Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
1.
Adv Mater ; : e2401538, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38738793

RESUMO

The identification of metastasis "seeds", isolated tumor cells (ITCs), is of paramount importance for the prognosis and tailored treatment of metastatic diseases. The conventional approach to clinical ITCs diagnosis through invasive biopsies is encumbered by the inherent risks of overdiagnosis and overtreatment. This underscores the pressing need for non-invasive ITCs detection methods that provide histopathological-level insights. Recent advancements in ultra-high-field (UHF) magnetic resonance imaging (MRI) have ignited hope for the revelation of minute lesions, including the elusive ITCs. Nevertheless, currently available MRI contrast agents are susceptible to magnetization-induced strong T2-decaying effects under UHF conditions, which compromises T1 MRI capability and further impedes the precise imaging of small lesions. Herein, we report a structural defect-enabled magnetic neutrality nanoprobe (MNN) distinguished by its paramagnetic properties featuring an exceptionally low magnetic susceptibility through atomic modulation, rendering it almost non-magnetic. This unique characteristic effectively mitigates T2-decaying effect while concurrently enhancing UHF T1 contrast. Under 9 T MRI, the MNN demonstrates an unprecedentedly low r2/r1 value (∼1.06), enabling non-invasive visualization of ITCs with an exceptional detection threshold of ∼0.16 mm. These high-performance MNNs unveil the domain of hitherto undetectable minute lesions, representing a significant advancement in UHF-MRI for diagnostic purposes and fostering comprehensive metastasis research. This article is protected by copyright. All rights reserved.

2.
J Control Release ; 369: 517-530, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38569942

RESUMO

Cancer cells rely on aerobic glycolysis and DNA repair signals to drive tumor growth and develop drug resistance. Yet, fine-tuning aerobic glycolysis with the assist of nanotechnology, for example, dampening lactate dehydrogenase (LDH) for cancer cell metabolic reprograming remains to be investigated. Here we focus on anaplastic thyroid cancer (ATC) as an extremely malignant cancer with the high expression of LDH, and develop a pH-responsive and nucleus-targeting platinum nanocluster (Pt@TAT/sPEG) to simultaneously targets LDH and exacerbates DNA damage. Pt@TAT/sPEG effectively disrupts LDH activity, reducing lactate production and ATP levels, and meanwhile induces ROS production, DNA damage, and apoptosis in ATC tumor cells. We found Pt@TAT/sPEG also blocks nucleotide excision repair pathway and achieves effective tumor cell killing. In an orthotopic ATC xenograft model, Pt@TAT/sPEG demonstrates superior tumor growth suppression compared to Pt@sPEG and cisplatin. This nanostrategy offers a feasible approach to simultaneously inhibit glycolysis and DNA repair for metabolic reprogramming and enhanced tumor chemotherapy.

3.
Acta Pharm Sin B ; 14(3): 1132-1149, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38486992

RESUMO

Cancer, a complex and heterogeneous disease, arises from genomic instability. Currently, DNA damage-based cancer treatments, including radiotherapy and chemotherapy, are employed in clinical practice. However, the efficacy and safety of these therapies are constrained by various factors, limiting their ability to meet current clinical demands. Metal nanoparticles present promising avenues for enhancing each critical aspect of DNA damage-based cancer therapy. Their customizable physicochemical properties enable the development of targeted and personalized treatment platforms. In this review, we delve into the design principles and optimization strategies of metal nanoparticles. We shed light on the limitations of DNA damage-based therapy while highlighting the diverse strategies made possible by metal nanoparticles. These encompass targeted drug delivery, inhibition of DNA repair mechanisms, induction of cell death, and the cascading immune response. Moreover, we explore the pivotal role of physicochemical factors such as nanoparticle size, stimuli-responsiveness, and surface modification in shaping metal nanoparticle platforms. Finally, we present insights into the challenges and future directions of metal nanoparticles in advancing DNA damage-based cancer therapy, paving the way for novel treatment paradigms.

4.
Nat Commun ; 15(1): 460, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38212655

RESUMO

Targeted assembly of nanoparticles in biological systems holds great promise for disease-specific imaging and therapy. However, the current manipulation of nanoparticle dynamics is primarily limited to organic pericyclic reactions, which necessitate the introduction of synthetic functional groups as bioorthogonal handles on the nanoparticles, leading to complex and laborious design processes. Here, we report the synthesis of tyrosine (Tyr)-modified peptides-capped iodine (I) doped CuS nanoparticles (CuS-I@P1 NPs) as self-catalytic building blocks that undergo self-propelled assembly inside tumour cells via Tyr-Tyr condensation reactions catalyzed by the nanoparticles themselves. Upon cellular internalization, the CuS-I@P1 NPs undergo furin-guided condensation reactions, leading to the formation of CuS-I nanoparticle assemblies through dityrosine bond. The tumour-specific furin-instructed intracellular assembly of CuS-I NPs exhibits activatable dual-modal imaging capability and enhanced photothermal effect, enabling highly efficient imaging and therapy of tumours. The robust nanoparticle self-catalysis-regulated in situ assembly, facilitated by natural handles, offers the advantages of convenient fabrication, high reaction specificity, and biocompatibility, representing a generalizable strategy for target-specific activatable biomedical imaging and therapy.


Assuntos
Nanopartículas , Neoplasias , Humanos , Furina , Fototerapia , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Nanopartículas/química , Catálise , Cobre/química
5.
Angew Chem Int Ed Engl ; 63(10): e202318948, 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38212253

RESUMO

Ultra-high field (UHF) magnetic resonance imaging (MRI) has emerged as a focal point of interest in the field of cancer diagnosis. Despite the ability of current paramagnetic or superparamagnetic smart MRI contrast agents to selectively enhance tumor signals in low-field MRI, their effectiveness at UHF remains inadequate due to inherent magnetism. Here, we report a ligand-mediated magnetism-conversion nanoprobe (MCNP) composed of 3-mercaptopropionic acid ligand-coated silver-gadolinium bimetallic nanoparticles. The MCNP exhibits a pH-dependent magnetism conversion from ferromagnetism to diamagnetism, facilitating tunable nanomagnetism for pH-activatable UHF MRI. Under neutral pH, the thiolate (-S- ) ligands lead to short τ'm and increased magnetization of the MCNPs. Conversely, in the acidic tumor microenvironment, the thiolate ligands are protonated and transform into thiol (-SH) ligands, resulting in prolonged τ'm and decreased magnetization of the MCNP, thereby enhancing longitudinal relaxivity (r1) values at UHF MRI. Notably, under a 9 T MRI field, the pH-sensitive changes in Ag-S binding affinity of the MCNP lead to a remarkable (>10-fold) r1 increase in an acidic medium (pH 5.0). In vivo studies demonstrate the capability of MCNPs to amplify MRI signal of hepatic tumors, suggesting their potential as a next-generation UHF-tailored smart MRI contrast agent.


Assuntos
Imageamento por Ressonância Magnética , Neoplasias , Humanos , Ligantes , Imageamento por Ressonância Magnética/métodos , Meios de Contraste , Concentração de Íons de Hidrogênio , Microambiente Tumoral
6.
Adv Sci (Weinh) ; 11(6): e2307389, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38064201

RESUMO

Cancer therapeutic vaccines are powerful tools for immune system activation and eliciting protective responses against tumors. However, their efficacy has often been hindered by weak and slow immune responses. Here, the authors introduce an immunization strategy employing senescent erythrocytes to facilitate the accumulation of immunomodulatory zinc-Alum/ovalbumin (ZAlum/OVA) nanovaccines within both the spleen and solid tumors by temporarily saturating liver macrophages. This approach sets the stage for boosted cancer metalloimmunotherapy through a cascade immune activation. The accumulation of ZAlum/OVA nanovaccines in the spleen substantially enhances autophagy-dependent antigen presentation in dendritic cells, rapidly initiating OVA-specific T-cell responses against solid tumors. Concurrently, ZAlum/OVA nanovaccines accumulated in the tumor microenvironment trigger immunogenic cell death, leading to the induction of individualized tumor-associated antigen-specific T cell responses and increased T cell infiltration. This erythrocyte-assisted cascade immune activation using ZAlum/OVA nanovaccines results in rapid and robust antitumor immunity induction, holding great potential for clinical cancer metalloimmunotherapy.


Assuntos
Compostos de Alúmen , Vacinas Anticâncer , Neoplasias , Humanos , Ovalbumina , Nanovacinas , Neoplasias/tratamento farmacológico , Apresentação de Antígeno , Zinco , Microambiente Tumoral
7.
Nanomedicine (Lond) ; 18(22): 1585-1606, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37830425

RESUMO

Phototherapeutics is gaining momentum as a mainstream treatment for cancer, with gold-semiconductor nanocomposites showing promise as potent phototherapeutic agents due to their structural tunability, biocompatibility and functional diversity. Such nanohybrids possess plasmonic characteristics in the presence of gold and the catalytic nature of semiconductor units, as well as the unexpected physicochemical properties arising from the contact interface. This perspective provides an overview of the latest research on gold-semiconductor nanocomposites for photodynamic, photothermal and photocatalytic therapy. The relationship between the spatial configuration of these nanohybrids and their practical performance was explored to deliver comprehensive insights and guidance for the design and fabrication of novel composite nanoplatforms to enhance the efficiency of phototherapeutics, promoting the development of nanotechnology-based advanced biomedical applications.


Assuntos
Neoplasias , Fotoquimioterapia , Humanos , Ouro/química , Fototerapia , Neoplasias/tratamento farmacológico , Semicondutores
8.
ACS Nano ; 17(18): 18548-18561, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37706454

RESUMO

Replication stress (RS) induced by DNA damage plays a significant role in conferring the anticancer effects of radiotherapy and is tightly associated with radioresistance of cancer cells. Amplification of RS represents an effective approach to improving the efficacy of radiotherapy, although the development of selective RS amplifiers remains an unexplored frontier. We herein present an RS nano amplifier (RSNA) consisting of a catalytic FePt nanoparticle loaded with the chemotherapeutic doxorubicin (DOX), which selectively exacerbates RS in cancer cells by promoting replication fork (RF) catastrophe. RSNA converts the excessive reactive oxygen species (ROS) in cancer cells into oxygen, enhancing the DNA-damaging effects of radiotherapy to create more template lesions that impede RF progression in coalition with DOX. After radiation, ROS scavenging by RSNA accelerates RF progression through damaged template strands, increasing the frequency of RF collapse into double-strand breaks. Moreover, pretreatment with RSNA accumulates cancer cells in the S phase, exposing more RFs to radiation-induced RS. These effects of RSNA convergently maximize RS in cancer cells, effectively overcoming the radioresistance of cancer cells without affecting normal cells. Our study demonstrates the feasibility of selectively amplifying RS to boost radiotherapy.


Assuntos
Neoplasias , Humanos , Espécies Reativas de Oxigênio , Divisão Celular , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Catálise , Dano ao DNA , Doxorrubicina/farmacologia
9.
Adv Healthc Mater ; 12(31): e2302111, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37699592

RESUMO

Photothermal immunotherapy (PTI) has emerged as a promising approach for cancer treatment, while its efficacy is often hindered by the immunosuppressive tumor microenvironment (TME). Here, this work presents a multifunctional platform for tumor PTI based on ruthenium nanocrystal-decorated mesoporous silica nanoparticles (RuNC-MSN). By precisely regulating the distance between RuNC on MSN, this work achieves a remarkable enhancement in surface plasmon resonance of RuNC, leading to a significant improvement in the photothermal efficiency of RuNC-MSN. Furthermore, the inherent catalase-like activity of RuNC-MSN enables effective modulation of the immunosuppressive TME, thereby facilitating an enhanced immune response triggered by the photothermal effect-mediated immunogenic cell death (ICD). As a result, RuNC-MSN exhibits superior PTI performance, resulting in pronounced inhibition of primary tumor and metastasis. This study highlights the rational design of PTI agents with coupling effect-enhanced surface plasmon resonance, enabling simultaneous induction of ICD and regulation of the immunosuppressive TME, thereby significantly boosting PTI efficacy.


Assuntos
Nanopartículas , Neoplasias , Rutênio , Humanos , Dióxido de Silício/farmacologia , Doxorrubicina/farmacologia , Ressonância de Plasmônio de Superfície , Neoplasias/patologia , Imunoterapia , Microambiente Tumoral , Linhagem Celular Tumoral
10.
ACS Appl Mater Interfaces ; 15(31): 37193-37204, 2023 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-37493513

RESUMO

Mesenchymal stem cell (MSC)-based therapy has emerged as a promising strategy for the treatment of spinal cord injury (SCI). However, the hostile microenvironment of SCI, which can adversely affect the survival and paracrine effect of the implanted MSCs, severely limits the therapeutic efficacy of this approach. Here, we report on a ceria nanozyme-integrated thermoresponsive in situ forming hydrogel (CeNZ-gel) that can enable dual enhancement of MSC viability and paracrine effect, leading to highly efficient spinal cord repair. The sol-gel transition property of the CeNZ-gel at body temperature ensures uniform coverage of the hydrogel in injured spinal cord tissues. Our results demonstrate that the CeNZ-gel significantly increases the viability of transplanted MSCs in the microenvironment by attenuating oxidative stress and, more importantly, promotes the secretion of angiogenic factors from MSCs by inducing autophagy of MSCs. The synergy between the oxidative stress-relieving effect of CeNZs and the paracrine effect of MSCs accelerates angiogenesis, nerve repair, and motor function recovery after SCI, providing an efficient strategy for MSC-based SCI therapy.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Traumatismos da Medula Espinal , Regeneração da Medula Espinal , Humanos , Hidrogéis/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Traumatismos da Medula Espinal/terapia
11.
Small ; 19(25): e2300736, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37029565

RESUMO

Cell cycle checkpoint activation promotes DNA damage repair, which is highly associated with the chemoresistance of various cancers including acute myeloid leukemia (AML). Selective cell cycle checkpoint inhibitors are strongly demanded to overcome chemoresistance, but remain unexplored. A selective nano cell cycle checkpoint inhibitor (NCCI: citric acid capped ultra-small iron oxide nanoparticles) that can catalytically inhibit the cell cycle checkpoint of AML to boost the chemotherapeutic efficacy of genotoxic agents is now reported. NCCI can selectively accumulate in AML cells and convert H2 O2 to • OH to cleave heat shock protein 90, leading to the degradation of ataxia telangiectasia and Rad3-related proteinand checkpoint kinase 1, and the subsequent dysfunction of the G2/M checkpoint. Consequently, NCCI revitalizes the anti-AML efficacy of cytarabine that is previously ineffective both in vitro and in vivo. This study offers new insights into designing selective cell cycle checkpoint inhibitors for biomedical applications.


Assuntos
Antineoplásicos , Pontos de Checagem do Ciclo Celular , Resistencia a Medicamentos Antineoplásicos , Leucemia Mieloide Aguda , Nanopartículas Magnéticas de Óxido de Ferro , Animais , Camundongos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Ácido Cítrico/química , Desenho de Fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Leucemia Mieloide Aguda/tratamento farmacológico , Nanopartículas Magnéticas de Óxido de Ferro/química , Linhagem Celular Tumoral
12.
J Control Release ; 357: 20-30, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36940774

RESUMO

Ferroptosis has recently become an attractive strategy to combat the chemoresistance of cancer cells, but the intracellular ferroptosis defense system greatly challenges the efficient ferroptosis induction. Herein, we report a ferrous metal-organic framework-based nanoagent (FMN) that inhibits the intracellular upstream glutathione synthesis and induces self-amplified ferroptosis of cancer cells, for reversing chemoresistance and boosting chemotherapy. The FMN is loaded with SLC7A11 siRNA (siSLC7A11) and chemotherapeutic doxorubicin (DOX), which shows enhanced tumor cell uptake and retention, thus ensuring the effective DOX delivery and tumor intracellular iron accumulation. Importantly, the FMN simultaneously catalyzes the iron-dependent Fenton reaction and triggers the siSLC7A11-mediated suppression of upstream glutathione synthesis for intracellularly self-amplified ferroptosis, which further inhibits P-glycoprotein activity for DOX retention, and regulates the expression of Bcl-2/Bax to reverse the apoptotic resistance state of tumor cells. The FMN-mediated ferroptosis is also demonstrated in ex vivo patient-derived tumor fragment platform. Consequently, FMN successfully reverses cancer chemoresistance and achieves a highly efficient in vivo therapeutic efficacy in MCF7/ADR tumor-bearing mice. Our study provides a self-amplified ferroptosis strategy via inhibiting intracellular upstream glutathione synthesis, which is effective to reverse cancer chemoresistance.


Assuntos
Ferroptose , Neoplasias , Animais , Camundongos , Resistencia a Medicamentos Antineoplásicos , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Ferro , RNA Interferente Pequeno , Glutationa , Linhagem Celular Tumoral
13.
Nat Commun ; 13(1): 7361, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36450764

RESUMO

Platinum (Pt) resistance in cancer almost inevitably occurs during clinical Pt-based chemotherapy. The spontaneous nucleotide-excision repair of cancer cells is a representative process that leads to Pt resistance, which involves the local DNA bending to facilitate the recruitment of nucleotide-excision repair proteins and subsequent elimination of Pt-DNA adducts. By exploiting the structural vulnerability of this process, we herein report a nuclease-mimetic Pt nanozyme that can target cancer cell nuclei and induce concurrent DNA platination and oxidative cleavage to overcome Pt drug resistance. We show that the Pt nanozyme, unlike cisplatin and conventional Pt nanoparticles, specifically induces the nanozyme-catalyzed cleavage of the formed Pt-DNA adducts by generating in situ reactive oxygen species, which impairs the damage recognition factors-induced DNA bending prerequisite for nucleotide-excision repair. The recruitment of downstream effectors of nucleotide-excision repair to DNA lesion sites, including xeroderma pigmentosum groups A and F, is disrupted by the Pt nanozyme in cisplatin-resistant cancer cells, allowing excessive accumulation of the Pt-DNA adducts for highly efficient cancer therapy. Our study highlights the potential benefits of applying enzymatic activities to the use of the Pt nanomedicines, providing a paradigm shift in DNA damaging chemotherapy.


Assuntos
Neoplasias , Platina , Platina/farmacologia , Adutos de DNA , Cisplatino/farmacologia , Endonucleases , Resistencia a Medicamentos Antineoplásicos , DNA , Estresse Oxidativo , Nucleotídeos , Neoplasias/tratamento farmacológico
14.
Nanoscale ; 14(47): 17483-17499, 2022 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-36413075

RESUMO

Ultrahigh-field magnetic resonance imaging (UHF-MRI) has been attracting tremendous attention in biomedical imaging owing to its high signal-to-noise ratio, superior spatial resolution, and fast imaging speed. However, at UHF-MRI, there is a lack of proper imaging probes that can impart superior imaging sensitivity of disease lesions because conventional contrast agents generally produce pronounced susceptibility artifacts and induce very strong T2 decay effects, thus hindering satisfactory imaging performance. This review focused on the recent development of high-performance nanoprobes that can improve the sensitivity and specificity of UHF-MRI. Firstly, the contrast enhancement mechanism of nanoprobes at UHF-MRI has been elucidated. In particular, the strategies for modulating nanoprobe performance, including size effects, metal alloying and magnetic-dopant effects, surface effects, and stimuli-response regulation, have been comprehensively discussed. Furthermore, we illustrate the remarkable advances in the design of UHF-MRI nanoprobes for medical diagnosis, such as early-stage primary tumor and metastasis imaging, angiography, and dynamic monitoring of biosignaling factors in vivo. Finally, we provide a summary and outlook on the development of cutting-edge UHF-MRI nanoprobes for advanced biomedical imaging.


Assuntos
Imageamento por Ressonância Magnética
15.
Adv Mater ; 34(41): e2206915, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35986645

RESUMO

Clinical immunotherapy of solid tumors elicits durable responses only in a minority of patients, largely due to the highly immunosuppressive tumor microenvironment (TME). Although rational combinations of vaccine adjuvants with inflammatory cytokines or immune agonists that relieve immunosuppression represent an appealing therapeutic strategy against solid tumors, there are unavoidable nonspecific toxicities due to the pleiotropy of cytokines and undesired activation of off-target cells. Herein, a Zn2+ doped layered double hydroxide (Zn-LDH) based immunomodulating adjuvant, which not only relieves immunosuppression but also elicits robust antitumor immunity, is reported. Peritumorally injected Zn-LDH sustainably neutralizes acidic TME and releases abundant Zn2+ , promoting a pro-inflammatory network composed of M1-tumor-associated macrophages, cytotoxic T cells, and natural-killer cells. Moreover, the Zn-LDH internalized by tumor cells effectively disrupts endo-/lysosomes to block autophagy and induces mitochondrial damage, and the released Zn2+ activates the cGas-STING signaling pathway to induce immunogenic cell death, which further promotes the release of tumor-associated antigens to induce antigen-specific cytotoxic T lymphocytes. Unprecedentedly, merely injection of Zn-LDH adjuvant, without using any cytotoxic inflammatory cytokines or immune agonists, significantly inhibits the growth, recurrence, and metastasis of solid tumors in mice. This study provides a rational bottom-up design of potent adjuvant for cancer metalloimmunotherapy against solid tumors.


Assuntos
Imunoterapia , Neoplasias , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico , Animais , Citocinas , Hidróxidos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/tratamento farmacológico , Nucleotidiltransferases , Microambiente Tumoral
16.
Natl Sci Rev ; 9(7): nwac080, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35832777

RESUMO

Although molecular imaging probes have the potential to non-invasively diagnose a tumor, imaging probes that can detect a tumor and simultaneously identify tumor malignancy remain elusive. Here, we demonstrate a potassium ion (K+) sensitive dual-mode nanoprobe (KDMN) for non-invasive tumor imaging and malignancy identification, which operates via a cascaded 'AND' logic gate controlled by inputs of magnetic resonance imaging (MRI) and fluorescence imaging (FI) signals. We encapsulate commercial K+ indicators into the hollow cavities of magnetic mesoporous silica nanoparticles, which are subsequently coated with a K+-selective membrane that exclusively permits the passage of K+ while excluding other cations. The KDMN can readily accumulate in tumors and enhance the MRI contrast after systemic administration. Spatial information of the tumor lesion is thus accessible via MRI and forms the first layer of the 'AND' gate. Meanwhile, the KDMN selectively captures K+ and prevents interference from other cations, triggering a K+-activated FI signal as the second layer of the 'AND' gate in the case of a malignant tumor with a high extracellular K+ level. This dual-mode imaging approach effectively eliminates false positive or negative diagnostic results and allows for non-invasive imaging of tumor malignancy with high sensitivity and accuracy.

17.
Nanomicro Lett ; 14(1): 101, 2022 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-35412159

RESUMO

The structural change-mediated catalytic activity regulation plays a significant role in the biological functions of natural enzymes. However, there is virtually no artificial nanozyme reported that can achieve natural enzyme-like stringent spatiotemporal structure-based catalytic activity regulation. Here, we report a sub-nanostructural transformable gold@ceria (STGC-PEG) nanozyme that performs tunable catalytic activities via near-infrared (NIR) light-mediated sub-nanostructural transformation. The gold core in STGC-PEG can generate energetic hot electrons upon NIR irradiation, wherein an internal sub-nanostructural transformation is initiated by the conversion between CeO2 and electron-rich state of CeO2-x, and active oxygen vacancies generation via the hot-electron injection. Interestingly, the sub-nanostructural transformation of STGC-PEG enhances peroxidase-like activity and unprecedentedly activates plasmon-promoted oxidase-like activity, allowing highly efficient low-power NIR light (50 mW cm-2)-activated photocatalytic therapy of tumors. Our atomic-level design and fabrication provide a platform to precisely regulate the catalytic activities of nanozymes via a light-mediated sub-nanostructural transformation, approaching natural enzyme-like activity control in complex living systems.

18.
Tissue Eng Regen Med ; 19(2): 205-219, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34674182

RESUMO

Cancer is one of the major diseases that threaten human life worldwide. Despite advances in cancer treatment techniques, such as radiation therapy, chemotherapy, targeted therapy, and immunotherapy, it is still difficult to cure cancer because of the resistance mechanism of cancer cells. Current understanding of tumor biology has revealed that resistance to these anticancer therapies is due to the tumor microenvironment (TME) represented by hypoxia, acidity, dense extracellular matrix, and immunosuppression. This review demonstrates the latest strategies for effective cancer treatment using functional nanoparticles that can modulate the TME. Indeed, preclinical studies have shown that functional nanoparticles can effectively modulate the TME to treat refractory cancer. This strategy of using TMEs with controllable functional nanoparticles is expected to maximize cancer treatment efficiency in the future by combining it with various modern cancer therapeutics.


Assuntos
Nanopartículas , Neoplasias , Matriz Extracelular , Humanos , Neoplasias/tratamento farmacológico , Microambiente Tumoral
19.
J Control Release ; 339: 547-552, 2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34478749

RESUMO

Self-assembly in nature creates matter with complex structures and unpredictable designs; disordered building blocks spontaneously organize into ordered structures to achieve specific functions. Self-assembly begins to play an important role in the design of advanced drug delivery as well. Though, the behavior of 'dynamic nanoassembly-based drug delivery systems' (DNDDS) in biological media and cells remains poorly understood, while this is highly critical for controlling spatiotemporal drug release from DNDDS in vivo. To deepen the understanding of tailor-made DNDDS, this contribution in the Oration - New Horizons section of the Journal of controlled Release aims to highlight nature-inspired designs, construction principles, and controllable functionalities of DNDDS and how they are triggered by endogenous and exogenous stimuli. Furthermore, biomedical applications of tailor-made DNDDS for accurate diagnosis and precise treatment of diseases, including tumors, neurological diseases, injuries and infections are discussed. Finally, current challenges and future perspectives of DNDDS are briefly outlined.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias , Liberação Controlada de Fármacos , Humanos , Neoplasias/tratamento farmacológico
20.
Sci Adv ; 7(40): eabj0534, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34586849

RESUMO

The transfer of mitochondria between cells has recently been revealed as a spontaneous way to protect the injured cells. However, the utilization of this natural transfer process for disease treatment is so far limited by its unsatisfactory transfer efficiency and selectivity. Here, we demonstrate that iron oxide nanoparticles (IONPs) can augment the intercellular mitochondrial transfer from human mesenchymal stem cells (hMSCs) selectively to diseased cells, owing to the enhanced formation of connexin 43­containing gap junctional channels triggered by ionized IONPs. In a mouse model of pulmonary fibrosis, the IONP-engineered hMSCs achieve a remarkable mitigation of fibrotic progression because of the promoted intercellular mitochondrial transfer, with no serious safety issues identified. The present study reports a potential method of using IONPs to enable hMSCs for efficient and safe transfer of mitochondria to diseased cells to restore mitochondrial bioenergetics.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA