Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Adv Healthc Mater ; : e2401199, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39054675

RESUMO

The development of nanovaccines capable of eliciting tumor-specific immune responses holds significant promise for tumor immunotherapy. However, many nanovaccine designs rely heavily on incorporating multiple adjuvants and carriers, increasing the biological hazards associated with these additional components. Here, this work introduces novel flexible nanocapsules (OVAnano) designed to mimic extracellular vesicles, primarily using the ovalbumin antigen and minimal polyethylenimine adjuvant components. These results show that the biomimetic flexible structure of OVAnano facilitates enhanced antigen uptake by dendritic cells (DCs), leading to efficient antigen and adjuvant release into the cytosol via endosomal escape, and ultimately, successful antigen cross-presentation by DCs. Furthermore, OVAnano modulates the intracellular nuclear factor kappa-B (NF-κB) signaling pathway, promoting DC maturation. The highly purified antigens in OVAnano demonstrate remarkable antigen-specific immunogenicity, triggering strong antitumor immune responses mediated by DCs. Therapeutic tumor vaccination studies have also shown that OVAnano administration effectively suppresses tumor growth in mice by inducing immune responses from CD8+ and CD4+ T cells targeting specific antigens, reducing immunosuppression by regulatory T cells, and boosting the populations of effector memory T cells. These findings underscore that the simple yet potent strategy of employing minimal flexible nanocapsules markedly enhances DC-mediated antitumor immunotherapy, offering promising avenues for future clinical applications.

2.
Acta Biomater ; 184: 313-322, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38897337

RESUMO

Antimicrobial phototherapy has gained recognition as a promising approach for addressing bacterial biofilms, however, its effectiveness is often impeded by the robust physical and chemical defenses of the biofilms. Traditional antibacterial nanoplatforms face challenges in breaching the extracellular polymeric substances barrier to efficiently deliver photosensitizers deep into biofilms. Moreover, the prevalent hypoxia within biofilms restricts the success of oxygen-reliant phototherapy. In this study, we engineered a soft mesoporous organosilica nanoplatform (SMONs) by incorporating polyethylene glycol (PEG), catalase (CAT), and indocyanine green (ICG), forming SMONs-PEG-CAT-ICG (SPCI). We compared the antimicrobial efficacy of SPCI with more rigid nanoplatforms. Our results demonstrated that unique flexible mechanical properties of SPCI enable it to navigate through biofilm barriers, markedly enhancing ICG penetration in methicillin-resistant Staphylococcus aureus (MRSA) biofilms. Notably, in a murine subcutaneous MRSA biofilm infection model, SPCI showed superior biofilm penetration and pharmacokinetic benefits over its rigid counterparts. The embedded catalase in SPCI effectively converts excess H2O2 present in infected tissues into O2, alleviating hypoxia and significantly boosting the antibacterial performance of phototherapy. Both in vitro and in vivo experiments confirmed that SPCI surpasses traditional rigid nanoplatforms in overcoming biofilm barriers, offering improved treatment outcomes for infections associated with bacterial biofilms. This study presents a viable strategy for managing bacterial biofilm-induced diseases by leveraging the unique attributes of a soft mesoporous organosilica-based nanoplatform. STATEMENT OF SIGNIFICANCE: This research introduces an innovative antimicrobial phototherapy soft nanoplatform that overcomes the inherent limitations posed by the protective barriers of bacterial biofilms. By soft nanoplatform with flexible mechanical properties, we enhance the penetration and delivery of photosensitizers into biofilms. The inclusion of catalase within this soft nanoplatform addresses the hypoxia in biofilms by converting hydrogen peroxide into oxygen in infected tissues, thereby amplifying the antibacterial effectiveness of phototherapy. Compared to traditional rigid nanoplatforms, this flexible nanoplatform not only promotes the delivery of therapeutic agents but also sets a new direction for treating bacterial biofilm infections, offering significant implications for future antimicrobial therapies.


Assuntos
Antibacterianos , Biofilmes , Catalase , Verde de Indocianina , Fármacos Fotossensibilizantes , Biofilmes/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/farmacocinética , Camundongos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química , Verde de Indocianina/farmacologia , Verde de Indocianina/química , Catalase/metabolismo , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/fisiologia , Fototerapia , Polietilenoglicóis/química , Polietilenoglicóis/farmacologia , Nanopartículas/química , Infecções Estafilocócicas/tratamento farmacológico , Permeabilidade , Feminino , Camundongos Endogâmicos BALB C
3.
Small ; : e2400927, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38726949

RESUMO

Due to the presence of spatial barriers, persistent bacteria, and excessive inflammation in bacteria biofilm-infected wounds, current nanoplatforms cannot effectively address these issues simultaneously during the therapeutic process. Herein, a novel biomimetic photothermal nanoplatform integrating silver and polydopamine nanoparticles (Ag/PDAs) that can damage biofilms, kill bacterial persisters, and reduce inflammation for wound treatment is presented. These findings reveal that Ag/PDAs exhibit a broad-spectrum antimicrobial activity through direct damage to the bacterial membrane structure. Additionally, Ag/PDAs demonstrate a potent photothermal conversion efficiency. When combined with near-infrared (NIR) irradiation, Ag/PDAs effectively disrupt the spatial structure of biofilms and synergistically eradicate the resident bacteria. Furthermore, Ag/PDAs show remarkable anti-inflammatory properties in counteracting bacterium-induced macrophage polarization. The in vivo results confirm that the topical application of Ag/PDAs significantly suppress Staphylococcus aureus biofilm-infected wounds in murine models, concurrently facilitating wound healing. This research provides a promising avenue for the eradication of bacterial biofilms and the treatment of biofilm-infected wounds.

4.
ACS Nano ; 18(23): 15204-15217, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38803167

RESUMO

The Pseudomonas aeruginosa biofilm in recalcitrant chronic lung infections not only develops high antimicrobial tolerance but also induces an aberrant host inflammatory response. The metabolic condition plays a vital role in both the antimicrobial susceptibility of bacteria and the inflammatory response of immune cells, thereby offering a potential therapeutic target. Herein, we described a metabolic modulation strategy by using ultrasound-responsive liposomal nanoparticles containing a sonosensitizer and a hypoxia-activated prodrug against biofilm-associated chronic lung infections. Under ultrasound stimulation, the sonosensitizer generates antibacterial reactive oxygen species by oxygen consumption. Subsequently, the oxygen consumption-mediated hypoxia not only induces the anaerobic metabolism of bacteria for antibiotic activation but also triggers the glycolysis pathway of immune cells for inflammatory activation. Such metabolic modulation strategy demonstrated efficient therapeutic efficacy for P. aeruginosa biofilm-induced chronic lung infections in mice models and provides a promising way for combating biofilm-associated chronic infections.


Assuntos
Antibacterianos , Biofilmes , Infecções por Pseudomonas , Pseudomonas aeruginosa , Animais , Antibacterianos/farmacologia , Antibacterianos/química , Pseudomonas aeruginosa/efeitos dos fármacos , Camundongos , Biofilmes/efeitos dos fármacos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/imunologia , Nanopartículas/química , Lipossomos/química , Doença Crônica , Espécies Reativas de Oxigênio/metabolismo , Pró-Fármacos/farmacologia , Pró-Fármacos/química
5.
Nano Lett ; 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38606881

RESUMO

Photocatalysis offers a direct, yet robust, approach to eradicate pathogenic bacteria. However, the practical implementation of photocatalytic disinfection faces a significant challenge due to low-efficiency photogenerated carrier separation and transfer. Here, we present an effective approach to improve photocatalytic disinfection performance by exploiting the pyro-phototronic effect through a synergistic combination of pyroelectric properties and photocatalytic processes. A set of comprehensive studies reveals that the temperature fluctuation-induced pyroelectric field promotes photoexcited carrier separation and transfer and thus facilitates the generation of reactive oxygen species and ultimately enhances photocatalytic disinfection performance. It is worth highlighting that the constructed film demonstrated an exceptional antibacterial efficiency exceeding 95% against pathogenic bacteria under temperature fluctuations and light irradiation. Moreover, the versatile modulation role of the pyro-phototronic effect in boosting photocatalytic disinfection was corroborated. This work paves the way for improving photocatalytic disinfection efficiency by harnessing the synergistic potential of various inherent material properties.

6.
Artigo em Inglês | MEDLINE | ID: mdl-38593051

RESUMO

In clinical settings, saliva has been established as a straightforward, noninvasive medium for diagnosing periodontitis. However, the precise diagnosis is often hampered by the absence of a specialized analyzer capable of detecting low concentrations of biomarkers typically found in saliva. In this study, we present a noninvasive, on-site screen-printed biomicrochip specifically engineered for the precise and sensitive quantification of lactate concentrations in saliva, a critical biomarker in the diagnosis of periodontitis. The microchip is constructed using a nanostructured ink formulation that includes MnFe@N-doped carbon nanotubes (MnFe@N-CNTs). These MnFe@N-CNTs exhibit a high degree of graphitization and low electrical resistance, significantly augmenting the electrocatalytic efficiency of the enzymatic reaction of lactate. This results in doubled sensitivity and a detection limit that surpasses those of the current advanced salivary assay methods. Remarkably, within just 30 s, the biomicrochip can quantitatively and precisely measure lactate concentrations in the saliva of 10 patients, which provides valuable insights into the severity of their periodontitis. This biosensor holds excellent potential for large-scale production and could broaden the scope of biomarker recognition, paving the way for the analysis of a wider range of oral diseases.

7.
Int J Nanomedicine ; 19: 263-280, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38226319

RESUMO

Background: Nanovaccines have emerged as a promising vaccination strategy, exhibiting their capacity to deliver antigens and adjuvants to elicit specific immune responses. Despite this potential, optimizing the design and delivery of nanovaccines remains a challenge. Methods: In this study, we engineered a dendritic mesoporous silica-based nanocarrier enveloped in a metal-phenolic network (MPN) layer containing divalent manganese ions and tannic acid (MSN@MT). This nanocarrier was tailored for antigen loading to serve as a nanovaccine, aiming to activate the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway in dendritic cells (DCs). Our experimental approach encompassed both cellular assays and mouse immunizations, allowing a comprehensive evaluation of the nanovaccine's impact on DC activation and its influence on the generation of antigen-specific T-cell responses. Results: MSN@MT demonstrated a remarkable enhancement in humoral and cellular immune responses in mice compared to control groups. This highlights the potential of MSN@MT to effectively trigger the cGAS-STING pathway in DCs, resulting in robust immune responses. Conclusion: Our study introduces MSN@MT, a unique nanocarrier incorporating divalent manganese ions and tannic acid, showcasing its exceptional ability to amplify immune responses by activating the cGAS-STING pathway in DCs. This innovation signifies a stride in refining nanovaccine design for potent immune activation.


Assuntos
Manganês , Nanovacinas , Polifenóis , Animais , Camundongos , Nucleotidiltransferases/genética , Íons , Células Dendríticas
8.
Adv Sci (Weinh) ; 11(10): e2307048, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38109089

RESUMO

Host immune systems serving as crucial defense lines are vital resisting mechanisms against biofilm-associated implant infections. Nevertheless, biofilms hinder the penetration of anti-bacterial species, inhibit phagocytosis of immune cells, and frustrate host inflammatory responses, ultimately resulting in the weakness of the host immune system for biofilm elimination. Herein, a cell-like construct is developed through encapsulation of erythrocyte membrane fragments on the surface of Fe3 O4 nanoparticle-fabricated microbubbles and then loaded with hydroxyurea (EMB-Hu). Under ultrasound (US) stimulation, EMB-Hu undergoes a stable oscillation manner to act in an "exocytosis" mechanism for disrupting biofilm, releasing agents, and enhancing penetration of catalytically generated anti-bacterial species within biofilms. Additionally, the US-stimulated "exocytosis" by EMB-Hu can activate pro-inflammatory macrophage polarization and enhance macrophage phagocytosis for clearance of disrupted biofilms. Collectively, this work has exhibited cell-like microbubbles with US-stimulated "exocytosis" mechanisms to overcome the biofilm barrier and signal macrophages for inflammatory activation, finally achieving favorable therapeutic effects against implant infections caused by methicillin-resistant Staphylococcus aureus (MRSA) biofilms.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Humanos , Microbolhas , Antibacterianos/farmacologia , Fagocitose , Macrófagos , Biofilmes , Complicações Pós-Operatórias
9.
Int J Nanomedicine ; 18: 4601-4616, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37600119

RESUMO

Background: Inflammatory bowel disease (IBD) is closely related to higher intracellular oxidative stress. Therefore, developing a novel method to scavenge the harmful reactive oxygen species (ROS) and alleviate colon inflammation to treat IBD is a promising strategy. Methods: CeO2@PDA-PEG (CeO2@PP) were synthesized by modifying ceria (CeO2) nanorods with polydopamine (PDA) and polyethylene glycol (PEG). The ROS scavenging ability of CeO2@PP was detected by using flow cytometry and confocal laser scanning microscope (CLSM). The anti-inflammatory ability of CeO2@PP was determined in vitro by treating lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. The biocompatibility of CeO2@PP was evaluated in vivo and in vitro. Moreover, the therapeutic effects of CeO2@PP in vivo were estimated in a dextran sulfate sodium salt (DSS)-induced colitis mouse model. Results: Physicochemical property results demonstrated that PDA and PEG modification endowed CeO2 nanorods with excellent dispersibility and colloidal stability. CeO2@PP maintained superior enzyme-like activity, including superoxide dismutase (SOD) and catalase (CAT), indicating antioxidant ability. Moreover, in vitro results showed that CeO2@PP with PDA promotes LPS-induced RAW 264.7 macrophages into M2-type polarization. In addition, in vitro and in vivo results showed that CeO2@PP have great biocompatibility and biosafety. Animal experiments have shown that CeO2@PP have excellent anti-inflammatory effects against DSS-induced colitis and effectively alleviated intestinal mucosal injury. Conclusion: The nanoplatform CeO2@PP possessed excellent antioxidant and anti-inflammatory properties for scavenging ROS and modulating macrophage polarization, which is beneficial for efficient colitis therapy.


Assuntos
Colite , Sequestradores de Radicais Livres , Indóis , Nanotubos , Polímeros , Espécies Reativas de Oxigênio , Indóis/química , Indóis/farmacologia , Indóis/uso terapêutico , Polímeros/química , Polímeros/farmacologia , Polímeros/uso terapêutico , Colite/tratamento farmacológico , Colite/patologia , Inflamação/tratamento farmacológico , Inflamação/patologia , Sequestradores de Radicais Livres/química , Sequestradores de Radicais Livres/farmacologia , Sequestradores de Radicais Livres/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Polaridade Celular , Cério , Animais , Camundongos , Células RAW 264.7 , Masculino , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
10.
J Nanobiotechnology ; 21(1): 236, 2023 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-37482608

RESUMO

Therapeutic tumor vaccines have attracted considerable attention in the past decade; they can induce tumor regression, eradicate minimal residual disease, establish lasting immune memory and avoid non-specific and adverse side effects. However, the challenge in the field of therapeutic tumor vaccines is ensuring the delivery of immune components to the lymph nodes (LNs) to activate immune cells. The clinical response rate of traditional therapeutic tumor vaccines falls short of expectations due to inadequate lymph node delivery. With the rapid development of nanotechnology, a large number of nanoplatform-based LN-targeting nanovaccines have been exploited for optimizing tumor immunotherapies. In addition, some nanovaccines possess non-invasive visualization performance, which is benefit for understanding the kinetics of nanovaccine exposure in LNs. Herein, we present the parameters of nanoplatforms, such as size, surface modification, shape, and deformability, which affect the LN-targeting functions of nanovaccines. The recent advances in nanoplatforms with different components promoting LN-targeting are also summarized. Furthermore, emerging LNs-targeting nanoplatform-mediated imaging strategies to both improve targeting performance and enhance the quality of LN imaging are discussed. Finally, we summarize the prospects and challenges of nanoplatform-based LN-targeting and /or imaging strategies, which optimize the clinical efficacy of nanovaccines in tumor immunotherapies.


Assuntos
Vacinas Anticâncer , Neoplasias , Humanos , Linfonodos , Neoplasias/terapia , Imunoterapia , Nanotecnologia
11.
Adv Sci (Weinh) ; 10(18): e2301339, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37088780

RESUMO

Dendritic cell (DC)-based cancer immunotherapy has exhibited remarkable clinical prospects because DCs play a central role in initiating and regulating adaptive immune responses. However, the application of traditional DC-mediated immunotherapy is limited due to insufficient antigen delivery, inadequate antigen presentation, and high levels of immunosuppression. To address these challenges, engineered biomaterials have been exploited to enhance DC-mediated immunotherapeutic effects. In this review, vital principal components that can enhance DC-mediated immunotherapeutic effects are first introduced. The parameters considered in the rational design of biomaterials, including targeting modifications, size, shape, surface, and mechanical properties, which can affect biomaterial optimization of DC functions, are further summarized. Moreover, recent applications of various engineered biomaterials in the field of DC-mediated immunotherapy are reviewed, including those serve as immune component delivery platforms, remodel the tumor microenvironment, and synergistically enhance the effects of other antitumor therapies. Overall, the present review comprehensively and systematically summarizes biomaterials related to the promotion of DC functions; and specifically focuses on the recent advances in biomaterial designs for DC activation to eradicate tumors. The challenges and opportunities of treatment strategies designed to amplify DCs via the application of biomaterials are discussed with the aim of inspiring the clinical translation of future DC-mediated cancer immunotherapies.


Assuntos
Imunoterapia , Neoplasias , Terapia de Imunossupressão , Apresentação de Antígeno , Materiais Biocompatíveis , Células Dendríticas , Neoplasias/terapia
12.
ACS Appl Mater Interfaces ; 15(15): 18720-18733, 2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-37018422

RESUMO

Implant infections are difficult to cure by traditional antibiotic therapy due to bacterial biofilm-induced antibiotic tolerance and impaired immune responses. To efficiently treat implant infections, therapeutic agents need to kill bacteria and regulate the inflammatory response of immune cells during the biofilm elimination process. Herein, multifunctional smart hollow Cu2MoS4 nanospheres (H-CMS NSs) with pH-responsive enzyme-like activities were prepared for self-adaptively eliminating biofilms and regulating the inflammation of macrophages in implant infections. During biofilm infection, the tissue microenvironment around implants is acidic. H-CMS NSs with oxidase (OXD)/peroxidase (POD)-like activities can catalyze reactive oxidative species (ROS) generation for directly killing bacteria and polarizing macrophages to a proinflammatory phenotype. Moreover, the POD-like activity and antibacterial property of H-CMS NSs can be further enhanced under ultrasound (US) irradiation. After the elimination of biofilms, the tissue microenvironment around implants shifts from acidic to neutral. H-CMS NSs show catalase (CAT)-like activity and eliminate excessive ROS, which polarizes macrophages to anti-inflammatory phenotype and promotes healing of infected tissue. This work provides a smart nanozyme with self-adaptive regulation of the antibiofilm activity and immune response by regulating ROS generation/elimination according to the different pathological microenvironments in implant infections during the different therapeutic stages.


Assuntos
Nanosferas , Humanos , Espécies Reativas de Oxigênio/farmacologia , Biofilmes , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Complicações Pós-Operatórias , Bactérias
13.
Mater Today Bio ; 19: 100559, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36798535

RESUMO

With the widespread prevalence of drug-resistant pathogens, traditional antibiotics have limited effectiveness and do not yield the desired outcomes. Recently, alternative antibacterial therapies based on ultrasound (US) have been explored to overcome the crisis of bacterial pathogens. Antimicrobial sonodynamic therapy (aSDT) offers an excellent solution that relies on US irradiation to produce reactive oxygen species (ROS) and achieve antibiotic-free mediated antimicrobial effects. In addition, aSDT possesses the advantage of superior tissue penetrability of US compared to light irradiation, demonstrating great feasibility in treating deep infections. Although existing conventional sonosensitizers can produce ROS for antimicrobial activity, some limitations, such as low penetration rate, nonspecific distribution and poor ROS production under hypoxic conditions, result in suboptimal sterilization in aSDT. Recently, emerging nanosonosensitizers have enormous advantages as high-performance agents in aSDT, which overcome the deficiencies of conventional sonosensitizers as described above. Thus, nanosonosensitizer-mediated aSDT has a bright future for the management of bacterial infections. This review classifies the current available nanosonosensitizers and provides an overview of the mechanisms, biomedical applications, recent advances and perspectives of aSDT.

14.
Adv Healthc Mater ; 12(17): e2203028, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36807733

RESUMO

Traditional dendritic cell (DC)-mediated immunotherapy is usually suppressed by weak immunogenicity in tumors and generally leads to unsatisfactory outcomes. Synergistic exogenous/endogenous immunogenic activation can provide an alternative strategy for evoking a robust immune response by promoting DC activation. Herein, Ti3 C2 MXene-based nanoplatforms (termed MXP) are prepared with high-efficiency near-infrared photothermal conversion and immunocompetent loading capacity to form endogenous/exogenous nanovaccines. Specifically, the immunogenic cell death of tumor cells induced by the photothermal effects of the MXP can generate endogenous danger signals and antigens release to boost vaccination for DC maturation and antigen cross-presentation. In addition, MXP can deliver model antigen ovalbumin (OVA) and agonists (CpG-ODN) as an exogenous nanovaccine (MXP@OC), which further enhances DC activation. Importantly, the synergistic strategy of photothermal therapy and DC-mediated immunotherapy by MXP significantly eradicates tumors and enhances adaptive immunity. Hence, the present work provides a two-pronged strategy for improving immunogenicity and killing tumor cells to achieve a favorable outcome in tumor patients.


Assuntos
Vacinas Anticâncer , Nanopartículas , Neoplasias , Humanos , Neoplasias/terapia , Apresentação de Antígeno , Antígenos/farmacologia , Imunoterapia , Células Dendríticas , Vacinas Anticâncer/farmacologia
15.
Sci Adv ; 9(4): eade5446, 2023 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-36696490

RESUMO

Efficient treatment of chronic lung infections caused by Pseudomonas aeruginosa biofilms is a great challenge because of drug tolerance and immune evasion issues. Here, we develop ultrasound-responsive catalytic microbubbles with biofilm elimination and immune activation properties to combat chronic lung infection induced by P. aeruginosa biofilms. In these microbubbles, piperacillin and Fe3O4 nanoparticles form a drug-loaded shell surrounding the air core. Under ultrasound stimulation, the microbubbles can physically disrupt the structure of biofilms and enhance the penetration of both Fe3O4 nanoparticles and piperacillin into the biofilm. Then, Fe3O4 nanoparticles chemically degrade the biofilm matrix and kill the bacteria with the assistance of piperacillin. Fe3O4 nanoparticles can activate the immune response for biofilm elimination by polarizing macrophages into a pro-inflammatory phenotype. These ultrasound-responsive catalytic microbubbles efficiently treat chronic lung infections in a mouse model by combining physical/chemical/antibiotic biofilm elimination and immune activation, thus providing a promising strategy for combating bacterial biofilm infections.


Assuntos
Infecções Bacterianas , Microbolhas , Animais , Camundongos , Biofilmes , Antibacterianos/farmacologia , Piperacilina/metabolismo , Pulmão , Pseudomonas aeruginosa
16.
Front Cell Dev Biol ; 10: 941750, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36092724

RESUMO

Head and neck squamous cell carcinoma (HNSCC) have a high incidence and mortality rate, and investigating the pathogenesis and potential therapeutic strategies of HNSCC is required for further progress. Immunotherapy is a considerable therapeutic strategy for HNSCC due to its potential to produce a broad and long-lasting antitumor response. However, immune escape, which involves mechanisms including dyregulation of cytokines, perturbation of immune checkpoints, and recruitment of inhibitory cell populations, limit the efficacy of immunotherapy. Currently, multiple immunotherapy strategies for HNSCC have been exploited, including immune checkpoint inhibitors, costimulatory agonists, antigenic vaccines, oncolytic virus therapy, adoptive T cell transfer (ACT), and epidermal growth factor receptor (EGFR)-targeted therapy. Each of these strategies has unique advantages, and the appropriate application of these immunotherapies in HNSCC treatment has significant value for patients. Therefore, this review comprehensively summarizes the mechanisms of immune escape and the characteristics of different immunotherapy strategies in HNSCC to provide a foundation and consideration for the clinical treatment of HNSCC.

17.
Nat Commun ; 13(1): 3875, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35790729

RESUMO

Traditional antibiotic treatment has limited efficacy for the drug-tolerant bacteria present in biofilms because of their unique metabolic conditions in the biofilm infection microenvironment. Modulating the biofilm infection microenvironment may influence the metabolic state of the bacteria and provide alternative therapeutic routes. In this study, photodynamic therapy is used not only to eradicate methicillin-resistant Staphylococcus aureus biofilms in the normoxic condition, but also to potentiate the hypoxic microenvironment, which induces the anaerobic metabolism of methicillin-resistant Staphylococcus aureus and activates the antibacterial activity of metronidazole. Moreover, the photodynamic therapy-activated chemotherapy can polarize the macrophages to a M2-like phenotype and promote the repair of the biofilm infected wounds in mice. This biofilm infection microenvironment modulation strategy, whereby the hypoxic microenvironment is potentiated to synergize photodynamic therapy with chemotherapy, provides an alternative pathway for efficient treatment of biofilm-associated infections.


Assuntos
Infecções Bacterianas , Staphylococcus aureus Resistente à Meticilina , Fotoquimioterapia , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Biofilmes , Camundongos
18.
Int J Nanomedicine ; 17: 2735-2750, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35769516

RESUMO

Background: Bacterial biofilm-related wound infections threaten human health due to the lack of efficient treatments. Therefore, developing a novel strategy for wound infection care is urgently needed. Methods: Cube-shaped Cu2WS4 nanocrystals (CWSNs) were successfully prepared via a microwave-assisted method. CWSNs, as photocatalysts, were first studied by using fluorescence spectroscopy for their ability to generate reactive oxygen species (ROS). The antibacterial and biofilm inhibition abilities of CWSNs were determined in vitro by using Staphylococcus aureus (S. aureus) as the model bacterium. Moreover, a CWSN gel was prepared and applied to treat S. aureus-infected wounds in mice. The toxicity of the CWSNs was evaluated through in vitro cell and in vivo animal experiments. Results: Studies on the properties of the CWSNs demonstrated that these nanomaterials can catalyze the generation of hydroxyl radicals (•OH) without the addition of H2O2 after visible-light irradiation, indicating their photocatalytic ability. Moreover, the in vitro experimental results showed that the CWSNs not only adhered to the surfaces of S. aureus to kill the bacteria, but also inhibited S. aureus biofilm formation. The in vivo study showed that the CWSN gel produced excellent antibacterial effects against S. aureus infected wounds in mice and effectively promoted wound healing. Furthermore, toxicity tests showed that the CWSNs have negligible toxicity in vitro and in vivo. Conclusion: This work provides a potential photocatalytic antibacterial nanoagent for efficient bacterial killing, inhibition of biofilms growth and wound infection treatment.


Assuntos
Nanopartículas , Infecções Estafilocócicas , Infecção dos Ferimentos , Animais , Antibacterianos/farmacologia , Biofilmes , Peróxido de Hidrogênio/farmacologia , Camundongos , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus , Infecção dos Ferimentos/microbiologia
19.
Clin Oral Investig ; 26(10): 6223-6233, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35672515

RESUMO

OBJECTIVES: To investigate the bacterial diversity in peri-implant plaques and the effect of periodontitis history on the occurrence of peri-implant mucositis. MATERIALS AND METHODS: Three groups of subgingival plaques were collected from peri-implant sulci in the first molar area. The three groups included healthy implants in patients without periodontitis (NH implant), healthy implants in patients with periodontitis history (PH implant), and peri-implant mucositis implants in patients with periodontitis history (PM implant). Subgingival plaques in periodontal pockets of contralateral natural first molars were also collected. Bacterial DNA was extracted and the V4 region of the 16S rDNA sequence was amplified and sequenced on an Illumina HiSeq platform. The operational taxonomic units obtained from amplicon sequencing were used to analyze the prevalence and identity of bacteria based on public databases and advanced techniques. RESULTS: Analysis of similarities indicated a significant difference in bacterial structures between the NH implant and PM implant groups. Additionally, a significantly higher relative abundance of the genera Actinomyces and Streptococcus was found in the samples of the NH implant group. The genera Fusobacterium and Prevotella could be considered as potential biomarkers for peri-implant mucositis. Moreover, more gram-negative anaerobic bacteria (Porphyromonas and Prevotella) were detected in the samples from patients with periodontitis history. CONCLUSIONS: The increased accumulation of Fusobacterium and Prevotella is associated with a higher risk of peri-implant mucositis. In addition, patients with periodontal history may be more likely to develop peri-implant mucositis. CLINICAL RELEVANCE: The increase in periodontal pathogens and the decrease in health-associated bacteria in patients with periodontitis history may be more likely to develop peri-implant mucositis. These results provide a bacteriological basis for the prevention and treatment of peri-implant mucositis in patients with periodontitis history.


Assuntos
Implantes Dentários , Placa Dentária , Microbiota , Mucosite , Peri-Implantite , Periodontite , Bactérias , DNA Bacteriano , DNA Ribossômico , Implantes Dentários/microbiologia , Placa Dentária/microbiologia , Humanos , Peri-Implantite/microbiologia , Periodontite/microbiologia
20.
Small ; 18(32): e2201108, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35734820

RESUMO

Nanovaccine-based immunotherapy (NBI) has the ability to initiate dendritic cell (DC)-mediated tumor-specific immune responses and maintain long-term antitumor immune memory. To date, the mechanism by which the mechanical properties of nanoparticles alter the functions of DCs in NBI remains largely unclear. Here, a soft mesoporous organosilica-based nanovaccine (SMONV) is prepared and the elasticity-dependent effect of the nanovaccine on the underlying DC-mediated immune responses is studied. It is found that the elasticity results in greater internalization of SMONV by DCs, followed by the induction of substantial cytosolic delivery of antigens via endosomal escape, leading to effective DC maturation and antigen cross-presentation. Impressively, elasticity enables SMONV to enhance lymphatic drainage of antigens in vivo, thus stimulating robust humoral and cellular immunity. The results from therapeutic tumor vaccination further reveal that subcutaneously administered SMONV effectively suppresses tumor growth in tumor-bearing mice by evoking antigen-specific CD8+ T-cell immune responses, mitigating regulatory T-cell-mediated immunosuppression, and increasing central memory and effector memory T-cell populations. Furthermore, combinatorial immunization with SMONV and anti-PD-L1 blocking antibodies results in an amplified therapeutic effect on tumor-bearing mice. These findings reveal the elastic effect of the nanovaccine on DC-mediated immune responses, and the prepared SMONV represents a facile and powerful strategy for antitumor immunotherapy.


Assuntos
Vacinas Anticâncer , Nanopartículas , Neoplasias , Animais , Antígenos , Linfócitos T CD8-Positivos , Células Dendríticas , Imunoterapia/métodos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...