Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Mol Cell ; 82(21): 4099-4115.e9, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36208627

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive hepatic lipid accumulation, which can progress to nonalcoholic steatohepatitis (NASH). Histone deacetylase Sirtuin 6 (SIRT6) regulates NAFLD by regulating metabolism-related gene expression, but an extrachromosomal role for SIRT6 in NAFLD development remains elusive. We investigated whether SIRT6 functions on NAFLD in the cytoplasm. We found that SIRT6 binds saturated fatty acids, especially palmitic acid. This binding leads to its nuclear export, where it deacetylates long-chain acyl-CoA synthase 5 (ACSL5), thereby facilitating fatty acid oxidation. High-fat diet-induced NAFLD is suppressed by ACSL5 hepatic overexpression but is exacerbated by its depletion. As confirmation, overexpression of a deacetylated ACSL5 mimic attenuated NAFLD in Sirt6 liver-specific knockout mice. Moreover, NASH-hepatic tissues from both patients and diet-fed mice exhibited significantly reduced cytoplasmic SIRT6 levels and increased ACSL5 acetylation. The SIRT6/ACSL5 signaling pathway has a critical role in NAFLD progression and might constitute an avenue for therapeutic intervention.


Subject(s)
Non-alcoholic Fatty Liver Disease , Sirtuins , Mice , Animals , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Acyl Coenzyme A/metabolism , Mice, Inbred C57BL , Liver/metabolism , Lipid Metabolism , Mice, Knockout , Fatty Acids/metabolism , Sirtuins/genetics , Sirtuins/metabolism , Cytoplasm/metabolism
2.
Nano Lett ; 24(29): 9017-9026, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39007530

ABSTRACT

The development of in situ tumor vaccines offers promising prospects for cancer treatment. Nonetheless, the generation of plenary autologous antigens in vivo and their codelivery to DC cells along with adjuvants remains a significant challenge. Herein, we developed an in situ tumor vaccine using a supramolecular nanoparticle/hydrogel composite (ANPMTO/ALCD) and a deformable nanoadjuvant (PPER848). The ANPMTO/ALCD composite consisted of ß-cyclodextrin-decorated alginate (Alg-g-CD) and MTO-encapsulated adamantane-decorated nanoparticles (ANPMTO) through supramolecular interaction, facilitating the long-term and sustained production of plenary autologous antigens, particularly under a 660 nm laser. Simultaneously, the produced autologous antigens were effectively captured by nanoadjuvant PPER848 and subsequently transported to lymph nodes and DC cells, benefiting from its optimized size and deformability. This in situ tumor vaccine can trigger a robust antitumor immune response and demonstrate significant therapeutic efficacy in inhibiting tumor growth, suppressing tumor metastasis, and preventing postoperative recurrence, offering a straightforward approach to programming in situ tumor vaccines.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines , Immunotherapy , Nanoparticles , Cancer Vaccines/chemistry , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Animals , Mice , Immunotherapy/methods , Nanoparticles/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Adjuvants, Immunologic/pharmacology , Hydrogels/chemistry , Humans , Cell Line, Tumor , Dendritic Cells/immunology , beta-Cyclodextrins/chemistry , Neoplasms/therapy , Neoplasms/immunology , Alginates/chemistry , Adamantane/chemistry , Adamantane/therapeutic use
3.
Nucleic Acids Res ; 48(6): 2982-3000, 2020 04 06.
Article in English | MEDLINE | ID: mdl-31970415

ABSTRACT

Genomic instability is an underlying hallmark of cancer and is closely associated with defects in DNA damage repair (DDR). Chromatin relaxation is a prerequisite for DDR, but how chromatin accessibility is regulated remains elusive. Here we report that the histone deacetylase SIRT6 coordinates with the chromatin remodeler CHD4 to promote chromatin relaxation in response to DNA damage. Upon DNA damage, SIRT6 rapidly translocates to DNA damage sites, where it interacts with and recruits CHD4. Once at the damage sites, CHD4 displaces heterochromatin protein 1 (HP1) from histone H3 lysine 9 trimethylation (H3K9me3). Notably, loss of SIRT6 or CHD4 leads to impaired chromatin relaxation and disrupted DNA repair protein recruitment. These molecular changes, in-turn, lead to defective homologous recombination (HR) and cancer cell hypersensitivity to DNA damaging agents. Furthermore, we show that SIRT6-mediated CHD4 recruitment has a specific role in DDR within compacted chromatin by HR in G2 phase, which is an ataxia telangiectasia mutated (ATM)-dependent process. Taken together, our results identify a novel function for SIRT6 in recruiting CHD4 onto DNA double-strand breaks. This newly identified novel molecular mechanism involves CHD4-dependent chromatin relaxation and competitive release of HP1 from H3K9me3 within the damaged chromatin, which are both essential for accurate HR.


Subject(s)
Chromatin/metabolism , DNA Repair , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Sirtuins/metabolism , Cell Line, Tumor , Cell Survival , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA Breaks, Double-Stranded , HEK293 Cells , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Mi-2 Nucleosome Remodeling and Deacetylase Complex/chemistry , Models, Biological , Protein Binding , Protein Domains
4.
Small ; : e2004879, 2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33289336

ABSTRACT

Blocking immune checkpoint pathways with an antibody or small interfering RNA (siRNA) has become a promising method to reactivate antitumor responses for cancer treatment. However, both blockade strategies achieve only temporary inhibition of these immune checkpoints. Herein, a photoswitched CRISPR/Cas9 system for genomic disruption of the PD-L1 gene is developed to achieve permanent blockade of the PD-1/PD-L1 pathway; this system is constructed by using a photoactivated self-degradable polyethyleneimine derivative and the plasmid pX330/sgPD-L1 (expression of the Cas9 protein and single-guide RNA targeting PD-L1). Under light irradiation, this photoswitched CRISPR/Cas9 system efficiently genetically disrupts the PD-L1 gene in not only bulk cancer cells but also cancer stem-like cells. As a result, the photoswitched CRISPR/Cas9 system significantly increases the infiltration of CD8+ T cells into tumor tissue, leading to effective activation of a T cell-mediated antitumor response against cancer cells and cancer stem-like cells. This study provides an alternative strategy to block the PD-1/PD-L1 pathway for efficacious immune checkpoint therapy.

5.
Small ; 16(16): e1907461, 2020 04.
Article in English | MEDLINE | ID: mdl-32187862

ABSTRACT

With the rapid growth in wearable electronics sensing devices, flexible sensing devices that monitor the human body have shown great promise in personalized healthcare. In the study, high-quality GaN pn junction microwire arrays with different aspect ratios and large-area uniformity are fabricated through an easy, repeatable fabrication process. The piezoelectric coefficient (d33 ) of GaN pn junction microwire arrays increases from 7.23 to 14.46 pm V-1 with the increasing of the aspect ratio, which is several times higher than that of GaN bulk material. Furthermore, flexible ultrasensitive strain sensor based on GaN microwires with the highest d33 is demonstrated to achieve the maximum open circuit voltage of 10.4 V, and presents excellent durability with stable output signals over 10 000 cycles with a response time of 50 ms. As a flexible and wearable sensor attached to the human skin, the GaN microwire pn junction arrays with such a high degree of uniformity can precisely monitor subtle human pulse and motions, which show great promise in future personalized healthcare.


Subject(s)
Heart Rate , Monitoring, Physiologic , Movement , Wearable Electronic Devices , Humans , Skin
6.
Nucleic Acids Res ; 46(15): 7716-7730, 2018 09 06.
Article in English | MEDLINE | ID: mdl-29982688

ABSTRACT

Linker histone H1 has a key role in maintaining higher order chromatin structure and genome stability, but how H1 functions in these processes is elusive. Here, we report that acetylation of lysine 85 (K85) within the H1 globular domain is a critical post-translational modification that regulates chromatin organization. H1K85 is dynamically acetylated by the acetyltransferase PCAF in response to DNA damage, and this effect is counterbalanced by the histone deacetylase HDAC1. Notably, an acetylation-mimic mutation of H1K85 (H1K85Q) alters H1 binding to the nucleosome and leads to condensed chromatin as a result of increased H1 binding to core histones. In addition, H1K85 acetylation promotes heterochromatin protein 1 (HP1) recruitment to facilitate chromatin compaction. Consequently, H1K85 mutation leads to genomic instability and decreased cell survival upon DNA damage. Together, our data suggest a novel model whereby H1K85 acetylation regulates chromatin structure and preserves chromosome integrity upon DNA damage.


Subject(s)
Chromatin/metabolism , DNA Damage , Genomic Instability , Histones/metabolism , Lysine/metabolism , A549 Cells , Acetylation , Cell Line, Tumor , Cell Survival/genetics , Chromatin/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , HCT116 Cells , HEK293 Cells , HeLa Cells , Histones/genetics , Humans , Lysine/genetics , Mutation , Nucleosomes/genetics , Nucleosomes/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
7.
Small ; 15(36): e1902022, 2019 09.
Article in English | MEDLINE | ID: mdl-31318147

ABSTRACT

The cell nucleus-targeted delivery of therapeutic agents plays a critical role in cancer therapy, since the biological target of many anticancer therapeutics is the cell nucleus. However, multiple physiological barriers limit the delivery efficiency of free drugs, resulting in unsatisfactory therapeutic effects. Herein, thioketal crosslinked polyphosphoester-based nanoparticles with a tumor acidity (pHe )-sensitive transactivator of transcription (TAT) peptide (DA-masked TAT-decorating reactive oxygen species (ROS)-sensitive Ce6/DOX-loaded hyperbranched nanoparticles (D TRCD)) are explored for cascade nucleus-targeted drug delivery. Following administration, D TRCD experiences prolonged circulation by masking the targeting effect of its TAT peptide and then achieves enhanced tumor cell uptake and improved translocation into the perinuclear region by reactivating the TAT targeting capability in tumor tissue. Subsequently, ROS generated by D TRCD under 660 nm laser not only disrupts the nuclear membrane to allow entry into the nuclei but also triggers intracellular release of the payload in the nuclei. As evidenced by in vivo experiments, such pHe /photo dual-sensitive polymeric nanocarriers offer remarkable therapeutic effects, efficiently suppressing tumor growth. This multistage cascade nucleus-targeted drug delivery concept provides new avenues to develop nucleus-targeted drug delivery systems.


Subject(s)
Cell Nucleus/metabolism , Nanoparticles/chemistry , Neoplasms/drug therapy , Polymers/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Drug Delivery Systems/methods , Humans , Hydrogen-Ion Concentration , Reactive Oxygen Species
8.
J Biol Chem ; 292(32): 13296-13311, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28655758

ABSTRACT

Sirtuin 7 (SIRT7), a member of the NAD+-dependent class III histone deacetylases, is involved in the regulation of various cellular processes and in resisting various stresses, such as hypoxia, low glucose levels, and DNA damage. Interestingly, SIRT7 is linked to the control of glycolysis, suggesting a role in glucose metabolism. Given the important roles of SIRT7, it is critical to clarify how SIRT7 activity is potentially regulated. It has been reported that some transcriptional and post-transcriptional regulatory mechanisms are involved. However, little is known how SIRT7 is regulated by the post-translational modifications. Here, we identified ubiquitin-specific peptidase 7 (USP7), a deubiquitinase, as a negative regulator of SIRT7. We showed that USP7 interacts with SIRT7 both in vitro and in vivo, and we further demonstrated that SIRT7 undergoes endogenous Lys-63-linked polyubiquitination, which is removed by USP7. Although the USP7-mediated deubiquitination of SIRT7 had no effect on its stability, the deubiquitination repressed its enzymatic activity. We also showed that USP7 coordinates with SIRT7 to regulate the expression of glucose-6-phosphatase catalytic subunit (G6PC), a gluconeogenic gene. USP7 depletion by RNA interference increased both G6PC expression and SIRT7 enzymatic activity. Moreover, SIRT7 targeted the G6PC promoter through the transcription factor ELK4 but not through forkhead box O1 (FoxO1). In summary, SIRT7 is a USP7 substrate and has a novel role as a regulator of gluconeogenesis. Our study may provide the basis for new clinical approaches to treat metabolic disorders related to glucose metabolism.


Subject(s)
Gene Expression Regulation, Enzymologic , Glucose-6-Phosphatase/metabolism , Promoter Regions, Genetic , Protein Processing, Post-Translational , Sirtuins/metabolism , Ubiquitin Thiolesterase/metabolism , ets-Domain Protein Elk-4/metabolism , Amino Acid Substitution , Cell Line, Tumor , Gene Deletion , Gluconeogenesis , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/genetics , HEK293 Cells , Humans , Hydrolysis , Lysine/metabolism , Mutation , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Subunits/genetics , Protein Subunits/metabolism , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sirtuins/antagonists & inhibitors , Sirtuins/genetics , Substrate Specificity , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination , ets-Domain Protein Elk-4/genetics
9.
Proc Natl Acad Sci U S A ; 111(29): 10684-9, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25009184

ABSTRACT

In mammalian cells, tumor suppressor p53 plays critical roles in the regulation of glucose metabolism, including glycolysis and oxidative phosphorylation, but whether and how p53 also regulates gluconeogenesis is less clear. Here, we report that p53 efficiently down-regulates the expression of phosphoenolpyruvate carboxykinase (PCK1) and glucose-6-phosphatase (G6PC), which encode rate-limiting enzymes in gluconeogenesis. Cell-based assays demonstrate the p53-dependent nuclear exclusion of forkhead box protein O1 (FoxO1), a key transcription factor that mediates activation of PCK1 and G6PC, with consequent alleviation of FoxO1-dependent gluconeogenesis. Further mechanistic studies show that p53 directly activates expression of the NAD(+)-dependent histone deacetylase sirtuin 6 (SIRT6), whose interaction with FoxO1 leads to FoxO1 deacetylation and export to the cytoplasm. In support of these observations, p53-mediated FoxO1 nuclear exclusion, down-regulation of PCK1 and G6PC expression, and regulation of glucose levels were confirmed in C57BL/J6 mice and in liver-specific Sirt6 conditional knockout mice. Our results provide insights into mechanisms of metabolism-related p53 functions that may be relevant to tumor suppression.


Subject(s)
Cell Nucleus/metabolism , Forkhead Transcription Factors/metabolism , Gluconeogenesis/genetics , Sirtuins/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Animals , Blood Glucose/metabolism , Down-Regulation/genetics , Forkhead Box Protein O1 , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , HCT116 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Protein Binding , Protein Transport , Sirtuins/genetics , Transcription, Genetic
10.
Genes Dis ; 11(5): 100949, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39071111

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL), a heterogeneous hematological malignancy, is caused by the developmental arrest of normal T-cell progenitors. The development of targeted therapeutic regimens is impeded by poor knowledge of the stage-specific aberrances in this disease. In this study, we performed multi-omics integration analysis, which included mRNA expression, chromatin accessibility, and gene-dependency database analyses, to identify potential stage-specific druggable targets and repositioned drugs for this disease. This multi-omics integration helped identify 29 potential pathological genes for T-ALL. These genes exhibited tissue-specific expression profiles and were enriched in the cell cycle, hematopoietic stem cell differentiation, and the AMPK signaling pathway. Of these, four known druggable targets (CDK6, TUBA1A, TUBB, and TYMS) showed dysregulated and stage-specific expression in malignant T cells and may serve as stage-specific targets in T-ALL. The TUBA1A expression level was higher in the early T cell precursor (ETP)-ALL cells, while TUBB and TYMS were mainly highly expressed in malignant T cells arrested at the CD4 and CD8 double-positive or single-positive stage. CDK6 exhibited a U-shaped expression pattern in malignant T cells along the naïve to maturation stages. Furthermore, mebendazole and gemcitabine, which target TUBA1A and TYMS, respectively, exerted stage-specific inhibitory effects on T-ALL cell lines, indicating their potential stage-specific antileukemic role in T-ALL. Collectively, our findings might aid in identifying potential stage-specific druggable targets and are promising for achieving more precise therapeutic strategies for T-ALL.

11.
Nat Nanotechnol ; 19(4): 545-553, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38216684

ABSTRACT

In some cancers mutant p53 promotes the occurrence, development, metastasis and drug resistance of tumours, with targeted protein degradation seen as an effective therapeutic strategy. However, a lack of specific autophagy receptors limits this. Here, we propose the synthesis of biomimetic nanoreceptors (NRs) that mimic selective autophagy receptors. The NRs have both a component for targeting the desired protein, mutant-p53-binding peptide, and a component for enhancing degradation, cationic lipid. The peptide can bind to mutant p53 while the cationic lipid simultaneously targets autophagosomes and elevates the levels of autophagosome formation, increasing mutant p53 degradation. The NRs are demonstrated in vitro and in a patient-derived xenograft ovarian cancer model in vivo. The work highlights a possible direction for treating diseases by protein degradation.


Subject(s)
Autophagy , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Proteolysis , Mutant Proteins/metabolism , Mutant Proteins/pharmacology , Cell Line, Tumor , Peptides/metabolism , Lipids/pharmacology
12.
Mol Genet Genomic Med ; 12(1): e2357, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38284445

ABSTRACT

BACKGROUND: Newborn screening (NBS) aims to detect congenital anomalies, and next-generation sequencing (NGS) has shown promise in this aspect. However, the NBS strategy for monogenic inherited diseases in China remains insufficient. METHODS: We developed a NeoEXOME panel comprising 601 genes that are relevant to the Chinese population found through extensive research on available databases. An interpretation system to grade the results into positive (high-risk, moderate-risk, and low-risk genotypes), negative, and carrier according to the American College of Medical Genetics (ACMG) guidelines was also developed. We validated the panel to evaluate its efficacy by using data from the "1000 Genomes Project" and conducted a pilot multicenter study involving 3423 neonates. RESULTS: The NGS positive rate in the 1000 Genomes Project was 7.6% (23/301), whereas the rate was 12.0% in the multicenter study, including 3249 recruited neonates. Notably, in 200 neonates, positive per conventional NBS, 58.5% (69/118) showed results consistent with NGS. In the remaining 3049 neonates showing negative results in conventional NBS, 271 (8.9%) were positive per NGS, and nine of them were clinically diagnosed with diseases in the follow-up. CONCLUSION: We successfully designed a NeoEXOME panel for targeted sequencing of monogenic inherited diseases in NBS. The panel demonstrated high performance in the Chinese population, particularly for the early detection of diseases with no biochemical markers.


Subject(s)
High-Throughput Nucleotide Sequencing , Neonatal Screening , Humans , Infant, Newborn , Pilot Projects , Exome Sequencing , Neonatal Screening/methods , Genotype , High-Throughput Nucleotide Sequencing/methods
13.
ACS Appl Mater Interfaces ; 15(19): 22843-22853, 2023 May 17.
Article in English | MEDLINE | ID: mdl-37133278

ABSTRACT

Cancer nanomedicine treatment aims to achieve highly specific targeting and localization to cancer cells. Coating of nanoparticles with cell membranes endows them with homologous cellular mimicry, enabling nanoparticles to acquire new functions and properties, including homologous targeting and long circulation in vivo, and can enhance internalization by homologous cancer cells. Herein, we fused a human-derived HCT116 colon cancer cell membrane (cM) with a red blood cell membrane (rM) to fabricate an erythrocyte-cancer cell hybrid membrane (hM). Oxaliplatin and chlorin e6 (Ce6) co-encapsulated reactive oxygen species-responsive nanoparticles (NPOC) were camouflaged by hM and obtained a hybrid biomimetic nanomedicine (denoted as hNPOC) for colon cancer therapy. hNPOC exhibited prolonged circulation time and recognized homologous targeting ability in vivo since both rM and HCT116 cM proteins were maintained on the hNPOC surface. hNPOC showed enhanced homologous cell uptake in vitro and considerable homologous self-localization in vivo, producing effective synergistic chemophotodynamic therapy efficacy under irradiation with a homologous HCT116 tumor compared to that with a heterologous tumor. Together, the biomimetic hNPOC nanoparticles showed prolonged blood circulation and preferential cancer cell-targeted function in vivo to provide a bioinspired strategy for chemophotodynamic synergistic therapy of colon cancer.


Subject(s)
Colonic Neoplasms , Nanoparticles , Humans , Bionics , Erythrocyte Membrane/metabolism , Phototherapy , Colonic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Cell Line, Tumor
14.
Adv Sci (Weinh) ; 10(7): e2204793, 2023 03.
Article in English | MEDLINE | ID: mdl-36698293

ABSTRACT

The passive diffusion performance of nanocarriers results in inefficient drug transport across multiple biological barriers and consequently cancer therapy failure. Here, a magnetically driven amoeba-like nanorobot (amNR) is presented for whole-process active drug transport. The amNR is actively extravasated from blood vessels and penetrated into deep tumor tissue through a magnetically driven deformation effect. Moreover, the acidic microenvironment of deep tumor tissue uncovers the masked targeting ligand of amNR to achieve active tumor cell uptake. Furthermore, the amNR rapidly releases the encapsulated doxorubicin (DOX) after alternating magnetic field application. The amNRs eventually deliver DOX into ≈92.3% of tumor cells and completely delay tumor growth with an inhibition rate of 96.1%. The deformable amNRs, with the assistance of magnetic field application, provide a facile strategy for whole-process active drug transport.


Subject(s)
Amoeba , Biological Transport , Doxorubicin , Magnetic Fields
15.
Biomaterials ; 302: 122339, 2023 11.
Article in English | MEDLINE | ID: mdl-37778054

ABSTRACT

Efficiently reawakening immune cells, including T cells and macrophages, to eliminate tumor cells is a promising strategy for cancer treatment, but remains a huge challenge nowadays. Herein, a nanoassembly formed by doxorubicin (DOX)-conjugated polyphosphoester (PP-(hDOX)) and CD47-targeting siRNA (siCD47) via electrostatic and π-π stacking interactions, termed as PP-(hDOX&siCD47), was developed to reawaken the T cell and macrophage-mediated anticancer activity. The PP-(hDOX&siCD47) could efficiently blockade antiphagocytic signal by downregulation of CD47 expression to reactive macrophage-mediated anticancer immunotherapy. Moreover, the conjugated DOX of PP-(hDOX&siCD47) can perform the chemotherapy towards tumor cells and also elicit the T cell-mediated anticancer immune response via immunogenic cell death (ICD) effect. Therefore, the PP-(hDOX&siCD47) treatment could significantly increase M1-like macrophages proportion and tumor infiltration of CD8+ T cells, while the proportions of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC) were considerably reduced in tumor tissue, eventually achieving significantly tumor growth inhibition. Overall, this study provides a simple siRNA and DOX codelivery approach to simultaneously elicit the macrophage- and T cell-mediated anticancer immune response for cancer therapy.


Subject(s)
CD47 Antigen , Neoplasms , RNA, Small Interfering/metabolism , CD8-Positive T-Lymphocytes/metabolism , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Immunotherapy , Macrophages/metabolism , Immunity , Cell Line, Tumor , Neoplasms/drug therapy
16.
Bioact Mater ; 27: 337-347, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37122898

ABSTRACT

The abnormal activation of epidermal growth factor receptor (EGFR) drives the development of non-small cell lung cancer (NSCLC). The EGFR-targeting tyrosine kinase inhibitor osimertinib is frequently used to clinically treat NSCLC and exhibits marked efficacy in patients with NSCLC who have an EGFR mutation. However, free osimertinib administration exhibits an inadequate response in vivo, with only ∼3% patients demonstrating a complete clinical response. Consequently, we designed a biomimetic nanoparticle (CMNP@Osi) comprising a polymeric nanoparticle core and tumor cell-derived membrane-coated shell that combines membrane-mediated homologous and molecular targeting for targeted drug delivery, thereby supporting a dual-target strategy for enhancing osimertinib efficacy. After intravenous injection, CMNP@Osi accumulates at tumor sites and displays enhanced uptake into cancer cells based on homologous targeting. Osimertinib is subsequently released into the cytoplasm, where it suppresses the phosphorylation of upstream EGFR and the downstream AKT signaling pathway and inhibits the proliferation of NSCLC cells. Thus, this dual-targeting strategy using a biomimetic nanocarrier can enhance molecular-targeted drug delivery and improve clinical efficacy.

17.
Leukemia ; 37(10): 1982-1993, 2023 10.
Article in English | MEDLINE | ID: mdl-37591940

ABSTRACT

TAL1+ T-cell acute lymphoblastic leukemia (T-ALL) is a distinct subtype of leukemia with poor outcomes. Through the cooperation of co-activators, including RUNX1, GATA3, and MYB, the TAL1 oncoprotein extends the immature thymocytes with autonomy and plays an important role in the development of T-ALL. However, this process is not yet well understood. Here, by investigating the transcriptome and prognosis of T-ALL from multiple cohorts, we found that S1PR3 was highly expressed in a subset of TAL1+ T-ALL (S1PR3hi TAL1+ T-ALL), which showed poor outcomes. Through pharmacological and genetic methods, we identified a specific survival-supporting role of S1P-S1PR3 in TAL1+ T-ALL cells. In T-ALL cells, TAL1-RUNX1 up-regulated the expression of S1PR3 by binding to the enhancer region of S1PR3 gene. With hyperactivated S1P-S1PR3, T-ALL cells grew rapidly, partly by activating the KRAS signal. Finally, we assessed S1PR3 inhibitor TY-52156 in T-ALL patient-derived xenografts (PDXs) mouse model. We found that TY-52156 attenuated leukemia progression efficiently and extended the lifespan of S1PR3hi TAL1+ T-ALL xenografts. Our findings demonstrate that S1PR3 plays an important oncogenic role in S1PR3hi TAL1+ T-ALL and may serve as a promising therapeutic target.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Animals , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Core Binding Factor Alpha 2 Subunit/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Thymocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics
18.
Nat Commun ; 13(1): 2038, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440570

ABSTRACT

Developing precise nanomedicines to improve the transport of anticancer drugs into tumor tissue and to the final action site remains a critical challenge. Here, we present a bioorthogonal in situ assembly strategy for prolonged retention of nanomedicines within tumor areas to act as drug depots. After extravasating into the tumor site, the slightly acidic microenvironment induces the exposure of cysteine on the nanoparticle surface, which subsequently undergoes a bioorthogonal reaction with the 2-cyanobenzothiazole group of another neighboring nanoparticle, enabling the formation of micro-sized drug depots to enhance drug retention and enrichment. This in situ nanoparticle assembly strategy remarkably improves the antimetastatic efficacy of extracellular-targeted drug batimastat, and also leads to the simultaneous enhanced retention and sustained release of multiple agents for combined cocktail chemoimmunotherapy to finally elicit a potent antitumor immune response. Such in situ assembly of nanomedicines represents a generalizable strategy towards extracellular drug delivery and cocktail chemoimmunotherapy.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Antineoplastic Agents/pharmacology , Drug Delivery Systems , Drug Liberation , Humans , Nanomedicine , Neoplasms/drug therapy , Neoplasms/pathology , Pharmaceutical Preparations , Tumor Microenvironment
19.
Genes Dis ; 9(6): 1466-1477, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36157504

ABSTRACT

Acute myeloid leukemia (AML) is a malignant hematological tumor with disordered oncogenes/tumor suppressor genes and limited treatments. The potent anti-cancer effects of bromodomain and extra-terminal domain (BET) inhibitors, targeting the key component of super enhancers, in early clinical trials on AML patients, implies the critical role of super enhancers in AML. Here, we review the concept and characteristic of super enhancer, and then summarize the current researches about super enhancers in AML pathogenesis, diagnosis and classification, followed by illustrate the potential super enhancer-related targets and drugs, and propose the future directions of super enhancers in AML. This information provides integrated insight into the roles of super enhancers in this disease.

20.
J Shanghai Jiaotong Univ Sci ; : 1-9, 2022 Dec 26.
Article in English | MEDLINE | ID: mdl-36588799

ABSTRACT

In response to the new round of COVID-19 outbreaks since March 2022, universities with high outbreak rates around the country have taken quarantine measures to contain the epidemic. Evidence from previous coronavirus outbreaks has shown that people under quarantine are at risk for mental health disorders. To better understand the impacts of this round of COVID-19 quarantine on domestic college students and their responses, we conducted a systematic survey to assess the stress and anxiety, and to evaluate effective measurements in this population. We searched relevant documents and literature, and designed a questionnaire from six aspects, including psychological status, epidemic situation, study, daily life, sports, and interpersonal communication, with 51 items in total. We sent the questionnaire on the Wenjuanxing Web platform, from April 2 to 8, 2022. We evaluated the mental status according to parts of the Generalized Anxiety Disorder-7 (GAD-7) and Depression Anxiety Stress Scales-21 (DASS-21), and investigated the influencing risk factors and countermeasures. Statistical analysis was performed by using the Chi-square test and multi-variable logistic regression. In total, 508 college respondents were recruited in our survey, and the pooled prevalence of mild anxiety (GAD score ≽ 5, or DASS-21 anxiety score ≽ 8) or stress (DASS-21 pressure score ≽ 14) caused by the new round of COVID-19 pandemic quarantine was 19.69% (100/508). The prevalence of the anxiety or stress in college students with COVID-19 quarantine between different genders, regions, and majors was not significantly different. Independent risk factors for the mild anxiety or stress of undergraduates by COVID-19 quarantine included learning efficiency or duration [OR = 1.36, 95%CI (1.14-1.62), P = 0.001], based on the combined analysis of Chi-square test analysis with multi-variable logistic regression analysis. Interestingly, the mental well-beings before COVID-19 epidemic quarantine [OR = 0.22, 95%CI (0.13-0.36), P < 0.0001], more low-intensity exercise [OR = 0.36, 95%CI (0.15-0.87), P = 0.02, high-intensity exercise as reference], and good sleep quality [OR = 0.14, 95%CI (0.07-0.30), P < 0.0001: OR = 0.42, 95%CI (0.30-0.59), P < 0.0001] are protective factors for alleviating the quarantine-caused anxiety or stress in Chinese college students for this round of COVID-19 epidemic quarantine. During the round of COVID-19 epidemic quarantine in 2022, a small number of college students have mild anxiety, affected by decreased learning efficiency or duration, which could be mitigated with low-intensity exercise and good sleep quality.

SELECTION OF CITATIONS
SEARCH DETAIL