Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
ACS Nano ; 18(9): 6990-7010, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38385433

RESUMEN

The clinical treatment efficacy for implant-associated infections (IAIs), particularly those caused by Methicillin-resistant Staphylococcus aureus (MRSA), remains unsatisfactory, primarily due to the formation of biofilm barriers and the resulting immunosuppressive microenvironment, leading to the chronicity and recurrence of IAIs. To address this challenge, we propose a light-induced immune enhancement strategy, synthesizing BSA@MnO2@Ce6@Van (BMCV). The BMCV exhibits precise targeting and adhesion to the S. aureus biofilm-infected region, coupled with its capacity to catalyze oxygen generation from H2O2 in the hypoxic and acidic biofilm microenvironment (BME), promoting oxygen-dependent photodynamic therapy efficacy while ensuring continuous release of manganese ions. Notably, targeted BMCV can penetrate biofilms, producing ROS that degrade extracellular DNA, disrupting the biofilm structure and impairing its barrier function, making it vulnerable to infiltration and elimination by the immune system. Furthermore, light-induced reactive oxygen species (ROS) around the biofilm can lyse S. aureus, triggering bacterium-like immunogenic cell death (ICD), releasing abundant immune costimulatory factors, facilitating the recognition and maturation of antigen-presenting cells (APCs), and activating adaptive immunity. Additionally, manganese ions in the BME act as immunoadjuvants, further amplifying macrophage-mediated innate and adaptive immune responses and reversing the immunologically cold BME to an immunologically hot BME. We prove that our synthesized BMCV elicits a robust adaptive immune response in vivo, effectively clearing primary IAIs and inducing long-term immune memory to prevent recurrence. Our study introduces a potent light-induced immunomodulatory nanoplatform capable of reversing the biofilm-induced immunosuppressive microenvironment and disrupting biofilm-mediated protective barriers, offering a promising immunotherapeutic strategy for addressing challenging S. aureus IAIs.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Humanos , Staphylococcus aureus , Peróxido de Hidrógeno/farmacología , Manganeso/uso terapéutico , Compuestos de Manganeso/farmacología , Especies Reactivas de Oxígeno/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Óxidos/farmacología , Biopelículas , Inmunidad , Terapia de Inmunosupresión , Oxígeno/farmacología , Antibacterianos/farmacología
2.
Int J Mol Sci ; 24(21)2023 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-37958705

RESUMEN

Trace elements and metals play critical roles in the normal functioning of the central nervous system (CNS), and their dysregulation has been implicated in neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). In a healthy CNS, zinc, copper, iron, and manganese play vital roles as enzyme cofactors, supporting neurotransmission, cellular metabolism, and antioxidant defense. Imbalances in these trace elements can lead to oxidative stress, protein aggregation, and mitochondrial dysfunction, thereby contributing to neurodegeneration. In AD, copper and zinc imbalances are associated with amyloid-beta and tau pathology, impacting cognitive function. PD involves the disruption of iron and manganese levels, leading to oxidative damage and neuronal loss. Toxic metals, like lead and cadmium, impair synaptic transmission and exacerbate neuroinflammation, impacting CNS health. The role of aluminum in AD neurofibrillary tangle formation has also been noted. Understanding the roles of these elements in CNS health and disease might offer potential therapeutic targets for neurodegenerative disorders. The Codex Alimentarius standards concerning the mentioned metals in foods may be one of the key legal contributions to safeguarding public health. Further research is needed to fully comprehend these complex mechanisms and develop effective interventions.


Asunto(s)
Enfermedad de Alzheimer , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Oligoelementos , Humanos , Cobre/uso terapéutico , Enfermedad de Alzheimer/patología , Manganeso/uso terapéutico , Oligoelementos/uso terapéutico , Zinc/uso terapéutico , Hierro/uso terapéutico , Metales/uso terapéutico , Enfermedades Neurodegenerativas/patología
3.
Nano Lett ; 23(22): 10350-10359, 2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-37930173

RESUMEN

Immunotherapies have shown high clinical success, however, the therapeutical efficacy is largely restrained by insufficient immune activation and an immunosuppressive microenvironment. Herein, we report tumor microenvironment (TME)-responsive manganese-enriched zinc peroxide nanoparticles (MONPs) for synergistic cancer immunotherapy by inducing the immunogenic death (ICD) of cancer cells and activating the stimulator of the interferon gene (STING) pathway. MONPs especially disassociate upon exposure to acidic tumor tissue and in situ generate •OH for the ICD effect. Moreover, Mn2+ activated the STING and synergistically induced the secretion of type I interferon and inflammatory cytokines for specific T cell responses. Meanwhile, MONPs relieved the immunosuppression of TME through decreasing Tregs and polarizing M2 macrophages to the M1 type to unleash a cascade adaptive immune response. In combination with the anti-PD-1 antibody, MONPs showed superior efficacy in inhibiting tumor growth and preventing lung metastasis. Our study demonstrates the feasibility of functional nanoparticles to amplify STING innate stimulation, showing a prominent strategy for cancer immunotherapy.


Asunto(s)
Neoplasias Pulmonares , Nanopartículas , Neoplasias , Humanos , Manganeso/uso terapéutico , Inmunoterapia , Nanopartículas/uso terapéutico , Microambiente Tumoral , Peróxidos , Zinc , Neoplasias/tratamiento farmacológico
4.
EBioMedicine ; 97: 104824, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37806287

RESUMEN

BACKGROUND: Huntington or Huntington's disease (HD) is an autosomal dominant neurodegenerative disease characterised by both progressive motor and cognitive dysfunction; its pathogenic mechanisms remain poorly understood and no treatment can currently slow, stop, or reverse its progression. There is some evidence of metallomic dysfunction in limited regions of the HD brain; we hypothesised that these alterations are more widespread than the current literature suggests and may contribute to pathogenesis in HD. METHODS: We measured the concentrations of eight essential metals (sodium, potassium, magnesium, calcium, iron, zinc, copper, and manganese) and the metalloid selenium across 11 brain regions in nine genetically confirmed, clinically manifest cases of HD and nine controls using inductively-coupled plasma mass spectrometry. Case-control differences were assessed by non-parametric Mann-Whitney U test (p < 0.05), risk ratios, E-values, and effect sizes. FINDINGS: We observed striking decreases in selenium levels in 11 out of 11 investigated brain regions in HD, with risk ratios and effect sizes ranging 2.3-9.0 and 0.7-1.9, respectively. Increased sodium/potassium ratios were observed in every region (risk ratio = 2.5-8.0; effect size = 1.2-5.8) except the substantia nigra (risk ratio = 0.25; effect size = 0.1). Multiple regions showed increased calcium and/or zinc levels, and localised decreases in iron, copper, and manganese were present in the globus pallidus, cerebellum, and substantia nigra, respectively. INTERPRETATION: The observed metallomic alterations in the HD brain may contribute to several pathogenic mechanisms, including mitochondrial dysfunction, oxidative stress, and blood-brain barrier dysfunction. Selenium supplementation may represent a potential, much-needed therapeutic pathway for the treatment of HD that would not require localised delivery in the brain due to the widespread presence of selenium deficiency in regions that show both high and low levels of neurodegeneration. FUNDING: In Acknowledgments, includes the Lee Trust, the Endocore Research Trust, Cure Huntington's Disease Initiative, the Oakley Mental Health Research Foundation, the Medical Research Council (MRC), the New Zealand Neurological Foundation, and others.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Selenio , Humanos , Enfermedad de Huntington/metabolismo , Selenio/metabolismo , Selenio/uso terapéutico , Cobre/metabolismo , Cobre/uso terapéutico , Manganeso/metabolismo , Manganeso/uso terapéutico , Enfermedades Neurodegenerativas/metabolismo , Calcio/metabolismo , Encéfalo/patología , Hierro/metabolismo , Zinc/metabolismo , Potasio/metabolismo , Sodio
5.
Biomaterials ; 302: 122321, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37722183

RESUMEN

Radiotherapy is an important therapeutic modality in the treatment of cancers. Nevertheless, the characteristics of the tumor microenvironment (TME), such as hypoxia and high glutathione (GSH), limit the efficacy of radiotherapy. Manganese-based (Mn-based) nanomaterials offer a promising prospect for sensitizing radiotherapy due to their good responsiveness to the TME. In this review, we focus on the mechanisms of radiosensitization of Mn-based nanosystems, including alleviating tumor hypoxia, increasing reactive oxygen species production, increasing GSH conversion, and promoting antitumor immunity. We further illustrate the applications of these mechanisms in cancer radiotherapy, including the development and delivery of radiosensitizers, as well as their combination with other therapeutic modalities. Finally, we summarize the application of Mn-based nanosystems as contrast agents in realizing precision therapy. Hopefully, the present review will provide new insights into the biological mechanisms of Mn-based nanosystems, as well as their applications in radiotherapy, in order to address the difficulties and challenges that remain in their clinical application in the future.


Asunto(s)
Nanoestructuras , Neoplasias , Fármacos Sensibilizantes a Radiaciones , Humanos , Manganeso/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Neoplasias/radioterapia , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
6.
J Am Anim Hosp Assoc ; 59(3): 152-157, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37167249

RESUMEN

Manganese is a common component of human joint supplements and may be a source of ingestion and subsequent toxicosis in dogs. Although hepatotoxicity secondary to manganese toxicosis has been reported in dogs before, no descriptions of successful management of manganese toxicosis has been reported in veterinary literature. A 5 yr old spayed female Shetland sheepdog and a 5 yr old female Shetland sheepdog were evaluated following accidental ingestion of a joint supplement. Consultation with a toxicologist revealed concern for manganese toxicosis resulting in hepatic injury. Both dogs developed subsequent acute liver injury, despite decontamination and initial management with N-acetylcysteine and cholestyramine. The patients were managed with calcium ethylenediaminetetraacetic acid, paraaminosalicylic acid, allopurinol, Vitamin E, ginkgo biloba, and S-adenosylmethionine/silybin. Liver values returned to normal in both dogs. Manganese exposure was confirmed with urine manganese analysis in one dog and fecal examination in the other dog. A previous case report detailed the fatal manganese toxicosis in a dog; this case report describes the successful management of severe acute hepatic injury secondary to manganese toxicosis. The combination of medications used above may be used for successful treatment of manganese toxicosis in dogs.


Asunto(s)
Enfermedades de los Perros , Manganeso , Humanos , Perros , Femenino , Animales , Manganeso/toxicidad , Manganeso/uso terapéutico , Enfermedades de los Perros/inducido químicamente , Enfermedades de los Perros/tratamiento farmacológico , Suplementos Dietéticos , Acetilcisteína/uso terapéutico , Hígado
7.
Viruses ; 15(2)2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36851604

RESUMEN

Foot-and-mouth disease (FMD) is an acute contagious disease of cloven-hoofed animals such as cattle, pigs, and sheep. Current emergency FMD vaccines are of limited use for early protection because their protective effect starts 7 days after vaccination. Therefore, antiviral drugs or additives are used to rapidly stop the spread of the virus during FMD outbreaks. Manganese (Mn2+) was recently found to be an important substance necessary for the host to protect against DNA viruses. However, its antiviral effect against RNA viruses remains unknown. In this study, we found that Mn2+ has antiviral effects on the FMD virus (FMDV) both in PK15 cells and mice. The inhibitory effect of Mn2+ on FMDV involves NF-κB activation and up-regulation of interferon-stimulated genes. Animal experiments showed that Mn2+ can be highly effective in protecting C57BL/6N mice from being infected with FMDV. Overall, we suggest Mn2+ as an effective antiviral additive for controlling FMDV infection.


Asunto(s)
Antivirales , Virus de la Fiebre Aftosa , Fiebre Aftosa , Manganeso , Animales , Bovinos , Ratones , Antivirales/farmacología , Antivirales/uso terapéutico , Interferones , Manganeso/farmacología , Manganeso/uso terapéutico , Ratones Endogámicos C57BL , Ovinos , Porcinos , Fiebre Aftosa/tratamiento farmacológico , Fiebre Aftosa/virología , Línea Celular
8.
Adv Mater ; 35(19): e2205409, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36121368

RESUMEN

As an essential micronutrient, manganese (Mn) participates in various physiological processes and plays important roles in host immune system, hematopoiesis, endocrine function, and oxidative stress regulation. Mn-based nanoparticles are considered to be biocompatible and show versatile applications in nanomedicine, in particular utilized in tumor immunotherapy in the following ways: 1) acting as a biocompatible nanocarrier to deliver immunotherapeutic agents for tumor immunotherapy; 2) serving as an adjuvant to regulate tumor immune microenvironment and enhance immunotherapy; 3) activating host's immune system through the cGAS-STING pathway to trigger tumor immunotherapy; 4) real-time monitoring tumor immunotherapy effect by magnetic resonance imaging (MRI) since Mn2+ ions are ideal MRI contrast agent which can significantly enhance the T1 -weighted MRI signal after binding to proteins. This comprehensive review focuses on the most recent progress of Mn-based nanoplatforms in tumor immunotherapy. The characteristics of Mn are first discussed to guide the design of Mn-based multifunctional nanoplatforms. Then the biomedical applications of Mn-based nanoplatforms, including immunotherapy alone, immunotherapy-involved multimodal synergistic therapy, and imaging-guided immunotherapy are discussed in detail. Finally, the challenges and future developments of Mn-based tumor immunotherapy are highlighted.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Manganeso/uso terapéutico , Nanopartículas/uso terapéutico , Imagen por Resonancia Magnética/métodos , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Inmunoterapia , Microambiente Tumoral
9.
Magnes Res ; 35(2): 39-50, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36354241

RESUMEN

Objective: To investigate the metabolic changes in rats with minimal hepatic encephalopathy (MHE) treated with oral magnesium sulphate administration. Materials and Methods: A total of 30 Sprague-Dawley rats were divided into a control group and MHE group (further divided into an MHE group and an MHE-Mg group treated with oral administration of 124 mg/kg/day magnesium sulphate). Morris water maze (MWM), Y maze and narrow beam walking (NBW) were used to evaluate cognitive and motor functions. Brain manganese and magnesium content were measured. The metabolic changes in rats with MHE were investigated using hydrogen-nuclear magnetic resonance. Metabolomic signatures were identified with enrichment and pathway analysis. Results: A significantly decreased number of entries into the MWM within the range of interest, longer latency and total time during NBW, and higher brain manganese content were found in rats with MHE. After magnesium sulphate treatment, the rats with MHE had better behavioural performance and lower brain manganese content. The 25 and 26 metabolomic signatures were identified in the cortex and striatum of rats with MHE. The pathway analysis revealed alanine, aspartate and glutamate metabolism as the major abnormal metabolic pathways associated with these metabolomic signatures. Conclusion: Alanine, aspartate and glutamate metabolism are major abnormal metabolic pathways in rats with MHE, which could be restored by magnesium sulphate treatment.


Asunto(s)
Encefalopatía Hepática , Animales , Ratas , Encefalopatía Hepática/tratamiento farmacológico , Encefalopatía Hepática/complicaciones , Encefalopatía Hepática/metabolismo , Sulfato de Magnesio/farmacología , Sulfato de Magnesio/uso terapéutico , Manganeso/metabolismo , Manganeso/uso terapéutico , Ácido Aspártico/metabolismo , Ácido Aspártico/uso terapéutico , Ratas Sprague-Dawley , Encéfalo/metabolismo , Espectroscopía de Resonancia Magnética , Administración Oral , Alanina/metabolismo , Alanina/uso terapéutico , Glutamatos/metabolismo , Glutamatos/uso terapéutico
10.
ACS Nano ; 16(10): 16909-16923, 2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36200692

RESUMEN

Cancer immunotherapy holds great promise but is generally limited by insufficient induction of anticancer immune responses. Here, a metal micellar nanovaccine is developed by the self-assembly of manganese (Mn), a stimulator of interferon genes (STING) agonist (ABZI) and naphthalocyanine (ONc) coordinated nanoparticles (ONc-Mn-A) in maleimide-modified Pluronic F127 (malF127) micelles. Owing to synergy between Mn and ABZI, the nanovaccine, termed ONc-Mn-A-malF127, elevates levels of interferon-ß (IFNß) by 324- and 8-fold in vivo, compared to use of Mn or ABZI alone. As such, the activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-STING pathway induces sufficient dendritic cell (DC) maturation, eventually resulting in the death of CD8+ T cell-sensitive tumors and CD8+ T cell-resistant tumors by simultaneously promoting cytotoxic CD8+ T cells and NK cells, respectively. Furthermore, with ONc used as a Mn chelator and an efficient photosensitizer, photoinduced immunogenic cell death (ICD) of tumor cells releases damage-associated molecular patterns (DAMPs) and neoantigens from dying primary tumor cells upon laser irradiation, which are captured in situ by malF127 in tumor cells and then transported to DCs. After laser treatment, in addition to the photothermal therapy, immune responses characterized by the level of IFNß are further elevated by another 4-fold. In murine cancer models, ICD-based metalloimmunotherapy using the ONc-Mn-A-malF127 nanovaccine in a single dose by intravenous injection achieved eradication of primary and distant tumors. Taken together, ONc-Mn-A-malF127 offers a nanoplatform to enhance anticancer efficacy by metalloimmunotherapy and photoinduced ICD based immunotherapy with strong abscopal effect.


Asunto(s)
Interferones , Neoplasias , Ratones , Animales , Interferones/metabolismo , Interferones/uso terapéutico , Micelas , Linfocitos T CD8-positivos , Manganeso/uso terapéutico , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Poloxámero , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/uso terapéutico , Neoplasias/tratamiento farmacológico , Inmunoterapia , Antivirales/uso terapéutico , Interferón beta/uso terapéutico , Maleimidas , Quelantes , Antígenos de Neoplasias
11.
ACS Nano ; 16(7): 10904-10917, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35797013

RESUMEN

Semimetallic nanomaterials as photothermal agents for bioimaging and cancer therapy have attracted tremendous interest. However, the poor photothermal stability, low biocompatibility, and single component limit their therapeutic efficiency in cancer treatment. Here, manganese-doped VSe2 semimetallic nanosheets were prepared and subsequently modified with chitosan (named VSe2/Mn-CS NSs) for combined enzyme catalytic and photothermal therapy. VSe2/Mn-CS NSs show high photothermal property with a photothermal conversion efficiency of 34.61% upon 808 nm near-infrared laser irradiation. In the tumor microenvironment, VSe2/Mn-CS NSs can convert endogenous H2O2 into lethal hydroxyl radicals (•OH) to induce cancer cell apoptosis. The interaction between glutathione (GSH) and Se-Se bonds in VSe2/Mn-CS NSs results in the depletion of GSH level, and the valence states transition of manganese ions is also beneficial for the GSH consumption. This dual depletion of GSH markedly enhances the peroxidase (POD) activity, leading to the high •OH production and the improved therapeutic effect. What is more, the T1-weighted magnetic resonance and photoacoustic imaging endow VSe2/Mn-CS NSs with the ability to guide and track the treatment process. Our study provides a research strategy for the application of semimetallic nanomaterials in cancer diagnosis and treatment.


Asunto(s)
Hipertermia Inducida , Metaloides , Neoplasias , Humanos , Manganeso/uso terapéutico , Peróxido de Hidrógeno , Glutatión , Hipertermia Inducida/métodos , Microambiente Tumoral , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Línea Celular Tumoral
12.
Biomaterials ; 286: 121572, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35580473

RESUMEN

Endogenous H2O2 sacrifices for diversified therapeutic reactions against tumor. However, the treatment outcome is not always satisfactory owing to the unsustainable H2O2 supply from tumor microenvironment (TME). Herein, a platinum (Pt) nanourchin-based multi-enzymatic platform (referred to PGMA) is established by surface conjugation of glucose oxidase (GOx) capped with manganese carbonyl (MnCO) and loading 3-amino-1,2,4-triazole (3-AT). The mild acidic and H2O2-rich TME can render the degradation of MnCO, followed by triggering the release of CO gas, 3-AT and Mn2+/3+. The resultant GOx exposure initiates intratumoral glucose depletion, which is promoted by the O2 replenishment through Pt-catalyzed decomposition of H2O2. Meanwhile, intracellular reactive oxygen species (ROS) level is elevated through Mn2+/3+ couple-mediated Fenton-like reaction. Hence, CO release-initiated gas therapy, glucose exhaustion-induced tumor starvation and ROS-triggered chemodynamic therapy are committed to realizing a combinatorial disruption effect on mitochondrial function. Importantly, the released 3-AT can inhibit the activity of endogenous catalase, which effectively elevates the intracellular H2O2 level to compensate its consumption and provides incremental reactant for cascade utilizations. Taken together, this study aims to emphasize the importance of intracellular H2O2 balance during H2O2-depleted therapeutic process, and affords a prime paradigm of applying this strategy for tumor treatment via mitochondrial dysfunction.


Asunto(s)
Peróxido de Hidrógeno , Neoplasias , Línea Celular Tumoral , Glucosa/metabolismo , Glucosa Oxidasa/metabolismo , Homeostasis , Humanos , Peróxido de Hidrógeno/metabolismo , Manganeso/uso terapéutico , Mitocondrias/metabolismo , Neoplasias/terapia , Platino (Metal)/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Microambiente Tumoral
13.
Int J Pharm ; 622: 121810, 2022 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-35580685

RESUMEN

In previous studies, we found that triphenylphosphine-modified doxorubicin (TPP-DOX) can effectively kill drug-resistant tumor cells, but its effect on sensitive tumor cells is weakened. In this research, with albumin from Bovine Serum (BSA) as a carrier, TPP-DOX@MnBSA (TD@MB) nanoparticles were prepared by co-loading TPP-DOX and manganese which can realize the combination of chemotherapy and chemodynamic therapy (CDT). The uniform and stable nano-spherical nanoparticle can promote drug uptake, achieve mitochondrial-targeted drug delivery, increase intracellular reactive oxygen species (ROS) and catalyze the production of highly toxic oxidative hydroxyl radicals (OH·), further inhibiting the growth of both sensitive and drug-resistant MCF-7 cells. Besides, TD@MB can down-regulate the stemness-related proteins and the metastasis-related proteins, potentially decreasing the tumor stemness and metastasis. In vivo experiment indicated that TD@MB was able to exert desired antitumor effect, good tumor targeting and biocompatibility.


Asunto(s)
Neoplasias de la Mama , Nanopartículas , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Catálisis , Línea Celular Tumoral , Doxorrubicina , Femenino , Humanos , Células MCF-7 , Manganeso/uso terapéutico , Nanopartículas/uso terapéutico
14.
J Mater Chem B ; 10(20): 3824-3833, 2022 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-35502611

RESUMEN

Chemodynamic therapy (CDT) and photodynamic therapy (PDT) based on intratumoral generation of reactive oxygen species (ROS) have been playing crucial roles in conquering tumors. However, the above therapeutic methods are still constrained by the overexpressed tumor glutathione (GSH) and intrinsic tumor resistance to conventional organic photosensitizers. Herein, lanthanide-doped nanoparticles (LDNPs) were coated with inorganic bimetallic copper and manganese silicate nanospheres (CMSNs) and modified with sodium alginate (SA) for second near-infrared (NIR-II, 1000-1700 nm) imaging-guided CDT and PDT. Interestingly, cross-relaxation (CR) pathways between Ce3+ and Ho3+ and CR between Ce3+ and Er3+ are fully exploited to enable dual-mode upconversion (UC) and NIR-II downconversion (DC) emissions of LDNPs under 980 nm laser excitation. UC emission can induce CMSNs to produce toxic singlet oxygen (1O2) for PDT, and the released Mn2+ and Cu+ ions caused by GSH-induced degradation of CMSNs can react with endogenous H2O2 to produce hydroxyl radical (˙OH) for CDT. Significantly, the ultrabright NIR-II DC emission endows the systems with exceptional optical imaging capabilities. All results affirm the potency of such an "all in one" theranostic nanomedicine integrating PDT, CDT and remarkable NIR-II imaging abilities accompanied by the function of modulating tumor microenvironment in cancer theranostics.


Asunto(s)
Nanosferas , Neoplasias , Fotoquimioterapia , Cobre/farmacología , Cobre/uso terapéutico , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/uso terapéutico , Manganeso/uso terapéutico , Nanomedicina , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Silicatos , Microambiente Tumoral
15.
ACS Biomater Sci Eng ; 8(5): 1930-1941, 2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35380774

RESUMEN

Inorganic nanomaterials showed great potential as drug carriers for chemotherapeutics molecules due to their biocompatible physical and chemical properties. A manganese-based inorganic nanomaterial manganese phosphate (MnP) had become a new drug carrier in cancer therapy. However, the approach for manganese phosphate preparation and drug integration is still confined in complex methods. Inspired by mimetic mineralization, we proposed a "one-step" method for the preparation of manganese phosphate-doxorubicin (DOX) nanomedicines (MnP-DOX) by manganese ion and DOX complexation. The structural characterization results revealed that the prepared MnP-DOX nanocomplexes were homogeneous with controlled sizes and shapes. More importantly, the MnP-DOX nanocomposites could significantly induce cancer inhibition in vitro and in vivo. The results indicated that the drug molecules were integrated into MnP nanocarriers by mimetic mineralization, which not only prevented the premature release of the drug but also reduced excessive modification. Moreover, the designed MnP-DOX complex showed high loading efficacy and pH-dependent degradation leading to drug release, achieving high efficiency for cancer chemotherapy in vitro and in vivo via a facile process. These achievements presented an approach to construct the manganese phosphate-based chemotherapy nanomedicines by mimetic mineralization for cancer therapy.


Asunto(s)
Nanocompuestos , Neoplasias , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Portadores de Fármacos/uso terapéutico , Humanos , Manganeso/química , Manganeso/uso terapéutico , Nanomedicina , Neoplasias/tratamiento farmacológico , Compuestos Organometálicos
16.
Drug Deliv Transl Res ; 12(11): 2678-2692, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35061221

RESUMEN

The limited tissue penetration depth and tumor hypoxic microenvironment have become the two pivotal obstacles that alleviate the antineoplastic efficacy in tumor photodynamic therapy (PDT). In the research, MnO2-decorated upconversion nanoparticles (UCSMn) have been designed to generate certain oxygen within the solid tumor, and also increase the light penetrating depth due to the optical conversion ability derived from upconversion nanoparticles. Furthermore, upconversion nanoparticles as the inner core are coated by mesoporous silica for the loading of curcumin as photosensitizer and chemotherapeutics, and then a MnO2 shell is proceeding to grow via redox method. When reaching the tumor tissue, the MnO2 nanoshells of UCSMn could be rapidly degraded into manganese ions (Mn2+) owing to the reaction with H2O2 in acidic tumor microenvironment, meanwhile producing oxygen and facilitating curcumin release. Once the tumor is illuminated by 980 nm light, the upconversion nanoparticles can transform the infrared light to visible light of 450 nm and 475.5 nm, which can be efficiently absorbed by curcumin, and then produce singlet oxygen to induce tumor cell apoptosis. Curcumin played a dual role which can not only be acted as a photosensitizer, but also a chemotherapeutic agent to further reinforce the antitumor activity. In short, the intelligent nanostructure has the potential to overcome the above-mentioned shortcomings existed in PDT and eventually do work well in the hypoxia tumors. MnO2-decorated upconversion nanoparticle to solve the tissue penetration and tumor hypoxic microenvironment for tumor photodynamic therapy.


Asunto(s)
Antineoplásicos , Curcumina , Nanopartículas , Neoplasias , Fotoquimioterapia , Antineoplásicos/química , Línea Celular Tumoral , Humanos , Peróxido de Hidrógeno , Manganeso/uso terapéutico , Compuestos de Manganeso/química , Compuestos de Manganeso/uso terapéutico , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Óxidos/química , Óxidos/uso terapéutico , Oxígeno/metabolismo , Oxígeno/uso terapéutico , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes , Dióxido de Silicio/química , Oxígeno Singlete/metabolismo , Oxígeno Singlete/uso terapéutico
17.
J Fluoresc ; 32(2): 593-601, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35015178

RESUMEN

Via the solvothermal reaction between Zn(II) or Mn(II) salts and 5-(3,4-dicarboxylphenoxy)nicotinic acid (H3L) ligand, a trifunctional N,O-building block having three diverse kinds of functional groups (O-ether, N-pyridyl and COOH), two new coordination polymers (CPs) could be generated, and their chemical formulae respectively are {[Mn3(L)2(H2O)2]·4H2O} (1) and {[Zn(HL)]·NMP} (2). The complex 2 based on Zn(II) possesses high efficiency of fluorescence quenching for the nitrophenol (2,4,6-trinitrophenol, TNP; 4-nitrophenol, 4-NP; 3-nitrophenol, 3-NP; 2-nitrophenol, 2-NP) in the aqueous solution. Furthermore, the treatment activity of compounds on the atherosclerosis was assessed, and relevant mechanism was investigated. First of all, the ELISA assay was used to measure the content of the inflammatory cytokines released into the plasma. Besides, the levels of the NF-κb signaling pathway in the vascular endothelial cells were measured with real time RT-PCR. The hemolysis test was conducted in this research to measure the biocompatibility of the new compound.


Asunto(s)
Aterosclerosis/sangre , Complejos de Coordinación/química , Manganeso/química , Polímeros/química , Zinc/química , Animales , Aterosclerosis/tratamiento farmacológico , Complejos de Coordinación/uso terapéutico , Citocinas/sangre , Células Endoteliales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Hemólisis , Humanos , Ligandos , Manganeso/uso terapéutico , FN-kappa B/metabolismo , Niacina/química , Nitrofenoles/química , Polímeros/uso terapéutico , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Espectrometría de Fluorescencia/métodos , Zinc/uso terapéutico
18.
J Hematol Oncol ; 14(1): 146, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34526097

RESUMEN

BACKGROUND: Our previous work showed that the anti-TGF-ß/PD-L1 bispecific antibody YM101 effectively overcame anti-PD-L1 resistance in immune-excluded tumor models. However, in immune-desert models, the efficacy of YM101 was limited. Bivalent manganese (Mn2+) is identified as a natural stimulator of interferon genes (STING) agonist, which might enhance cancer antigen presentation and improve the therapeutic effect of YM101. METHODS: The effect of Mn2+ on STING pathway was validated by western blotting and enzyme-linked immunosorbent assay. Dendritic cell (DC) maturation was measured by flow cytometry. The synergistic effect between Mn2+ and YM101 in vitro was determined by one-way mixed lymphocyte reaction, CFSE dilution assay, and cytokine detection. The in vivo antitumor effect of Mn2+ plus YM101 therapy was assessed in CT26, EMT-6, H22, and B16 tumor models. Flow cytometry, RNA-seq, and immunofluorescent staining were adopted to investigate the alterations in the tumor microenvironment. RESULTS: Mn2+ could activate STING pathway and promote the maturation of human and murine DC. The results of one-way mixed lymphocyte reaction showed that Mn2+ synergized YM101 in T cell activation. Moreover, in multiple syngeneic murine tumor models, Mn2+ plus YM101 therapy exhibited a durable antitumor effect and prolonged the survival of tumor-bearing mice. Relative to YM101 monotherapy and Mn2+ plus anti-PD-L1 therapy, Mn2+ plus YM101 treatment had a more powerful antitumor effect and a broader antitumor spectrum. Mechanistically, Mn2+ plus YM101 strategy simultaneously regulated multiple components in the antitumor immunity and drove the shift from immune-excluded or immune-desert to immune-inflamed tumors. The investigation in the TME indicated Mn2+ plus YM101 strategy activated innate and adaptive immunity, enhanced cancer antigen presentation, and upregulated the density and function of tumor-infiltrating lymphocytes. This normalized TME and reinvigorated antitumor immunity contributed to the superior antitumor effect of the combination therapy. CONCLUSION: Combining Mn2+ with YM101 has a synergistic antitumor effect, effectively controlling tumor growth and prolonging the survival of tumor-bearing mice. This novel cocktail strategy has the potential to be a universal regimen for inflamed and non-inflamed tumors.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Manganeso/uso terapéutico , Neoplasias/terapia , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Inmunoterapia/métodos , Ratones Endogámicos BALB C , Neoplasias/inmunología , Factor de Crecimiento Transformador beta/inmunología
19.
Environ Sci Pollut Res Int ; 28(46): 65474-65486, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34322790

RESUMEN

Nanotechnology is a rich field with infinite possibilities of drug designs for cancer treatment. We aimed to biosynthesize manganese nanoparticles (Mn NPs) using Lactobacillus helveticus to investigate its anticancer synergistic effect with low-dose gamma radiation on HCC-induced rats. Diethylnitrosamine (DEN) (20 mg/kg BW, 5 times a week for 6 weeks) induced HCC in rats. Rats received Mn NPs (5 mg/kg BW/day) by gastric gavage over 4 weeks concomitant with single dose of gamma radiation (γ-R) (0.25 Gy). Characterization, cytotoxicity, and anticancer activity of Mn NPs were evaluated. DEN-induced significant liver dysfunction (alanine transaminase activity ALT, total proteins, and albumin levels) associated with significant increase in lipid peroxidation levels with reduction in super oxide dismutase activity. Furthermore, DEN intoxication is sponsored for remarkable increase in levels of Alfa-fetoprotein, tumor necrosis factor α, vascular endothelial growth factor, and transforming growth factor beta with remarkable decrease in caspase 3 and cytochrome c. Treatment with Mn NPs (4.98-11.58 nm) and single dose gamma radiation evoked significant repair in ALT, total protein, and albumin accompanied with balanced oxidative status, diminished inflammatory biomarkers, angiogenic factor, and growth factor with restoration in apoptotic factors. Mn NPs revealed obvious in vitro cytotoxic activity against HepG2 cell line in a dose-dependent manner. Our findings were well appreciated with the histopathological study. In conclusion, a new approach of the single or combined use of Mn NPs with low-dose γ-radiation regimens as promising paradigm for HCC treatment is recommended.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas Experimentales/terapia , Nanopartículas del Metal/uso terapéutico , Animales , Carcinoma Hepatocelular/terapia , Dietilnitrosamina , Rayos gamma , Hígado , Manganeso/uso terapéutico , Ratas , Factor A de Crecimiento Endotelial Vascular
20.
ACS Appl Mater Interfaces ; 13(4): 4861-4873, 2021 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-33471499

RESUMEN

A combination treatment strategy that relies on the synergetic effects of different therapeutic approaches has been considered to be an effective method for cancer therapy. Herein, a chemotherapeutic drug (doxorubicin, Dox) and a manganese ion (Mn2+) were co-loaded into regenerated silk fibroin-based nanoparticles (NPs), followed by the surface conjugation of phycocyanin (PC) to construct tumor microenvironment-activated nanococktails. The resultant PC-Mn@Dox-NPs showed increased drug release rates by responding to various stimulating factors (acidic pH, hydrogen peroxide (H2O2), and glutathione), revealing that they could efficiently release the payloads (Dox and Mn2+) in tumor cells. The released Dox could not only inhibit the growth of tumor cells but also generated a large amount of H2O2. The elevated H2O2 was decomposed into the highly harmful hydroxyl radicals and oxygen through an Mn2+-mediated Fenton-like reaction. Furthermore, the generated oxygen participated in photodynamic therapy (PDT) and produced abundant singlet oxygen. Our investigations demonstrate that these PC-Mn@Dox-NPs exhibit multiple bioresponsibilities and favorable biosafety. By integrating Dox-induced chemotherapy, Mn2+-mediated chemodynamic therapy, and PC-based PDT via cascade reactions, PC-Mn@Dox-NPs achieved enhanced in vitro and in vivo anticancer efficacies compared to all the mono- or dual-therapeutic approaches. These findings reveal that PC-Mn@Dox-NPs can be exploited as a promising nanococktail for cascade reaction-mediated synergistic cancer treatment.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Manganeso/administración & dosificación , Neoplasias/tratamiento farmacológico , Fármacos Fotosensibilizantes/administración & dosificación , Ficocianina/administración & dosificación , Animales , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/uso terapéutico , Bombyx/química , Cationes Bivalentes/administración & dosificación , Cationes Bivalentes/farmacología , Cationes Bivalentes/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Portadores de Fármacos/química , Fibroínas/química , Glutatión/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Concentración de Iones de Hidrógeno , Manganeso/farmacología , Manganeso/uso terapéutico , Ratones , Nanopartículas/química , Neoplasias/metabolismo , Neoplasias/patología , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Ficocianina/farmacología , Ficocianina/uso terapéutico , Microambiente Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA