Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
ACS Nano ; 18(12): 8971-8987, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38497600

RESUMO

Immune checkpoint blockade (ICB) therapy has been approved for breast cancer (BC), but clinical response rates are limited. Recent studies have shown that commensal microbes colonize a variety of tumors and are closely related to the host immune system response. Here, we demonstrated that Fusobacterium nucleatum (F.n), which is prevalent in BC, creates an immunosuppressive tumor microenvironment (ITME) characterized by a high-influx of myeloid cells that hinders ICB therapy. Administering the antibiotic metronidazole in BC can deplete F.n and remodel the ITME. To prevent an imbalance in the systemic microbiota caused by antibiotic administration, we designed a biomimetic nanovehicle for on-site antibiotic delivery inspired by F.n homing to BC. Additionally, ferritin-nanocaged doxorubicin was coloaded into this nanovehicle, as immunogenic chemotherapy has shown potential for synergy with ICB. It has been demonstrated that this biomimetic nanovehicle can be precisely homed to BC and efficiently eliminate intratumoral F.n without disrupting the diversity and abundance of systemic microbiota. This ultimately remodels the ITME, improving the therapeutic efficacy of the PD-L1 blocker with a tumor inhibition rate of over 90% and significantly extending the median survival of 4T1 tumor-bearing mice.


Assuntos
Fusobacterium nucleatum , Neoplasias , Animais , Camundongos , Antígeno B7-H1 , Biomimética , Antibacterianos , Imunossupressores , Microambiente Tumoral
2.
Research (Wash D C) ; 7: 0315, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38357697

RESUMO

The ALPK1 (alpha-kinase 1)-TIFA (TRAF-interacting protein with fork head-associated domain)-TRAF6 signaling pathway plays a pivotal role in regulating inflammatory processes, with TIFA and TRAF6 serving as key molecules in this cascade. Despite its significance, the functional mechanism of TIFA-TRAF6 remains incompletely understood. In this study, we unveil that TIFA undergoes liquid-liquid phase separation (LLPS) induced by ALPK1 in response to adenosine diphosphate (ADP)-ß-D-manno-heptose (ADP-Hep) recognition. The phase separation of TIFA is primarily driven by ALPK1, the pT9-FHA domain, and the intrinsically disordered region segment. Simultaneously, TRAF6 exhibits phase separation during ADP-Hep-induced inflammation, a phenomenon observed consistently across various inflammatory signal pathways. Moreover, TRAF6 is recruited within the TIFA condensates, facilitating lysine (K) 63-linked polyubiquitin chain synthesis. The subsequent recruitment, enrichment, and activation of downstream effectors within these condensates contribute to robust inflammatory signal transduction. Utilizing a novel chemical probe (compound 22), our analysis demonstrates that the activation of the ALPK1-TIFA-TRAF6 signaling pathway in response to small molecules necessitates the phase separation of TIFA. In summary, our findings reveal TIFA as a sensor for upstream signals, initiating the LLPS of itself and downstream proteins. This process results in the formation of membraneless condensates within the ALPK1-TIFA-TRAF6 pathway, suggesting potential applications in therapeutic biotechnology development.

3.
Adv Mater ; 36(14): e2307923, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38174840

RESUMO

In vivo optical imaging of trace biomarkers in residual microtumors holds significant promise for cancer prognosis but poses a formidable challenge. Here, a novel hydrogel sensor is designed for ultrasensitive and specific imaging of the elusive biomarker. This hydrogel sensor seamlessly integrates a molecular beacon nanoprobe with fibroblasts, offering both high tissue retention capability and an impressive signal-to-noise ratio for imaging. Signal amplification is accomplished through exonuclease I-mediated biomarker recycling. The resulting hydrogel sensor sensitively detects the biomarker carcinoembryonic antigen with a detection limit of 1.8 pg mL-1 in test tubes. Moreover, it successfully identifies residual cancer nodules with a median diameter of less than 2 mm in mice bearing partially removed primary triple-negative breast carcinomas (4T1). Notably, this hydrogel sensor is proven effective for the sensitive diagnosis of invasive tumors in post-surgical mice with infiltrating 4T1 cells, leveraging the role of fibroblasts in locally enriching tumor cells. Furthermore, the residual microtumor is rapidly photothermal ablation by polydopamine-based nanoprobe under the guidance of visualization, achieving ≈100% suppression of tumor recurrence and lung metastasis. This work offers a promising alternative strategy for visually detecting residual microtumors, potentially enhancing the prognosis of cancer patients following surgical interventions.


Assuntos
Hidrogéis , Neoplasias , Humanos , Camundongos , Animais
4.
Nat Commun ; 14(1): 7722, 2023 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-38001092

RESUMO

Mutations in mitochondrial DNA (mtDNA) play critical roles in many human diseases. In vivo visualization of cells bearing mtDNA mutations is important for resolving the complexity of these diseases, which remains challenging. Here we develop an integrated nano Cas12a sensor (InCasor) and show its utility for efficient imaging of mtDNA mutations in live cells and tumor-bearing mouse models. We co-deliver Cas12a/crRNA, fluorophore-quencher reporters and Mg2+ into mitochondria. This process enables the activation of Cas12a's trans-cleavage by targeting mtDNA, which efficiently cleave reporters to generate fluorescent signals for robustly sensing and reporting single-nucleotide variations (SNVs) in cells. Since engineered crRNA significantly increase Cas12a's sensitivity to mismatches in mtDNA, we can identify tumor tissue and metastases by visualizing cells with mutant mtDNAs in vivo using InCasor. This CRISPR imaging nanoprobe holds potential for applications in mtDNA mutation-related basic research, diagnostics and gene therapies.


Assuntos
Sistemas CRISPR-Cas , Neoplasias , Humanos , Animais , Camundongos , Sistemas CRISPR-Cas/genética , Mutação , DNA Mitocondrial/genética , Mitocôndrias/genética , Neoplasias/genética
5.
ACS Nano ; 17(20): 20120-20134, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37819165

RESUMO

Programmed death-ligand 1 (PD-L1) on tumor-derived small extracellular vesicles (EVs) is a biomarker for prediction of the immunotherapy response. However, conventional bulk measurement can hardly analyze the expression of PD-L1 on individual tumor-derived EVs. Herein, a method for localized imaging of tumor-derived individual EVs PD-L1 (LITIE) is developed. In this assay, EVs in plasma were directly captured on a biochip. Then the liposome-mediated membrane fusion strategy was used to image miR-21 in EVs to discriminate miR-21-positive EVs from the whole EVs populations. Subsequently, the primer exchange reaction (PER) is applied to generate localized and amplified fluorescent signals for imaging PD-L1 on identified tumor-derived EVs. When applied in clinical sample tests, the LITIE assay could effectively distinguish breast cancer patients from healthy donors or patients with benign tumors. Interestingly, in a mice melanoma model, the LITIE assay showed the ability to predict immunotherapy response even before drug treatment. Thus, we think the strategy of measuring individual tumor-derived EVs PD-L1 could serve as an alternative way for screening clinical responders suitable for immunotherapy.


Assuntos
Vesículas Extracelulares , Melanoma , MicroRNAs , Animais , Camundongos , Humanos , Antígeno B7-H1/metabolismo , Imunoterapia/métodos , Melanoma/diagnóstico por imagem , Melanoma/terapia , Melanoma/metabolismo , Vesículas Extracelulares/metabolismo , MicroRNAs/metabolismo
6.
Sci Adv ; 9(39): eadi1965, 2023 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-37756407

RESUMO

Precise killing of tumor cells without affecting surrounding normal cells is a challenge. Mitochondrial DNA (mtDNA) mutations, a common genetic variant in cancer, can directly affect metabolic homeostasis, serving as an ideal regulatory switch for precise tumor therapy. Here, we designed a mutation-induced drug release system (MIDRS), using the single-nucleotide variation (SNV) recognition ability and trans-cleavage activity of Cas12a to convert tumor-specific mtDNA mutations into a regulatory switch for intracellular drug release, realizing precise tumor cell killing. Using Ce6 as a model drug, MIDRS enabled organelle-level photodynamic therapy, triggering innate and adaptive immunity simultaneously. In vivo evaluation showed that MIDRSMT could identify tumor tissue carrying SNVs in mtDNA in unilateral, bilateral, and heterogeneous tumor models, producing an excellent antitumor effect (~82.6%) without affecting normal cells and thus resulting in a stronger systemic antitumor immune response. Additionally, MIDRS was suitable for genotype-specific precision drug release of chemotherapeutic drugs. This strategy holds promise for mutation-specific personalized tumor treatment approaches.


Assuntos
DNA Mitocondrial , Mitocôndrias , Liberação Controlada de Fármacos , Mutação , Mitocôndrias/genética , DNA Mitocondrial/genética , Genótipo
7.
ACS Nano ; 17(17): 16923-16934, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37606317

RESUMO

Multidrug resistance (MDR) is a major cause of chemotherapy failure in oncology, and gene therapy is an excellent measure to reverse MDR. However, conventional gene therapy only modulates the expression of MDR-associated proteins but hardly affects their existing function, thus limiting the efficiency of tumor treatment. Herein, we designed a photoactivated DNA nanodrug (MCD@TMPyP4@DOX) to improve tumor chemosensitivity through the downregulation of MDR-related genes and mitochondria-targeted photodynamic therapy (PDT). The self-assembled DNA nanodrug encodes the mucin 1 (MUC1) aptamer and the cytochrome C (CytC) aptamer to facilitate its selective targeting to the mitochondria in tumor cells; the encoded P-gp DNAzyme can specifically cleave the substrate and silence MDR1 mRNA with the help of Mg2+ cofactors. Under near-infrared (NIR) light irradiation, PDT generates reactive oxygen species (ROS) that precisely damage the mitochondria of tumor cells and break single-stranded DNA (ssDNA) to activate MCD@TMPyP4@DOX self-disassembly for release of DOX and DNAzyme. We have demonstrated that this multifunctional DNA nanodrug has high drug delivery capacity and biosafety. It enables downregulation of P-gp expression while reducing the ATP on which P-gp pumps out drugs, improving the latency of gene therapy and synergistically reducing DOX efflux to sensitize tumor chemotherapy. We envision that this gene-modulating DNA nanodrug based on damaging mitochondria is expected to provide an important perspective for sensitizing tumor chemotherapy.


Assuntos
DNA Catalítico , Nanopartículas , Resistencia a Medicamentos Antineoplásicos , DNA , DNA de Cadeia Simples , Terapia Genética , Mitocôndrias , Nanopartículas/uso terapêutico
8.
ACS Nano ; 17(15): 15025-15043, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37481734

RESUMO

CRISPR/Cas9 systems have great potential to achieve sophisticated gene therapy and cell engineering by editing multiple genomic loci. However, to achieve efficient multiplex gene editing, the delivery system needs adequate capacity to transfect all CRISPR/Cas9 RNA species at the required stoichiometry into the cytosol of each individual cell. Herein, inspired by biomineralization in nature, we develop an all-in-one biomimetic mineralized CRISPR/Cas9 RNA delivery system. This system allows for precise control over the coencapsulation ratio between Cas9 mRNA and multiple sgRNAs, while also exhibiting a high RNA loading capacity. In addition, it enhances the storage stability of RNA at 4 °C for up to one month, and the surface of the nanoparticles can be easily functionalized for precise targeting of RNA nanoparticles in vivo at nonliver sites. Based on the above characteristics, as a proof-of-concept, our system was able to achieve significant gene-editing at each target gene (Survivin: 31.9%, PLK1: 24.41%, HPV: 23.2%) and promote apoptosis of HeLa cells in the mouse model, inhibiting tumor growth without obvious off-target effects in liver tissue. This system addresses various challenges associated with multicomponent RNA delivery in vivo, providing an innovative strategy for the RNA-based CRISPR/Cas9 gene editing.


Assuntos
Edição de Genes , Nanopartículas , Camundongos , Animais , Humanos , Sistemas CRISPR-Cas/genética , RNA , Células HeLa , Biomimética , RNA Guia de Sistemas CRISPR-Cas
9.
Nat Commun ; 14(1): 4511, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37500633

RESUMO

Postoperative tumor recurrence and metastases often lead to cancer treatment failure. Here, we develop a local embedded photodynamic immunomodulatory DNA hydrogel for early warning and inhibition of postoperative tumor recurrence. The DNA hydrogel contains PDL1 aptamers that capture and enrich in situ relapsed tumor cells, increasing local ATP concentration to provide a timely warning signal. When a positive signal is detected, local laser irradiation is performed to trigger photodynamic therapy to kill captured tumor cells and release tumor-associated antigens (TAA). In addition, reactive oxygen species break DNA strands in the hydrogel to release encoded PDL1 aptamer and CpG, which together with TAA promote sufficient systemic antitumor immunotherapy. In a murine model where tumor cells are injected at the surgical site to mimic tumor recurrence, we find that the hydrogel system enables timely detection of tumor recurrence by enriching relapsed tumor cells to increase local ATP concentrations. As a result, a significant inhibitory effect of approximately 88.1% on recurrent tumors and effectively suppressing metastasis, offering a promising avenue for timely and effective treatment of postoperative tumor recurrence.


Assuntos
Hidrogéis , Recidiva Local de Neoplasia , Humanos , Animais , Camundongos , Recidiva Local de Neoplasia/prevenção & controle , Antígenos de Neoplasias , DNA , Trifosfato de Adenosina , Linhagem Celular Tumoral
10.
ACS Nano ; 17(14): 13746-13759, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37438324

RESUMO

Mitochondria-specific photosensitizer accumulation is highly recommended for photodynamic therapy and mitochondrial DNA (mtDNA) oxidative damage-based innate immunotherapy but remains challenging. 5-Aminolevulinic acid (ALA), precursor of photosensitizer protoporphyrin IX (PpIX), can induce the exclusive biosynthesis of PpIX in mitochondria. Nevertheless, its photodynamic effect is limited by the intracellular biotransformation of ALA in tumors. Here, we report a photosensitizer metabolism-regulating strategy using ALA/DNAzyme-co-loaded nanoparticles (ALA&Dz@ZIF-PEG) for mitochondria-targeting photodynamic immunotherapy. The zeolitic imidazolate framework (ZIF-8) nanoparticles can be disassembled and release large amounts of zinc ions (Zn2+) within tumor cells. Notably, Zn2+ can relieve tumor hypoxia for promoting the conversion of ALA to PpIX. Moreover, Zn2+ acts as a cofactor of rationally designed DNAzyme for silencing excessive ferrochelatase (FECH; which catalyzes PpIX into photoinactive Heme), cooperatively promoting the exclusive accumulation of PpIX in mitochondria via the "open source and reduced expenditure" manner. Subsequently, the photodynamic effects derived from PpIX lead to the damage and release of mtDNA and activate the innate immune response. In addition, the released Zn2+ further enhances the mtDNA/cGAS-STING pathway mediated innate immunity. The ALA&Dz@ZIF-PEG system induced 3 times more PpIX accumulation than ALA-loaded liposome, significantly enhancing tumor regression in xenograft tumor models.


Assuntos
DNA Catalítico , Nanopartículas , Fotoquimioterapia , Humanos , Fármacos Fotossensibilizantes/farmacologia , DNA Catalítico/metabolismo , Linhagem Celular Tumoral , Ácido Aminolevulínico/metabolismo , Ácido Aminolevulínico/farmacologia , Mitocôndrias , DNA Mitocondrial/metabolismo , DNA Mitocondrial/farmacologia , Imunoterapia , Protoporfirinas
11.
Adv Sci (Weinh) ; 10(18): e2300127, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37088730

RESUMO

Liver fibrosis is a progressive histological manifestation that happens in almost all chronic liver diseases. An unabated liver fibrosis may eventually develop into liver cirrhosis or hepatocellular carcinoma. Yet, the strategy for reversal of liver fibrosis is still limited. Herein, a biomimetic nano-regulator (P-ZIF8-cirDNAzyme) is developed to affect both collagen synthesis and degradation in liver to remodel collagen microenvironment. It is found that Zn (II) interference can efficiently inhibit collagen synthesis in activated hepatic stellate cells (aHSC) by inactivating proline 4 hydroxylase and affecting many fibrosis-related signaling pathways. Meanwhile, Zn (II)-dependent circular DNAzymes (cirDNAzymes) are used to efficiently silence tissue inhibitors of metalloproteinase-1, accelerating the degradation of collagen. They act in concert to recover the balance between collagen deposition and degradation. Additionally, ZIF-8-cirDNAzyme is coated by platelet membrane (PM) for precisely targeting aHSC via PM's inflammatory tropism and CD62p-CD44 interaction. In carbon tetrachloride-induced fibrotic mice, P-ZIF-8-cirDNAzyme shows a potent anti-fibrotic effect, greatly reducing the expression of collagen by 73.12% and restoring liver function nearly to normal. This work proposes a prospective platform enabling ion interference and gene silencing, collectively acting in aHSC for reversal of liver fibrosis.


Assuntos
Biomimética , Neoplasias Hepáticas , Animais , Camundongos , Cirrose Hepática/tratamento farmacológico , Colágeno , Microambiente Tumoral
12.
J Control Release ; 356: 272-287, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36870541

RESUMO

Abundant cancer-associated fibroblasts (CAFs) in highly fibrotic breast cancer constitute an immunosuppressive barrier for T cell activity and are closely related to the failure of immune checkpoint blockade therapy (ICB). Inspired by the similar antigen-processing capacity of CAFs to professional antigen-presenting cells (APCs), a "turning foes to friends" strategy is proposed by in situ engineering immune-suppressed CAFs into immune-activated APCs for improving response rates of ICB. To achieve safe and specific CAFs engineering in vivo, a thermochromic spatiotemporal photo-controlled gene expression nanosystem was developed by self-assembly of molten eutectic mixture, chitosan andfusion plasmid. After photoactivatable gene expression, CAFs could be engineered as APCs via co-stimulatory molecule (CD86) expression, which effectively induced activation and proliferation of antigen-specific CD8 + T cells. Meanwhile, engineered CAFs could also secrete PD-L1 trap protein in situ for ICB, avoiding potential autoimmune-like disorders caused by "off-target" effects of clinically applied PD-L1 antibody. The study demonstrated that the designed nanosystem could efficiently engineer CAFs, significantly enhance the percentages of CD8+ T cells (4-folds), result in about 85% tumor inhibition rate and 83.3% survival rate at 60 days in highly fibrotic breast cancer, further inducing long-term immune memory effects and effectively inhibiting lung metastasis.


Assuntos
Neoplasias da Mama , Fibroblastos Associados a Câncer , Neoplasias Pulmonares , Humanos , Feminino , Inibidores de Checkpoint Imunológico/metabolismo , Antígeno B7-H1 , Fibroblastos Associados a Câncer/metabolismo , Imunoterapia , Neoplasias Pulmonares/metabolismo , Neoplasias da Mama/metabolismo , Microambiente Tumoral
13.
ACS Sens ; 8(2): 565-575, 2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-36722721

RESUMO

Exosomal miRNAs play a critical role in cancer biology and could be potential biomarkers for cancer diagnosis. However, due to the low abundance of miRNAs in the exosomes, recognizing and detecting disease-associated exosomal miRNAs in an easy-to-operate way remain a challenge. Herein, we used a liposome-mediated membrane fusion strategy (MFS) to transfect CRISPR/Cas13a into exosomes, termed MFS-CRISPR, directly measuring exosomal miRNAs in plasma. Using the MFS-CRISPR platform for detection of the exosomal miR-21, we achieve a linear range spanning four orders of magnitude (104-108 particles/mL) and the method is able to detect the exosomal miR-21 in as low as 1.2 × 103 particles/mL. The liposome-mediated MFS could confine fluorescent signals in fused vesicles, which can be used for exosome heterogeneity analysis. Moreover, MFS-CRISPR assay was evaluated by measuring clinical samples, and the difference of miR-21 expression of breast cancer patients and healthy donors was significant. Because of high sensitivity and simplicity, the proposed method could have promising clinical potential for cancer diagnosis and treatment monitoring.


Assuntos
Neoplasias da Mama , MicroRNAs , Humanos , Feminino , MicroRNAs/análise , Lipossomos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Neoplasias da Mama/diagnóstico , Transfecção
14.
Small Methods ; 7(2): e2201624, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36609885

RESUMO

Deoxyribonucleic acid (DNA) methylation is one of the epigenetic characteristics that result in heritable and revisable phenotype changes but without sequence changes in DNA. Aberrant methylation occurring at a specific locus was reported to be associated with cancers, insulin resistance, obesity, Alzheimer's disease, Parkinson's disease, etc. Therefore, locus-specific DNA methylation can serve as a valuable biomarker for disease diagnosis and therapy. Recently, Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas systems are applied to develop biosensors for DNA, ribonucleic acid, proteins, and small molecules detection. Because of their highly specific binding ability and signal amplification capacity, CRISPR-Cas assisted biosensor also serve as a potential tool for locus-specific detection of DNA methylation. In this perspective, based on the detection principle, a detailed classification and comprehensive discussion of recent works about the latest advances in locus-specific detection of DNA methylation using CRISPR-Cas systems are provided. Furthermore, current challenges and future perspectives of CRISPR-based locus-specific detection of DNA methylation are outlined.


Assuntos
Técnicas Biossensoriais , Sistemas CRISPR-Cas , Metilação de DNA , RNA , DNA
15.
Clin Cancer Res ; 29(1): 183-196, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36240005

RESUMO

PURPOSE: Despite approval of B-cell lymphoma (BCL)-2 inhibitor venetoclax for certain hematologic malignancies, its broader clinical benefit is curtailed by resistance. Our study aimed to determine if treatment with novel anticancer agents targeting BCL-2 and mouse double minute 2 (MDM2) could overcome venetoclax resistance in preclinical models. EXPERIMENTAL DESIGN: Venetoclax-sensitive and venetoclax-resistant acute myeloid leukemia (AML) and acute lymphoblastic leukemia cells and xenograft models were used to evaluate antitumor effects and underlying mechanisms associated with combined BCL-2 inhibitor lisaftoclax (APG-2575) and MDM2 inhibitor alrizomadlin (APG-115). RESULTS: The combination exhibited synergistic antiproliferative and apoptogenic activities in TP53 wild-type AML cell lines in vitro. This synergy was further exemplified by deep antitumor responses and prolonged survival in AML cell line-derived and patient-derived xenograft models. Interestingly, the combination treatment resensitized (to apoptosis) venetoclax-resistant cellular and mouse models established via chronic drug exposure or genetically engineered with clinically relevant BCL-2 gene mutations. Synergistic effects in reducing cellular viability and proliferation were also demonstrated in primary samples of patients with venetoclax-resistant AML treated with lisaftoclax and alrizomadlin ex vivo. Mechanistically, alrizomadlin likely primes cancer cells to BCL-2 inhibition-induced cellular apoptosis by downregulating expression of antiapoptotic proteins myeloid cell leukemia-1 and BCL-extra-large and upregulating pro-death BCL-2-associated X protein. CONCLUSIONS: Lisaftoclax in combination with alrizomadlin overcomes venetoclax resistance mediated by various mechanisms, including BCL-2 mutations. In addition, we posit further, putative molecular mechanisms. Our data rationalize clinical development of this treatment combination in patients with diseases that are insensitive or resistant to venetoclax.


Assuntos
Antineoplásicos , Leucemia Mieloide Aguda , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Animais , Camundongos , Proteínas Proto-Oncogênicas c-bcl-2 , Linhagem Celular Tumoral , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Antineoplásicos/uso terapêutico , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Apoptose , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
16.
ACS Appl Mater Interfaces ; 14(39): 44183-44198, 2022 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-36165393

RESUMO

Immunotherapy is emerging as a paradigm-shifting modality for treatment cancer. However, systemic administration of immunomodulators is usually accompanied by extra-tumor toxicity and adverse immune effects. Precise delivery of immunomodulators with a highly controllable system may provide a solution for this issue. Here, we developed a photocontrolled DNA nanomedicine for localized delivery of DNA immunomodulators to enhance membrane-targeted photodynamic immunotherapy. Specifically, the DNA nanomedicine is composed of long tandemly repeated functional DNA sequences (PDL1 aptamers and CpG) with a photosensitizer (TMPyP4) inserted into the DNA structure, providing high drug-loading capacity. By blocking the surface PDL1 aptamer with a pHLIP-modified cDNA, the DNA nanomedicine does not induce any obvious immune response and can be specifically delivered and anchored to the tumor membrane. Under localized irradiation, photodynamically generated reactive oxygen species (ROS) causes breakage of DNA sequences, which triggers the collapse of the nanostructure and release of internal DNA immunomodulators. Under localized illumination, photodynamically generated ROS can cause DNA sequence breaks, triggering the collapse of nanostructures and the release of internal DNA immunomodulators thus enhancing membrane-targeted photodynamic immunotherapy. We have demonstrated that the developed DNA nanomedicine can drive efficient immune responses in tumor tissue without perceptibly interfering off-tumor immunity, resulting in efficient antitumor treatment while mitigating systemic toxicity.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Linhagem Celular Tumoral , DNA , DNA Complementar , Humanos , Fatores Imunológicos , Imunoterapia , Nanopartículas/química , Neoplasias/tratamento farmacológico , Oligonucleotídeos , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Espécies Reativas de Oxigênio
17.
J Nanobiotechnology ; 20(1): 429, 2022 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-36175999

RESUMO

Despite many nano-based strategies devoted to delivering cisplatin for tumor therapy, its clinical benefits are compromised by poor tissue penetration and limited DNA adducts formation of the drug. Herein, a cisplatin loading nanomotor based janus structured Ag-polymer is developed for cisplatin delivery of deeper tissue and increased DNA adducts formation. The nanomotor displayed a self-propelled tumor penetration fueled by hydrogen peroxide (H2O2) in tumor tissues, which is catalytically decomposed into a large amount of oxygen bubbles by Ag nanoparticles (NPs). Notably, cisplatin could elevate the intracellular H2O2 level through cascade reactions, further promote the degradation of Ag NPs accompanied with the Ag+ release, which could downregulate intracellular Cl- through the formation of AgCl precipitate, thereby enhancing cisplatin dechlorination and Pt-DNA formation. Moreover, polymer can also inhibit the activity of ALKBH2 (a Fe2+-dependent DNA repair enzyme) by chelating intracellular Fe2+ to increase the proportion of irreparable Pt-DNA cross-links. It is found that deep tissue penetration, as well as the increased formation and maintenance of Pt-DNA adducts induced by the nanomotor afford 80% of tumor growth inhibition with negligible toxicity. This work provides an important perspective of resolving chemotherapeutic barriers for boosting cisplatin therapy.


Assuntos
Antineoplásicos , Nanopartículas Metálicas , Neoplasias , Antineoplásicos/uso terapêutico , Cisplatino/farmacologia , Cisplatino/uso terapêutico , DNA/metabolismo , Adutos de DNA/uso terapêutico , Humanos , Peróxido de Hidrogênio , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Oxigênio , Polímeros/uso terapêutico , Prata/uso terapêutico
18.
Clin Transl Oncol ; 24(11): 2166-2174, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35789980

RESUMO

PURPOSE: This study was designed to explore the role of COPZ1 in breast cancer as well as discuss its specific reaction mechanism. METHODS: With the help of RT-qPCR and western blot, the expression of BMI1 and COPZ1 were measured. Then, the proliferation, colony formation and apoptosis were evaluated by CCK-8, colony formation and TUNEL assays, separately. Luciferase reporter assay and ChIP were applied to assess the relative activity of COPZ1 promoter as well as its binding with BMI1. Moreover, western blot was utilized to measure the expression of proliferation-, apoptosis- and autophagy-related proteins. RESULTS: According to GEPIA2 database, COPZ1 was upregulated in breast cancer tissues and was associated with the poor prognosis (P = 0.03). Results obtained from RT-qPCR and western blot verified that COPZ1 expression was greatly increased at both mRNA and protein levels in breast cancer cells as compared to control cells (P < 0.05 or P < 0.001). COPZ1 knockdown inhibited the proliferation, induced the autophagy and promoted the apoptosis of breast cancer cells. HumanTFDB predicted the binding sites of BMI1 and COPZ1. The increased relative luciferase activity of COPZ1 promoter following BMI1 overexpression (P < 0.001) and the binding of BMI1 with COPZ1 promoter indicated that BMI1 could activate COPZ1. Further experiments suggested that the effects of COPZ1 knockdown on the proliferation, apoptosis and autophagy of breast cancer cells were reversed by BMI1 overexpression, implying that BMI1 promoted the proliferation and repressed the autophagy of breast cancer cells via activating COPZ1. CONCLUSIONS: To sum up, BMI1 exhibited promotive effects on the malignant progression of breast cancer through the activation of COPZ1. These findings might offer a preliminary theoretical basis for COPZ1 participation in autophagy in breast cancer cells.


Assuntos
Neoplasias da Mama , MicroRNAs , Apoptose/genética , Autofagia , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/metabolismo , Proteínas Relacionadas à Autofagia/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Proteína Coatomer , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , Complexo Repressor Polycomb 1/genética , RNA Mensageiro , Sincalida/metabolismo
19.
Front Genet ; 13: 852708, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35801082

RESUMO

Background: Bladder cancer (BLCA) is among the most frequent types of cancer. Patients with BLCA have a significant recurrence rate and a poor post-surgery survival rate. Recent research has found a link between tumor immune cell infiltration (ICI) and the prognosis of BLCA patients. However, the ICI's picture of BLCA remains unclear. Methods: Common gene expression data were obtained by combining the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) expression databases. Two computational algorithms were proposed to unravel the ICI landscape of BLCA patients. The R package "limma" was applied to find differentially expressed genes (DEGs). ICI patterns were defined by the unsupervised clustering method. Principal-component analysis (PCA) was used to calculate the ICI score. In addition, the combined ICI score and tumor burden mutation (TMB) were utilized to assess BLCA patients' prognosis. The predictive value of ICI scores was verified by different clinical characteristics. Results: A total of 569 common gene expression data were retrieved from TCGA and GEO cohorts. CD8+ T cells were found to have a substantial positive connection with activated memory CD4+ T cells and immune score. On the contrary, CD8+ T cells were found to have a substantial negative connection with macrophages M0. Thirty-eight DEGs were selected. Two ICI patterns were defined by the unsupervised clustering method. Patients of BLCA were separated into two groups. The high ICI score group exhibited a better outcome than the low ICI score one (p < 0.001). Finally, the group with a high tumor mutation burden (TMB) as well as a high ICI score had the best outcome. (p < 0.001). Conclusions: Combining TMB and ICI scores resulted in a more accurate survival prediction, suggesting that ICI scores could be used as a prognostic marker for BLCA patients.

20.
J Nanobiotechnology ; 20(1): 313, 2022 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-35794596

RESUMO

Metastasis is one of the main causes of failure in the treatment of triple-negative breast cancer (TNBC). Abnormally estrogen level and activated platelets are the key driving forces for TNBC metastasis. Herein, an "ion/gas" bioactive nanogenerator (termed as IGBN), comprising a copper-based MOF and loaded cisplatin-arginine (Pt-Arg) prodrug is developed for metastasis-promoting tumor microenvironment reprogramming and TNBC therapy. The copper-based MOF not only serves as a drug carrier, but also specifically produces Cu2+ in tumors, which catalytic oxidizing estrogen to reduce estrogen levels in situ. Meanwhile, the rationally designed Pt-Arg prodrug reduced into cisplatin to significantly promote the generation of H2O2 in the tumor, then permitting self-augmented cascade NO gas generation by oxidizing Arg through a H2O2 self-supplied way, thus blocking platelet activation in tumor. We clarified that IGBN inhibited TNBC metastasis through local estrogen deprivation and platelets blockade, affording 88.4% inhibition of pulmonary metastasis in a 4T1 mammary adenocarcinoma model. Notably, the locally copper ion interference, NO gas therapy and cisplatin chemotherapy together resulted in an enhanced therapeutic efficacy in primary tumor ablation without significant toxicity. This "ion/gas" bioactive nanogenerator offers a robust and safe strategy for TNBC therapy.


Assuntos
Estruturas Metalorgânicas , Pró-Fármacos , Neoplasias de Mama Triplo Negativas , Cisplatino/farmacologia , Cobre , Estrogênios , Humanos , Peróxido de Hidrogênio , Estruturas Metalorgânicas/farmacologia , Pró-Fármacos/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA