Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Immunity ; 50(1): 137-151.e6, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30650373

ABSTRACT

Fever is an evolutionarily conserved response that confers survival benefits during infection. However, the underlying mechanism remains obscure. Here, we report that fever promoted T lymphocyte trafficking through heat shock protein 90 (Hsp90)-induced α4 integrin activation and signaling in T cells. By inducing selective binding of Hsp90 to α4 integrins, but not ß2 integrins, fever increased α4-integrin-mediated T cell adhesion and transmigration. Mechanistically, Hsp90 bound to the α4 tail and activated α4 integrins via inside-out signaling. Moreover, the N and C termini of one Hsp90 molecule simultaneously bound to two α4 tails, leading to dimerization and clustering of α4 integrins on the cell membrane and subsequent activation of the FAK-RhoA pathway. Abolishment of Hsp90-α4 interaction inhibited fever-induced T cell trafficking to draining lymph nodes and impaired the clearance of bacterial infection. Our findings identify the Hsp90-α4-integrin axis as a thermal sensory pathway that promotes T lymphocyte trafficking and enhances immune surveillance during infection.


Subject(s)
Fever/immunology , HSP90 Heat-Shock Proteins/metabolism , Integrin alpha4/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/immunology , T-Lymphocytes/immunology , Animals , Bacterial Load , Cell Adhesion , Cell Movement , Dimerization , Focal Adhesion Kinase 1/metabolism , Immunologic Surveillance , Integrin alpha4/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Signal Transduction , rhoA GTP-Binding Protein/metabolism
2.
EMBO J ; 42(13): e112542, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37218505

ABSTRACT

Lipid droplets (LDs) form inter-organelle contacts with the endoplasmic reticulum (ER) that promote their biogenesis, while LD contacts with mitochondria enhance ß-oxidation of contained fatty acids. Viruses have been shown to take advantage of lipid droplets to promote viral production, but it remains unclear whether they also modulate the interactions between LDs and other organelles. Here, we showed that coronavirus ORF6 protein targets LDs and is localized to the mitochondria-LD and ER-LD contact sites, where it regulates LD biogenesis and lipolysis. At the molecular level, we find that ORF6 inserts into the LD lipid monolayer via its two amphipathic helices. ORF6 further interacts with ER membrane proteins BAP31 and USE1 to mediate ER-LDs contact formation. Additionally, ORF6 interacts with the SAM complex in the mitochondrial outer membrane to link mitochondria to LDs. In doing so, ORF6 promotes cellular lipolysis and LD biogenesis to reprogram host cell lipid flux and facilitate viral production.


Subject(s)
Coronavirus , Coronavirus/metabolism , Endoplasmic Reticulum/metabolism , Lipid Droplets/metabolism , Lipolysis , Fatty Acids/metabolism
3.
Proc Natl Acad Sci U S A ; 120(29): e2218973120, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37428928

ABSTRACT

Antibiotics are among the most used weapons in fighting microbial infections and have greatly improved the quality of human life. However, bacteria can eventually evolve to exhibit antibiotic resistance to almost all prescribed antibiotic drugs. Photodynamic therapy (PDT) develops little antibiotic resistance and has become a promising strategy in fighting bacterial infection. To augment the killing effect of PDT, the conventional strategy is introducing excess ROS in various ways, such as applying high light doses, high photosensitizer concentrations, and exogenous oxygen. In this study, we report a metallacage-based PDT strategy that minimizes the use of ROS by jointly using gallium-metal organic framework rods to inhibit the production of bacterial endogenous NO, amplify ROS stress, and enhance the killing effect. The augmented bactericidal effect was demonstrated both in vitro and in vivo. This proposed enhanced PDT strategy will provide a new option for bacterial ablation.


Subject(s)
Photochemotherapy , Humans , Reactive Oxygen Species/pharmacology , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacteria
4.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34531300

ABSTRACT

The Down syndrome cell adhesion molecule (DSCAM) belongs to the immunoglobulin superfamily (IgSF) and plays important roles in neural development. It has a large ectodomain, including 10 Ig-like domains and 6 fibronectin III (FnIII) domains. Previous data have shown that DSCAM can mediate cell adhesion by forming homophilic dimers between cells and contributes to self-avoidance of neurites or neuronal tiling, which is important for neural network formation. However, the organization and assembly of DSCAM at cell adhesion interfaces has not been fully understood. Here we combine electron microscopy and other biophysical methods to characterize the structure of the DSCAM-mediated cell adhesion and generate three-dimensional views of the adhesion interfaces of DSCAM by electron tomography. The results show that mouse DSCAM forms a regular pattern at the adhesion interfaces. The Ig-like domains contribute to both trans homophilic interactions and cis assembly of the pattern, and the FnIII domains are crucial for the cis pattern formation as well as the interaction with the cell membrane. By contrast, no obvious assembly pattern is observed at the adhesion interfaces mediated by mouse DSCAML1 or Drosophila DSCAMs, suggesting the different structural roles and mechanisms of DSCAMs in mediating cell adhesion and neural network formation.


Subject(s)
Cell Adhesion Molecules/chemistry , Cell Adhesion , Down Syndrome/pathology , Drosophila Proteins/chemistry , Neurogenesis , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Down Syndrome/genetics , Down Syndrome/metabolism , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Mice , Neurites
5.
PLoS Pathog ; 17(7): e1009746, 2021 07.
Article in English | MEDLINE | ID: mdl-34297778

ABSTRACT

HCV cell-culture system uses hepatoma-derived cell lines for efficient virus propagation. Tumor cells cultured in glucose undergo active aerobic glycolysis, but switch to oxidative phosphorylation for energy production when cultured in galactose. Here, we investigated whether modulation of glycolysis in hepatocytes affects HCV infection. We showed HCV release, but not entry, genome replication or virion assembly, is significantly blocked when cells are cultured in galactose, leading to accumulation of intracellular infectious virions within multivesicular body (MVB). Blockade of the MVB-lysosome fusion or treatment with pro-inflammatory cytokines promotes HCV release in galactose. Furthermore, we found this glycometabolic regulation of HCV release is mediated by MAPK-p38 phosphorylation. Finally, we showed HCV cell-to-cell transmission is not affected by glycometabolism, suggesting that HCV cell-to-supernatant release and cell-to-cell transmission are two mechanistically distinct pathways. In summary, we demonstrated glycometabolism regulates the efficiency and route of HCV release. We proposed HCV may exploit the metabolic state in hepatocytes to favor its spread through the cell-to-cell transmission in vivo to evade immune response.


Subject(s)
Hepacivirus/physiology , Hepatitis C/virology , Hepatocytes/metabolism , Hepatocytes/virology , Virus Release/physiology , Cell Line, Tumor , Humans
6.
PLoS Pathog ; 15(5): e1007759, 2019 05.
Article in English | MEDLINE | ID: mdl-31116791

ABSTRACT

Hepatitis C virus (HCV) is a member of Hepacivirus and belongs to the family of Flaviviridae. HCV infects millions of people worldwide and may lead to cirrhosis and hepatocellular carcinoma. HCV envelope proteins, E1 and E2, play critical roles in viral cell entry and act as major epitopes for neutralizing antibodies. However, unlike other known flaviviruses, it has been challenging to study HCV envelope proteins E1E2 in the past decades as the in vitro expressed E1E2 heterodimers are usually of poor quality, making the structural and functional characterization difficult. Here we express the ectodomains of HCV E1E2 heterodimer with either an Fc-tag or a de novo designed heterodimeric tag and are able to isolate soluble E1E2 heterodimer suitable for functional and structural studies. Then we characterize the E1E2 heterodimer by electron microscopy and model the structure by the coevolution based modeling strategy with Rosetta, revealing the potential interactions between E1 and E2. Moreover, the E1E2 heterodimer is applied to examine the interactions with the known HCV receptors, neutralizing antibodies as well as the inhibition of HCV infection, confirming the functionality of the E1E2 heterodimer and the binding profiles of E1E2 with the cellular receptors. Therefore, the expressed E1E2 heterodimer would be a valuable target for both viral studies and vaccination against HCV.


Subject(s)
Hepacivirus/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Antibodies, Neutralizing/metabolism , HEK293 Cells , Hepatitis C/genetics , Hepatitis C/metabolism , Hepatitis C/virology , Humans , Protein Conformation , Protein Multimerization , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/genetics , Viral Envelope Proteins/genetics , Virus Internalization
7.
Proc Natl Acad Sci U S A ; 115(37): 9246-9251, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150416

ABSTRACT

Cell-cell adhesion is important for cell growth, tissue development, and neural network formation. Structures of cell adhesion molecules have been widely studied by crystallography, revealing the molecular details of adhesion interfaces. However, due to technical limitations, the overall structure and organization of adhesion molecules at cell adhesion interfaces has not been fully investigated. Here, we combine electron microscopy and other biophysical methods to characterize the structure of cell-cell adhesion mediated by the cell adhesion molecule Sidekick (Sidekick-1 and Sidekick-2) and obtain 3D views of the Sidekick-mediated adhesion interfaces as well as the organization of Sidekick molecules between cell membranes by electron tomography. The results suggest that the Ig-like domains and the fibronectin III (FnIII) domains of Sidekicks play different roles in cell adhesion. The Ig-like domains mediate the homophilic transinteractions bridging adjacent cells, while the FnIII domains interact with membranes, resulting in a tight adhesion interface between cells that may contribute to the specificity and plasticity of cell-cell contacts during cell growth and neural development.


Subject(s)
Cell Membrane , Electron Microscope Tomography , Immunoglobulin G , Membrane Proteins , Animals , Cell Adhesion/physiology , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane/ultrastructure , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Immunoglobulin G/ultrastructure , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Proteins/ultrastructure , Mice , Protein Domains
8.
Proc Natl Acad Sci U S A ; 113(47): 13438-13443, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821726

ABSTRACT

Clearance of dead cells is critical for maintaining homeostasis and prevents autoimmunity and inflammation. When cells undergo apoptosis and necrosis, specific markers are exposed and recognized by the receptors on phagocytes. DEC205 (CD205) is an endocytotic receptor on dendritic cells with antigen presentation function and has been widely used in immune therapies for vaccine generation. It has been shown that human DEC205 recognizes apoptotic and necrotic cells in a pH-dependent fashion. However, the natural ligand(s) of DEC205 remains unknown. Here we find that keratins are the cellular ligands of human DEC205. DEC205 binds to keratins specifically at acidic, but not basic, pH through its N-terminal domains. Keratins form intermediate filaments and are important for maintaining the strength of cells and tissues. Our results suggest that keratins also function as cell markers of apoptotic and necrotic cells and mediate a pH-dependent pathway for the immune recognition of dead cells.


Subject(s)
Antigens, CD/metabolism , Apoptosis , Dendritic Cells/metabolism , Keratins/metabolism , Lectins, C-Type/metabolism , Minor Histocompatibility Antigens/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, CD/chemistry , Glycoside Hydrolases/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Jurkat Cells , Keratins/chemistry , Lectins, C-Type/chemistry , Ligands , Mice, Inbred C57BL , Minor Histocompatibility Antigens/chemistry , Necrosis , Protein Binding , Receptors, Cell Surface/chemistry
9.
Proc Natl Acad Sci U S A ; 112(23): 7237-42, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26039988

ABSTRACT

Dendritic cells play important roles in regulating innate and adaptive immune responses. DEC205 (CD205) is one of the major endocytotic receptors on dendritic cells and has been widely used for vaccine generation against viruses and tumors. However, little is known about its structure and functional mechanism. Here we determine the structure of the human DEC205 ectodomain by cryoelectron microscopy. The structure shows that the 12 extracellular domains form a compact double ring-shaped conformation at acidic pH and become extended at basic pH. Biochemical data indicate that the pH-dependent conformational change of DEC205 is correlated with ligand binding and release. DEC205 only binds to apoptotic and necrotic cells at acidic pH, whereas live cells cannot be recognized by DEC205 at either acidic or basic conditions. These results suggest that DEC205 is an immune receptor that recognizes apoptotic and necrotic cells specifically through a pH-dependent mechanism.


Subject(s)
Antigens, CD/physiology , Dendritic Cells/cytology , Hydrogen-Ion Concentration , Lectins, C-Type/physiology , Receptors, Cell Surface/physiology , Antigens, CD/chemistry , Antigens, CD/ultrastructure , Cryoelectron Microscopy , HEK293 Cells , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/ultrastructure , Minor Histocompatibility Antigens , Mutagenesis , Necrosis , Protein Conformation , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/ultrastructure
10.
Nat Commun ; 15(1): 631, 2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38245527

ABSTRACT

Lipid droplets (LDs) are dynamic lipid storage organelles that can be degraded by autophagy machinery to release neutral lipids, a process called lipophagy. However, specific receptors and regulation mechanisms for lipophagy remain largely unknown. Here, we identify that ATG14, the core unit of the PI3KC3-C1 complex, also targets LD and acts as an autophagic receptor that facilitates LD degradation. A negative regulator, Syntaxin18 (STX18) binds ATG14, disrupting the ATG14-ATG8 family members interactions and subverting the PI3KC3-C1 complex formation. Knockdown of STX18 activates lipophagy dependent on ATG14 not only as the core unit of PI3KC3-C1 complex but also as the autophagic receptor, resulting in the degradation of LD-associated anti-viral protein Viperin. Furthermore, coronavirus M protein binds STX18 and subverts the STX18-ATG14 interaction to induce lipophagy and degrade Viperin, facilitating virus production. Altogether, our data provide a previously undescribed mechanism for additional roles of ATG14 in lipid metabolism and virus production.


Subject(s)
Lipid Droplets , Lipid Metabolism , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Proteins/metabolism , Autophagy/physiology
11.
Adv Sci (Weinh) ; 11(17): e2309491, 2024 May.
Article in English | MEDLINE | ID: mdl-38380490

ABSTRACT

The regeneration of bone defects in diabetic patients still faces challenges, as the intrinsic healing process is impaired by hyperglycemia. Inspired by the discovery that the endoplasmic reticulum (ER) is in a state of excessive stress and dysfunction under hyperglycemia, leading to osteogenic disorder, a novel engineered exosome is proposed to modulate ER homeostasis for restoring the function of mesenchymal stem cells (MSCs). The results indicate that the constructed engineered exosomes efficiently regulate ER homeostasis and dramatically facilitate the function of MSCs in the hyperglycemic niche. Additionally, the underlying therapeutic mechanism of exosomes is elucidated. The results reveal that exosomes can directly provide recipient cells with SHP2 for the activation of mitophagy and elimination of mtROS, which is the immediate cause of ER dysfunction. To maximize the therapeutic effect of engineered exosomes, a high-performance hydrogel with self-healing, bioadhesive, and exosome-conjugating properties is applied to encapsulate the engineered exosomes for in vivo application. In vivo, evaluation in diabetic bone defect repair models demonstrates that the engineered exosomes delivering hydrogel system intensively enhance osteogenesis. These findings provide crucial insight into the design and biological mechanism of ER homeostasis-based tissue-engineering strategies for diabetic bone regeneration.


Subject(s)
Bone Regeneration , Endoplasmic Reticulum , Exosomes , Homeostasis , Hydrogels , Mesenchymal Stem Cells , Exosomes/metabolism , Bone Regeneration/physiology , Bone Regeneration/genetics , Animals , Homeostasis/physiology , Hydrogels/chemistry , Mice , Endoplasmic Reticulum/metabolism , Mesenchymal Stem Cells/metabolism , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Experimental/metabolism , Osteogenesis/physiology , Disease Models, Animal , Tissue Engineering/methods , Male , Humans
12.
Plant Physiol ; 158(1): 264-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22100644

ABSTRACT

Primexine deposition and plasma membrane undulation are the initial steps of pollen wall formation. However, little is known about the genes involved in this important biological process. Here, we report a novel gene, NO PRIMEXINE AND PLASMA MEMBRANE UNDULATION (NPU), which functions in the early stage of pollen wall development in Arabidopsis (Arabidopsis thaliana). Loss of NPU function causes male sterility due to a defect in callose synthesis and sporopollenin deposition, resulting in disrupted pollen in npu mutants. Transmission electronic microscopy observation demonstrated that primexine deposition and plasma membrane undulation are completely absent in the npu mutants. NPU encodes a membrane protein with two transmembrane domains and one intracellular domain. In situ hybridization analysis revealed that NPU is strongly expressed in microspores and the tapetum during the tetrad stage. All these results together indicate that NPU plays a vital role in primexine deposition and plasma membrane undulation during early pollen wall development.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/physiology , Cell Membrane/metabolism , Gametogenesis, Plant/genetics , Membrane Proteins/metabolism , Arabidopsis Proteins/genetics , Biopolymers , Carotenoids , Cell Membrane/genetics , Cloning, Molecular , Gene Expression Regulation, Plant , Glucans/biosynthesis , Meiosis , Membrane Proteins/genetics , Mutation , Plant Infertility/genetics , Pollen/genetics , Pollen/growth & development , Pollen/metabolism , Protein Structure, Tertiary
13.
Adv Sci (Weinh) ; 10(25): e2302136, 2023 09.
Article in English | MEDLINE | ID: mdl-37400369

ABSTRACT

Tissue regeneration is regulated by morphological clues of implants in bone defect repair. Engineered morphology can boost regenerative biocascades that conquer challenges such as material bioinertness and pathological microenvironments. Herein, a correlation between the liver extracellular skeleton morphology and the regenerative signaling, namely hepatocyte growth factor receptor (MET), is found to explain the mystery of rapid liver regeneration. Inspired by this unique structure, a biomimetic morphology is prepared on polyetherketoneketone (PEKK) via femtosecond laser etching and sulfonation. The morphology reproduces MET signaling in macrophages, causing positive immunoregulation and optimized osteogenesis. Moreover, the morphological clue activates an anti-inflammatory reserve (arginase-2) to translocate retrogradely from mitochondria to the cytoplasm due to the difference in spatial binding of heat shock protein 70. This translocation enhances oxidative respiration and complex II activity, reprogramming the metabolism of energy and arginine. The importance of MET signaling and arginase-2 in the anti-inflammatory repair of biomimetic scaffolds is also verified via chemical inhibition and gene knockout. Altogether, this study not only provides a novel biomimetic scaffold for osteoporotic bone defect repair that can simulate regenerative signals, but also reveals the significance and feasibility of strategies to mobilize anti-inflammatory reserves in bone regeneration.


Subject(s)
Bone Regeneration , Inflammation , Liver , Macrophages , Osseointegration , Osteoporosis , Tissue Scaffolds , Animals , Female , Mice , Rats , Cell Respiration , Energy Metabolism , Inflammation/prevention & control , Liver/cytology , Liver/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Osteoporosis/metabolism , Proto-Oncogene Proteins c-met/metabolism , Rats, Sprague-Dawley , Signal Transduction , Tissue Scaffolds/chemistry
14.
Acta Biomater ; 150: 380-390, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-35917910

ABSTRACT

Infections caused by drug-resistant bacteria pose a great threat to human health. Non-antibiotic-dependent antibacterial strategies have become the focus of research. Among them, chemical dynamic treatment-based (CDT) therapeutic systems, which catalyze the production of hydroxyl radicals by enzymes, have achieved tremendous success for antibacterial purposes. However, limited kinetics of the Fenton reaction, poor permeability, and short half-life of hydroxyl radicals compromise the antibacterial effects of CDT. In addition, difficulties in the early diagnosis of infection lead to drug abuse and delayed treatment. Herein, a polydopamine coated ferrous sulfide theranostic platform adsorbing a hypochlorite responsive probe with photothermal treatment (PTT) enhanced CDT was synthesized. The probe component was used for the early diagnosis of infection. PTT not only inactivated bacteria by hyperthermia but also accelerated the Fenton reaction to produce more ·OH. In vitro antibacterial experiments demonstrated that the multifunctional theranostic platform has a broad antibacterial spectrum, including methicillin-resistant Staphylococcus aureus (MRSA), drug-resistant Escherichia coli (DR E. coli), and Pseudomonas aeruginosa (P. aeruginosa). In addition, in vivo antibacterial experiments demonstrated that nanoparticles could effectively rescue S. aureus-infected full-thickness skin defects with negligible cytotoxicity. This study proposes an efficient and multifunctional theranostic platform for bacterial infection, providing an effective synergistic antibacterial strategy for the treatment of antibiotic resistance. STATEMENT OF SIGNIFICANCE: An infection responsive theranostic platform (ClO- probe@FeS@PDA) is prepared. ·CDT is enhanced prominently by PTT at a relative low temperature. · FeS@PDA exhibits good antibacterial performance against drug resistant bacteria in vitro and in vivo.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcus aureus , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria , Escherichia coli , Humans , Indoles , Phototherapy , Polymers , Precision Medicine , Theranostic Nanomedicine
15.
Front Cardiovasc Med ; 9: 800185, 2022.
Article in English | MEDLINE | ID: mdl-35369285

ABSTRACT

Background: Diabetic cardiomyopathy (DCM) is a complex multifaceted disease responsible for elevated heart failure (HF) morbidity and mortality in patients with diabetes mellitus (DM). Patients with DCM exhibit subclinical diastolic dysfunction, progression toward systolic impairment, and abnormal electrophysiology. Hypoglycemia events that occur spontaneously or due to excess insulin administration threaten the lives of patients with DM-with the increased risk of sudden death. However, the molecular underpinnings of this fatal disease remain to be elucidated. Methods and Results: Here, we used the established streptozotocin-induced DCM murine model to investigate how hypoglycemia aggravates DCM progression. We confirmed connexin 43 (Cx43) dissociation from cell-cell interaction and accumulation at mitochondrial inner membrane both in the cardiomyocytes of patients with DM and DCM murine. Here, we observed that cardiac diastolic function, induced by chronic hyperglycemia, was further aggravated upon hypoglycemia challenge. Similar contractile defects were recapitulated using neonatal mouse ventricular myocytes (NMVMs) under glucose fluctuation challenges. Using immunoprecipitation mass spectrometry, we identified and validated that hypoglycemia challenge activates the mitogen-activated protein kinase kinase (MAPK kinase) (MEK)/extracellular regulated protein kinase (ERK) and inhibits phosphoinositide 3-kinase (PI3K)/Akt pathways, which results in Cx43 phosphorylation by Src protein and translocation to mitochondria in cardiomyocytes. To determine causality, we overexpressed a mitochondrial targeting Cx43 (mtCx43) using adeno-associated virus serotype 2 (AAV2)/9. At normal blood glucose levels, mtCx43 overexpression recapitulated cardiac diastolic dysfunction as well as aberrant electrophysiology in vivo. Our findings give support for therapeutic targeting of MEK/ERK/Src and PI3K/Akt/Src pathways to prevent mtCx43-driven DCM. Conclusion: DCM presents compensatory adaptation of mild mtCx43 accumulation, yet acute hypoglycemia challenges result in further accumulation of mtCx43 through the MEK/ERK/Src and PI3K/Akt/Src pathways. We provide evidence that Cx43 mislocalization is present in hearts of patients with DM hearts, STZ-induced DCM murine model, and glucose fluctuation challenged NMVMs. Mechanistically, we demonstrated that mtCx43 is responsible for inducing aberrant contraction and disrupts electrophysiology in cardiomyocytes and our results support targeting of mtCx43 in treating DCM.

16.
Nat Commun ; 13(1): 3553, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35729111

ABSTRACT

Candida auris is a multidrug-resistant human fungal pathogen responsible for nosocomial outbreaks worldwide. Although considerable progress has increased our understanding of the biological and clinical aspects of C. auris, its interaction with the host immune system is only now beginning to be investigated in-depth. Here, we compare the innate immune responses induced by C. auris BJCA001 and Candida albicans SC5314 in vitro and in vivo. Our results indicate that C. auris BJCA001 appears to be less immunoinflammatory than C. albicans SC5314, and this differential response correlates with structural features of the cell wall.


Subject(s)
Candida , Candidiasis , Antifungal Agents/pharmacology , Candida albicans , Candida auris , Candidiasis/microbiology , Humans , Immunity, Innate , Microbial Sensitivity Tests
17.
Adv Mater ; 33(45): e2103593, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34553427

ABSTRACT

Current treatments for diabetic ulcers (DUs) remain unsatisfactory due to the risk of bacterial infection and impaired angiogenesis during the healing process. The increased degradation of polyubiquitinated hypoxia-inducible factor-1α (HIF-1α) compromises wound healing efficacy. Therefore, the maintenance of HIF-1α protein stability might help treat DU. Nitric oxide (NO) is an intrinsic biological messenger that functions as a ubiquitination flow repressor and antibacterial agent; however, its clinical application in DU treatment is hindered by the difficulty in controlling NO release. Here, an intelligent near-infrared (NIR)-triggered NO nanogenerator (SNP@MOF-UCNP@ssPDA-Cy7/IR786s, abbreviated as SNP@UCM) is presented. SNP@UCM represses ubiquitination-mediated proteasomal degradation of HIF-1α by inhibiting its interaction with E3 ubiquitin ligases under NIR irradiation. Increased HIF-1α expression in endothelial cells by SNP@UCM enhances angiogenesis in wound sites, promoting vascular endothelial growth factor (VEGF) secretion and cell proliferation and migration. SNP@UCM also enables early detection of wound infections and ROS-mediated killing of bacteria. The potential clinical utility of SNP@UCM is further demonstrated in infected full-thickness DU model under NIR irradiation. SNP@UCM is the first reported HIF-1α-stabilizing advanced nanomaterial, and further materials engineering might offer a facile, mechanism-based method for clinical DU management.


Subject(s)
Biocompatible Materials/chemistry , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nitric Oxide/metabolism , Wound Healing , Biocompatible Materials/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Diabetic Foot/microbiology , Diabetic Foot/pathology , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry , Infrared Rays , Metal Nanoparticles/chemistry , Metal-Organic Frameworks/chemistry , Neovascularization, Physiologic/drug effects , Nitroprusside/chemistry , Precision Medicine , Protein Stability/drug effects , Reactive Oxygen Species/metabolism , Staphylococcus aureus/drug effects , Ubiquitination , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects
18.
Front Cell Dev Biol ; 9: 744838, 2021.
Article in English | MEDLINE | ID: mdl-34621753

ABSTRACT

Optic atrophy1 (OPA1) is crucial for inner mitochondrial membrane (IMM) fusion and essential for maintaining crista structure and mitochondrial morphology. Optic atrophy and hearing impairment are the most prevalent clinical features associated with mutations in the OPA1 gene, but the function of OPA1 in hearing is still unknown. In this study, we examined the ability of Opa1 to protect against cisplatin-induced cochlear cell death in vitro and in vivo. Our results revealed that knockdown of Opa1 affects mitochondrial function in HEI-OC1 and Neuro 2a cells, as evidenced by an elevated reactive oxygen species (ROS) level and reduced mitochondrial membrane potential. The dysfunctional mitochondria release cytochrome c, which triggers apoptosis. Opa1 expression was found to be significantly reduced after cell exposed to cisplatin in HEI-OC1 and Neuro 2a cells. Loss of Opa1 aggravated the apoptosis and mitochondrial dysfunction induced by cisplatin treatment, whereas overexpression of Opa1 alleviated cisplatin-induced cochlear cell death in vitro and in explant. Our results demonstrate that overexpression of Opa1 prevented cisplatin-induced ototoxicity, suggesting that Opa1 may play a vital role in ototoxicity and/or mitochondria-associated cochlear damage.

19.
Sci Rep ; 10(1): 18313, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110205

ABSTRACT

Porphyromonas gingivalis is a causative agent in the onset and progression of periodontal disease. This study aims to investigate the effects of quercetin, a natural plant product, on P. gingivalis virulence properties including gingipain, haemagglutinin and biofilm formation. Antimicrobial effects and morphological changes of quercetin on P. gingivalis were detected. The effects of quercetin on gingipains activities and hemolytic, hemagglutination activities were evaluated using chromogenic peptides and sheep erythrocytes. The biofilm biomass and metabolism with different concentrations of quercetin were assessed by the crystal violet and MTT assay. The structures and thickness of the biofilms were observed by confocal laser scanning microscopy. Bacterial cell surface properties including cell surface hydrophobicity and aggregation were also evaluated. The mRNA expression of virulence and iron/heme utilization was assessed using real time-PCR. Quercetin exhibited antimicrobial effects and damaged the cell structure. Quercetin can inhibit gingipains, hemolytic, hemagglutination activities and biofilm formation at sub-MIC concentrations. Molecular docking analysis further indicated that quercetin can interact with gingipains. The biofilm became sparser and thinner after quercetin treatment. Quercetin also modulate cell surface hydrophobicity and aggregation. Expression of the genes tested was down-regulated in the presence of quercetin. In conclusion, our study demonstrated that quercetin inhibited various virulence factors of P. gingivalis.


Subject(s)
Anti-Bacterial Agents/pharmacology , Periodontal Diseases/microbiology , Porphyromonas gingivalis/drug effects , Quercetin/pharmacology , Biofilms/drug effects , Gingipain Cysteine Endopeptidases/metabolism , Hemagglutinins/metabolism , Humans , Microbial Sensitivity Tests , Microscopy, Confocal , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/pathogenicity , Porphyromonas gingivalis/ultrastructure , Virulence/drug effects
20.
Nat Commun ; 10(1): 564, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718482

ABSTRACT

During meiotic prophase I, telomeres attach to and move on the nuclear envelope (NE), regulating chromosome movement to promote homologous pairing. Meiosis-specific proteins TERB1, TERB2 and MAJIN play a key role in this process. Here, we report the crystal structures of human TERB1-TERB2 and TERB2-MAJIN subcomplexes. Specific disruption of the TERB1-TERB2 or the TERB2-MAJIN interaction in the mouse Terb2 gene abolishes the telomere attachment to the NE and causes aberrant homologous pairing and disordered synapsis. In addition, depletion of SUN1 also partially disrupts the telomere-NE connection. We propose that the telomere-TRF1-TERB1-TERB2-MAJIN-NE interaction network and the telomere-LINC complex connection are likely two separate but cooperative pathways to stably recruit telomeres to the NE in meiosis prophase I. Our work provides a molecular model of the connection between telomeres and the NE and reveals the correlation between aberrant synapsis and the defective telomere attachment to the NE.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Membrane Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Telomere-Binding Proteins/metabolism , Telomere/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line , Female , Humans , Male , Meiosis , Membrane Proteins/genetics , Mice , Mice, Knockout , Nuclear Proteins/genetics , Telomere-Binding Proteins/genetics , Telomeric Repeat Binding Protein 1
SELECTION OF CITATIONS
SEARCH DETAIL