Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Adv Sci (Weinh) ; : e2402039, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38828705

ABSTRACT

Exploring efficient and low-toxicity radiosensitizers to break through the bottleneck of radiation tolerance, immunosuppression and poor prognosis remains one of the critical developmental challenges in radiotherapy. Nanoheterojunctions, due to their unique physicochemical properties, have demonstrated excellent radiosensitization effects in radiation energy deposition and in lifting tumor radiotherapy inhibition. Herein, they doped selenium (Se) into prussian blue (PB) to construct a nano-heterojunction (Se@PB), which could promote the increase of Fe2+/Fe3+ ratio and conversion of Se to a high valence state with Se introduction. The Fe2+-Se-Fe3+ electron transfer chain accelerates the rate of electron transfer on the surface of the nanoparticles, which in turn endows it with efficient X-ray energy transfer and electron transport capability, and enhances radiotherapy physical sensitivity. Furthermore, Se@PB induces glutathione (GSH) depletion and Fe2+ accumulation through pro-Fenton reaction, thereby disturbs the redox balance in tumor cells and enhances biochemical sensitivity of radiotherapy. As an excellent radiosensitizer, Se@PB effectively enhances X-ray induced mitochondrial dysfunction and DNA damage, thereby promotes cell apoptosis and synergistic cervical cancer radiotherapy. This study elucidates the radiosensitization mechanism of Se-doped nanoheterojunction from the perspective of the electron transfer chain and biochemistry reaction, which provides an efficient and low-toxic strategy in radiotherapy.

2.
Angew Chem Int Ed Engl ; : e202404822, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687056

ABSTRACT

Autophagy could play suppressing role in cancer therapy by facilitating release of tumor antigens from dying cells and inducing immunogenic cell death (ICD). Therefore, discovery and rational design of more effective inducers of cytotoxic autophagy is expected to develop new strategies for finding innovative drugs for precise and successful cancer treatment. Herein, we develop MoO3-x nanowires (MoO3-x NWs) with high oxygen vacancy and strong photothermal responsivity to ablate tumors through hyperthermia, thus promote the induction of cytotoxic autophagy and severe ICD. As expected, the combination of MoO3-x NWs and photothermal therapy (PTT) effectively induces autophagy to promote the release of tumor antigens from the ablated cells, and induces the maturation and antigen presentation of dendritic cells (DCs), subsequently activates cytotoxic T lymphocytes (CTLs)-mediated adaptive immunity. Furthermore, the combination treatment of MoO3-x NWs with immune checkpoint blockade of PD-1 could promote the tumor-associated macrophages (TAMs) polarization into tumor-killing M1 macrophages, inhibit infiltration of Treg cells at tumor sites, and alleviate immunosuppression in the tumor microenvironment, finally intensify the anti-tumor activity in vivo. This study provides a strategy and preliminary elucidation of the mechanism of using MoO3-x nanowires with high oxygen vacancy to induce autophagy and thus enhance photothermal immunotherapy.

3.
Small ; : e2310118, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38506599

ABSTRACT

The combination of ferroptosis and innovative tumor therapy methods offers another promising answer to the problem of tumors. In order to generate effective ferroptosis in tumor cells, iron-based nanomaterials are commonly utilized to introduce foreign iron as a trigger for ferroptosis. However, this usually necessitates the injection of larger doses of iron into the body. These exogenous iron increases are likely to create concealed concerns for symptoms such as liver damage and allergy. Herein, an iron-free radiosensitizer is introduced, oxygen-vacancy-rich MnO2 nanoflowers (ovs-MnO2 ), that promotes ferroptosis and modifies the tumor microenvironment to assist radiotherapy. ovs-MnO2 with enriched oxygen vacancies on the surface induces the release of intracellular free iron (Fe2+ ), which functions as an activator of Fenton reaction and enhances the accumulation of intracellular reactive oxygen species. On the other hand, Fe2+ also triggers the ferroptosis and promotes the accumulation of lipid peroxides. Subsequently, the depletion of glutathione and accumulation of lipid peroxidation in tumor cells leads to the inactivation of glutathione peroxidase 4 (GPX4) and ferroptosis, thereby enhancing the therapeutic efficacy of radiotherapy. The nanoplatform provides a novel strategy for generating novel nanomedicines for ferroptosis-assisted radiotherapy.

4.
Biomaterials ; 303: 122358, 2023 12.
Article in English | MEDLINE | ID: mdl-37951099

ABSTRACT

Scavenging free radicals and reducing inflammatory reaction to relieve the secondary damage are important issues in the spinal cord injury (SCI) therapeutic strategy. Nanozymes attract more attention in the drug development of SCI due to the high stability, long-lasting catalytic capacity, and multienzyme-like properties. Herein, we constructed a Rapamycin (Rapa)-loaded and hollow mesoporous Prussian blue (HMPB)-based nanozyme (RHPAzyme) to realize the combined antioxidation and anti-inflammation combination therapy of SCI. Furthermore, activated cell penetrating peptide (ACPP) is modified onto nanozyme to endow the effectively ability of lesion area-targeting. This RHPAzyme exhibits ROS scavenging capacity with the transformation of Fe2+/Fe3+ valance and cyanide group of HMPB to achieve multienzyme-like activity. As expected, RHPAzyme scavenges the ROS overproduction and reduces inflammation in oxygen-glucose deprivation (OGD)-induced damage via inhibiting MAPK/AKT signaling pathway. Furtherly, RHPAzyme exhibits the combined antioxidant and anti-inflammatory activity in vivo, which can effectively alleviate neuronal damage and promote motor function recovery in SCI mice. Overall, this study demonstrates the RHPAzyme induces an effective treatment of SCI by inhibiting oxygen-mediated cell apoptosis and suppressing inflammation-induced injury, thus reduces the nervous impairment and promotes motor function recovery.


Subject(s)
Sirolimus , Spinal Cord Injuries , Rats , Mice , Animals , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Inflammation/drug therapy , Inflammation/metabolism , Antioxidants/metabolism , Oxygen/metabolism , Spinal Cord/pathology
5.
ACS Nano ; 17(14): 14053-14068, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37428976

ABSTRACT

Easy recurrence and strong treatment side effects significantly limit the clinical treatment of allergic dermatitis. The human trace element selenium (Se) plays essential roles in redox regulation through incorporation into selenoproteins in the form of 21st necessary amino acid selenocysteine, to participates in the pathogenesis and intervention of chronic inflammatory diseases. Therefore, based on the safe and elemental properties of Se, we construct a facile-synthesis strategy for antiallergic selenium nanoparticles (LET-SeNPs), and scale up the production by employing a spray drying method with lactose (Lac-LET-SeNPs) or maltodextrin (Mal-LET-SeNPs) as encapsulation agents realizing larger scale production and a longer storage time. As expected, these as-prepared LET-SeNPs could effectively activate the Nrf2-Keap1 signaling pathway to enhance the expression of antioxidative selenoprotein at mRNA and protein levels, then inhibit mast cell activation to achieve efficient antiallergic activity. Interestingly, LET-SeNPs undergo metabolism to seleno-amino acids to promote biosynthesis of selenoproteins, which could suppress ROS-induced cyclooxygenase-2 (COX-2) and MAPKs activation to suppress the release of histamine and inflammatory cytokines. Allergic mouse and Macaca fascicularis models further confirm that LET-SeNPs could increase the Se content and selenoprotein expression in the skin, decrease mast cells activation and inflammatory cells infiltration, and finally exhibit the high therapeutic effects on allergic dermatitis. Taken together, this study not only constructs facile large-scale synthesis of translational Se nanomedicine to break through the bottleneck problem of nanomaterials but also sheds light on its application in the intervention and treatment of allergies.


Subject(s)
Anti-Allergic Agents , Dermatitis , Nanoparticles , Selenium , Humans , Mice , Animals , Selenium/chemistry , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Selenoproteins/metabolism , Nanoparticles/chemistry , Dermatitis/drug therapy
6.
Stroke Vasc Neurol ; 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37328278

ABSTRACT

BACKGROUND: Nanoparticles (NPs) are a class of substances that can be loaded with therapeutic agents delivered to specific areas. In our earlier research, we identified a neuron-derived circular RNA (circRNA), circular oxoglutarate dehydrogenase (CircOGDH), as a promising therapeutic target for acute ischaemic stroke. This study dedicated to explore a prospective preliminary strategy of CircOGDH-based NP delivered to the ischaemic penumbra region in middle cerebral artery occlusion/reperfusion (MCAO/R) mice. METHODS: Immunofluorescence in primary cortex neurons and in vivo fluorescence imaging revealed endocytosis of Poly(lactide-co-glycolide) (PLGA) poly amidoamine(PAMAM)@CircOGDH small interfering RNA (siRNA) NPs. Western blotting analysis and CCK8 assay were performed to evaluate the apoptotic level in ischaemic neurons treated with PLGA-PAMAM@CircOGDH siRNA NPs. Quantitative reverse transcription PCR experiments, mice behaviour test, T2 MRI analysis, Nissl and TdT-mediated dUTP nick end labeling (TUNEL) co-staining were performed to evaluate the apoptosis level of ischaemic penumbra neurons in MCAO/R mice. Biosafety evaluation of NPs in MCAO/R mice was detected by blood routine examination, liver and kidney function examination and HE staining. RESULTS: PLGA-PAMAM@CircOGDH siRNA NPs were successfully assembled. Endocytosis of PLGA-PAMAM@CircOGDH siRNA NPs in ischaemic neurons alleviated neuronal apoptotic level in vitro and in vivo. Furthermore, mice behaviour test showed that the neurological defects of MCAO/R mice were significantly alleviated after the tail injection of PLGA-PAMAM@CircOGDH siRNA NPs, and no toxic effects were observed. CONCLUSION: In conclusion, our results suggest that PLGA-PAMAM@CircOGDH siRNA NPs can be delivered to the ischaemic penumbra region and alleviate neuron apoptosis in MCAO/R mice and in ischaemic neurons; therefore, our study provides a desirable approach for using circRNA-based NPs for the treatment of ischaemic stroke.

7.
Microorganisms ; 11(2)2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36838291

ABSTRACT

Oilseed rape is sensitive to soil phosphorus deficiencies. In contrast, white lupin is widely used as a model plant because it has efficient phosphorus utilization. Therefore, soil fertility and microbial composition in the rhizospheres of oilseed rapes and root exudate metabolites were compared under monocropping and intercropping systems. The main purpose was to explore whether the phosphorus absorption of rapeseed can be promoted by intercropping with white lupine. In comparison with oilseed rape monoculture (RR), the results showed that the contents of soil-available phosphorus, microbial biomass and phosphorus in the rhizospheres of oilseed rapes in the intercropping system (RL) were all higher than those of RR. Meanwhile, in comparison with RR, not only phosphorus-solubilizing bacteria, such as Streptomyces, Actinomadura and Bacillus, but also phosphorus-solubilizing fungi, such as Chaetomium, Aspergillus, Penicillium, were enriched in the rhizospheres of the oilseed rape under the RL system. Moreover, more abundant soil bacterial functions, organic acids and metabolites were also detected in root exudates of the oilseed rapes under the RL system. All of the above results suggest that soil phosphorus availability in the rhizospheres of oilseed rape could be improved by intercropping with white lupin. Additionally, soil phosphorus-solubilizing microorganisms, that are enriched in the rhizospheres of oilseed rapes under RL systems, have an important function in the improvement of phosphorus absorption of rapeseed by intercropping with white lupin.

8.
Adv Sci (Weinh) ; 10(8): e2202519, 2023 03.
Article in English | MEDLINE | ID: mdl-36683155

ABSTRACT

Low persistence, metabolic dysfunction in microenvironment, and tumor-derived immunosuppression of Natural killer (NK) cells in patients are greatly limited the successful clinical application of NK cell-based cancer immunotherapy. Interestingly, herein that human serum albumin-encapsulated black phosphorus quantum dots (BPQDs@HSA) can effectively augment antitumor efficacy of clinical patients-derived NK cell immunotherapy is found. As the donor of phosphate group, BPQDs@HSA binds with the protein of phosphatidylinositol 4-phosphate 5-kinase type-1 gamma (PIP5K1A) and activates the downstream PI3K-Akt and mTOR signaling pathways to reprogram cell metabolism of glycolysis and further promote the oxidative phosphorylation, sequentially maintains the cell viability and immunity of NK cells. And multiomics analysis is therefore conducted to reveal the underlying immunoregulation mechanisms, and that BPQDs@HSA can interact with the Toll-like receptor (TLR) on the NK cell surface and increase the expression level of mTOR, and thus activate downstream NF-κB signalling pathways to regulate cytokine secretion and enhance immune tumoricidal is found. BPQDs@HSA can also enhance immune surveillance, relieve immune suppression, and inhibit tumor immune escape. Collectively, this study not only demonstrates a successful strategy for nanomedicine-potentiated immune-cancer therapy, but also sheds light on the understanding of interface between nanomedicine and immune cells activation.


Subject(s)
Neoplasms , Quantum Dots , Humans , Phosphorus , Phosphatidylinositol 3-Kinases , Killer Cells, Natural , Immunotherapy , Neoplasms/pathology , TOR Serine-Threonine Kinases , Tumor Microenvironment
9.
Biomater Sci ; 11(1): 339-340, 2022 Dec 20.
Article in English | MEDLINE | ID: mdl-36444841

ABSTRACT

Correction for 'Selenium-driven enhancement of synergistic cancer chemo-/radiotherapy by targeting nanotherapeutics' by Xinxin Liu et al., Biomater. Sci., 2021, 9, 4691-4700, https://doi.org/10.1039/d1bm00348h.

10.
J Nanobiotechnology ; 20(1): 401, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36064356

ABSTRACT

BACKGROUND: Cancer cell membrane-camouflaged nanotechnology for metal complex can enhance its biocompatibility and extend the effective circulation time in body. The ruthenium polypyridyl complex (RuPOP) has extensive antitumor activity, but it still has disadvantages such as poor biocompatibility, lack of targeting, and being easily metabolized by the organism. Cancer cell membranes retain a large number of surface antigens and tumor adhesion molecules CD47, which can be used to camouflage the metal complex and give it tumor homing ability and high biocompatibility. RESULTS: Therefore, this study provides an electrostatic adsorption method, which uses the electrostatic interaction of positive and negative charges between RuPOP and cell membranes to construct a cancer cell membrane-camouflaged nano-platform (RuPOP@CM). Interestingly, RuPOP@CM maintains the expression of surface antigens and tumor adhesion molecules, which can inhibit the phagocytosis of macrophage, reduce the clearance rate of RuPOP, and increase effective circulation time, thus enhancing the accumulation in tumor sites. Besides, RuPOP@CM can enhance the activity of cellular immune response and promote the production of inflammatory cytokines including TNF-α, IL-12 and IL-6, which is of great significance in treatment of tumor. On the other hand, RuPOP@MCM can produce intracellular ROS overproduction, thereby accelerating the apoptosis and cell cycle arrest of tumor cells to play an excellent antitumor effect in vitro and in vivo. CONCLUSION: In brief, engineering cancer cell membrane-camouflaged metal complex is a potential strategy to improve its biocompatibility, biological safety and antitumor effects.


Subject(s)
Breast Neoplasms , Ruthenium , Antigens, Surface/metabolism , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Membrane/metabolism , Female , Humans
11.
Elife ; 112022 08 15.
Article in English | MEDLINE | ID: mdl-35969037

ABSTRACT

Knockout (KO) mouse models play critical roles in elucidating biological processes behind disease-associated or disease-resistant traits. As a presumed consequence of gene KO, mice display certain phenotypes. Based on insight into the molecular role of said gene in a biological process, it is inferred that the particular biological process causally underlies the trait. This approach has been crucial towards understanding the basis of pathological and/or advantageous traits associated with Mertk KO mice. Mertk KO mice suffer from severe, early-onset retinal degeneration. MERTK, expressed in retinal pigment epithelia, is a receptor tyrosine kinase with a critical role in phagocytosis of apoptotic cells or cellular debris. Therefore, early-onset, severe retinal degeneration was described to be a direct consequence of failed MERTK-mediated phagocytosis of photoreceptor outer segments by retinal pigment epithelia. Here, we report that the loss of Mertk alone is not sufficient for retinal degeneration. The widely used Mertk KO mouse carries multiple coincidental changes in its genome that affect the expression of a number of genes, including the Mertk paralog Tyro3. Retinal degeneration manifests only when the function of Tyro3 is concomitantly lost. Furthermore, Mertk KO mice display improved anti-tumor immunity. MERTK is expressed in macrophages. Therefore, enhanced anti-tumor immunity was inferred to result from the failure of macrophages to dispose of cancer cell corpses, resulting in a pro-inflammatory tumor microenvironment. The resistance against two syngeneic mouse tumor models observed in Mertk KO mice is not, however, phenocopied by the loss of Mertk alone. Neither Tyro3 nor macrophage phagocytosis by alternate genetic redundancy accounts for the absence of anti-tumor immunity. Collectively, our results indicate that context-dependent epistasis of independent modifier alleles determines Mertk KO traits.


Subject(s)
Retinal Degeneration , Alleles , Animals , Disease Models, Animal , Mice , Mice, Knockout , Phagocytosis/genetics , Phenotype , Proto-Oncogene Proteins/genetics , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Pigments , c-Mer Tyrosine Kinase/genetics , c-Mer Tyrosine Kinase/metabolism
12.
Ren Fail ; 44(1): 1227-1235, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35848372

ABSTRACT

BACKGROUND: The systemic inflammatory response index (SIRI), a novel inflammation maker, has proven to be associated with prognostic outcomes in various diseases. However, few studies have been conducted assessing how SIRI may influence outcomes of patients on peritoneal dialysis (PD). Herein, we assessed the predictive value of SIRI on mortality all-cause mortality, including cardiovascular disease (CVD) in PD patients. METHODS: A total of 646 PD patients were enrolled in this study. PD patients received regular PD treatments at the Zhujiang Hospital from 1 January 2011 to 31 December 2018. SIRI values could be computed as follows: neutrophil count × monocyte count/lymphocyte count. Patients were divided into two groups according to the median level of SIRI. Cox regression analysis and Kaplan-Meier methods were applied to analyze the relationship between SIRI and mortality outcomes in PD patients. RESULTS: During the median 31-month follow-up period, 97 (15.0%) PD patients died from all-causes, and 47 (49.0%) died of CVD. Kaplan-Meier analyses revealed that a high SIRI corresponded to the high mortality of all-cause deaths, including CVD (both p < 0.001) in patients on PD. After adjusting for potential confounders, the higher SIRI level was significantly associated with an increased all-cause mortality (HR: 2.007, 95% CI: 1.304-3.088, p = 0.002) and cardiovascular mortality (HR: 2.847, 95% CI: 1.445-5.608, p = 0.002). CONCLUSIONS: SIRI was a promising predictor of mortality in PD patients, with a higher SIRI corresponding to increased risk of mortality.


Subject(s)
Cardiovascular Diseases , Peritoneal Dialysis , Cardiovascular Diseases/etiology , Humans , Inflammation , Prognosis , Retrospective Studies
13.
Biomaterials ; 287: 121620, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35709558

ABSTRACT

Disrupting redox homeostasis in the tumor microenvironment (TME), like excessive H2O2, glutathione (GSH) and weak acidity, has been proved as an effective tumor therapeutic strategy. Herein, we constructed a TME-responsive nanozyme, DOX@HMSN/Mn3O4(R), with reversible Mn3+/Mn2+ transition in situ triggered by TME to perturb the intrinsic redox homeostasis and catalyze reactive oxygen species (ROS) overproduction. In addition, this nanozyme could react with excess GSH in TME to produce GSSG, resulting in the consumption of reducing agents to suppress ROS clearance. Density functional theory calculations further confirmed that the nanozyme mainly exhibited the oxidase-like activity to catalyze the formation of hydroxyl radicals from O2, thus strengthening the oxidation environment in the TME. Combined with radiotherapy, the high-energy X-ray could excite the outer-layer electrons in the nanozyme, forming photoelectrons that participate in the oxidase-like enzymatic reaction, thus intensifying ROS accumulation and amplifying the radio-/chemotherapeutic efficacy.

14.
ACS Appl Mater Interfaces ; 14(24): 27651-27665, 2022 Jun 22.
Article in English | MEDLINE | ID: mdl-35675569

ABSTRACT

Rapid glycolysis of tumor cells produces excessive lactate to trigger acidification of the tumor microenvironment (TME), leading to the formation of immunosuppressive TME and tumor-associated macrophage (TAM) dysfunction. Therefore, reprogramming TAMs by depleting lactate with nanodrugs is expected to serve as an effective means of tumor-targeted immunotherapy. Herein, we report the use of lactic acid dehydrogenase (LDH)-mimicking SnSe nanosheets (SnSe NSs) loaded with a carbonic anhydrase IX (CAIX) inhibitor to reconstruct an acidic and immunosuppressive TME. As expected, this nanosystem could reprogram the TAM to achieve M1 macrophage activation and could also restore the potent tumor-killing activity of macrophages while switching their metabolic mode from mitochondrial oxidative phosphorylation to glycolysis. In addition, the repolarizing effect of SnSe NSs on macrophages was validated in a coculture model of bone marrow-derived macrophages, in three patient-derived malignant pleural effusion and in vivo mouse model. This study proposes a feasible therapeutic strategy for depleting lactate and thus ameliorating acidic TME employing Se-containing nanosheets, which could further amply the effects of TAM-based antitumor immunotherapy.


Subject(s)
Neoplasms , Tumor-Associated Macrophages , Animals , Humans , Immunotherapy , L-Lactate Dehydrogenase , Lactic Acid/metabolism , Mice , Neoplasms/pathology , Tumor Microenvironment
15.
Biomaterials ; 285: 121549, 2022 06.
Article in English | MEDLINE | ID: mdl-35567998

ABSTRACT

Bone metastasis is the major cause of cancer-related morbidity and mortality. To avoid further osteolysis, current treatment ideas focus on tumor cell and the inhibition of osteoclast. Herein, zeolitic imidazolate framework-8-capped Cu2-XSe composite nanoplatform (ICG@Cu2-XSe-ZIF-8) is developed for chemodynamic therapy (CDT) and photothermal therapy (PTT) treatment of malignant breast cancer bone tumors. The rational design of ZIF-8 encapsulation greatly reduces the accumulation of Cu2-XSe to damage the normal cells. Under acidic microenvironment in tumor, ZIF-8 is cleaved to release Cu2-XSe, which can subsequently degrade into Cu (+) and Cu (2+) ions to initiate a Fenton-like reaction inducing CDT. Meanwhile, Cu2-XSe is used to be an effective photothermal transduction agent for exerting the PTT effect. What's more, the selenium element in Cu2-XSe can regulates selenoprotein to inhibit tumor cells and osteoclasts. Of note, the hyperthermia induced by PTT can further enhance the CDT effect in tumor, achieving a synergistic PTT/CDT effect. Based on these advantages, ICG@Cu2-XSe-ZIF-8 effectively suppresses the tumor cells in bone tissue, and reduces the erosion of bone tissue via suppressing osteoclastogenesis. In conclusion, this study demonstrates the potential action mechanism of ZIF-8-capped nanomedicine against osteolysis in bone metastasis.


Subject(s)
Bone Neoplasms , Breast Neoplasms , Hyperthermia, Induced , Metal-Organic Frameworks , Nanoparticles , Osteolysis , Bone Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Humans , Metal-Organic Frameworks/therapeutic use , Osteogenesis , Osteolysis/drug therapy , Tumor Microenvironment
16.
ESC Heart Fail ; 9(4): 2399-2406, 2022 08.
Article in English | MEDLINE | ID: mdl-35419980

ABSTRACT

AIM: Current study was to evaluate relationship between baseline serum lipoprotein (a) [Lp(a)] level and prognosis in patients with heart failure with reduced ejection fraction (HFrEF) and to explore whether the relationship would be modified by baseline high-sensitivity C-reactive protein (Hs-CRP) level. METHODS AND RESULTS: This is an observational prospective study. HFrEF patients from outpatient clinic were consecutively recruited (n = 362). Based on Lp(a) cutoff (30 mg/dL), patients were divided into normal and high Lp(a) groups; and based on Hs-CRP cutoff (3 mg/dL), patients were divided into low-degree and high-degree groups. The 1 year rate of HF rehospitalization was similar between these two groups (22.7% vs. 24.1%, P = 0.18), while the 1 year rate of cardiovascular mortality was higher in Lp(a) ≥ 30 mg/dL versus Lp(a) < 30 mg/dL groups (20.3% vs. 13.3%, P = 0.009), as was composite endpoint (44.4% vs. 36.0%, P < 0.001). After adjusting for covariates, elevated Lp(a) level remained associated with a higher risk of cardiovascular mortality [hazard ratio (HR) 1.22 and 95% confidence interval (CI) 1.04-1.64, P = 0.02] and composite endpoint (HR 1.38 and 95% CI 1.16-2.01, P = 0.006). In Hs-CRP ≥ 3 mg/dL group, elevated Lp(a) level was associated with HF rehospitalization, cardiovascular mortality, and composite endpoint, which was not observed in Hs-CRP < 3 mg/dL group. The association was greater for cardiovascular mortality (P-interaction = 0.04) and composite endpoint (P-interaction = 0.02) in Hs-CRP ≥ 3 mg/dL versus Hs-CRP < 3 mg/dL groups. CONCLUSION: Elevated Lp(a) level is associated with higher risk of cardiovascular mortality in HFrEF patients, which might be due to enhanced systemic inflammation.


Subject(s)
Heart Failure , Ventricular Dysfunction, Left , C-Reactive Protein , Heart Failure/epidemiology , Humans , Lipoprotein(a) , Prognosis , Prospective Studies , Stroke Volume
17.
Dalton Trans ; 51(12): 4836-4842, 2022 Mar 22.
Article in English | MEDLINE | ID: mdl-35257123

ABSTRACT

We report here a simple but efficient "ship-in-a-bottle" synthetic strategy for increasing the stability and luminescence performance of LOPs by embedding them into mesoporous silica nanoparticles (MSNs). Three types of hybrid materials, i.e. Eu8L12@MSNs, Eu8L12@MSNs-NH2 and Eu8L12@MSNs-biotin, have been prepared and characterized by FT-IR, TGA, SEM and TEM. Photo-optical measurements confirmed that the photoluminescence quantum yields in water have been greatly improved from 5.50% for pristine Eu8L12 to 44.04% for Eu8L12@MSNs-biotin, along with fast disassembly for the former and the optical performance has been maintained for the latter under acidic conditions (pH = 4). Moreover, compared to Eu8L12, Eu8L12@MSNs and Eu8L12@MSNs-NH2, the biotin-modified hybrid material Eu8L12@MSNs-biotin has exhibited much enhanced fluorescence-imaging ability toward the MDA-MB-231 human breast cancer cells, with significantly reduced dosage of the complex. Our work provides a useful strategy for the functionalization of multinuclear lanthanide organic assemblies toward their biosocial applications.


Subject(s)
Coordination Complexes/chemistry , Lanthanoid Series Elements/chemistry , Luminescence , Nanoparticles/chemistry , Optical Imaging , Silicon Dioxide/chemistry , Cell Line, Tumor , Humans , Particle Size , Porosity , Surface Properties
18.
Biomaterials ; 281: 121371, 2022 02.
Article in English | MEDLINE | ID: mdl-35063740

ABSTRACT

Discovery of effective chemical sensitizers to synergize with natural killer cells immunotherapy is urgently desired to overcome its unsatisfactory efficacy in clinic. Herein, we design a series of ruthenium (Ru) polypyridyl complex to systematically explore their potentials in facilitating NK cells treatment. Intriguingly, the chemical structure greatly determines the activity of Ru complexes, while only RuPOP effectively regulates the immuno-suppressors and target proteins within tumor cells. This unique property contributes to its good capability in enhancing the sensitivity of MDA-MB-231 cells to NK cells from cancer patients. Furthermore, besides directly damaging tumor cells, RuPOP pretreatment together with NK cells can also induce robust ROS generation, activate multiple apoptosis-related receptors like TNF-R1, DR5, Fas and maximize the interactions between NK and tumor cells via up-regulating NKG2D and its multiple ligands to trigger caspase 3-dependent apoptosis. Moreover, the combination treatment exhibits high in vivo therapeutic efficacy against breast tumor through boosting the infiltration of NK cells and reducing the protumoral capability of myeloid-derived suppressor cells (MDSC). This study sheds lights for designing metal complexes to potentiate NK cells immunotherapy with clear action mechanisms and provides important information for developing more effective adoptive cell transfer therapy in clinic.


Subject(s)
Ruthenium , Triple Negative Breast Neoplasms , Cell Line, Tumor , Humans , Immunotherapy , Immunotherapy, Adoptive , Killer Cells, Natural , Ruthenium/pharmacology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/therapy
19.
Cancer Immunol Immunother ; 71(1): 97-109, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34028568

ABSTRACT

Cyclophosphamide plus fludarabine (C/F) are currently used to improve the expansion and effectiveness of adoptive cell therapy (ACT). However, these chemotherapeutics cause pan-leukopenia and adverse events, suggesting that safer and more effective conditioning treatments are needed to improve ACT outcomes. Previously, we reported that varlilumab, a CD27-targeting antibody, mediates Treg -preferential T cell depletion, CD8-T cell dominant costimulation, and systemic immune activation in hCD27 transgenic mice and cancer patients. We reasoned that the activities induced by varlilumab may provide an effective conditioning regimen for ACT. Varlilumab pretreatment of hCD27 +/+mCD27 - /- mice resulted in prominent proliferation of transferred T cells isolated from wild-type mice. These studies uncovered a critical role for CD27 signaling for the expansion of transferred T cells, as transfer of T cells from CD27 deficient mice or treatment with a CD70 blocking antibody greatly reduced their proliferation. In this model, varlilumab depletes endogenous hCD27+/+ T cells and blocks their subsequent access to CD70, allowing for more CD70 costimulation available to the mCD27 +/+ transferred T cells. CD27-targeted depletion led to a greater expansion of transferred T cells compared to C/F conditioning and resulted in longer median survival and more cures than C/F conditioning in the E.G7 tumor model receiving OT-I cell therapy. We propose that translation of this work could be achieved through engineering of T cells for ACT to abrogate varlilumab binding but preserve CD70 ligation. Thus, varlilumab could be an option to chemotherapy as a conditioning regimen for ACT.


Subject(s)
Adoptive Transfer , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal/chemistry , Neoplasms/therapy , T-Lymphocytes/cytology , Tumor Necrosis Factor Receptor Superfamily, Member 7/chemistry , Animals , CD27 Ligand/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Line, Tumor , Cell Proliferation , Immune System , Immunotherapy , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neoplasms/metabolism , Signal Transduction , Transplantation Conditioning , Treatment Outcome
20.
ACS Nano ; 16(1): 431-452, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-34958560

ABSTRACT

Designing translational antioxidative agents that could scavenge free radicals produced during reperfusion in brain ischemia stroke and alleviate neurologic damage is the main objective for ischemic stroke treatment. Herein, we explored and simply synthesized a biomimic and translational Mn3O4 nanoenzyme (HSA-Mn3O4) to constrain ischemic stroke reperfusion-induced nervous system injury. This nanosystem exhibits reduced levels of inflammation and prolonged circulation time and potent ROS scavenging activities. As expected, HSA-Mn3O4 effectively inhibits oxygen and glucose deprivation-mediated cell apoptosis and endoplasmic reticulum stress and demonstrates neuroprotective capacity against ischemic stroke and reperfusion injury of brain tissue. Furthermore, HSA-Mn3O4 effectively releases Mn ions and promotes the increase of superoxide dismutase 2 activity. Therefore, HSA-Mn3O4 inhibits brain tissue damage by restraining cell apoptosis and endoplasmic reticulum stress in vivo. Taken together, this study not only sheds light on design of biomimic and translational nanomedicine but also reveals the neuroprotective action mechanisms against ischemic stroke and reperfusion injury.


Subject(s)
Brain Ischemia , Ischemic Stroke , Neuroprotective Agents , Reperfusion Injury , Stroke , Humans , Endoplasmic Reticulum Stress , Ischemic Stroke/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Reperfusion Injury/drug therapy , Brain Ischemia/drug therapy , Neurons , Stroke/drug therapy , Apoptosis , Antioxidants/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...