic therapy (PDT) is significantly restricted by the poor water solubility and limited treatment depth. Therefore, a novel AIEgens-involved therapeutic platform with good permeability and bioavailability is urgently required. Herein, supramolecular chemistry is combined with the AIEgen bis-pyrene (BP) to construct a peptide-AIEgen hybrid nanosystem (PAHN). After intravenous injection, the versatile nanoplatform not only improved the hydrophilicity of BP but also achieved stratified targeting from tumor to mitochondrial and induced mitochondrial dysfunction, thus activating caspase-3 upregulation. Then, sonodynamic therapy (SDT), an alternative modality with high tissue penetrability, is performed to evoke reactive oxygen species (ROS) generation for BP. More importantly, since the hydrophilic shell is separated from the nanosystem by the specific cleavage of caspase-3, the resulting decrease in hydrophilicity induced tight self-aggregation of PAHN residues in situ, further allowing more absorbed energy to be used for ROS generation under ultrasound irradiation and enhancing SDT efficacy. Moreover, severe oxidative stress resulting from ROS imbalance in the mitochondria initiates the immunogenic cell death process, thus evoking antitumor immunogenicity. This PAHN provides prospective ideas into AIE-involved antitumor therapy and design of peptide-AIEgens hybrids.


Assuntos
Fotoquimioterapia , Caspase 3 , Espécies Reativas de Oxigênio , Estudos Prospectivos , Fotoquimioterapia/métodos , Peptídeos
9.
Int J Nanomedicine ; 17: 4547-4565, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36199475

RESUMO

Background: Sonodynamic therapy (SDT) has been regarded as a novel therapeutic modality for killing tumors. However, the hypoxic tumor microenvironment, especially deep-seated tumors distant from blood vessels, severely restricts therapeutic efficacy due to the oxygen-dependent manner of SDT. Methods: Herein, we report a novel ultrasonic cavitation effect-based therapeutic modality that is able to facilitate the hypoxia-tolerant SDT for inducing hypoxic tumor death. A tLyP-1 functionalized liposomes is fabricated, composed of hematoporphyrin monomethyl ether gadolinium as the sonosentizer and perfluoropentane (PFP) as the acoustic environment regulator. Moreover, the tLyP-1 functioned liposomes could achieve active tumor homing and effective deep-penetrating into hypoxic tumors. Upon low intensity focused ultrasound (LIFU) irradiation, the acoustic droplet vaporization effect of PFP induced fast liquid-to-gas transition and quick bubbles explosion to generate hydroxyl radicals, efficiently promoting cell death in both normoxic and hypoxic microenvironment (acting as deep-penetration nanobomb, DPNB). Results: The loading of PFP is proved to significantly enhance the therapeutic efficacy of hypoxic tumors. In particular, these DPNB can also act as ultrasound, photoacoustic, magnetic resonance, and near-infrared fluorescence tetramodal imaging agents for guiding the therapeutic process. Conclusion: This study is the first report involving that liquid-to-gas transition based SDT has the potential to combat hypoxic tumors.


Assuntos
Neoplasias , Terapia por Ultrassom , Linhagem Celular Tumoral , Gadolínio , Humanos , Hipóxia/diagnóstico por imagem , Hipóxia/terapia , Lipossomos , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Neoplasias/terapia , Oxigênio , Microambiente Tumoral , Terapia por Ultrassom/métodos
10.
Nutrients ; 14(16)2022 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-36014803

RESUMO

Background: Numerous studies have investigated the effects of the supplementation of fructooligosaccharides (FOS) on the number of bacteria in the gut that are good for health, but the results have been inconsistent. Additionally, due to its high fermentability, supplementation of FOS may be associated with adverse gastrointestinal symptoms such as bloating and flatulence. Therefore, we assessed the effects of FOS interventions on the composition of gut microbiota and gastrointestinal symptoms in a systematic review and meta-analysis. Design: All randomized controlled trials published before 10 July 2022 that investigated the effects of FOS supplementation on the human gut microbiota composition and gastrointestinal symptoms and met the selection criteria were included in this study. Using fixed or random-effects models, the means and standard deviations of the differences between the two groups before and after the intervention were combined into weighted mean differences using 95% confidence intervals (CIs). Results: Eight studies containing 213 FOS supplements and 175 controls remained in this meta-analysis. Bifidobacterium spp. counts significantly increased during FOS ingestion (0.579, 95% CI: 0.444−0.714) in comparison with that of the control group. Subgroup analysis showed greater variation in Bifidobacterium spp. in adults (0.861, 95% CI: 0.614−1.108) than in infants (0.458, 95% CI: 0.297−0.619). The increase in Bifidobacterium spp. counts were greater in the group with an intervention duration greater than 4 weeks (0.841, 95% CI: 0.436−1.247) than an intervention time less than or equal to four weeks (0.532, 95% CI: 0.370−0.694), and in the group with intervention doses > 5 g (1.116, 95% CI: 0.685−1.546) the counts were higher than those with doses ≤ 5 g (0.521, 95% CI: 0.379−0.663). No differences in effect were found between FOS intervention and comparators in regard to the abundance of other prespecified bacteria or adverse gastrointestinal symptoms. Conclusions: This is the first meta-analysis to explore the effect of FOS on gut microbiota and to evaluate the adverse effects of FOS intake on the gastrointestinal tract. FOS supplementation could increase the number of colonic Bifidobacterium spp. while higher dose (7.5−15 g/d) and longer duration (>4 weeks) showed more distinct effects and was well tolerated.


Assuntos
Microbioma Gastrointestinal , Adulto , Bactérias , Bifidobacterium , Suplementos Nutricionais , Humanos , Lactente , Oligossacarídeos/farmacologia , Ensaios Clínicos Controlados Aleatórios como Assunto
11.
Theranostics ; 12(6): 2987-3006, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35401832

RESUMO

Rationale: Multidrug resistance (MDR) and metastasis of breast cancer remain major hurdles in clinical anticancer therapy. The unsatisfactory outcome is largely due to insufficient cytotoxicity of chemotherapeutic agents and limited immunogenic cell death (ICD). On the other hand, efflux proteins, especially P-glycoprotein (P-gp), can recognize and promote the efflux of drugs from tumor cells. Methods: In this study, silver nanoparticles (Ag NPs) and peptide- functionalized doxorubicin (PDOX) were used to prepare a theranostic nanocomposite (Ag-TF@PDOX), which induced organelle-mediated immunochemotherapy and drug efflux protein inhibition in drug-resistant breast cancer cells (MCF-7/ADR) via a strategy based on endoplasmic reticulum (ER) stress and cell-nucleus penetration. Results: The silver nanoparticle-triggered persistent activation of ER stress synergizes with chemotherapy to enhance cytotoxicity and stimulate the ICD effect. It has the potential to enhance chemosensitivity by downregulating of P-gp expression due to the increased production of ATP-consuming chaperones. In addition, the novel peptide (CB5005), which not only penetrates the cell membrane but also has a nuclear localization sequence, is conjugated to DOX to improve both cellular internalization and intranuclear accumulation. Moreover, surface TA-Fe3+ engineering endows the nanocomposite with ATP-responsive disassembly and ATP depletion properties to improve biocompatibility and decrease ATP-dependent drug efflux. Ag-TF@PDOX has potential as a dual-mode (PAI/MRI) contrast-enhanced agent for realizing theranostic guidance. Conclusion: This theranostic nanocomposite greatly restricts the growth of drug-resistant breast tumors and activates a strong immune response as well, providing an opportunity for the development of therapeutics that reverse tumor MDR and metastasis at the subcellular level.


Assuntos
Neoplasias da Mama , Nanopartículas Metálicas , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Doxorrubicina/química , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Estresse do Retículo Endoplasmático , Feminino , Humanos , Células MCF-7 , Prata/metabolismo
12.
Small ; 18(13): e2107809, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35143709

RESUMO

The efficacy of immune checkpoint inhibition in inducing death of cancer cells is affected by the immunosuppressive "cold" tumor microenvironment, which results in a poor response by the patient's antitumor immune system. However, the immunomodulatory effects of immunogenic cell death in response to irritation by heat energy and reactive oxygen species (ROS) can switch the tumor microenvironment from "cold" to "hot." This study has developed a nanoadjuvant for immune therapy using iron tungsten oxide (FeWOx)-based nanosheets with surface PEGylation (FeWOx-PEG). This FeWOx-PEG nanoadjuvant serves as a chemodynamic reagent via the Fenton reaction and acts as a photosensitizer for photodynamic and photothermal therapy under near-infrared II laser irradiation; however, it could also be used to augment tumor-infiltrating T-cells and provoke a systemic antitumor immune response by combining the immunogenic cell death triggered by ROS and photothermal therapy with the immune checkpoint blockade. This research demonstrates that application of the FeWOx-PEG nanoadjuvant under the guidance of magnetic resonance/computed tomography/photoacoustic imaging can eliminate the primary tumor and suppress the growth of distant tumors.


