Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Small ; : e2401397, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898735

RESUMO

Pyroptosis, an inflammatory cell death, plays a pivotal role in activating inflammatory response, reversing immunosuppression and enhancing anti-tumor immunity. However, challenges remain regarding how to induce pyroptosis efficiently and precisely in tumor cells to amplify anti-tumor immunotherapy. Herein, a pH-responsive polydopamine (PDA) nanocluster, perfluorocarbon (PFC)@octo-arginine (R8)-1-Hexadecylamine (He)-porphyrin (Por)@PDA-gambogic acid (GA)-cRGD (R-P@PDA-GC), is rationally design to augment phototherapy-induced pyroptosis and boost anti-tumor immunity through a two-input programmed cascade therapy. Briefly, oxygen doner PFC is encapsulated within R8 linked photosensitizer Por and He micelles as the core, followed by incorporation of GA and cRGD peptides modified PDA shell, yielding the ultimate R-P@PDA-GC nanoplatforms (NPs). The pH-responsive NPs effectively alleviate hypoxia by delivering oxygen via PFC and mitigate heat resistance in tumor cells through GA. Upon two-input programmed irradiation, R-P@PDA-GC NPs significantly enhance reactive oxygen species production within tumor cells, triggering pyroptosis via the Caspase-1/GSDMD pathway and releasing numerous inflammatory factors into the TME. This leads to the maturation of dendritic cells, robust infiltration of cytotoxic CD8+ T and NK cells, and diminution of immune suppressor Treg cells, thereby amplifying anti-tumor immunity.

2.
ACS Nano ; 18(20): 12870-12884, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38727063

RESUMO

Epirubicin (EPI) alone can trigger mildly protective autophagy in residual tumor cells, resulting in an immunosuppressive microenvironment. This accelerates the recurrence of residual tumors and leads to antiprogrammed death ligand 1 (anti-PD-1)/PD-L1 therapy resistance, posing a significant clinical challenge in tumor immunotherapy. The combination of checkpoint inhibitors targeting the PD-1/PD-L1 pathway and amplifying autophagy presents an innovative approach to tumor treatment, which can prevent tumor immune escape and enhance therapeutic recognition. Herein, we aimed to synthesize a redox-triggered autophagy-induced nanoplatform with SA&EA-induced PD-L1 inhibition. The hyaluronic acid (HA) skeleton and arginine segment promoted active nanoplatform targeting, cell uptake, and penetration. The PLGLAG peptide was cleaved by overexpressing matrix metalloproteinase-2 (MMP-2) in the tumor microenvironment, and the PD-L1 inhibitor D-PPA was released to inhibit tumor immune escape. The intense autophagy inducers, STF-62247 and EPI, were released owing to the cleavage of disulfide bonds influenced by the high glutathione (GSH) concentration in tumor cells. The combination of EPI and STF induced apoptosis and autophagic cell death, effectively eliminating a majority of tumor cells. This indicated that the SA&EA nanoplatform has better therapeutic efficacy than the single STF@AHMPP and EPI@AHMPTP groups. This research provided a way to set up a redox-triggered autophagy-induced nanoplatform with PD-L1 inhibition to enhance chemo-immunotherapy.


Assuntos
Autofagia , Antígeno B7-H1 , Imunoterapia , Nanopartículas , Oxirredução , Autofagia/efeitos dos fármacos , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Animais , Humanos , Camundongos , Nanopartículas/química , Microambiente Tumoral/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais
3.
Adv Sci (Weinh) ; : e2401214, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38647420

RESUMO

Deep penetration and downregulation of heat shock protein (HSP) expression in multimodal synergistic therapy are promising approaches for curing cancer in clinical trials. However, free small-molecule drugs and most drug vehicles have a low delivery efficiency deep into the tumor owing to poor drug penetration and hypoxic conditions at the tumor site. In this study, the objective is to use reactive oxygen species (ROS)-responsive supramolecular gels co-loaded with the photosensitizer Zn(II) phthalocyanine tetrasulfonic acid (ZnPCS4) and functionalized tetrahedral DNA (TGSAs) (G@P/TGSAs) to enhance deep tissue and cell penetration and block the HSP90 pathway for chemo- photodynamic therapy (PDT) - photothermal therapy (PTT) trimodal synergistic therapy. The (G@P/TGSAs) are injected in situ into the tumor to release ZnPCS4 and TGSAs under high ROS concentrations originating from both the tumor and PDT. TGSAs penetrate deeply into tumor tissues and augment photothermal therapy by inhibiting the HSP90 pathway. Proteomics show that HSP-related proteins and molecular chaperones are inhibited/activated, inhibiting the HSP90 pathway. Simultaneously, the TGSA-regulated apoptotic pathway is activated. In vivo study demonstrates efficient tumor penetration and excellent trimodal synergistic therapy (45% tumor growth inhibition).

4.
Colloids Surf B Biointerfaces ; 237: 113871, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38547796

RESUMO

Highly toxic reactive oxygen species (ROS), crucial in inducing apoptosis and ferroptosis, are pivotal for cell death pathways in cancer therapy. However, the effectiveness of ROS-related tumor therapy is impeded by the limited intracellular ROS and substrates, coupled with the presence of abundant ROS scavengers like glutathione (GSH). In this research, we developed acid-responsive, iron-coordinated polymer nanoparticles (PPA/TF) encapsulating a mitochondrial-targeting drug alpha-tocopheryl succinate (α-TOS) for enhanced synergistic tumor treatment. The imidazole grafted micelles exhibit prolonged blood circulation and improve the delivery efficiency of the hydrophobic drug α-TOS. Additionally, PPA's design aids in delivering Fe3+, supplying ample iron ions for chemodynamic therapy (CDT) and ferroptosis through the attachment of imidazole groups to Fe3+. In the tumor's weakly acidic intracellular environment, PPA/TF facilitates pH-responsive drug release. α-TOS specifically targets mitochondria, generating ROS and replenishing those depleted by the Fenton reaction. Moreover, the presence of Fe3+ in PPA/TF amplifies ROS upregulation, promotes GSH depletion, and induces oxidative damage and ferroptosis, effectively inhibiting tumor growth. This research presents an innovative ROS-triggered amplification platform that optimizes CDT and ferroptosis for effective cancer treatment.


Assuntos
Ferroptose , Neoplasias , Humanos , Espécies Reativas de Oxigênio/metabolismo , Micelas , Linhagem Celular Tumoral , Polímeros/química , Neoplasias/patologia , Ferro/química , Concentração de Íons de Hidrogênio , Imidazóis/farmacologia , Peróxido de Hidrogênio/química , Glutationa/metabolismo , Microambiente Tumoral
5.
J Am Chem Soc ; 146(1): 1185-1195, 2024 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-38148611

RESUMO

Patients treated with Pt-based anticancer drugs (PtII) often experience severe side effects and are susceptible to cancer recurrence due to the limited bioavailability of PtII and tumor-induced immunosuppression. The exposure of phosphatidylserine on the cell's outer surface induced by PtII results in profound immunosuppression through the binding of phosphatidylserine to its receptors on immune cells. Here, we report a novel approach for enhanced cancer chemoimmunotherapy, where a novel nuclear-targeting lipid PtIV prodrug amphiphile was used to deliver a small interfering RNA (siXkr8) to simultaneously amplify Pt-DNA adducts and reduce the level of exposure of phosphatidylserine. This drug delivery vehicle is engineered by integrating the PtIV prodrug with self-assembly performance and siXkr8 into a lipid nanoparticle, which shows tumor accumulation, cancer cell nucleus targeting, and activatable in a reduced microenvironment. It is demonstrated that nuclear-targeting lipid PtIV prodrug increases the DNA cross-linking, resulting in increased Pt-DNA adduct formation. The synergistic effects of the PtIV prodrug and siXkr8 contribute to the improvement of the tumor immune microenvironment. Consequently, the increased Pt-DNA adducts and immunogenicity effectively inhibit primary tumor growth and prevent tumor recurrence. These results underscore the potential of utilizing the nuclear-targeting lipid PtIV prodrug amphiphile to enhance Pt-DNA adduct formation and employing siXkr8 to alleviate immunosuppression during chemotherapy.


Assuntos
Antineoplásicos , Neoplasias , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Adutos de DNA , Fosfatidilserinas , RNA Interferente Pequeno , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , RNA de Cadeia Dupla , Linhagem Celular Tumoral , Cisplatino , Microambiente Tumoral
6.
J Mater Chem B ; 12(2): 413-435, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38112639

RESUMO

Ferroptosis is a new form of non-apoptotic programmed cell death. Due to its effectiveness in cancer treatment, there are increasing studies on the application of nanoparticles based on ferroptosis in cancer therapy. In this paper, we present a summary of the latest progress in nanoparticles based on ferroptosis for effective tumor therapy. We also describe the combined treatment of ferroptosis with other therapies, including chemotherapy, radiotherapy, phototherapy, immunotherapy, and gene therapy. This summary of drug delivery systems based on ferroptosis aims to provide a basis and inspire opinions for researchers concentrating on exploring this field. Finally, we present some prospects and challenges for the application of nanotherapies to clinical treatment by promoting ferroptosis in cancer cells.


Assuntos
Ferroptose , Nanopartículas , Neoplasias , Terapia Combinada , Imunoterapia , Fototerapia , Neoplasias/tratamento farmacológico
7.
ACS Appl Mater Interfaces ; 15(29): 34436-34450, 2023 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-37415554

RESUMO

Tumors have become the biggest obstacle to human health, and there are various treatment methods at present. Photothermal therapy (PTT) is usually ineffective and does not inhibit tumor progression due to the inability of the lasers to penetrate deeply. Therefore, most existing studies chose a 1064 nm laser with stronger penetrating power; meanwhile, studies have shown that the inclusion of harmful free radicals can significantly improve the antitumor efficacy. Herein, TiO nanosheets (NSs) were creatively prepared and encapsulated with an alkyl radical generator {2,2'-azobis[2-(2-imidazoline-2-yl)propane] dihydrochloride, [AIPH]} in sodium alginate (ALG) hydrogel for effective tumor killing by PTT and pairing with dangerous free radicals. TiO NSs were obtained by the liquid-phase exfoliation method, together with AIPH, which were in situ coencapsulated multifunctional hydrogels formed by the combination of Ca2+ and ALG. This ALG hydrogel could enrich TiO NSs and AIPH at the tumor site for a long time, and through the excellent photothermal properties of TiO NSs, AIPH could slowly and effectively generate alkyl radicals at the tumor site, which, in turn, gave it a better antitumor effect compared with that of TiO NSs in the deep hypoxic environment of the tumor. The AIPH + TiO + ALG hydrogel has distinctive anticancer capabilities based on the results of both in vivo and in vitro experiments. This material also has good biosafety. By combining PTT and free radical treatment, this work provides a novel therapeutic method to achieve oxygen-independent free radical production and enhance therapeutic efficacy.


Assuntos
Hidrogéis , Neoplasias , Humanos , Hidrogéis/química , Terapia Fototérmica , Fototerapia , Neoplasias/tratamento farmacológico , Termodinâmica , Radicais Livres/uso terapêutico , Linhagem Celular Tumoral
8.
Int J Pharm ; 641: 122987, 2023 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-37207860

RESUMO

Diabetic retinopathy (DR) is one of the serious complications of diabetes, which has become the fourth leading cause of vision loss worldwide. Current treatment of DR relies on intravitreal injections of antiangiogenic agents, which has made considerable achievements in reducing visual impairment. However, long-term invasive injections require advanced technology and can lead to poor patient compliance as well as the incidence of ocular complications including bleeding, endophthalmitis, retinal detachment and others. Hence, we developed non-invasive liposomes (EA-Hb/TAT&isoDGR-Lipo) for efficiency co-delivery of ellagic acid and oxygen, which can be administered intravenously or by eye drops. Among that, ellagic acid (EA), as an aldose reductase inhibitor, could remove excessive reactive oxygen species (ROS) induced by high glucose for preventing retinal cell apoptosis, as well as reduce retinal angiogenesis through the blockage of VEGFR2 signaling pathway; carried oxygen could ameliorate DR hypoxia, and further enhanced the anti-neovascularization efficacy. Our results showed that EA-Hb/TAT&isoDGR-Lipo not only effectively protected retinal cells from high glucose-induced damage, but also inhibited VEGF-induced vascular endothelial cells migration, invasion, and tube formation in vitro. In addition, in a hypoxic cell model, EA-Hb/TAT&isoDGR-Lipo could reverse retinal cell hypoxia, thereby reducing the expression of VEGF. Significantly, after being administered as an injection or eye drops, EA-Hb/TAT&isoDGR-Lipo obviously ameliorated the structure (central retinal thickness and retinal vascular network) of retina by eliminating ROS and down-regulating the expression of GFAP, HIF-1α, VEGF and p-VEGFR2 in a DR mouse model. In summary, EA-Hb/TAT&isoDGR-Lipo holds great potentials in improvement of DR, which provides a novel approach for the treatment of DR.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Neovascularização Retiniana , Camundongos , Animais , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/metabolismo , Retinopatia Diabética/prevenção & controle , Neovascularização Retiniana/tratamento farmacológico , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/prevenção & controle , Lipossomos/farmacologia , Ácido Elágico/metabolismo , Ácido Elágico/farmacologia , Ácido Elágico/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células Endoteliais/metabolismo , Retina/metabolismo , Hipóxia , Glucose/farmacologia , Soluções Oftálmicas/farmacologia
9.
J Colloid Interface Sci ; 645: 986-996, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37179196

RESUMO

Autophagy inducers increase the sensitivity of tumor cells to chemotherapeutic drugs and enhance anti-tumor efficacy. An autophagy-induced intracellular signaling fractional nano-drug system was constructed for the co-delivery of the autophagy inducer rapamycin (RAPA) and the anti-tumor drug 9-nitro-20(S)-camptothecin (9-NC). Link peptides, including cathepsin B-sensitive peptides (Ala-Leu-Ala-Leu, ALAL), nucleus-targeting peptides (TAT, sequence: YGRKKRRQRRR), and chrysin (CHR)-modified hydrophobic biodegradable polymers (poly(-caprolactone)) (PCL), were grafted onto hyaluronic acid (HA) to yield two amphiphiles, HA-ALAL-PCL-CHR (CPAH) and HA-ALAL-TAT-PCL-CHR (CPTAH). Spherical RAPA- and 9-NC-loaded micelles were obtained by the self-assembly of amphiphiles comprising CPAH and RAPA and CPTAH and 9-NC. In this fractional nano-drug system, RAPA was released earlier than 9-NC, as CPAH as a RAPA carrier lacked a nucleus-targeting TAT (unlike CPTAH as an 9-NC carrier). RAPA induced autophagy in tumor cells and improved their sensitivity, whereas the secondary nucleus-targeting micelles directly delivered 9-NC to the nucleus, considerably improving anti-tumor efficacy. Immunofluorescence staining, acridine orange (AO) staining, and western blotting results demonstrated that the system induced a high level of autophagy in combination chemotherapy. The proposed system possesses a high level of cytotoxicity in vitro and in vivo and provides a potential method for enhancing anti-tumor efficacy in clinical settings.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Micelas , Antineoplásicos/química , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Portadores de Fármacos/química , Peptídeos/farmacologia , Nanopartículas/uso terapêutico , Autofagia , Linhagem Celular Tumoral
10.
Nanoscale Adv ; 5(5): 1271-1290, 2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36866253

RESUMO

Ferroptosis, an iron-dependent mode of regulated cell death, is induced by lipid peroxidation, whose occurrence and execution are primarily controlled by metabolism of iron, lipids, amino acids and glutathione. In recent years, the fast-growing studies of ferroptosis in cancer have promoted its application in cancer therapy. So, this review focuses on the feasibility and characteristics of initiating ferroptosis for cancer therapy, as well as the main mechanism of ferroptosis. And various emerging strategies of cancer therapy based on ferroptosis are then highlighted to describe their design, mechanism of action, and anticancer applications. In addition ferroptosis in diverse cancer types is summarized, some considerations for the research of various preparations that can cause ferroptosis are introduced, and this emerging field is discussed in terms of its challenges and future development directions.

11.
Small ; 19(24): e2207898, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36932938

RESUMO

Chemotherapeutics can induce immunogenic cell death (ICD) by triggering autophagy and mediate antitumor immunotherapy. However, using chemotherapeutics alone can only cause mild cell-protective autophagy and be incapable of inducing sufficient ICD efficacy. The participation of autophagy inducer is competent to enhance autophagy, so the level of ICD is promoted and the effect of antitumor immunotherapy is highly increased. Herein, tailor-made autophagy cascade amplification polymeric nanoparticles STF@AHPPE are constructed to enhance tumor immunotherapy. Arginine (Arg), polyethyleneglycol-polycaprolactone, and epirubicin (EPI) are grafted onto hyaluronic acid (HA) via disulfide bond to form the AHPPE nanoparticles and autophagy inducer STF-62247 (STF) is loaded. When STF@AHPPE nanoparticles target to tumor tissues and efficiently enter into tumor cells with the help of HA and Arg, the high glutathione concentration leads to the cleavage of disulfide bond and the release of EPI and STF. Finally, STF@AHPPE induces violent cytotoxic autophagy and strong ICD efficacy. As compared to AHPPE nanoparticles, STF@AHPPE nanoparticles kill the most tumor cells and show the more obvious ICD efficacy and immune activation ability. This work provides a novel strategy for combining tumor chemo-immunotherapy with autophagy induction.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Humanos , Antineoplásicos/farmacologia , Autofagia , Imunoterapia , Dissulfetos/farmacologia , Linhagem Celular Tumoral , Microambiente Tumoral
12.
J Mater Chem B ; 11(2): 415-429, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36512437

RESUMO

Ferroptosis, a type of programmed cell death induced by the iron-dependent lipid hydroperoxide pathway, has attracted widespread attention. However, Fenton response-dependent ferroptosis has many limitations, such as insufficient reaction conditions in the tumor micro-environment. Here, we propose an all-in-one phototherapy nanoplatform consisting of iron-polydopamine (Fe-PDA), a folic acid-modified red blood cell membrane (FA-RBCm), and epirubicin (EPI), namely, Fe-PDA-EPI@FA-RBCm NPs, to achieve enhanced photothermal-ferroptosis effects via overcoming the limitations of the Fenton-like reaction. The results showed that the synthesized biomimetic nanoparticles could decompose hydrogen peroxide (H2O2) to generate hydroxyl radicals (˙OH), and further induce the non-apoptotic ferroptosis pathway. After irradiation with near-infrared (NIR) light, the uptake of Fe-PDA-EPI@FA-RBCm NPs by cells could be effectively promoted, and it presented impressive in vitro and in vivo photothermal properties. In vitro and in vivo results showed that laser irradiation could enhance ferroptosis by promoting the production of reactive oxygen species (ROS) and lipid peroxides, down-regulating the expression of glutathione peroxidase 4 (GPX4), and reducing the mitochondrial membrane potential. Furthermore, the photothermal-promoted ferroptosis and apoptosis pathways (photothermal therapy and chemotherapy) exhibited outstanding synergistic antitumor efficacy in vitro and in vivo, with an in vivo tumor inhibition rate as high as 76.95%. In conclusion, the construction of tumor-targeted biomimetic nanocarriers utilizing the advantageous properties of RBCm has been investigated as a potential anticancer strategy.


Assuntos
Ferroptose , Nanopartículas , Neoplasias , Peróxido de Hidrogênio/farmacologia , Nanopartículas/uso terapêutico , Apoptose , Epirubicina/farmacologia , Ferro/farmacologia
13.
Biomacromolecules ; 23(12): 5202-5212, 2022 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-36287618

RESUMO

Poor antitumor drug penetration into tumor tissues is a global challenge in clinical cancer treatment. Here, we reported a smart multistage "Trojan Horse"-inspired bovine serum albumin (BSA)-coated liposome (HBM), including the mimics of capsid and secondary BSA-coated polymeric nanoparticles (NPs) for enhancing tumor penetration and antitumor efficacy. These drug-loaded polymeric NPs possess a capsid-like component, a well-distributed nanostructure (size: 190.1 ± 4.98 nm, PDI: 0.259), and an excellent drug loading content (15.85 ± 1.36%). Meaningfully, after the smart multistage BSA-coated liposome targeted the tumor tissue, the mimics of capsid were "taken off" under the condition of tumor-specific enzymes, releasing "Heart" BSA-modified secondary NPs to increase the ability to penetrate tumor cells for enhancing antitumor efficacy. As expected, the HBM efficiently achieves high drug penetration into PAN02 tumor cells. Moreover, compared to free DOX and HM (HBM without BSA) NPs, DOX/HBM NPs exhibited the strongest tumor penetration and the highest cytotoxicity against PAN02 tumor cells both in vitro (IC50 = 0.141 µg/mL) and in vivo. This smart multistage "Trojan Horse"-inspired BSA-coated liposome should provide a new hathpace for further development of polymeric NPs in clinical treatment.


Assuntos
Nanopartículas , Neoplasias , Humanos , Soroalbumina Bovina , Lipossomos/uso terapêutico , Portadores de Fármacos/uso terapêutico , Neoplasias/tratamento farmacológico , Linhagem Celular Tumoral
14.
Mol Pharm ; 19(9): 3439-3449, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35994700

RESUMO

The combined delivery of chemotherapeutics with checkpoint inhibitors of the PD-1/PD-L1 pathway provides a new approach for cancer treatment. Small-molecule peptide inhibitors possess short production cycle, low immunogenicity, and fewer side effects; however, their potential in cancer therapy is hampered by the rapid biodegradation and a nanocarrier is needed for efficient drug delivery. Herein, anticancer drug doxorubicin (DOX) and PD-L1 inhibitor peptide P-12 are co-loaded by a lipid polymer nanocomplex based on poly(lactic-co-glycolic acid) (PLGA) and DSPE-PEG. Octaarginine (R8)-conjugated DSPE-PEG renders the LPN efficient internalization by cancer cells. The optimal nanomedicine LPN-30-R82K@DP shows a diameter of 125 nm and a DOX and P-12 loading content of 5.0 and 6.2%, respectively. LPN-30-R82K@DP exhibits good physiological stability and enhanced cellular uptake by cancer cells. It successfully induces immunogenic cell death and PD-L1 blockade in CT26 cancer cells. The in vivo antitumor study further suggests that co-loaded nanomedicine efficiently suppresses CT26 tumor growth and elicits antitumor immune response. This study manifests that lipid polymer nanocomplexes are promising drug carriers for the efficient chemo-immunotherapy of cancer.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Doxorrubicina/química , Imunoterapia , Lipídeos/química , Nanopartículas/química , Neoplasias/tratamento farmacológico , Polímeros/química
15.
Regen Biomater ; 9: rbac032, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35668924

RESUMO

Combination therapy is extensively developed for cancer treatment in recent years due to its high efficiency. Herein, we constructed a nanocomposite based on gold nanorods (GNRs) and drug-loaded tetrahedral DNA nanostructures (TDN) for chemo-photothermal combinational therapy. Anti-tumor drug doxorubicin (DOX) was loaded via the insertion within GC base pairs of TDN. The aptamer AS1411 was attached to the apex of TDN (ATDN) to target tumor cells. The DOX-loaded DNA tetrahedron (ATDN-DOX) was compressed by the GNRs coated with PEI (GNRs@ATDN-DOX) to realize the photothermal function and lysosome escape. GNRs under the illumination of 808 nm infrared laser showed high photothermal conversion and stability due to the protection of PEI layer. The drug-loading capacity of ATDN-DOX was as high as 314 DOX molecules in per ATDN. The positive charge of PEI in GNRs@ATDN-DOX nanocomposites was utilized to achieve excellent cell penetration and induce proton sponge effect for lysosomal escape. The nanocomposites presented HeLa and 4T1 cells targeting and resulted in efficient anticancer activity.

16.
Mol Neurobiol ; 59(4): 2246-2257, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35066763

RESUMO

Accumulating evidence indicates that the continuous and intense nociceptive from inflamed tissue may increase the excitability of spinal dorsal horn neurons, which can signal back and modulate peripheral inflammation. Previous studies have demonstrated that spinal interleukin (IL)-33 contributes to the hyperexcitability of spinal dorsal horn neurons. This study was undertaken to investigate whether spinal IL-33 can also influence a peripheral inflammatory response in a rat model of arthritis. Lentivirus-delivered short hairpin RNA targeting IL-33 (LV-shIL-33) was constructed for gene silencing. Rats with adjuvant-induced arthritis (AIA) were injected intrathecally with LV-shIL-33 3 days before the complete Freund's adjuvant (CFA) injection. During an observation period of 21 days, pain-related behavior and inflammation were assessed. In addition, the expression of spinal proinflammatory cytokines and the activation of spinal extracellular signal-regulated kinase (ERK) and nuclear factor-κB (NF-κB) pathways were evaluated on 9 days after CFA treatment. The existence of tissue injury or inflammation in rats with AIA resulted in the upregulation of spinal IL-33, which is predominantly expressed in neurons, astrocytes, and oligodendrocytes. Intrathecal administration of LV-shIL-33 significantly alleviated hyperalgesia, paw swelling, and joint destruction, and attenuated the expression of proinflammatory cytokines [IL-6, IL-1ß, and tumor necrosis factor-α (TNF-α)], as well as the activation of ERK and NF-κB/p65 in the spinal cord. Our data suggest that spinal IL-33 contributes to the development of both peripheral inflammation and hyperalgesia. Thus, interference with IL-33 at the spinal level might represent a novel therapeutic target for painful inflammatory disorders.


Assuntos
Artrite , Hiperalgesia , Animais , Artrite/patologia , Citocinas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Adjuvante de Freund/efeitos adversos , Adjuvante de Freund/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Inflamação/metabolismo , Interleucina-33/metabolismo , Interleucina-33/farmacologia , NF-kappa B/metabolismo , Ratos , Medula Espinal/patologia
17.
J Mater Chem B ; 10(3): 438-449, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-34951442

RESUMO

A peptide/DNA nanocomplex was developed for the targeted delivery of chemotherapeutics and photosensitizers to cancer cells for efficient combination therapy. The chemotherapeutic drug doxorubicin (DOX) and the photosensitizer 5,10,15,20-tetra-(1-methylpyridine-4-yl)-porphyrin (TMPyP4) were physically incorporated by an aptamer (AS1411)-modified tetrahedral DNA nanostructure, where the tetrahedral DNA and aptamer-induced G-quadruplex provide binding sites of DOX and TMPyP4. The co-loaded 3A-TDN/DT displayed a targeted uptake by HeLa cancer cells through the high affinity and specificity between AS1411 and nucleolin, a protein overexpressed on many types of cancer cells. A polycationic polymer, mPEG-PAsp(TECH), was synthesized to complex with the DNA nanostructure to efficiently escape from lysosomes via the proton sponge effect upon the enhanced internalization by tumor cells. Under the irradiation of 660 nm laser light, TMPyP4 induced an upregulation of intracellular reactive oxygen species, which combined with DOX to fulfill the efficient inhibition of HeLa cells. Our study demonstrated a biocompatible peptide/DNA composite nanoplatform for combinational cancer therapy via the targeted delivery of therapeutic agents and efficient lysosomal escape.


Assuntos
Antineoplásicos/farmacologia , DNA/química , Portadores de Fármacos/química , Nanoestruturas/química , Peptídeos/química , Fármacos Fotossensibilizantes/farmacologia , Células 3T3 , Animais , Antineoplásicos/química , Aptâmeros de Nucleotídeos/química , Doxorrubicina/química , Doxorrubicina/farmacologia , Tratamento Farmacológico , Células HeLa , Humanos , Luz , Lisossomos/metabolismo , Camundongos , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/efeitos da radiação , Porfirinas/química , Porfirinas/farmacologia , Porfirinas/efeitos da radiação , Oxigênio Singlete/metabolismo
18.
J Nanobiotechnology ; 19(1): 412, 2021 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-34876145

RESUMO

Recently, DNA nanostructures with vast application potential in the field of biomedicine, especially in drug delivery. Among these, tetrahedral DNA nanostructures (TDN) have attracted interest worldwide due to their high stability, excellent biocompatibility, and simplicity of modification. TDN could be synthesized easily and reproducibly to serve as carriers for, chemotherapeutic drugs, nucleic acid drugs and imaging probes. Therefore, their applications include, but are not restricted to, drug delivery, molecular diagnostics, and biological imaging. In this review, we summarize the methods of functional modification and application of TDN in cancer treatment. Also, we discuss the pressing questions that should be targeted to increase the applicability of TDN in the future.


Assuntos
DNA , Sistemas de Liberação de Medicamentos , Nanoestruturas , Neoplasias/tratamento farmacológico , Animais , DNA/química , DNA/uso terapêutico , Humanos , Camundongos , Nanoestruturas/química , Nanoestruturas/uso terapêutico
19.
Anesth Analg ; 133(5): 1321-1330, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34524124

RESUMO

BACKGROUND: The maternal pain threshold gradually increases during pregnancy, especially in late pregnancy. A series of mechanisms underlying pregnancy-induced analgesia have been reported. However, these mechanisms are still not completely clear, and the underlying molecular mechanisms need further investigation. We examined the relationship between the antinociceptive effect and the expression level of programmed cell death ligand-1 (PD-L1) during pregnancy and further observed the changes in pain thresholds and expression levels of cytokines in late-pregnant mice before and after blockade of PD-L1 or programmed cell death-1 (PD-1). METHODS: Part 1: Female mice were assigned to 3 groups (nonpregnant, late-pregnant, and postpartum). Part 2: Late-pregnant mice were assigned to 3 treatment groups (control [phosphate buffer solution], RMP1-14 [mouse anti-PD-1 antibody], and soluble PD-1 [sPD-1]). Behavioral testing (mechanical and thermal) and tissue (serum and spinal cord) analysis were performed on all groups. PD-L1, interleukin (IL)-10, tumor necrosis factor-α (TNF-α), and IL-6 expression levels in tissue were examined via reverse transcription-polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and Western blot analysis. RESULTS: The mechanical and thermal pain thresholds were significantly increased in late pregnancy and decreased after delivery. PD-L1 expression was also elevated in late pregnancy and decreased after delivery. In addition, in the late stage of gestation, the maternal inflammatory microenvironment was dominated by anti-inflammatory factors. After administration of RMP1-14 or sPD-1, the pain thresholds of late-pregnant mice were significantly reduced. In late-pregnant mice, the high level of IL-10 was obviously reduced, and the low levels of TNF-α and IL-6 were elevated. CONCLUSIONS: The PD-L1/PD-1 pathway mediates pregnancy-induced analgesia, partially via the regulation of cytokines.


Assuntos
Antígeno B7-H1/metabolismo , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Limiar da Dor , Dor/prevenção & controle , Receptor de Morte Celular Programada 1/metabolismo , Medula Espinal/metabolismo , Animais , Comportamento Animal , Feminino , Camundongos , Dor/metabolismo , Dor/fisiopatologia , Gravidez , Transdução de Sinais , Medula Espinal/fisiopatologia
20.
Nanoscale ; 13(21): 9577-9589, 2021 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-33998643

RESUMO

Improving the deep penetration of nanoparticles and realizing the combination of chemotherapy and immunotherapy have become a promising strategy for cancer treatment. Herein, a nuclear-targeted tetrahedral DNA nanostructure (NLS-TDNs, NT) was synthesized to construct matrix metalloproteinase (MMP-2) sensitive hydrogels as delivery vehicles with co-loaded disulfide cross-linked polyethyleneimine (PSP)/nuclear-targeted tetrahedral DNA (NLS-TDNs, NT)/doxorubicin (DOX) nanoparticles (NPs) (PSP/NT/DOX NPs and PNT/DOX NPs) and an immune adjuvant imiquimod (R837) to realize a combination of chemotherapy and immunization for metastatic breast cancer. Due to the membrane-breaking ability of the PNT/DOX NPs, the nanoparticles could effectively achieve deep penetration into tumor tissues, and the in situ generation of tumor-associated antigens by PNT/DOX elicited a strong immune response in the presence of R837, achieving a chemo-immune combination therapy of breast cancer, inducing the maturation of dendritic cells (DCs) and secretion of related cytokines, such as interleukin-6 (IL-6), interleukin-12 (IL-12p70) and tumor necrosis factor (TNF-α) in vitro. The combination significantly promoted the proportions of cytotoxic T cells (CD8+ CTL) and cytotoxic T cells/regulatory T cells (CD8+ CTL/Treg) (5.52% and 11.46%, respectively) and the secretion of cytokines, which cooperatively eradicated primary tumor growth (the tumor growth inhibition (TGI) value was 78.3%) and inhibited the tumor from metastasizing effectively in vivo. Our study provided the basis for activating the antitumor immune system to realize chemo-immunotherapy and tumor metastasis therapy.


Assuntos
Neoplasias da Mama , Nanopartículas , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Humanos , Hidrogéis , Metaloproteinases da Matriz
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA