LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 123

Search options

  1. Article ; Online: Pro-Death or Pro-Survival: Contrasting Paradigms on Nanomaterial-Induced Autophagy and Exploitations for Cancer Therapy.

    Zhang, Yunjiao / Zhang, Li / Gao, Jinhao / Wen, Longping

    Accounts of chemical research

    2019  Volume 52, Issue 11, Page(s) 3164–3176

    Abstract: Autophagy is a critical lysosome-mediated cellular degradation process for the clearance of damaged organelles, obsolete proteins, and invading pathogens and plays important roles in the pathogenesis and treatment of human diseases including cancer. ... ...

    Abstract Autophagy is a critical lysosome-mediated cellular degradation process for the clearance of damaged organelles, obsolete proteins, and invading pathogens and plays important roles in the pathogenesis and treatment of human diseases including cancer. While not a cell death process per se, autophagy is nevertheless intimately linked to a cell's live/die decision. Basal autophagy, operating constitutively at low levels in essentially every mammalian cell, is vital for maintaining cellular homeostasis and promotes cell survival. On the other hand, elevated level of autophagy is frequently observed in cells responding to a physical, chemical, or biological stress. This "induced" autophagy, a hallmark under a variety of pathological and pathophysiological conditions, may be either pro-death or pro-survival, two contrasting paradigms for cell fate determination. Research in our laboratory and other groups around the world over the last 15 years has revealed nanomaterials as a unique class of autophagy inducers, with the capability of elevating the cellular autophagy to extremely high levels. In this Account we focus on the contrasting cell fate decision impacted by nanomaterial-induced autophagy. First, we give a brief introduction to nanomaterial-induced autophagy and summarize our current understanding on how it affects a cell's live/die decision. Autophagy induced by nanomaterials, in most cases, promotes cell death, but a significant number of nanomaterials are also able to elicit pro-survival autophagy. Although not a common feature, some nanomaterials may induce pro-death autophagy in one cell type while eliciting pro-survival autophagy in a different cell type. The ability to control the level of the induced autophagy, and furthermore its pro-death/pro-survival nature, is critically important for nanomedicine. Second, we discuss several possible mechanistic insights on the pro-death/pro-survival decision for nanomaterial-induced autophagy. "Disrupted" autophagic processes, with a "block" or perhaps "diversion" at the various stages, may be a characteristic hallmark for nanomaterial-induced autophagy, rendering it intrinsically pro-death in nature. On the other hand, autophagy-mediated upregulation and activation of pro-survival factors or signaling pathways, overriding the intrinsic pro-death nature, may be a common mechanism for nanomaterial-induced pro-survival autophagy. In addition, cargo degradation and reactive oxygen species may also play important roles in the pro-death/pro-survival decision impacted by nanomaterial-induced autophagy. Finally, we focus on the situation where nanomaterials induce autophagy in cancer cells and summarize the different strategies in exploiting the pro-death or pro-survival nature of nanomaterial-induced autophagy to enhance the various modalities of cancer therapy, including direct cancer cell killing, chemotherapy and radiotherapy, photothermal therapy, and integrated diagnosis and therapy. While the details vary, the basic principle is simple and straightforward. If the induced autophagy is pro-death, maximize it. Otherwise, inhibit it. Effective exploitation of nanomaterial-induced autophagy has the potential to become a new weapon in our ever-increasing arsenal to fight cancer, particularly difficult-to-treat and drug-resistant cancer.
    MeSH term(s) Animals ; Autophagy/drug effects ; Cell Survival/drug effects ; Lysosomes/metabolism ; Nanomedicine ; Nanostructures/chemistry ; Nanostructures/therapeutic use ; Neoplasms/drug therapy ; Neoplasms/metabolism ; Neoplasms/pathology
    Language English
    Publishing date 2019-10-17
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 1483291-4
    ISSN 1520-4898 ; 0001-4842
    ISSN (online) 1520-4898
    ISSN 0001-4842
    DOI 10.1021/acs.accounts.9b00397
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Lgr5 in cancer biology: functional identification of Lgr5 in cancer progression and potential opportunities for novel therapy.

    Xu, Liangliang / Lin, Weiping / Wen, Longping / Li, Gang

    Stem cell research & therapy

    2019  Volume 10, Issue 1, Page(s) 219

    Abstract: Cancer remains one of the leading lethal diseases worldwide. Identifying biomarkers of cancers might provide insights into the strategies for the development of novel targeted anti-cancer therapies. Leucine-rich repeat-containing G protein-coupled ... ...

    Abstract Cancer remains one of the leading lethal diseases worldwide. Identifying biomarkers of cancers might provide insights into the strategies for the development of novel targeted anti-cancer therapies. Leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5) has been recently discovered as a candidate marker of cancer stem cell populations. Aberrant increased expression of Lgr5 may represent one of the most common molecular alterations in some human cancers, leading to long-term potentiation of canonical Wnt/β-catenin signaling. On the other hand, however, Lgr5-mediated suppression in canonical Wnt/β-catenin signaling has also been reported in certain cancers, such as B cell malignancies. Until now, therapeutic approaches targeting Lgr5-associated signaling axis are not yet clinically available. Increasing evidence have indicated that endogenous Lgr5
    MeSH term(s) Biomarkers, Tumor/genetics ; Biomarkers, Tumor/metabolism ; Disease Progression ; Humans ; Neoplasms/therapy ; Neoplastic Stem Cells/cytology ; Receptors, G-Protein-Coupled/chemistry ; Receptors, G-Protein-Coupled/genetics ; Receptors, G-Protein-Coupled/metabolism ; Wnt Signaling Pathway
    Chemical Substances Biomarkers, Tumor ; LGR5 protein, human ; Receptors, G-Protein-Coupled
    Language English
    Publishing date 2019-07-29
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 2548671-8
    ISSN 1757-6512 ; 1757-6512
    ISSN (online) 1757-6512
    ISSN 1757-6512
    DOI 10.1186/s13287-019-1288-8
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: MDSC-targeting gold nanoparticles enhance PD-1 tumor immunotherapy by inhibiting NLRP3 inflammasomes.

    Zhu, Yangyang / Chen, Pin / Hu, Bochuan / Zhong, Suqin / Yan, Kai / Wu, Yu / Li, Shanshan / Yang, Yinyin / Xu, Zexin / Lu, Yutong / Ouyang, Ying / Bao, Hui / Gu, Weiguang / Wen, Longping / Zhang, Yunjiao

    Biomaterials

    2024  Volume 307, Page(s) 122533

    Abstract: Myeloid-derived suppressor cells (MDSCs) play a crucial role in the immune escape mechanisms that limit the efficacy of immunotherapeutic strategies. In the tumor microenvironment, NLRP3 inflammasome-driven Interleukin-1β (IL-1β) production serves to ... ...

    Abstract Myeloid-derived suppressor cells (MDSCs) play a crucial role in the immune escape mechanisms that limit the efficacy of immunotherapeutic strategies. In the tumor microenvironment, NLRP3 inflammasome-driven Interleukin-1β (IL-1β) production serves to dampen antitumor immune responses, promoting tumor growth, progression, and immunosuppression. In this study, we revealed that gold nanoparticles (Au NPs) with a size of 30 nm disrupted NLRP3 inflammasome, but not other inflammasomes, in bone marrow-derived macrophages through abrogating NLRP3-NEK7 interactions mediated by reactive oxygen species (ROS). Density functional theory (DFT) calculations provided insights into the mechanism underlying the exceptional ROS scavenging capabilities of Au NPs. Additionally, when coupled with H6, a small peptide targeting MDSCs, Au NPs demonstrated the capacity to effectively reduce IL-1β levels and diminish the MDSCs population in tumor microenvironment, leading to enhanced T cell activation and increased immunotherapeutic efficacy in mouse tumor models that are sensitive and resistant to PD-1 inhibition. Our findings unraveled a novel approach wherein peptide-modified Au NPs relieved the suppressive impact of the tumor microenvironment by inhibiting MDSCs-mediated IL-1β release, which is the first time reported the employing a nanostrategy at modulating MDSCs to reverse the immunosuppressive microenvironment and may hold promise as a potential therapeutic agent for cancer immunotherapy.
    MeSH term(s) Mice ; Animals ; Inflammasomes ; NLR Family, Pyrin Domain-Containing 3 Protein ; Myeloid-Derived Suppressor Cells ; Gold ; Programmed Cell Death 1 Receptor ; Reactive Oxygen Species ; Metal Nanoparticles ; Neoplasms ; Immunotherapy ; Tumor Microenvironment
    Chemical Substances Inflammasomes ; NLR Family, Pyrin Domain-Containing 3 Protein ; Gold (7440-57-5) ; Programmed Cell Death 1 Receptor ; Reactive Oxygen Species
    Language English
    Publishing date 2024-03-13
    Publishing country Netherlands
    Document type Journal Article
    ZDB-ID 603079-8
    ISSN 1878-5905 ; 0142-9612
    ISSN (online) 1878-5905
    ISSN 0142-9612
    DOI 10.1016/j.biomaterials.2024.122533
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Nanoreceptors promote mutant p53 protein degradation by mimicking selective autophagy receptors.

    Huang, Xiaowan / Cao, Ziyang / Qian, Jieying / Ding, Tao / Wu, Yanxia / Zhang, Hao / Zhong, Suqin / Wang, Xiaoli / Ren, Xiaoguang / Zhang, Wang / Xu, Youcui / Yao, Guangyu / Wang, Xingwu / Yang, Xianzhu / Wen, Longping / Zhang, Yunjiao

    Nature nanotechnology

    2024  Volume 19, Issue 4, Page(s) 545–553

    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 ...

    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.
    MeSH term(s) Humans ; Tumor Suppressor Protein p53/genetics ; Tumor Suppressor Protein p53/metabolism ; Proteolysis ; Mutant Proteins/metabolism ; Mutant Proteins/pharmacology ; Cell Line, Tumor ; Autophagy ; Peptides/metabolism ; Lipids/pharmacology
    Chemical Substances Tumor Suppressor Protein p53 ; Mutant Proteins ; Peptides ; Lipids
    Language English
    Publishing date 2024-01-12
    Publishing country England
    Document type Journal Article
    ZDB-ID 2254964-X
    ISSN 1748-3395 ; 1748-3387
    ISSN (online) 1748-3395
    ISSN 1748-3387
    DOI 10.1038/s41565-023-01562-5
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Autophagy regulation as a promising approach for improving cancer immunotherapy.

    Ishimwe, Nestor / Zhang, Wenbin / Qian, Jieying / Zhang, Yunjiao / Wen, Longping

    Cancer letters

    2020  Volume 475, Page(s) 34–42

    Abstract: Autophagy plays a critical role in intracellular metabolism and maintaining cellular homeostasis. Certain tumor cells present a higher basal autophagy rate and autophagy inhibition can lead to impaired metabolic dysfunction in autophagy-dependent tumor ... ...

    Abstract Autophagy plays a critical role in intracellular metabolism and maintaining cellular homeostasis. Certain tumor cells present a higher basal autophagy rate and autophagy inhibition can lead to impaired metabolic dysfunction in autophagy-dependent tumor cells. Autophagy status in immune cells dictates their fate and response to antigen; however, autophagy in immune cells may be beneficial or detrimental depending on the developmental stage of the cell and more specifically its degree of differentiation. Autophagy-deficient hosts present variations in many metabolites, proteins and enzymes that may have tumor-promoting or -inhibiting effects. The centrality of autophagy in the metabolism of some cancers and immune cells poses as a critical target whose mechanisms must be further unraveled to optimize patient response and prevent tumor recurrence.
    MeSH term(s) Animals ; Autophagy ; Homeostasis ; Humans ; Immunotherapy/methods ; Neoplasms/immunology ; Neoplasms/pathology ; Neoplasms/therapy ; Tumor Microenvironment/immunology
    Language English
    Publishing date 2020-01-31
    Publishing country Ireland
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 195674-7
    ISSN 1872-7980 ; 0304-3835
    ISSN (online) 1872-7980
    ISSN 0304-3835
    DOI 10.1016/j.canlet.2020.01.034
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: 2D-CuPd nanozyme overcome tamoxifen resistance in breast cancer by regulating the PI3K/AKT/mTOR pathway.

    Jiang, Wenwei / Zhong, Suqin / Chen, Ziying / Qian, Jieying / Huang, Xiaowan / Zhang, Hao / Wen, Longping / Zhang, Yunjiao / Yao, Guangyu

    Biomaterials

    2022  Volume 294, Page(s) 121986

    Abstract: Tamoxifen is the most commonly used treatment for estrogen-receptor (ER) positive breast cancer patients, but its efficacy is severely hampered by resistance. PI3K/AKT/mTOR pathway inhibition was proven to augment the benefit of endocrine therapy and ... ...

    Abstract Tamoxifen is the most commonly used treatment for estrogen-receptor (ER) positive breast cancer patients, but its efficacy is severely hampered by resistance. PI3K/AKT/mTOR pathway inhibition was proven to augment the benefit of endocrine therapy and exhibited potential for reversing tamoxifen-induced resistance. However, the vast majority of PI3K inhibitors currently approved for clinical use are unsatisfactory in terms of safety and efficacy. We developed two-dimensional CuPd (2D-CuPd) nanosheets with oxidase and peroxidase nanozyme activities to offer a novel solution to inhibit the activity of the PI3K/AKT/mTOR pathway. 2D-CuPd exhibit superior dual nanozyme activities converting hydrogen peroxide accumulated in drug-resistant cells into more lethal hydroxyl radicals while compensating for the insufficient superoxide anion produced by tamoxifen. The potential clinical utility was further demonstrated in an orthotopically implanted tamoxifen-resistant PDX breast cancer model. Our results reveal a novel nanozyme ROS-mediated protein mechanism for the regulation of the PI3K subunit, illustrate the cellular pathways through which increased p85β protein expression contributes to tamoxifen resistance, and reveal p85β protein as a potential therapeutic target for overcoming tamoxifen resistance. 2D-CuPd is the first reported nanomaterial capable of degrading PI3K subunits, and its high performance combined with further materials engineering may lead to the development of nanozyme-based tumor catalytic therapy.
    MeSH term(s) Female ; Humans ; Antineoplastic Agents, Hormonal/pharmacology ; Antineoplastic Agents, Hormonal/therapeutic use ; Breast Neoplasms/pathology ; Cell Line, Tumor ; Drug Resistance, Neoplasm ; Phosphatidylinositol 3-Kinases/metabolism ; Proto-Oncogene Proteins c-akt/metabolism ; Tamoxifen/pharmacology ; Tamoxifen/therapeutic use ; TOR Serine-Threonine Kinases/metabolism ; Copper ; Lead ; Nanostructures
    Chemical Substances Antineoplastic Agents, Hormonal ; MTOR protein, human (EC 2.7.1.1) ; Phosphatidylinositol 3-Kinases (EC 2.7.1.-) ; Proto-Oncogene Proteins c-akt (EC 2.7.11.1) ; Tamoxifen (094ZI81Y45) ; TOR Serine-Threonine Kinases (EC 2.7.11.1) ; Copper (789U1901C5) ; Lead (2P299V784P)
    Language English
    Publishing date 2022-12-30
    Publishing country Netherlands
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 603079-8
    ISSN 1878-5905 ; 0142-9612
    ISSN (online) 1878-5905
    ISSN 0142-9612
    DOI 10.1016/j.biomaterials.2022.121986
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article: Myosin Light-Chain Kinase Inhibitors Attenuate Nanoparticles-Induced Autophagy and Cytotoxicity by Suppression Endocytosis.

    Wang, Yanhong / Liu, Yiming / Zhou, Wei / Lin, Jun / Wen, Longping

    Journal of nanoscience and nanotechnology

    2019  Volume 19, Issue 7, Page(s) 3792–3797

    Abstract: Myosin light-chain kinase (MLCK) is a widely known kinase that controls the contraction of muscle cells. Whether MLCK is involved in the endocytosis of nanoparticles (NPs) has not been reported yet. NPs attract interest in many fields; most of them can ... ...

    Abstract Myosin light-chain kinase (MLCK) is a widely known kinase that controls the contraction of muscle cells. Whether MLCK is involved in the endocytosis of nanoparticles (NPs) has not been reported yet. NPs attract interest in many fields; most of them can induce autophagy and cytotoxicity after entering cells via endocytosis. In this study, we found that MLCK inhibitors, ML-7 and ML-9, abolished the endocytosis of silver NPs and yttrium oxide NPs in HeLa cells, and subsequently attenuated the autophagy induction and cytotoxicity caused by the NPs treatment. This is the first report that MLCK inhibitors acted as an endocytosis inhibitor and abolished the cell entry of NPs, which provided a new method for inhibiting the endocytosis of NPs.
    MeSH term(s) Autophagy ; Endocytosis ; HeLa Cells ; Humans ; Myosin-Light-Chain Kinase ; Myosins ; Nanoparticles/toxicity
    Chemical Substances Myosin-Light-Chain Kinase (EC 2.7.11.18) ; Myosins (EC 3.6.4.1)
    Language English
    Publishing date 2019-02-09
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ISSN 1533-4880
    ISSN 1533-4880
    DOI 10.1166/jnn.2019.16324
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Crizotinib Nanomicelles Synergize with Chemotherapy through Inducing Proteasomal Degradation of Mutp53 Proteins

    Yi, Tianxiang / Qian, Jieying / Ye, Yayi / Zhang, Hao / Jin, Xin / Wang, Meimei / Yang, Zhenyu / Zhang, Wang / Wen, Longping / Zhang, Yunjiao

    ACS Applied Materials & Interfaces. 2022 Dec. 29, v. 15, no. 1 p.511-523

    2022  

    Abstract: TP53 missense mutations that express highly stabilized mutant p53 protein (mutp53) driving tumorigenesis have been witnessed in a considerable percentage of human cancers. The attempt to induce degradation of mutp53 has thus been an attractive strategy ... ...

    Abstract TP53 missense mutations that express highly stabilized mutant p53 protein (mutp53) driving tumorigenesis have been witnessed in a considerable percentage of human cancers. The attempt to induce degradation of mutp53 has thus been an attractive strategy to realize precise antitumor therapy, but currently, there has been no FDA-approved medication for mutp53 cancer. Herein, we discovered a small molecule compound crizotinib, an FDA-approved antitumor drug, exhibited outstanding mutp53-degrading capability. Crizotinib induced ubiquitination-mediated proteasomal degradation of wide-spectrum mutp53 but not the wild-type p53 protein. Degradation of mutp53 by crizotinib eliminated mutp53-conferred gain-of-function (GOF), leading to reduced cell proliferation, migration, demise, and cell cycle arrest, as well as enhanced sensitivity to doxorubicin-elicited killing in mutp53 cancer. To alleviate the side effects and improve the therapeutic effect, we adopted poly­(ethylene glycol)-polylactide-co-glycolide (PEG-PLGA) nanomicelles to deliver the hydrophobic drugs doxorubicin and crizotinib, demonstrating that crizotinib nanomicelles effectively enhanced doxorubicin-elicited anticancer efficacy in a p53Y²²⁰C pancreatic cancer in vitro and in vivo via mutp53 degradation induced by crizotinib, manifesting its promising application in clinical practice. Our work therefore revealed that crizotinib exerted significant synergistic chemotherapy with doxorubicin and suggested a novel combination therapeutic strategy for targeting p53 cancer in further clinical application.
    Keywords cancer therapy ; carcinogenesis ; cell cycle checkpoints ; cell proliferation ; doxorubicin ; drug therapy ; gain-of-function mutation ; humans ; hydrophobicity ; micelles ; mutants ; pancreatic neoplasms ; mutant p53 ; crizotinib ; PEG−PLGA nanomicelles ; chemosensitization
    Language English
    Dates of publication 2022-1229
    Size p. 511-523.
    Publishing place American Chemical Society
    Document type Article ; Online
    ISSN 1944-8252
    DOI 10.1021/acsami.2c18020
    Database NAL-Catalogue (AGRICOLA)

    More links

    Kategorien

  9. Article ; Online: Nickle-cobalt alloy nanocrystals inhibit activation of inflammasomes.

    Lin, Jun / Dong, Liang / Liu, Yi-Ming / Hu, Yi / Jiang, Chen / Liu, Ke / Liu, Liu / Song, Yong-Hong / Sun, Mei / Xiang, Xing-Cheng / Qu, Kun / Lu, Yang / Wen, Long-Ping / Yu, Shu-Hong

    National science review

    2023  Volume 10, Issue 8, Page(s) nwad179

    Abstract: Activation of inflammasomes-immune system receptor sensor complexes that selectively activate inflammatory responses-has been associated with diverse human diseases, and many nanomedicine studies have reported that structurally and chemically diverse ... ...

    Abstract Activation of inflammasomes-immune system receptor sensor complexes that selectively activate inflammatory responses-has been associated with diverse human diseases, and many nanomedicine studies have reported that structurally and chemically diverse inorganic nanomaterials cause excessive inflammasome activation. Here, in stark contrast to reports of other inorganic nanomaterials, we find that nickel-cobalt alloy magnetic nanocrystals (NiCo NCs) actually inhibit activation of NLRP3, NLRC4 and AIM2 inflammasomes. We show that NiCo NCs disrupt the canonical inflammasome ASC speck formation process by downregulating the lncRNA
    Language English
    Publishing date 2023-06-26
    Publishing country China
    Document type Journal Article
    ZDB-ID 2745465-4
    ISSN 2053-714X ; 2053-714X
    ISSN (online) 2053-714X
    ISSN 2053-714X
    DOI 10.1093/nsr/nwad179
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article: Bacteria-based multiplex system eradicates recurrent infections with drug-resistant bacteria via photothermal killing and protective immunity elicitation.

    Xu, Youcui / Wu, Yi / Hu, Yi / Xu, Mengran / Liu, Yanyan / Ding, Yuting / Chen, Jing / Huang, Xiaowan / Wen, Longping / Li, Jiabin / Zhu, Chen

    Biomaterials research

    2023  Volume 27, Issue 1, Page(s) 27

    Abstract: Background: The high mortality associated with drug-resistant bacterial infections is an intractable clinical problem resulting from the low susceptibility of these bacteria to antibiotics and the high incidence of recurrent infections.: Methods: ... ...

    Abstract Background: The high mortality associated with drug-resistant bacterial infections is an intractable clinical problem resulting from the low susceptibility of these bacteria to antibiotics and the high incidence of recurrent infections.
    Methods: Herein, a photosynthetic bacteria-based multiplex system (Rp@Al) composed of natural Rhodopseudomonas palustris (Rp) and Food and Drug Administration-approved aluminum (Al) adjuvant, was developed to combat drug-resistant bacterial infections and prevent their recurrence. We examined its photothermal performance and in vitro and in vivo antibacterial ability; revealed its protective immunomodulatory effect; verified its preventative effect on recurrent infections; and demonstrated the system's safety.
    Results: Rp@Al exhibits excellent photothermal properties with an effective elimination of methicillin-resistant Staphylococcus aureus (MRSA). In addition, Rp@Al enhances dendritic cell activation and further triggers a T helper 1 (T
    Conclusions: This innovative multiplex system, with superior photothermal and immunomodulatory effects, presents great potential for the treatment and prevention of drug-resistant bacterial infections.
    Language English
    Publishing date 2023-04-06
    Publishing country England
    Document type Journal Article
    ZDB-ID 2775188-0
    ISSN 2055-7124 ; 1226-4601
    ISSN (online) 2055-7124
    ISSN 1226-4601
    DOI 10.1186/s40824-023-00363-0
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top