Assuntos
Antígeno B7-H1 , Morte Celular Imunogênica , Linhagem Celular Tumoral , Humanos , Imunoterapia , Fototerapia , Terapia Fototérmica
13.
J Nanobiotechnology ; 19(1): 200, 2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34225744

RESUMO

BACKGROUND: Recent studies have demonstrated that multidrug resistance (MDR) is a critical factor in the low efficacy of cancer chemotherapy. The main mechanism of MDR arises from the overexpression of P-glycoprotein (P-gp), which actively enhances drug efflux and limits the effectiveness of chemotherapeutic agents. RESULTS: In this study, we fabricated a "combo" nanoagent equipping with triple synergistic strategies for enhancing antitumor efficacy against MDR cells. Tumor homing-penetrating peptide endows the nanosystem with targeting and penetrating capabilities in the first stage of tumor internalization. The abundant amine groups of polyethylenimine (PEI)-modified nanoparticles then trigger a proton sponge effect to promote endo/lysosomal escape, which enhances the intracellular accumulation and retention of anticancer drugs. Furthermore, copper tetrakis(4-carboxyphenyl)porphyrin (CuTCPP) encapsulated in the nanosystem, effectively scavenges endogenous glutathione (GSH) to reduce the detoxification mediated by GSH and sensitize the cancer cells to drugs, while simultaneously serving as a photoacoustic imaging (PAI) contrast agent for image visualization. Moreover, we also verify that these versatile nanoparticles in combination with PD-1/PD-L1 blockade therapy can not only activate immunological responses but also inhibit P-gp expression to obliterate primary and metastatic tumors. CONCLUSION: This work shows a significant enhancement in therapeutic efficacy against MDR cells and syngeneic tumors by using multiple MDR reversing strategies compared to an equivalent dose of free paclitaxel.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/farmacologia , Nanopartículas/uso terapêutico , Animais , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Cobre , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Tratamento Farmacológico , Feminino , Compostos Heterocíclicos , Humanos , Lisossomos , Células MCF-7 , Metaloporfirinas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanomedicina , Nanopartículas/química , Compostos Organofosforados , Paclitaxel/farmacologia
14.
Biomaterials ; 271: 120710, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33610047

RESUMO

Fenton reaction-mediated chemodynamic therapy (CDT), which destroys tumor cells by converting H2O2 into cytotoxic hydroxyl radical (OH) and singlet oxygen (1O2) species, is a promising field. However, Fenton-based CDT is severely impaired by the inappropriate tumor environment associated with undesirable intratumoral acidity and insufficient H2O2 supply in tumor microenvironment (TME). Therefore, a strategy that can address these concerns is highly desired and beneficial for boosting such treatment. Herein, a magnetic nanoreactor system (denoted as poly (lactic-co-glycolic acid) (PLGA)-superparamagnetic iron oxide (SPIO)&vitamin C (Vc) was constructed with Vc in the core, SPIO on the shell, and PLGA as the building carrier. Upon low-intensity focused ultrasound irradiation, on-demand Vc release can locally decompose into H2O2, which can generate a favorable condition for facilitating SPIO-based Fenton-like reaction and result in continuous O2 and OH/1O2 generation. The TME modulation-augmented CDT by this nanoreactor based on the reinforced Fenton reaction tremendously improved the antitumor outcomes, especially under increased accumulation contributed by magnetic targeting combined with enhanced permeability and retention effect. Moreover, the explosive production of oxygen can be monitored by real-time photoacoustic imaging, offering a noninvasive means to forecast the treatment efficacy. Therefore, this established microenvironment modulation strategy for augmenting Fenton reaction-based CDT paves a new avenue to realize highly efficient cancer theranostics.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Humanos , Peróxido de Hidrogênio , Radical Hidroxila , Neoplasias/tratamento farmacológico , Espécies Reativas de Oxigênio , Microambiente Tumoral
15.
J Am Chem Soc ; 143(5): 2413-2422, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33507066

RESUMO

The development of activatable photosensitizers to allow for the reversible control of singlet oxygen (1O2) production for photodynamic therapy (PDT) faces great challenges. Fortunately, the flourishing field of supramolecular biotechnology provides more effective strategies for activatable PDT systems. Here, we developed a new reversible PDT on a switch that controls the 1O2 generation of self-assembled albumin nanotheranostics in vitro and in vivo. A new molecular design principle of aggregation-induced self-quenching photochromism and albumin on-photoswitching was demonstrated using a new asymmetric, synthetic diarylethene moiety DIA. The photosensitizer porphyrin and DIA were incorporated as building blocks in a glutaraldehyde-induced covalent albumin cross-linking nanoplatform, HSA-DIA-porphyrin nanoparticles (NPs). More importantly, the excellent photoswitching property of DIA enables the resultant nanoplatform to act as a facile, switchable strategy for photodynamic-immunotherapy.


Assuntos
Albuminas/metabolismo , Hidrocarbonetos Aromáticos/química , Hidrocarbonetos Aromáticos/farmacologia , Imunoterapia/métodos , Fotoquimioterapia/métodos , Oxigênio Singlete/metabolismo , Linhagem Celular Tumoral , Humanos , Hidrocarbonetos Aromáticos/metabolismo
16.
Theranostics ; 11(4): 1953-1969, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33408791

RESUMO

Photodynamic therapy (PDT) efficacy in cancer cells is affected by sub-physiological hypoxia caused by dysregulated and "chaotic" tumor microvasculature. However, current traditional O2-replenishing strategies are undergoing their own intrinsic deficiencies. In addition, resistance mechanisms activated during PDT also lead the present situation far from satisfactory. Methods: We propose a nitric oxide (NO)-based theranostic nanoplatform by using biocompatible poly-lactic-co-glycolic acid nanoparticles (PLGA NPs) as carriers, in which the outer polymeric layer embeds chlorin e6 (Ce6) and incorporates L-Arginine (L-Arg). This nanoplatform (L-Arg@Ce6@P NPs) can reduce hyperactive O2 metabolism of tumor cells by NO-mediated mitochondrial respiration inhibition, which should raise endogenous O2 tension to counteract hypoxia. Furthermore, NO can also hinder oxidative phosphorylation (OXPHOS) which should cause intracellular adenosine triphosphate (ATP) depletion, inhibiting tumor cells proliferation and turning cells more sensitive to PDT. Results: When the L-Arg@Ce6@P NPs accumulate in solid tumors by the enhanced permeability and retention (EPR) effect, locally released L-Arg is oxidized by the abundant H2O2 to produce NO. In vitro experiments suggest that NO can retard hypoactive O2 metabolism and save intracellular O2 for enhancing PDT efficacy under NIR light irradiation. Also, lower intracellular ATP hinders proliferation of DNA, improving PDT sensitization. PDT phototherapeutic efficacy increased by combining these two complementary strategies in vitro/in vivo. Conclusion: We show that this NO-based nanoplatform can be potentially used to alleviate hypoxia and sensitize tumor cells to amplify the efficacy of phototherapy guided by photoacoustic (PA) imaging.


Assuntos
Trifosfato de Adenosina/metabolismo , Neoplasias da Mama/tratamento farmacológico , Nanopartículas/administração & dosagem , Óxido Nítrico/metabolismo , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/administração & dosagem , Animais , Apoptose , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Fármacos Fotossensibilizantes/química , Poliésteres/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Theranostics ; 11(6): 2670-2690, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33456566

RESUMO

Glucocorticoids are widely used in the treatment of nephritis, however, its dose-dependent side effects, such as the increased risk of infection and metabolic disturbances, hamper its clinical use. This study reports a visualized podocyte-targeting and focused ultrasound responsive glucocorticoid nano-delivery system (named as Dex/PFP@LIPs-BMS-α), which specific delivers dexamethasone (Dex) to podocyte targets and reduces systemic side effects. Methods: The glucocorticoid nano-delivery system was synthesized by a lipid thin film and a simple facile acoustic-emulsification method. This glucocorticoid nano-delivery system used BMS-470539 (BMS-α), a synthetic compound, as a "navigator" to specifically identify and target the melanocortin-1 receptor (MC-1R) on podocytes. The loaded perfluoropentane (PFP) realizes the directed "explosion effect" through ultrasound-targeted microbubble destruction (UTMD) technology under the coordination of low intensity focused ultrasound (LIFU) to completely release Dex. Results: Both in vitro and in vivo experiments have demonstrated that Dex/PFP@LIPs-BMs-α accurately gathered to podocyte targets and improved podocyte morphology. Moreover, in vivo, proteinuria and serum creatinine levels were significantly reduced in the group treated with Dex/PFP@LIPs-BMS-α, and no severe side effects were detected. Furthermore, Dex/PFP@LIPs-BMS-α, with capabilities of ultrasound, photoacoustic and fluorescence imaging, provided individualized visual guidance and the monitoring of treatment. Conclusion: This study provides a promising strategy of Dex/PFP@LIPs-BMS-α as effective and safe against immune-associated nephropathy.


Assuntos
Glucocorticoides/farmacologia , Nefropatias/tratamento farmacológico , Podócitos/efeitos dos fármacos , Animais , Células Cultivadas , Creatinina/sangue , Dexametasona/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Fluorocarbonos/farmacologia , Humanos , Imidazóis/farmacologia , Nefropatias/sangue , Nefropatias/metabolismo , Masculino , Podócitos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Melanocortina/metabolismo , Ultrassonografia/métodos
18.
Food Sci Nutr ; 8(10): 5402-5409, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33133542

RESUMO

To satisfy the demand of industrial production, selecting strains suitable for fermentation initiation is necessary. In this study, the effects of mixed-starter culture including Leuconostoc, Lactobacillus, and Weissella on the quality of Sichuan pickle were discussed. Results showed that mixed-starter culture can accelerate fermentation and had the highest efficiency for nitrite degradation, that is, the maximum nitrite concentration was 8.97 g/kg on day 1 and decreased to 1.88 mg/kg after 3 days. The mixed-starter culture improved the sensory properties of pickles, which easily produced acids but had reduced amounts of total acids. The pickle products fermented by the mixed-starter culture contained increased lactic acid (17.52 g/kg), mannitol (0.62%), umami (35.85), and sweet (11.36) amino acids on day 4. The strains Weissella paramesenteroides C2-2 and Lactobacillus brevis ZP11-2 grew well in the mixed-starter culture fermentation.

19.
Biomater Sci ; 8(23): 6561-6578, 2020 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-33231593

RESUMO

Phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), has shown great promise for cancer treatment in many preclinical studies. This study reports a nanoreactor designed for an enhanced mild temperature phototherapy which utilizes multiple mechanisms including simultaneous glucose consumption, oxygen supply, glutathione (GSH) depletion and heat-resistance relief. The nanoreactor is prepared using an Fe-doped polydiaminopyridine (Fe-PDAP) nanozyme with an intrinsic catalase-like activity coloaded with glucose oxidase (GOx) and indocyanine green (ICG). Evidence shows that glucose plays a vital role in tumor progression. Initiated by the breakdown of glucose into gluconic acid and H2O2 by GOx, Fe-PDAP promotes reoxygenation by catalyzing the reaction-supplied and tumor cell-supplied H2O2 into O2, which then enhances the O2-dependent PDT. Moreover, Fe-PDAP depletes GSH in tumor cells for more efficient reactive oxygen species (ROS) production. Meanwhile, the heat resistance of tumor cells is relieved by GOx-induced glucose exhaustion and heat shock protein (HSP) reduction, improving the efficiency of PTT. In particular, the nanoreactor also serves as a contrast agent for fluorescence, photoacoustic, and magnetic resonance multimodal imaging. Consequently, this nanoreactor efficiently inhibits tumor growth through mild temperature phototherapy under multimodal imaging guidance, resulting in successful tumor ablation with minimal systemic toxicity.


Assuntos
Peróxido de Hidrogênio , Nanotecnologia , Fototerapia , Animais , Linhagem Celular Tumoral , Imagem Multimodal , Temperatura
20.
ACS Nano ; 14(10): 12652-12667, 2020 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-32986406

RESUMO

Organic-inorganic hybrid materials have drawn increasing attention as photothermal agents in tumor therapy due to the advantages of green synthesis, high loading efficiency of hydrophobic drugs, facile incorporation of theranostic iron, and excellent photothermal efficiency without inert components or additives. Herein, we proposed a strategy for biomimetic engineering-mediated enhancement of photothermal performance in the tumor microenvironment (TME). This strategy is based on the specific characteristics of organic-inorganic hybrid materials and endows these materials with homologous targeting ability and photothermal stability in the TME. The hybrid materials perform the functions of cancer cells to target homolytic tumors (acting as "artificial nanotargeted cells (ANTC)"). Inspired by the pH-dependent disassembly behaviors of tannic acid (TA) and ferric ion (FeIII) and subsequent attenuation of photothermal performance, cancer cell membranes were self-deposited onto the surfaces of protoporphyrin-encapsulated TA and FeIII nanoparticles to achieve ANTC with TME-stable photothermal performance and tumor-specific phototherapy. The resulting ANTC can be used as contrast agents for concurrent photoacoustic imaging, magnetic resonance imaging, and photothermal imaging to guide the treatment. Importantly, the high loading efficiency of protoporphyrin enables the initiation of photodynamic therapy to enhance photothermal therapeutic efficiency, providing antitumor function with minimal side effects.


Assuntos
Hipertermia Induzida , Nanopartículas , Animais , Linhagem Celular Tumoral , Compostos Férricos , Camundongos , Camundongos Endogâmicos BALB C , Imagem Multimodal , Fototerapia , Nanomedicina Teranóstica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA