Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 526
Filtrer
1.
Adv Drug Deliv Rev ; : 115458, 2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-39383997

RÉSUMÉ

Emerging studies have disclosed the pivotal role of cancer-associated microbiota in supporting cancer development, progression and dissemination, with the in-depth comprehending of tumor microenvironment. In particular, certain invasive bacteria that hide in various cells within the tumor tissues can render assistance to tumor growth and invasion through intricate mechanisms implicated in multiple branches of cancer biology. Thus, tumor-resident intracellular microbes are anticipated as next-generation targets for oncotherapy. This review is intended to delve into these internalized bacteria-driven cancer-promoting mechanisms and explore diversified antimicrobial therapeutic strategies to counteract the detrimental impact caused by these intruders, thereby improving therapeutic benefit of antineoplastic therapy.

2.
J Immunother Cancer ; 12(9)2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39242117

RÉSUMÉ

BACKGROUND: In allogeneic-hematopoietic stem cell transplantation for acute myeloid leukemia (AML), donor T cells combat leukemia through the graft-versus-leukemia (GVL) effect, while they also pose a risk of triggering life-threatening graft-versus-host disease (GVHD) by interacting with recipient cells. The onset of GVHD hinges on the interplay between donor T cells and recipient antigen-presenting cells (APCs), sparking T-cell activation. However, effective methods to balance GVHD and GVL are lacking. METHODS: In our study, we crafted nanocapsules by layering polycationic aminated gelatin and polyanionic alginate onto the surface of T cells, examining potential alterations in their fundamental physiological functions. Subsequently, we established an AML mouse model and treated it with transplantation of bone marrow cells (BMCs) combined with encapsulated T cells to investigate the GVL and anti-GVHD effects of encapsulated T cells. In vitro co-culture was employed to probe the effects of encapsulation on immune synapses, co-stimulatory molecules, and tumor-killing pathways. RESULTS: Transplantation of BMCs combined with donor T cells selectively encapsulated onto AML mice significantly alleviates GVHD symptoms while preserving essential GVL effects. Encapsulated T cells exerted their immunomodulatory effects by impeding the formation of immune synapses with recipient APCs, thereby downregulating co-stimulatory signals such as CD28-CD80, ICOS-ICOSL, and CD40L-CD40. Recipient mice receiving encapsulated T-cell transplantation exhibited a marked increase in donor Ly-5.1-BMC cell numbers, accompanied by unaltered in vivo expression levels of perforin and granzyme B. While transient inhibition of donor T-cell cytotoxicity in the tumor microenvironment was observed in vitro following single-cell nanoencapsulation, subsequent restoration to normal antitumor activity ensued, attributed to selective permeability of encapsulated vesicle shells and material degradation. Moreover, the expression of apoptotic proteins and FAS-FAS ligand pathway at normal levels was still observed in leukemia tumor cells. CONCLUSIONS: Encapsulated donor T cells effectively mitigate GVHD while preserving the GVL effect by minimizing co-stimulatory signaling with APCs through early immune isolation. Subsequent degradation of nanocapsules restores T-cell cytotoxic efficacy against AML cells, mediated by cytotoxic pathways. Using transplant-encapsulated T cells offers a promising strategy to suppress GVHD while preserving the GVL effect.


Sujet(s)
Modèles animaux de maladie humaine , Maladie du greffon contre l'hôte , Leucémie aigüe myéloïde , Lymphocytes T , Animaux , Leucémie aigüe myéloïde/thérapie , Leucémie aigüe myéloïde/immunologie , Souris , Maladie du greffon contre l'hôte/immunologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Humains , Réaction du greffon contre la leucémie , Nanocapsules/composition chimique
3.
Nano Lett ; 24(38): 11976-11984, 2024 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-39270053

RÉSUMÉ

Elevated production of extracellular matrix (ECM) in tumor stroma is a critical obstacle for drug penetration. Here we demonstrate that ATP-citrate lyase (ACLY) is significantly upregulated in cancer-associated fibroblasts (CAFs) to produce tumor ECM. Using a self-assembling nanoparticle-design approach, a carrier-free nanoagent (CFNA) is fabricated by simply assembling NDI-091143, a specific ACLY inhibitor, and doxorubicin (DOX) or paclitaxel (PTX), the first-line chemotherapeutic drug, via multiple noncovalent interactions. After arriving at the CAFs-rich tumor site, NDI-091143-mediated ACLY inhibition in CAFs can block the de novo synthesis of fatty acid, thereby dampening the fatty acid-involved energy metabolic process. As the lack of enough energy, the energetic CAFs will be in a dispirited state that is unable to produce abundant ECM, thereby significantly improving drug perfusion in tumors and enhancing the efficacy of chemotherapy. Such a simple drug assembling strategy aimed at CAFs' ACLY-mediated metabolism pathway presents the feasibility of stromal matrix reduction to potentiate chemotherapy.


Sujet(s)
ATP citrate (pro-S)-lyase , Fibroblastes associés au cancer , Doxorubicine , Paclitaxel , Fibroblastes associés au cancer/effets des médicaments et des substances chimiques , Fibroblastes associés au cancer/métabolisme , Fibroblastes associés au cancer/anatomopathologie , Doxorubicine/pharmacologie , Doxorubicine/composition chimique , Humains , Paclitaxel/pharmacologie , Paclitaxel/usage thérapeutique , Animaux , Souris , ATP citrate (pro-S)-lyase/métabolisme , ATP citrate (pro-S)-lyase/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Nanoparticules/composition chimique , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques
4.
Adv Mater ; : e2408473, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39212208

RÉSUMÉ

Treatment of osteomyelitis is clinically challenging with low therapeutic efficacy and high risk of recurrence owing to the immunosuppressive microenvironment. Existing therapies are limited by drug concentration and single regulatory effect on the immune network, and emphasize the role of anti-inflammatory effects in reducing osteoclast rather than the role of proinflammatory effects in accelerating infection clearance, which is not conducive to complete bacteria elimination and recurrence prevention. Herein, a direct-current triboelectric nanogenerator (DC-TENG) is established to perform antibacterial effects and modulate immunological properties of infectious microenvironments of osteomyelitis through electrical stimulation, namely triboelectric immunotherapy. Seeing from the results, the triboelectric immunotherapy successfully activates polarization to proinflammatory (M1) macrophages in vitro, accompanied by satisfying direct antibacterial effects. The antibacterial and osteogenic abilities of triboelectric immunotherapy are verified in rat cranial osteomyelitis models. The effects on the polarization and differentiation of immune-related cells in vivo are investigated by establishing in situ tibial osteomyelitis models and immunosurveillance models in C57 mice respectively, indicating the ability of activating immunity and producing immunological memory for in situ infection and secondary recurrence, thus accelerating healing and preventing relapse. This study provides an efficient, long-acting, multifunctional, and wearable triboelectric immunotherapy strategy for drug-free osteomyelitis treatment systems.

5.
Parasitol Res ; 123(8): 302, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39158739

RÉSUMÉ

Penicillidia dufourii (Westwood 1834) is a specialized parasite categorized under family Nycteribiidae that prefers to parasitize the body surface of various bats under the genus Myotis. Many species of the family Nycteribiidae are carriers of various pathogens; however, research on P. dufourii remains scarce, and studies on its molecular identification and population genetic structure are still lacking. In this study, the complete mitochondrial genome of P. dufourii was elucidated for the first time using Illumina sequencing. The mitochondrial genome is 15,354 bp in size and encodes approximately 37 genes, including 13 protein-coding genes, 22 tRNA genes, 2 rRNA genes, and 1 control region. Analysis of 13 protein-coding genes revealed that UUA, UCA, CGA, and GGA were the most common codons, while nad4L had the fastest evolutionary rate and cox1 the slowest. Phylogenetic analysis based on the mitochondrial genome indicated that P. dufourii is clustered with other species of the family Nycteribiidae and is most closely related to Nycteribia parvula and Phthiridium szechuanum.


Sujet(s)
Diptera , Génome mitochondrial , Phylogenèse , Animaux , Génome mitochondrial/génétique , Diptera/génétique , Diptera/classification , Analyse de séquence d'ADN
6.
ACS Nano ; 18(34): 23497-23507, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39146387

RÉSUMÉ

Colorectal cancer (CRC) is a major global health concern, and the development of effective treatment strategies is crucial. Enzyme prodrug therapy (EPT) shows promise in combating tumors but faces challenges in achieving sustained expression of therapeutic enzymes and optimal biological distribution. To address these issues, a fungi-triggered in situ chemotherapeutics generator (named as SC@CS@5-FC) was constructed via oral delivery of a prodrug (5-fluorocytosine, 5-FC) for the treatment of orthotopic colorectal tumor. When SC@CS@5-FC targets the tumor through tropism by Saccharomyces cerevisiae (SC), the chemotherapeutic generator could be degraded under abundant hyaluronidase (HAase) in the tumor microenvironment by an enzyme-responsive gate to release prodrug (5-FC). And nontoxic 5-FC was catalyzed to toxic chemotherapy drug 5-fluorouracil (5-FU) by cytosine deaminase (CD) of SC. Meanwhile, SC and zinc-coordinated chitosan nanoparticles could be used as immune adjuvants to activate antigen-presenting cells and further enhance the therapeutic effect. Our results demonstrated that SC@CS@5-FC could effectively inhibit tumor growth and prolong mouse survival in an orthotopic colorectal cancer model. This work utilizes living SC as a dynamotor and positioning system for the chemotherapeutic generator SC@CS@5-FC, providing a strategy for oral enzyme prodrug therapy for the treatment of orthotopic colorectal.


Sujet(s)
Tumeurs colorectales , Flucytosine , Fluorouracil , Immunothérapie , Promédicaments , Saccharomyces cerevisiae , Promédicaments/composition chimique , Promédicaments/pharmacologie , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Animaux , Souris , Humains , Flucytosine/pharmacologie , Flucytosine/composition chimique , Administration par voie orale , Fluorouracil/pharmacologie , Fluorouracil/composition chimique , Fluorouracil/administration et posologie , Cytosine deaminase/métabolisme , Chitosane/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Hyaluronoglucosaminidase/métabolisme , Souris de lignée BALB C , Nanoparticules/composition chimique , Tests de criblage d'agents antitumoraux
7.
Nano Lett ; 24(33): 10362-10371, 2024 Aug 21.
Article de Anglais | MEDLINE | ID: mdl-39133195

RÉSUMÉ

Adoptive cell therapies for solid tumors are usually limited by off-target antigens, incapable tissue infiltration, and cell function exhaustion. In contrast, bacterial cells possess the inherent competencies of preferential tumor targeting, deep tissue penetration, and high intratumoral bioactivity and represent promising alternatives to overcome these challenges. Here, a sialic-acid-responsive regulatory gene circuit is engineered into Escherichia coli MG1655 to express cytolysin of hemolysin E (HlyE). Furthermore, sialidases are bioorthogonally decorated onto the surface of azido-functionalized bioengineered bacteria for recognizing tumor sialoglycans and cleaving their sialosides into free sialic acids. As chemical inducers, sialic acids feedbackingly activate the bacterial gene circuit to produce HlyE and lyse tumor cells. This study mimics the tumor antigen-induced cytotoxin production and cell lysis that occurs in chimeric antigen receptor T (CAR-T) cells yet surmounts the intrinsic limitations of adoptive cell therapies. Moreover, sialidase-mediated tumor cell desialylation also reverses the immunosuppressive effect of glycoimmune checkpoints and further improves the therapeutic effect of solid tumors.


Sujet(s)
Escherichia coli , Tumeurs , Sialidase , Sialidase/génétique , Sialidase/métabolisme , Humains , Escherichia coli/génétique , Animaux , Tumeurs/thérapie , Souris , Lignée cellulaire tumorale , Hémolysines/composition chimique , Récepteurs chimériques pour l'antigène/immunologie , Immunothérapie adoptive
8.
Nat Commun ; 15(1): 7096, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39154092

RÉSUMÉ

The intratumor microbiome imbalance in pancreatic cancer promotes a tolerogenic immune response and triggers immunotherapy resistance. Here we show that Lactobacillus rhamnosus GG probiotics, outfitted with a gallium-polyphenol network (LGG@Ga-poly), bolster immunotherapy in pancreatic cancer by modulating microbiota-immune interactions. Upon oral administration, LGG@Ga-poly targets pancreatic tumors specifically, and selectively eradicates tumor-promoting Proteobacteria and microbiota-derived lipopolysaccharides through a gallium-facilitated disruption of bacterial iron respiration. This elimination of intratumor microbiota impedes the activation of tumoral Toll-like receptors, thus reducing immunosuppressive PD-L1 and interleukin-1ß expression by tumor cells, diminishing immunotolerant myeloid populations, and improving the infiltration of cytotoxic T lymphocytes in tumors. Moreover, LGG@Ga-poly hampers pancreatic tumor growth in both preventive and therapeutic contexts, and amplifies the antitumor efficacy of immune checkpoint blockade in preclinical cancer models in female mice. Overall, we offer evidence that thoughtfully designed biomaterials targeting intratumor microbiota can efficaciously augment immunotherapy for the challenging pancreatic cancer.


Sujet(s)
Gallium , Lacticaseibacillus rhamnosus , Microbiote , Tumeurs du pancréas , Polyphénols , Probiotiques , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/microbiologie , Animaux , Probiotiques/administration et posologie , Souris , Femelle , Humains , Lacticaseibacillus rhamnosus/immunologie , Polyphénols/pharmacologie , Microbiote/immunologie , Microbiote/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Immunothérapie/méthodes , Souris de lignée C57BL , Antigène CD274/métabolisme , Antigène CD274/immunologie , Lymphocytes T cytotoxiques/immunologie
9.
ACS Nano ; 18(32): 21077-21090, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39088785

RÉSUMÉ

Porphyromonas gingivalis has been demonstrated to have the strongest association with periodontitis. Within the host, P. gingivalis relies on acquiring iron and heme through the aggregation and lysis of erythrocytes, which are important factors in the growth and virulence of P. gingivalis. Additionally, the excess obtained heme is deposited on the surface of P. gingivalis, protecting the cells from oxidative damage. Based on these biological properties of the interaction between P. gingivalis and erythrocytes, this study developed an erythrocyte membrane nanovesicle loaded with gallium porphyrins to mimic erythrocytes. The nanovesicle can target and adhere with P. gingivalis precisely, being lysed and utilized by P. gingivalis as erythrocytes. Ingested gallium porphyrin replaces iron porphyrin in P. gingivalis, causing intracellular metabolic disruption. Deposited porphyrin generates a large amount of reactive oxygen species (ROS) under blue light, causing oxidative damage, and its lethality is enhanced by bacterial metabolic disruption, synergistically killing P. gingivalis. Our results demonstrate that this strategy can target and inhibit P. gingivalis, reduce its invasion of epithelial cells, and alleviate the progression of periodontitis.


Sujet(s)
Érythrocytes , Parodontite , Porphyrines , Porphyromonas gingivalis , Porphyromonas gingivalis/effets des médicaments et des substances chimiques , Porphyromonas gingivalis/métabolisme , Porphyromonas gingivalis/composition chimique , Parodontite/microbiologie , Parodontite/traitement médicamenteux , Parodontite/anatomopathologie , Érythrocytes/effets des médicaments et des substances chimiques , Érythrocytes/métabolisme , Humains , Porphyrines/composition chimique , Porphyrines/pharmacologie , Animaux , Espèces réactives de l'oxygène/métabolisme , Gallium/composition chimique , Gallium/pharmacologie , Souris , Antibactériens/pharmacologie , Antibactériens/composition chimique , Matériaux biomimétiques/composition chimique , Matériaux biomimétiques/pharmacologie
10.
Adv Mater ; 36(36): e2405673, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39022876

RÉSUMÉ

Immunogenic cell death (ICD) often results in the production and accumulation of adenosine (ADO), a byproduct that negatively impacts the therapeutic effect as well as facilitates tumor development and metastasis. Here, an innovative strategy is elaborately developed to effectively activate ICD while avoiding the generation of immunosuppressive adenosine. Specifically, ZIF-90, an ATP-responsive consumer, is synthesized as the core carrier to encapsulate AB680 (CD73 inhibitor) and then coated with an iron-polyphenol layer to prepare the ICD inducer (AZTF), which is further grafted onto prebiotic bacteria via the esterification reaction to obtain the engineered biohybrid (Bc@AZTF). Particularly, the designed Bc@AZTF can actively enrich in tumor sites and respond to the acidic tumor microenvironment to offload AZTF nanoparticles, which can consume intracellular ATP (iATP) content and simultaneously inhibit the ATP-adenosine axis to reduce the accumulation of adenosine, thereby alleviating adenosine-mediated immunosuppression and strikingly amplifying ICD effect. Importantly, the synergy of anti-PD-1 (αPD-1) with Bc@AZTF not only establishes a collaborative antitumor immune network to potentiate effective tumoricidal immunity but also activates long-lasting immune memory effects to manage tumor recurrence and rechallenge, presenting a new paradigm for ICD treatment combined with adenosine metabolism.


Sujet(s)
Adénosine triphosphate , Adénosine , Mort cellulaire immunogène , Immunothérapie , Adénosine/composition chimique , Adénosine triphosphate/métabolisme , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Souris , Animaux , Lignée cellulaire tumorale , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/anatomopathologie , Nanoparticules/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique
11.
ACS Biomater Sci Eng ; 10(8): 4701-4715, 2024 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-38959418

RÉSUMÉ

The utilization of traditional therapies (TTS), such as chemotherapy, reactive oxygen species-based therapy, and thermotherapy, to induce immunogenic cell death (ICD) in tumor cells has emerged as a promising strategy for the activation of the antitumor immune response. However, the limited effectiveness of most TTS in inducing the ICD effect of tumors hinders their applications in combination with immunotherapy. To address this challenge, various intelligent strategies have been proposed to strengthen the immune activation effect of these TTS, and then achieve synergistic antitumor efficacy with immunotherapy. These strategies primarily focus on augmenting the tumor ICD effect or facilitating the antigen (released by the ICD tumor cells) presentation process during TTS, and they are systematically summarized in this review. Finally, the existing bottlenecks and prospects of TTS in the application of tumor immune regulation are also discussed.


Sujet(s)
Immunothérapie , Tumeurs , Humains , Tumeurs/immunologie , Tumeurs/thérapie , Immunothérapie/méthodes , Animaux , Mort cellulaire immunogène/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme
12.
Molecules ; 29(13)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38999079

RÉSUMÉ

Transition-metal-based oxygen evolution reaction (OER) catalysts have attracted widespread attention due to their inexpensive prices, unique layered structures, and rich active sites. Currently, designing low-cost, sustainable, and simple synthesis methods is essential for the application of transition-metal-based catalysts. Here, magnetic field (MF)-assisted chemical corrosion, as a novel technology, is adopted to construct superior OER electrocatalysts. The produced Ni(Fe)(OH)2-Fe2O3 electrode exhibits an overpotential of 272 mV at a current density of 100 mA cm-2, presenting a 64 mV reduction compared to the electrode without an MF. The experimental results indicate that an MF can induce the directional growth of Fe2O3 rods and reduce their accumulation. In addition, an external MF is beneficial for the lattice dislocation of the obtained catalysts, which can increase the surface free energy, thus reducing the activation energy and accelerating the electrochemical reaction kinetics. This work effectively combines a magnetic field with chemical corrosion and electrochemical energy, which offers a novel strategy for the large-scale development of environmentally friendly and superior electrocatalysts.

13.
Langmuir ; 40(31): 16605-16614, 2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39039962

RÉSUMÉ

Despite its significant potential in various disease treatments and diagnostics, microbiotherapy is consistently plagued by multiple limitations ranging from manufacturing challenges to in vivo functionality. Inspired by the strategy involving nonproliferating yet metabolically active microorganisms, we report an intracellular gelation approach that can generate a synthetic polymer network within bacterial cells to solve these challenges. Specifically, poly(ethylene glycol dimethacrylate) (PEGDA, 700 Da) monomers are introduced into the bacterial cytosol through a single cycle of freeze-thawing followed by the initiation of intracellular free radical polymerization by UV light to create a macromolecular PEGDA gel within the bacterial cytosol. The molecular crowding resulting from intracytoplasmic gelation prohibits bacterial division and confers robust resistance to simulated gastrointestinal fluids and bile acids while retaining the ability to secrete functional proteins. Biocompatibility assessments demonstrate that the nondividing gelatinized bacteria are effective in alleviating systemic inflammation triggered by intravenous Escherichia coli injection. Furthermore, the therapeutic efficacy of gelatinized Lactobacillus rhamnosus in colitis mice provides additional support for this approach. Collectively, intracellular gelation indicates a universal strategy to manufacture next-generation live biotherapeutics for advanced microbiotherapy.


Sujet(s)
Escherichia coli , Polyéthylène glycols , Animaux , Souris , Escherichia coli/effets des médicaments et des substances chimiques , Polyéthylène glycols/composition chimique , Gels/composition chimique , Modèles animaux de maladie humaine , Colite/traitement médicamenteux , Colite/induit chimiquement , Méthacrylates/composition chimique
14.
Adv Healthc Mater ; : e2401118, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38979865

RÉSUMÉ

Bacteria, especially drug-resistant strains, can quickly cause wound infections, leading to delayed healing and fatal risk in clinics. With the growing need for alternative antibacterial approaches that rely less on antibiotics or eliminate their use altogether, a novel antibacterial hydrogel named Ovtgel is developed. Ovtgel is formulated by chemically crosslinking thiol-modified ovotransferrin (Ovt), a member of the transferrin family found in egg white, with olefin-modified agarose through thiol-ene click chemistry. Ovt is designed to sequester ferric ions essential for bacterial survival and protect wound tissues from damages caused by the reactive oxygen species (ROS) generated in Fenton reactions. Experimental data have shown that Ovtgel significantly enhances wound healing by inhibiting bacterial growth and shielding tissues from ROS-induced harms. Unlike traditional antibiotics, Ovtgel targets essential trace elements required for bacterial survival in the host environment, preventing the development of drug resistance in pathogenic bacteria. Ovtgel exhibits excellent biocompatibility due to the homology of Ovt to mammalian transferrin. This hydrogel has the potential to serve as an effective antibiotic-free solution for combating bacterial infections.

15.
Int J Biol Sci ; 20(8): 2922-2942, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904021

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and the production of autoantibodies. Previous studies have indicated an association between high-salt diets (HSD) and an increased risk of RA, yet the underlying mechanisms remain unclear. Macrophage pyroptosis, a pro-inflammatory form of cell death, plays a pivotal role in RA. In this study, we demonstrate that HSD exacerbates the severity of arthritis in collagen-induced arthritis (CIA) mice, correlating with macrophage infiltration and inflammatory lesions. Given the significant alterations observed in macrophages from CIA mice subjected to HSD, we specifically investigate the impact of HSD on macrophage responses in the inflammatory milieu of RA. In our in vitro experiments, pretreatment with NaCl enhances LPS-induced pyroptosis in RAW.264.7 and THP-1 cells through the p38 MAPK/NF-κB signaling pathway. Subsequent experiments reveal that Slc6a12 inhibitors and SGK1 silencing inhibit sodium-induced activation of macrophage pyroptosis and the p38 MAPK/NF-κB signaling pathway, whereas overexpression of the SGK1 gene counteracts the effect of sodium on macrophages. In conclusion, our findings verified that high salt intake promotes the progression of RA and provided a detailed elucidation of the activation of macrophage pyroptosis induced by sodium transportation through the Slc6a12 channel.


Sujet(s)
Polyarthrite rhumatoïde , Macrophages , Protein-Serine-Threonine Kinases , Pyroptose , Animaux , Souris , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Macrophages/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Chlorure de sodium/pharmacologie , Cellules RAW 264.7 , Humains , Mâle , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Arthrite expérimentale/métabolisme , Transduction du signal , p38 Mitogen-Activated Protein Kinases/métabolisme , Souris de lignée DBA
16.
ACS Nano ; 18(20): 13266-13276, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38709874

RÉSUMÉ

One key challenge in postoperative glioblastoma immunotherapy is to guarantee a potent and durable T-cell response, which is restricted by the immunosuppressive microenvironment within the lymph nodes (LNs). Here, we develop an in situ sprayed exosome-cross-linked gel that acts as an artificial LN structure to directly activate the tumor-infiltrating T cells for prevention of glioma recurrence. Briefly, this gel is generated by a bio-orthogonal reaction between azide-modified chimeric exosomes and alkyne-modified alginate polymers. Particularly, these chimeric exosomes are generated from dendritic cell (DC)-tumor hybrid cells, allowing for direct and robust T-cell activation. The gel structure with chimeric exosomes as cross-linking points avoids the quick clearance by the immune system and thus prolongs the durability of antitumor T-cell immunity. Importantly, this exosome-containing immunotherapeutic gel provides chances for ameliorating functions of antigen-presenting cells (APCs) through accommodating different intracellular-acting adjuvants, such as stimulator of interferon genes (STING) agonists. This further enhances the antitumor T-cell response, resulting in the almost complete elimination of residual lesions after surgery. Our findings provide a promising strategy for postsurgical glioma immunotherapy that warrants further exploration in the clinical arena.


Sujet(s)
Exosomes , Glioblastome , Immunothérapie , Noeuds lymphatiques , Exosomes/composition chimique , Glioblastome/thérapie , Glioblastome/immunologie , Glioblastome/anatomopathologie , Humains , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/anatomopathologie , Animaux , Souris , Gels/composition chimique , Cellules dendritiques/immunologie , Lymphocytes T/immunologie , Lignée cellulaire tumorale , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Tumeurs du cerveau/anatomopathologie , Souris de lignée C57BL
17.
Adv Mater ; 36(25): e2402532, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38563503

RÉSUMÉ

Due to inherent differences in cellular composition and metabolic behavior with host cells, tumor-harbored bacteria can discriminatorily affect tumor immune landscape. However, the mechanisms by which intracellular bacteria affect antigen presentation process between tumor cells and antigen-presenting cells (APCs) are largely unknown. The invasion behavior of attenuated Salmonella VNP20009 (VNP) into tumor cells is investigated and an attempt is made to modulate this behavior by modifying positively charged polymers on the surface of VNP. It is found that non-toxic chitosan oligosaccharide (COS) modified VNP (VNP@COS) bolsters the formation of gap junction between tumor cells and APCs by enhancing the ability of VNP to infect tumor cells. On this basis, a bacterial biohybrid is designed to promote in situ antigen cross-presentation through intracellular bacteria induced gap junction. This bacterial biohybrid also enhances the expression of major histocompatibility complex class I molecules on the surface of tumor cells through the incorporation of Mdivi-1 coupled with VNP@COS. This strategic integration serves to heighten the immunogenic exposure of tumor antigens; while, preserving the cytotoxic potency of T cells. A strategy is proposed to precisely controlling the function and local effects of microorganisms within tumors.


Sujet(s)
Présentation d'antigène , Chitosane , Jonctions communicantes , Salmonella , Humains , Chitosane/composition chimique , Lignée cellulaire tumorale , Jonctions communicantes/métabolisme , Salmonella/immunologie , Animaux , Cross-priming , Souris , Oligosaccharides/composition chimique , Tumeurs/immunologie , Tumeurs/anatomopathologie , Cellules présentatrices d'antigène/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité de classe I/immunologie
18.
Nat Commun ; 15(1): 3343, 2024 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-38637580

RÉSUMÉ

Pathogenic gut microbiota is responsible for a few debilitating gastrointestinal diseases. While the host immune cells do produce extracellular vesicles to counteract some deleterious effects of the microbiota, the extracellular vesicles are of insufficient doses and at unreliable exposure times. Here we use mechanical stimulation of hydrogel-embedded macrophage in a bioelectronic controller that on demand boost production of up to 20 times of therapeutic extracellular vesicles to ameliorate the microbes' deleterious effects in vivo. Our miniaturized wireless bioelectronic system termed inducible mechanical activation for in-situ and sustainable generating extracellular vesicles (iMASSAGE), leverages on wireless electronics and responsive hydrogel to impose mechanical forces on macrophages to produce extracellular vesicles that rectify gut microbiome dysbiosis and ameliorate colitis. This in vivo controllable extracellular vesicles-produced system holds promise as platform to treat various other diseases.


Sujet(s)
Colite , Vésicules extracellulaires , Microbiome gastro-intestinal , Microbiote , Humains , Microbiome gastro-intestinal/physiologie , Hydrogels/pharmacologie , Dysbiose
19.
Nano Lett ; 24(15): 4691-4701, 2024 Apr 17.
Article de Anglais | MEDLINE | ID: mdl-38588212

RÉSUMÉ

Tumor cells exhibit heightened glucose (Glu) consumption and increased lactic acid (LA) production, resulting in the formation of an immunosuppressive tumor microenvironment (TME) that facilitates malignant proliferation and metastasis. In this study, we meticulously engineer an antitumor nanoplatform, denoted as ZLGCR, by incorporating glucose oxidase, LA oxidase, and CpG oligodeoxynucleotide into zeolitic imidazolate framework-8 that is camouflaged with a red blood cell membrane. Significantly, ZLGCR-mediated consumption of Glu and LA not only amplifies the effectiveness of metabolic therapy but also reverses the immunosuppressive TME, thereby enhancing the therapeutic outcomes of CpG-mediated antitumor immunotherapy. It is particularly important that the synergistic effect of metabolic therapy and immunotherapy is further augmented when combined with immune checkpoint blockade therapy. Consequently, this engineered antitumor nanoplatform will achieve a cooperative tumor-suppressive outcome through the modulation of metabolism and immune responses within the TME.


Sujet(s)
Tumeurs , Microenvironnement tumoral , Humains , Immunothérapie , Radioimmunothérapie , Glucose , Glucose oxidase , Immunosuppresseurs , Acide lactique , Tumeurs/thérapie , Lignée cellulaire tumorale
20.
Nano Lett ; 24(12): 3801-3810, 2024 Mar 27.
Article de Anglais | MEDLINE | ID: mdl-38477714

RÉSUMÉ

The effectiveness of various cancer therapies for solid tumors is substantially limited by the highly hypoxic tumor microenvironment (TME). Here, a microalgae-integrated living hydrogel (ACG gel) is developed to concurrently enhance hypoxia-constrained tumor starvation therapy and immunotherapy. The ACG gel is formed in situ following intratumoral injection of a biohybrid fluid composed of alginate, Chlorella sorokiniana, and glucose oxidase, facilitated by the crossing-linking between divalent ions within tumors and alginate. The microalgae Chlorella sorokiniana embedded in ACG gel generate abundant oxygen through photosynthesis, enhancing glucose oxidase-catalyzed glucose consumption and shifting the TME from immunosuppressive to immunopermissive status, thus reducing the tumor cell energy supply and boosting antitumor immunity. In murine 4T1 tumor models, the ACG gel significantly suppresses tumor growth and effectively prevents postoperative tumor recurrence. This study, leveraging microalgae as natural oxygenerators, provides a versatile and universal strategy for the development of oxygen-dependent tumor therapies.


Sujet(s)
Chlorella , Microalgues , Tumeurs , Animaux , Souris , Hydrogels , Glucose oxidase , Photosynthèse , Hypoxie , Oxygène , Immunothérapie , Alginates , Microenvironnement tumoral
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE