Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
1.
Theranostics ; 14(11): 4375-4392, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113803

RESUMEN

Rationale: Autism spectrum disorder (ASD) represents a complex neurodevelopmental condition lacking specific pharmacological interventions. Given the multifaced etiology of ASD, there exist no effective treatment for ASD. Rapamycin (RAPA) can activate autophagy by inhibiting the mTOR pathway and has exhibited promising effects in treating central nervous system disorders; however, its limited ability to cross the blood-brain barrier (BBB) has hindered its clinical efficacy, leading to substantial side effects. Methods: To address this challenge, we designed a drug delivery system utilizing red blood cell membrane (CM) vesicles modified with SS31 peptides to enhance the brain penetration of RAPA for the treatment of autism. Results: The fabricated SCM@RAPA nanoparticles, with an average diameter of 110 nm, exhibit rapid release of RAPA in a pathological environment characterized by oxidative stress. In vitro results demonstrate that SCM@RAPA effectively activate cellular autophagy, reduce intracellular ROS levels, improve mitochondrial function, thereby ameliorating neuronal damage. SS31 peptide modification significantly enhances the BBB penetration and rapid brain accumulation of SCM@RAPA. Notably, SCM@RAPA nanoparticles demonstrate the potential to ameliorate social deficits, improve cognitive function, and reverse neuronal impairments in valproic acid (VPA)-induced ASD models. Conclusions: The therapeutic potential of SCM@RAPA in managing ASD signifies a paradigm shift in autism drug treatment, holding promise for clinical interventions in diverse neurological conditions.


Asunto(s)
Trastorno del Espectro Autista , Autofagia , Barrera Hematoencefálica , Nanopartículas , Estrés Oxidativo , Sirolimus , Sirolimus/administración & dosificación , Sirolimus/farmacología , Estrés Oxidativo/efectos de los fármacos , Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/metabolismo , Animales , Autofagia/efectos de los fármacos , Nanopartículas/química , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Ratones , Humanos , Sistemas de Liberación de Medicamentos/métodos , Modelos Animales de Enfermedad , Masculino , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Biomimética/métodos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Péptidos/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Ácido Valproico/administración & dosificación , Ácido Valproico/farmacología
2.
J Control Release ; 374: 50-60, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39111599

RESUMEN

Corneal neovascularization (CNV) is a major cause of blindness worldwide. However, the recent drug treatment is limited by repeated administration and low drug bioavailability. In this work, SU6668 (an inhibitor of receptor tyrosine kinases) and indocyanine green (ICG) are loaded onto poly(lactic-co-glycolic acid) (PLGA) nanoparticles, and then coated with anti-VEGFR2 single chain antibody (AbVr2 scFv) genetically engineered cell membrane vesicles. The nanomedicine is delivered via eye drops, and the hyperthermia induced by laser irradiation could block the blood vessels. Meanwhile, the photothermal effect can also cause the degradation of nanomaterials and release chemotherapeutic drugs in the blocked area, thereby continuously inhibit the neovascularization. Furthermore, SU6668 could inhibit the expression of heat shock protein 70 (HSP70), promoting the cell death induced by photothermal effect. In conclusion, the combination of photothermal and chemotherapy drugs provides a novel, effective and safe approach for the treatment of CNV.


Asunto(s)
Neovascularización de la Córnea , Verde de Indocianina , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Nanomedicina Teranóstica , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Verde de Indocianina/administración & dosificación , Verde de Indocianina/farmacocinética , Nanomedicina Teranóstica/métodos , Humanos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/química , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/farmacocinética , Liberación de Fármacos , Proteínas HSP70 de Choque Térmico , Biomimética , Terapia Fototérmica/métodos
3.
J Control Release ; 373: 699-712, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39089504

RESUMEN

Adapting the mechanical strength between the implant materials and the brain tissue is crucial for the postoperative treatment of glioblastoma. However, no related study has been reported. Herein, we report an injectable lipoic acid­iron (LA-Fe) hydrogel (LFH) that can adapt to the mechanical strength of various brain tissues, including human brain tissue, by coordinating Fe3+ into a hybrid hydrogel of LA and its sodium salt (LANa). When LFH, which matches the mechanical properties of mouse brain tissue (337 ± 8.06 Pa), was injected into the brain resection cavity, the water content of the brain tissue was maintained at a normal level (77%). Similarly, LFH did not induce the activation or hypertrophy of glial astrocytes, effectively preventing brain edema and scar hyperplasia. Notably, LFH spontaneously degrades in the interstitial fluid, releasing LA and Fe3+ into tumor cells. The redox couples LA/DHLA (dihydrolipoic acid, reduction form of LA in cells) and Fe3+/Fe2+ would regenerate each other to continuously provide ROS to induce ferroptosis and activate immunogenic cell death. As loaded the anti-PDL1, anti-PDL1@LFH further enhanced the efficacy of tumor-immunotherapy and promoted tumor ferroptosis. The injectable hydrogel that adapted the mechanical strength of tissues shed a new light for the tumor postoperative treatment.


Asunto(s)
Neoplasias Encefálicas , Encéfalo , Glioblastoma , Hidrogeles , Ácido Tióctico , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Animales , Hidrogeles/administración & dosificación , Hidrogeles/química , Ácido Tióctico/química , Ácido Tióctico/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Humanos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular Tumoral , Ratones , Hierro/química , Inyecciones , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Ferroptosis/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C
4.
Int J Pharm ; 662: 124493, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39048042

RESUMEN

Sepsis is a life-threatening syndrome resulting from an imbalanced immune response to severe infections. Despite advances in nanomedicines, effective treatments for sepsis are still lacking. Herein, vancomycin free base (VCM)-loaded dual functionalized biomimetic liposomes based on a novel TLR4-targeting peptide (P3) and hyaluronic acid (HA) (HA-P3-Lipo) were developed to enhance sepsis therapy. The nanocarrier revealed appropriate physicochemical parameters, good stability, and biocompatibility. The release of VCM from HA-P3-Lipo was found to be sustained with 76 % VCM released in 48 h. The biomimicry was elucidated by in silico tools and MST and results confirmed strong binding between the system and TLR4. Furthermore, HA-P3-Lipo revealed 2-fold enhanced antibacterial activity against S. aureus, sustained antibacterial activity against MRSA over 72 h and 5-fold better MRSA biofilm inhibition compared to bare VCM. Bacterial-killing kinetics and flow cytometry confirmed the superiority of HA-P3-Lipo in eliminating MRSA faster than VCM. The in vivo potential of the nanocarrier was elucidated in an MRSA-induced sepsis mice model, and the results confirmed the superiority of HA-P3-Lipo compared to free VCM in eliminating bacteria and down-regulating the proinflammatory markers. Therefore, HA-P3-Lipo exhibits potential as a promising novel multi-functional nanosystem against sepsis and could significantly contribute to the transformation of sepsis therapy.


Asunto(s)
Antibacterianos , Ácido Hialurónico , Liposomas , Staphylococcus aureus Resistente a Meticilina , Péptidos , Sepsis , Vancomicina , Ácido Hialurónico/química , Animales , Sepsis/tratamiento farmacológico , Sepsis/microbiología , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Antibacterianos/química , Ratones , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Vancomicina/administración & dosificación , Vancomicina/farmacología , Vancomicina/química , Péptidos/química , Péptidos/farmacología , Péptidos/administración & dosificación , Liberación de Fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Receptor Toll-Like 4/metabolismo , Biopelículas/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Masculino , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Materiales Biomiméticos/administración & dosificación , Células RAW 264.7
5.
J Control Release ; 373: 172-188, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38972639

RESUMEN

Ovarian cancer is one of the deadliest cancers, and combined chemo- and immunotherapies are potential strategies to combat it. However, the anti-cancer efficacy of the combined therapies may be limited by the non-selective co-delivery of chemotherapy and immunotherapy. Herein, a combined chemo- and immunotherapy is designed to selectively target ovarian tumor (ID8) cells and dendritic cells (DCs) using ID8 cell membrane (IM) and bacterial outer membrane vesicles (OMVs), respectively. Doxorubicin (DOX) and Ovalbumin (OVA) peptide (OVA257-264) are chosen as model chemotherapy and immunotherapy agents, respectively. A DNA nanocube capable of easily loading DOX or OVA257-264 is chosen as the carrier. Firstly, the DNA nanocube is used to load DOX or OVA257-264 to prepare cube-DOX or cube-OVA. This nanocube was then encapsulated with IM to form IM@Cube-DOX and with OMV to form OMV@Cube-OVA. IM@Cube-DOX can be selectively taken up by ID8 cells, leading to effective cell killing, while OMV@Cube-OVA targets and activates DC2.4 cells in vitro. Both IM@Cube-DOX and OMV@Cube-OVA show increased accumulation at ID8 tumors in C57BL/6 mice. Combined IM@Cube-DOX + OMV@Cube-OVA therapy demonstrates better anti-tumor efficacy than non-selective delivery methods such as OMV@(Cube-DOX + Cube-OVA) or IM@(Cube-DOX + Cube-OVA) in ID8-OVA tumor-bearing mice. In conclusion, this study demonstrates a biomimetic delivery strategy that enables selective drug delivery to tumor cells and DCs, thereby enhancing the anti-tumor efficacy of combined chemo- and immunotherapy through the selective delivery strategy.


Asunto(s)
Células Dendríticas , Doxorrubicina , Inmunoterapia , Ratones Endogámicos C57BL , Nanomedicina , Neoplasias Ováricas , Femenino , Animales , Neoplasias Ováricas/terapia , Neoplasias Ováricas/tratamiento farmacológico , Doxorrubicina/administración & dosificación , Doxorrubicina/uso terapéutico , Doxorrubicina/química , Inmunoterapia/métodos , Línea Celular Tumoral , Nanomedicina/métodos , Células Dendríticas/inmunología , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Humanos , Ratones , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Biomimética/métodos , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación
6.
Int J Pharm ; 661: 124426, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38972519

RESUMEN

In recent years, the use of arsenic trioxide (ATO) in the context of ovarian cancer chemotherapy has attracted significant attention. However, ATO's limited biocompatibility and the occurrence of severe toxic side effects hinder its clinical application. A nanoparticle (NP) drug delivery system using ATO as a therapeutic agent is reported in this study. Achieving a synergistic effect by combining starvation therapy, chemodynamic therapy, and chemotherapy for the treatment of ovarian cancer was the ultimate goal of this system. This nanotechnology-based drug delivery system (NDDS) introduced arsenic-manganese complexes into cancer cells, leading to the subsequent release of lethal arsenic ions (As3+) and manganese ions (Mn2+). The acidic microenvironment of the tumor facilitated this process, and MR imaging offered real-time monitoring of the ATO dose distribution. Simultaneously, to produce reactive oxygen species that induced cell death through a Fenton-like reaction, Mn2+ exploited the surplus of hydrogen peroxide (H2O2) within tumor cells. Glucose oxidase-based starvation therapy further supported this mechanism, which restored H2O2 and lowered the cellular acidity. Consequently, this approach achieved self-enhanced chemodynamic therapy. Homologous targeting of the NPs was facilitated through the use of SKOV3 cell membranes that encapsulated the NPs. Hence, the use of a multimodal NDDS that integrated ATO delivery, therapy, and monitoring exhibited superior efficacy and biocompatibility compared with the nonspecific administration of ATO. This approach presents a novel concept for the diagnosis and treatment of ovarian cancer.


Asunto(s)
Antineoplásicos , Trióxido de Arsénico , Nanopartículas , Neoplasias Ováricas , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Humanos , Concentración de Iones de Hidrógeno , Línea Celular Tumoral , Trióxido de Arsénico/administración & dosificación , Trióxido de Arsénico/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacología , Nanopartículas/química , Manganeso/química , Manganeso/administración & dosificación , Peróxido de Hidrógeno , Especies Reactivas de Oxígeno/metabolismo , Ratones , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación , Ratones Desnudos , Sistemas de Liberación de Medicamentos/métodos , Sistema de Administración de Fármacos con Nanopartículas/química , Glucosa Oxidasa/administración & dosificación , Ratones Endogámicos BALB C
7.
J Control Release ; 371: 484-497, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38851537

RESUMEN

The precise and targeted delivery of therapeutic agents to the lesion sites remains a major challenge in treating brain diseases represented by ischemic stroke. Herein, we modified liposomes with mesenchymal stem cells (MSC) membrane to construct biomimetic liposomes, termed MSCsome. MSCsome (115.99 ± 4.03 nm) exhibited concentrated accumulation in the cerebral infarcted hemisphere of mice with cerebral ischemia-reperfusion injury, while showing uniform distribution in the two cerebral hemispheres of normal mice. Moreover, MSCsome exhibited high colocalization with damaged nerve cells in the infarcted hemisphere, highlighting its advantageous precise targeting capabilities over liposomes at both the tissue and cellular levels. Leveraging its superior targeting properties, MSCsome effectively delivered Dl-3-n-butylphthalide (NBP) to the injured hemisphere, making a single-dose (15 mg/kg) intravenous injection of NBP-encapsulated MSCsome facilitate the recovery of motor functions in model mice by improving the damaged microenvironment and suppressing neuroinflammation. This study underscores that the modification of the MSC membrane notably enhances the capacity of liposomes for precisely targeting the injured hemisphere, which is particularly crucial in treating cerebral ischemia-reperfusion injury.


Asunto(s)
Benzofuranos , Sistemas de Liberación de Medicamentos , Liposomas , Células Madre Mesenquimatosas , Daño por Reperfusión , Animales , Daño por Reperfusión/terapia , Masculino , Benzofuranos/administración & dosificación , Isquemia Encefálica/terapia , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Trasplante de Células Madre Mesenquimatosas/métodos
8.
J Control Release ; 372: 531-550, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38851535

RESUMEN

Recovery and survival following traumatic brain injury (TBI) depends on optimal amelioration of secondary injuries at lesion site. Delivering mitochondria-protecting drugs to neurons may revive damaged neurons at sites secondarily traumatized by TBI. Pioglitazone (PGZ) is a promising candidate for TBI treatment, limited by its low brain accumulation and poor targetability to neurons. Herein, we report a ROS-responsive nanosystem, camouflaged by hybrid membranes of platelets and engineered extracellular vesicles (EVs) (C3-EPm-|TKNPs|), that can be used for targeted delivery of PGZ for TBI therapy. Inspired by intrinsic ability of macrophages for inflammatory chemotaxis, engineered M2-like macrophage-derived EVs were constructed by fusing C3 peptide to EVs membrane integrator protein, Lamp2b, to confer them with ability to target neurons in inflamed lesions. Platelets provided hybridized EPm with capabilities to target hemorrhagic area caused by trauma via surface proteins. Consequently, C3-EPm-|PGZ-TKNPs| were orientedly delivered to neurons located in the traumatized hemisphere after intravenous administration, and triggered the release of PGZ from TKNPs via oxidative stress. The current work demonstrate that C3-EPm-|TKNPs| can effectively deliver PGZ to alleviate mitochondrial damage via mitoNEET for neuroprotection, further reversing behavioral deficits in TBI mice. Our findings provide proof-of-concept evidence of C3-EPm-|TKNPs|-derived nanodrugs as potential clinical approaches against neuroinflammation-related intracranial diseases.


Asunto(s)
Plaquetas , Lesiones Traumáticas del Encéfalo , Exosomas , Neuronas , Especies Reactivas de Oxígeno , Animales , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Plaquetas/metabolismo , Masculino , Exosomas/metabolismo , Ratones , Péptidos/administración & dosificación , Péptidos/química , Ratones Endogámicos C57BL , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/química , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Sistemas de Liberación de Medicamentos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Biomimética
9.
Drug Resist Updat ; 75: 101098, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38833804

RESUMEN

Breakthroughs in actual clinical applications have begun through vaccine-based cancer immunotherapy, which uses the body's immune system, both humoral and cellular, to attack malignant cells and fight diseases. However, conventional vaccine approaches still face multiple challenges eliciting effective antigen-specific immune responses, resulting in immunotherapy resistance. In recent years, biomimetic nanovaccines have emerged as a promising alternative to conventional vaccine approaches by incorporating the natural structure of various biological entities, such as cells, viruses, and bacteria. Biomimetic nanovaccines offer the benefit of targeted antigen-presenting cell (APC) delivery, improved antigen/adjuvant loading, and biocompatibility, thereby improving the sensitivity of immunotherapy. This review presents a comprehensive overview of several kinds of biomimetic nanovaccines in anticancer immune response, including cell membrane-coated nanovaccines, self-assembling protein-based nanovaccines, extracellular vesicle-based nanovaccines, natural ligand-modified nanovaccines, artificial antigen-presenting cells-based nanovaccines and liposome-based nanovaccines. We also discuss the perspectives and challenges associated with the clinical translation of emerging biomimetic nanovaccine platforms for sensitizing cancer cells to immunotherapy.


Asunto(s)
Células Presentadoras de Antígenos , Vacunas contra el Cáncer , Inmunoterapia , Nanopartículas , Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/inmunología , Inmunoterapia/métodos , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Nanopartículas/administración & dosificación , Células Presentadoras de Antígenos/inmunología , Biomimética/métodos , Materiales Biomiméticos/administración & dosificación , Animales , Liposomas , Nanovacunas
10.
Int J Nanomedicine ; 19: 6177-6199, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38911498

RESUMEN

Purpose: Ginsenoside Rg3 (Rg3) and Panax notoginseng saponins (PNS) can be used for ischemic stroke treatment, however, the lack of targeting to the ischemic region limits the therapeutic effect. To address this, we leveraged the affinity of macrophage membrane proteins for inflamed brain microvascular endothelial cells to develop a macrophage membrane-cloaked liposome loaded with Rg3 and PNS (MM-Lip-Rg3/PNS), which can precisely target brain lesion region through intranasal administration. Methods: MM-Lip-Rg3/PNS was prepared by co-extrusion method and was performed by characterization, stability, surface protein, and morphology. The cellular uptake, immune escape ability, and blood-brain barrier crossing ability of MM-Lip-Rg3/PNS were studied in vitro. The in vivo brain targeting, biodistribution and anti-ischemic efficacy of MM-Lip-Rg3/PNS were evaluated in MACO rats, and we determined the diversity of the nasal brain pathway through the olfactory nerve blockade model in rats. Finally, the pharmacokinetics and brain targeting index of MM-Lip-Rg3/PNS were investigated. Results: Our results indicated that MM-Lip-Rg3/PNS was spherical with a shell-core structure. MM-Lip-Rg3/PNS can avoid mononuclear phagocytosis, actively bind to inflammatory endothelial cells, and have the ability to cross the blood-brain barrier. Moreover, MM-Lip-Rg3/PNS could specifically target ischemic sites, even microglia, increase the cumulative number of drugs in the brain, improve the inflammatory environment of the brain, and reduce the infarct size. By comparing olfactory nerve-blocking rats with normal rats, it was found that there are direct and indirect pathways for nasal entry into the brain. Pharmacokinetics demonstrated that MM-Lip-Rg3/PNS exhibited stronger brain targeting and prolonged drug half-life. Conclusion: MM-Lip-Rg3/PNS might contribute to the accumulation of Rg3 and PNS in the ischemic brain area to improve treatment efficacy. This biomimetic nano-drug delivery system provides a new and promising strategy for the treatment of ischemic stroke.


Asunto(s)
Administración Intranasal , Barrera Hematoencefálica , Ginsenósidos , Accidente Cerebrovascular Isquémico , Liposomas , Macrófagos , Animales , Liposomas/química , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Ratas , Masculino , Ginsenósidos/farmacocinética , Ginsenósidos/química , Ginsenósidos/administración & dosificación , Ginsenósidos/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Ratas Sprague-Dawley , Distribución Tisular , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/administración & dosificación , Saponinas/farmacocinética , Saponinas/química , Saponinas/administración & dosificación , Saponinas/farmacología , Ratones
11.
Sci Adv ; 10(19): eadm9561, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38718119

RESUMEN

Lactic acid (LA) accumulation in the tumor microenvironment poses notable challenges to effective tumor immunotherapy. Here, an intelligent tumor treatment microrobot based on the unique physiological structure and metabolic characteristics of Veillonella atypica (VA) is proposed by loading Staphylococcus aureus cell membrane-coating BaTiO3 nanocubes (SAM@BTO) on the surface of VA cells (VA-SAM@BTO) via click chemical reaction. Following oral administration, VA-SAM@BTO accurately targeted orthotopic colorectal cancer through inflammatory targeting of SAM and hypoxic targeting of VA. Under in vitro ultrasonic stimulation, BTO catalyzed two reduction reactions (O2 → •O2- and CO2 → CO) and three oxidation reactions (H2O → •OH, GSH → GSSG, and LA → PA) simultaneously, effectively inducing immunogenic death of tumor cells. BTO catalyzed the oxidative coupling of VA cells metabolized LA, effectively disrupting the immunosuppressive microenvironment, improving dendritic cell maturation and macrophage M1 polarization, and increasing effector T cell proportions while decreasing regulatory T cell numbers, which facilitates synergetic catalysis and immunotherapy.


Asunto(s)
Compuestos de Bario , Materiales Biomiméticos , Neoplasias Colorrectales , Terapia de Inmunosupresión , Nanotubos , Robótica , Titanio , Microambiente Tumoral , Veillonella , Materiales Biomiméticos/administración & dosificación , Catálisis , Neoplasias Colorrectales/tratamiento farmacológico , Staphylococcus aureus , Nanotubos/química , Titanio/administración & dosificación , Titanio/farmacología , Compuestos de Bario/administración & dosificación , Compuestos de Bario/farmacología , Membrana Celular/química , Administración Oral , Oxidación-Reducción , Terapia de Inmunosupresión/métodos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ácido Láctico/metabolismo , Humanos , Línea Celular Tumoral
12.
J Control Release ; 370: 556-569, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38697316

RESUMEN

The treatment dilemma of triple-negative breast cancer (TNBC) revolves around drug resistance and metastasis. Cancer-associated fibroblasts (CAFs) contribute to cisplatin (Cis) resistance and further metastasis in TNBC, making TNBC a difficult-to-treat disease. The dense stromal barrier which restricts drug delivery, invasive phenotype of tumor cells, and immunosuppressive tumor microenvironment (TME) induced by CAFs serve as three "shields" for TNBC against Cis therapy. Here, we designed a silybin-loaded biomimetic nanoparticle coated with anisamide-modified red blood cell membrane (ARm@SNP) as a "nanospear" for CAFs-targeting, which could shatter the "shields" and significantly exhibit inhibitory effect on 4T1 cells in combination with Cis both in vitro and in vivo. The ARm@SNP/Cis elicited 4T1 tumor growth arrest and destroyed three "shields" as follows: disintegrating the stromal barrier by inhibiting blood vessels growth and the expression of fibronectin; decreasing 4T1 cell invasion and metastasis by affecting the TGF-ß/Twist/EMT pathway which impeded EMT activation; reversing the immunosuppressive microenvironment by increasing the activity and infiltration of immunocompetent cells. Based on CAFs-targeting, ARm@SNP reversed the resistance of Cis, remodeled the TME and inhibited invasion and metastasis while significantly improving the therapeutic effect of Cis on 4T1 tumor-bearing mice, providing a promising approach for treating intractable TNBC.


Asunto(s)
Antineoplásicos , Fibroblastos Asociados al Cáncer , Cisplatino , Ratones Endogámicos BALB C , Nanopartículas , Neoplasias de la Mama Triple Negativas , Microambiente Tumoral , Animales , Cisplatino/administración & dosificación , Cisplatino/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Femenino , Microambiente Tumoral/efectos de los fármacos , Nanopartículas/química , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación , Humanos , Ratones , Biomimética/métodos
13.
Int J Pharm ; 659: 124261, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38782155

RESUMEN

The progression of renal fibrosis to end-stage renal disease (ESRD) is significantly influenced by transforming growth factor-beta (TGF-beta) signal pathway. This study aimed to develop nanoparticles (PMVs@PLGA complexes) with platelet membrane camouflage, which can transport interfering RNA to target and regulate the TGF-ß1 pathway in damaged renal tissues. The aim is to reduce the severity of acute kidney injury and to reduce fibrosis in chronic kidney disease. Hence, we formulated PMVs@TGF-ß1-siRNA NP complexes and employed them for both in vitro and in vivo therapy. From the experimental findings we know that the PMVs@siRNA NPs could effectively target the kidneys in unilateral ureteral obstruction (UUO) mice and ischemia/reperfusion injury (I/R) mice. In animal models of treatment, PMVs@siRNA NP complexes effectively decreased the expression of TGF-ß1 and mitigated inflammation and fibrosis in the kidneys by blocking the TGF-ß1/Smad3 pathway. Therefore, these PMVs@siRNA NP complexes can serve as a promising biological delivery system for treating kidney diseases.


Asunto(s)
Fibrosis , Nanopartículas , ARN Interferente Pequeño , Factor de Crecimiento Transformador beta1 , Animales , ARN Interferente Pequeño/administración & dosificación , Factor de Crecimiento Transformador beta1/metabolismo , Masculino , Ratones , Plaquetas/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/prevención & control , Riñón/metabolismo , Riñón/patología , Riñón/efectos de los fármacos , Ratones Endogámicos C57BL , Obstrucción Ureteral/terapia , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/química , Inflamación/tratamiento farmacológico , Modelos Animales de Enfermedad , Humanos , Proteína smad3/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Lesión Renal Aguda/prevención & control
14.
J Control Release ; 371: 204-215, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38810704

RESUMEN

Cuproptosis, a newly discovered mechanism of inducing tumor cell death, primarily relies on the intracellular accumulation of copper ions. The utilization of Cu-based nanomaterials to induce cuproptosis holds promising prospects in future biomedical applications. However, the presence of high levels of glutathione (GSH) within tumor cells hinders the efficacy of cuproptosis. In this study, we have developed a BPTES-loaded biomimetic Cu-doped polypyrrole nanoparticles (CuP) nanosystem (PCB) for enhanced cuproptosis and immune modulation. PCB comprises an internal BPTES and CuP core and an external platelet membrane (PM) that facilitates active targeting to tumor sites following intravenous administration. Subsequently, PCB effectively suppresses glutaminase (GLS1) activity, thereby reducing GSH content. Moreover, CuP catalyze intracellular H2O2, amplifying oxidative stress while simultaneously inducing dihydrolipoyl transacetylase (DLAT) oligomerization through released Cu2+, resulting in cuproptosis. PCB not only inhibits primary tumors but also exhibits inhibitory effects on abscopal tumors. This work represents the first instance where GLS inhibition has been employed to enhance cuproptosis and immunotherapy. It also provides valuable insights into further investigations on cuproptosis.


Asunto(s)
Materiales Biomiméticos , Neoplasias de la Mama , Cobre , Glutamina , Inmunoterapia , Nanopartículas , Polímeros , Pirroles , Cobre/química , Polímeros/química , Nanopartículas/química , Nanopartículas/administración & dosificación , Animales , Femenino , Pirroles/administración & dosificación , Pirroles/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Humanos , Inmunoterapia/métodos , Línea Celular Tumoral , Glutamina/metabolismo , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación , Ratones Endogámicos BALB C , Glutaminasa/metabolismo , Glutaminasa/antagonistas & inhibidores , Ratones , Glutatión/metabolismo
15.
J Pharmacol Exp Ther ; 389(3): 289-300, 2024 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-38580449

RESUMEN

Invasive bacterial infections and sepsis are persistent global health concerns, complicated further by the escalating threat of antibiotic resistance. Over the past 40 years, collaborative endeavors to improve the diagnosis and critical care of septic patients have improved outcomes, yet grappling with the intricate immune dysfunction underlying the septic condition remains a formidable challenge. Anti-inflammatory interventions that exhibited promise in murine models failed to manifest consistent survival benefits in clinical studies through recent decades. Novel therapeutic approaches that target bacterial virulence factors, for example with monoclonal antibodies, aim to thwart pathogen-driven damage and restore an advantage to the immune system. A pioneering technology addressing this challenge is biomimetic nanoparticles-a therapeutic platform featuring nanoscale particles enveloped in natural cell membranes. Borne from the quest for a durable drug delivery system, the original red blood cell-coated nanoparticles showcased a broad capacity to absorb bacterial and environmental toxins from serum. Tailoring the membrane coating to immune cell sources imparts unique characteristics to the nanoparticles suitable for broader application in infectious disease. Their capacity to bind both inflammatory signals and virulence factors assembles the most promising sepsis therapies into a singular, pathogen-agnostic therapeutic. This review explores the ongoing work on immune cell-coated nanoparticle therapeutics for infection and sepsis. SIGNIFICANCE STATEMENT: Invasive bacterial infections and sepsis are a major global health problem made worse by expanding antibiotic resistance, meaning better treatment options are urgently needed. Biomimetic cell-membrane-coated nanoparticles are an innovative therapeutic platform that deploys a multifaceted mechanism to action to neutralize microbial virulence factors, capture endotoxins, and bind excessive host proinflammatory cytokines, seeking to reduce host tissue injury, aid in microbial clearance, and improve patient outcomes.


Asunto(s)
Infecciones Bacterianas , Materiales Biomiméticos , Nanomedicina , Sepsis , Humanos , Animales , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Sepsis/microbiología , Nanomedicina/métodos , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/inmunología , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/uso terapéutico , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , Biomimética/métodos , Nanopartículas
16.
J Drug Target ; 32(6): 606-623, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38656224

RESUMEN

Atherosclerosis (AS) is considered to be one of the major causes of cardiovascular disease. Its pathological microenvironment is characterised by increased production of reactive oxygen species, lipid oxides, and excessive inflammatory factors, which accumulate at the monolayer endothelial cells in the vascular wall to form AS plaques. Therefore, intervention in the pathological microenvironment would be beneficial in delaying AS. Researchers have designed biomimetic nanomedicines with excellent biocompatibility and the ability to avoid being cleared by the immune system through different therapeutic strategies to achieve better therapeutic effects for the characteristics of AS. Biomimetic nanomedicines can further enhance delivery efficiency and improve treatment efficacy due to their good biocompatibility and ability to evade clearance by the immune system. Biomimetic nanomedicines based on therapeutic strategies such as neutralising inflammatory factors, ROS scavengers, lipid clearance and integration of diagnosis and treatment are versatile approaches for effective treatment of AS. The review firstly summarises the targeting therapeutic strategy of biomimetic nanomedicine for AS in recent 5 years. Biomimetic nanomedicines using cell membranes, proteins, and extracellular vesicles as carriers have been developed for AS.


Asunto(s)
Aterosclerosis , Biomimética , Sistemas de Liberación de Medicamentos , Nanomedicina , Humanos , Aterosclerosis/tratamiento farmacológico , Nanomedicina/métodos , Biomimética/métodos , Sistemas de Liberación de Medicamentos/métodos , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo
17.
Drug Deliv Transl Res ; 14(6): 1432-1457, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38117405

RESUMEN

Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common clinical critical diseases with high morbidity and mortality. Especially since the COVID-19 outbreak, the mortality rates of critically ill patients with ARDS can be as high as 60%. Therefore, this problem has become a matter of concern to respiratory critical care. To date, the main clinical measures for ALI/ARDS are mechanical ventilation and drug therapy. Although ventilation treatment reduces mortality, it increases the risk of hyperxemia, and drug treatment lacks safe and effective delivery methods. Therefore, novel therapeutic strategies for ALI/ARDS are urgently needed. Developments in nanotechnology have allowed the construction of a safe, efficient, precise, and controllable drug delivery system. However, problems still encounter in the treatment of ALI/ARDS, such as the toxicity, poor targeting ability, and immunogenicity of nanomaterials. Cell-derived biomimetic nanodelivery drug systems have the advantages of low toxicity, long circulation, high targeting, and high bioavailability and show great therapeutic promises for ALI/ARDS owing to their acquired cellular biological features and some functions. This paper reviews ALI/ARDS treatments based on cell membrane biomimetic technology and extracellular vesicle biomimetic technology, aiming to achieve a significant breakthrough in ALI/ARDS treatments.


Asunto(s)
Lesión Pulmonar Aguda , Nanopartículas , Síndrome de Dificultad Respiratoria , Humanos , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Nanopartículas/administración & dosificación , Lesión Pulmonar Aguda/tratamiento farmacológico , Materiales Biomiméticos/química , Materiales Biomiméticos/administración & dosificación , Sistemas de Liberación de Medicamentos , COVID-19 , Biomimética , Tratamiento Farmacológico de COVID-19 , Animales
18.
Nat Commun ; 12(1): 7264, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34907171

RESUMEN

Antibodies targeting costimulatory receptors of T cells have been developed for the activation of T cell immunity in cancer immunotherapy. However, costimulatory molecule expression is often lacking in tumor-infiltrating immune cells, which can impede antibody-mediated immunotherapy. Here, we hypothesize that delivery of costimulatory receptor mRNA to tumor-infiltrating T cells will enhance the antitumor effects of antibodies. We first design a library of biomimetic nanoparticles and find that phospholipid nanoparticles (PL1) effectively deliver costimulatory receptor mRNA (CD137 or OX40) to T cells. Then, we demonstrate that the combination of PL1-OX40 mRNA and anti-OX40 antibody exhibits significantly improved antitumor activity compared to anti-OX40 antibody alone in multiple tumor models. This treatment regimen results in a 60% complete response rate in the A20 tumor model, with these mice being resistant to rechallenge by A20 tumor cells. Additionally, the combination of PL1-OX40 mRNA and anti-OX40 antibody significantly boosts the antitumor immune response to anti-PD-1 + anti-CTLA-4 antibodies in the B16F10 tumor model. This study supports the concept of delivering mRNA encoding costimulatory receptors in combination with the corresponding agonistic antibody as a strategy to enhance cancer immunotherapy.


Asunto(s)
Materiales Biomiméticos/administración & dosificación , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Nanopartículas/administración & dosificación , ARN Mensajero/administración & dosificación , Linfocitos T/inmunología , Animales , Materiales Biomiméticos/química , Sistemas de Liberación de Medicamentos , Glucolípidos/administración & dosificación , Glucolípidos/química , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Nanopartículas/química , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Fosfolípidos/administración & dosificación , Fosfolípidos/química , ARN Mensajero/química , Receptores OX40/antagonistas & inhibidores , Receptores OX40/genética , Receptores OX40/inmunología , Receptores OX40/metabolismo , Linfocitos T/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
19.
J Nanobiotechnology ; 19(1): 457, 2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-34963466

RESUMEN

High doses of radiation can cause serious side effects and efficient radiosensitizers are urgently needed. To overcome this problem, we developed a biomimetic nanozyme system (CF) by coating pyrite (FeS2) into tumor-derived exosomes for enhanced low-dose radiotherapy (RT). CF system give FeS2 with immune escape and homologous targeting abilities. After administration, CF with both glutathione oxidase (GSH-OXD) and peroxidase (POD) activities can significantly lower the content of GSH in tumor tissues and catalyze intracellular hydrogen peroxide (H2O2) to produce a large amount of ·OH for intracellular redox homeostasis disruption and mitochondria destruction, thus reducing RT resistance. Experiments in vivo and in vitro showed that combining CF with RT (2 Gy) can provide a substantial suppression of tumor proliferation. This is the first attempt to use exosomes bionic FeS2 nanozyme for realizing low-dose RT, which broaden the prospects of nanozymes.


Asunto(s)
Materiales Biomiméticos/administración & dosificación , Enzimas/administración & dosificación , Nanoestructuras/administración & dosificación , Neoplasias/radioterapia , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Materiales Biomiméticos/farmacología , Línea Celular Tumoral , Enzimas/química , Enzimas/metabolismo , Exosomas/química , Exosomas/inmunología , Glutatión/metabolismo , Peróxido de Hidrógeno/metabolismo , Evasión Inmune , Hierro/administración & dosificación , Hierro/química , Ratones , Mitocondrias/efectos de los fármacos , Nanoestructuras/química , Neoplasias/metabolismo , Oxidación-Reducción/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Dosificación Radioterapéutica , Sulfuros/administración & dosificación , Sulfuros/química
20.
ACS Appl Mater Interfaces ; 13(48): 56988-56999, 2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34806359

RESUMEN

Vascular embolization provides an effective approach for the treatment of hemorrhage, aneurysms, and other vascular abnormalities. However, current embolic materials, such as metallic coils and liquid embolic agents, are limited by their inability to provide safe, consistent, and controlled embolization. Here, we report an injectable hydrogel that can remain at the injection site and subsequently undergo in situ covalent crosslinking, leading to the formation of a dual-crosslinking network (DCN) hydrogel for endovascular embolization. The DCN hydrogel is simple to prepare, easy to deploy via needles and catheters, and mechanically stable at the target injection site, thereby avoiding embolization of nontarget vessels. It possesses efficient hemostatic activity and good biocompatibility. The DCN hydrogel is also clearly visible under X-ray imaging, thereby allowing for targeted embolization. In vivo tests in a rabbit artery model demonstrates that the DCN hydrogel is effective in achieving immediate embolization of the target artery with long-term occlusion by inducing luminal fibrosis. Collectively, the DCN hydrogel provides a viable, biocompatible, and cost-effective alternative to existing embolic materials with clinical translation potential for endovascular embolization.


Asunto(s)
Arterias/efectos de los fármacos , Materiales Biomiméticos/farmacología , Reactivos de Enlaces Cruzados/farmacología , Embolización Terapéutica , Fibrosis/tratamiento farmacológico , Hidrogeles/farmacología , Animales , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/química , Células Cultivadas , Reactivos de Enlaces Cruzados/administración & dosificación , Reactivos de Enlaces Cruzados/química , Humanos , Hidrogeles/administración & dosificación , Hidrogeles/química , Ensayo de Materiales , Ratones , Estructura Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA