Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63.562
Filter
1.
Subcell Biochem ; 104: 1-16, 2024.
Article in English | MEDLINE | ID: mdl-38963480

ABSTRACT

The global emergence of multidrug resistance (MDR) in gram-negative bacteria has become a matter of worldwide concern. MDR in these pathogens is closely linked to the overexpression of certain efflux pumps, particularly the resistance-nodulation-cell division (RND) efflux pumps. Inhibition of these pumps presents an attractive and promising strategy to combat antibiotic resistance, as the efflux pump inhibitors can effectively restore the potency of existing antibiotics. AcrAB-TolC is one well-studied RND efflux pump, which transports a variety of substrates, therefore providing resistance to a broad spectrum of antibiotics. To develop effective pump inhibitors, a comprehensive understanding of the structural aspect of the AcrAB-TolC efflux pump is imperative. Previous studies on this pump's structure have been limited to individual components or in vitro determination of fully assembled pumps. Recent advancements in cellular cryo-electron tomography (cryo-ET) have provided novel insights into this pump's assembly and functional mechanism within its native cell membrane environment. Here, we present a summary of the structural data regarding the AcrAB-TolC efflux pump, shedding light on its assembly pathway and operational mechanism.


Subject(s)
Anti-Bacterial Agents , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Carrier Proteins/metabolism , Carrier Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/chemistry , Cryoelectron Microscopy , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry
2.
Methods Enzymol ; 700: 217-234, 2024.
Article in English | MEDLINE | ID: mdl-38971601

ABSTRACT

Sphingomyelin is postulated to form clusters with glycosphingolipids, cholesterol and other sphingomyelin molecules in biomembranes through hydrophobic interaction and hydrogen bonds. These clusters form submicron size lipid domains. Proteins that selectively binds sphingomyelin and/or cholesterol are useful to visualize the lipid domains. Due to their small size, visualization of lipid domains requires advanced microscopy techniques in addition to lipid binding proteins. This Chapter describes the method to characterize plasma membrane sphingomyelin-rich and cholesterol-rich lipid domains by quantitative microscopy. This Chapter also compares different permeabilization methods to visualize intracellular lipid domains.


Subject(s)
Cholesterol , Sphingomyelins , Sphingomyelins/chemistry , Sphingomyelins/metabolism , Cholesterol/chemistry , Cholesterol/metabolism , Humans , Animals , Membrane Microdomains/metabolism , Membrane Microdomains/chemistry , Microscopy/methods , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Membrane/metabolism , Cell Membrane/chemistry
3.
J Coll Physicians Surg Pak ; 34(7): 811-816, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38978246

ABSTRACT

OBJECTIVE: To investigate the expression level of pyruvate kinase M1 (PKM1) in patients with acute myeloid leukaemia (AML) as well as its clinical significance. STUDY DESIGN: A case-control study. Place and Duration of the Study: Department of Haematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China, from January 2013 to 2023. METHODOLOGY: The expression levels of PKM1 and pyruvate kinase m2 (PKM2) in the bone marrow of 65 AML patients (excluding M3) and 31 healthy volunteers were determined using reverse transcription-quantitative polymerase chain reaction (RT-qPCR), a method that measures fluorescence in real-time. The associations between PKM1, PKM2 expressions, clinical parameters, and the survival and prognosis of AML patients were analysed. RESULTS: AML patients showed higher PKM1 expression compared to controls. The area under the curve (AUC) of the receiver operating characteristics (ROC) was 0.65 (p = 0.017). PKM1 expression was correlated with peripheral blood leukocyte count (r = -0.276, p = 0.026), CCAAT enhancer-binding protein alpha CEBPA mutation (r = -0.306, p = 0.014), and chemotherapy-induced response (r = -0.292, p = 0.018). Patients with high PKM1 expression had a lower remission rate (p = 0.019) and long-term survival rate (p = 0.034) than those with low PKM1 expression. Patients with AML showed a rise in PKM2 levels; however, the variation was not statistically significant (p >0.05). CONCLUSION: PKM1 expression is upregulated in AML and patients with high PKM1 expression have a lower survival rate. KEY WORDS: PKM1, Acute myeloid leukaemia, Clinical prognosis.


Subject(s)
Carrier Proteins , Leukemia, Myeloid, Acute , Membrane Proteins , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Humans , Leukemia, Myeloid, Acute/genetics , Male , Female , Middle Aged , Case-Control Studies , Adult , Thyroid Hormones/blood , Thyroid Hormones/metabolism , Prognosis , Carrier Proteins/genetics , Carrier Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , Aged , China/epidemiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Young Adult
4.
Nat Commun ; 15(1): 5761, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982055

ABSTRACT

While protein aggregation's association with aging and age-related diseases is well-established, the specific proteins involved and whether dissolving them could alleviate aging remain unclear. Our research addresses this gap by uncovering the role of PKM2 aggregates in aging. We find that PKM2 forms aggregates in senescent cells and organs from aged mice, impairing its enzymatic activity and glycolytic flux, thereby driving cells into senescence. Through a rigorous two-step small molecule library screening, we identify two compounds, K35 and its analog K27, capable of dissolving PKM2 aggregates and alleviating senescence. Further experiments show that treatment with K35 and K27 not only alleviate aging-associated signatures but also extend the lifespan of naturally and prematurely aged mice. These findings provide compelling evidence for the involvement of PKM2 aggregates in inducing cellular senescence and aging phenotypes, and suggest that targeting these aggregates could be a promising strategy for anti-aging drug discovery.


Subject(s)
Aging , Cellular Senescence , Thyroid Hormone-Binding Proteins , Animals , Aging/metabolism , Mice , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Carrier Proteins/metabolism , Glycolysis , Thyroid Hormones/metabolism , Protein Aggregates , Pyruvate Kinase/metabolism , Mice, Inbred C57BL , Male
5.
Methods Mol Biol ; 2821: 65-70, 2024.
Article in English | MEDLINE | ID: mdl-38997480

ABSTRACT

Conjugation to carrier proteins is necessary for peptides to be able to induce antibody formation when injected into animals together with a suitable adjuvant. This is usually performed by conjugation in solution followed by mixing with the adjuvant. Alternatively, the carrier may be adsorbed onto a solid support followed by activation and conjugation with the peptide by solid-phase chemistry. Different reagents can be used for conjugation through peptide functional groups (-SH, -NH2, -COOH), and various carrier proteins may be used depending on the peptides and the intended use of the antibodies. The solid phase may be an ion exchange matrix, from which the conjugate can subsequently be eluted and mixed with adjuvant. Alternatively, the adjuvant aluminum hydroxide may be used as the solid-phase matrix, whereupon the carrier is immobilized and conjugated with peptide. The resulting adjuvant-carrier-peptide complexes may then be used directly for immunization.


Subject(s)
Peptides , Peptides/chemistry , Animals , Adjuvants, Immunologic/chemistry , Aluminum Hydroxide/chemistry , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Solid-Phase Synthesis Techniques/methods
6.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39000596

ABSTRACT

The adaption of plants to stressful environments depends on long-distance responses in plant organs, which themselves are remote from sites of perception of external stimuli. Jasmonic acid (JA) and its derivatives are known to be involved in plants' adaptation to salinity. However, to our knowledge, the transport of JAs from roots to shoots has not been studied in relation to the responses of shoots to root salt treatment. We detected a salt-induced increase in the content of JAs in the roots, xylem sap, and leaves of pea plants related to changes in transpiration. Similarities between the localization of JA and lipid transfer proteins (LTPs) around vascular tissues were detected with immunohistochemistry, while immunoblotting revealed the presence of LTPs in the xylem sap of pea plants and its increase with salinity. Furthermore, we compared the effects of exogenous MeJA and salt treatment on the accumulation of JAs in leaves and their impact on transpiration. Our results indicate that salt-induced changes in JA concentrations in roots and xylem sap are the source of accumulation of these hormones in leaves leading to associated changes in transpiration. Furthermore, they suggest the possible involvement of LTPs in the loading/unloading of JAs into/from the xylem and its xylem transport.


Subject(s)
Carrier Proteins , Cyclopentanes , Oxylipins , Pisum sativum , Plant Leaves , Plant Proteins , Plant Roots , Xylem , Oxylipins/metabolism , Cyclopentanes/metabolism , Cyclopentanes/pharmacology , Pisum sativum/metabolism , Pisum sativum/drug effects , Plant Proteins/metabolism , Xylem/metabolism , Plant Roots/metabolism , Carrier Proteins/metabolism , Plant Leaves/metabolism , Biological Transport , Plant Growth Regulators/metabolism
7.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39013281

ABSTRACT

We previously identified talin rod domain-containing protein 1 (TLNRD1) as a potent actin-bundling protein in vitro. Here, we report that TLNRD1 is expressed in the vasculature in vivo. Its depletion leads to vascular abnormalities in vivo and modulation of endothelial cell monolayer integrity in vitro. We demonstrate that TLNRD1 is a component of the cerebral cavernous malformations (CCM) complex through its direct interaction with CCM2, which is mediated by a hydrophobic C-terminal helix in CCM2 that attaches to a hydrophobic groove on the four-helix domain of TLNRD1. Disruption of this binding interface leads to CCM2 and TLNRD1 accumulation in the nucleus and actin fibers. Our findings indicate that CCM2 controls TLNRD1 localization to the cytoplasm and inhibits its actin-bundling activity and that the CCM2-TLNRD1 interaction impacts endothelial actin stress fiber and focal adhesion formation. Based on these results, we propose a new pathway by which the CCM complex modulates the actin cytoskeleton and vascular integrity.


Subject(s)
Hemangioma, Cavernous, Central Nervous System , Human Umbilical Vein Endothelial Cells , Humans , Animals , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemangioma, Cavernous, Central Nervous System/pathology , Hemangioma, Cavernous, Central Nervous System/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Endothelial Cells/metabolism , Focal Adhesions/metabolism , Carrier Proteins/metabolism , Carrier Proteins/genetics , Stress Fibers/metabolism , Actins/metabolism , Actin Cytoskeleton/metabolism , Protein Binding , Mice , Cell Nucleus/metabolism , Talin
8.
Exp Dermatol ; 33(7): e15145, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39046322

ABSTRACT

Acne vulgaris, rosacea, and hidradenitis suppurativa are enduring inflammatory skin conditions that frequently manifest with akin clinical attributes, posing a considerable challenge for their distinctive diagnosis. While these conditions do exhibit certain resemblances, they also demonstrate distinct underlying pathophysiological mechanisms and treatment modalities. Delving into both the molecular parallels and disparities among these three disorders can yield invaluable insights for refined diagnostics, effective management, and targeted therapeutic interventions. In this report, we present a comparative analysis of transcriptomic data across these three diseases, elucidating differentially expressed genes and enriched pathways specific to each ailment, as well as those shared among them. Specifically, we identified multiple zinc-binding proteins (SERPINA1, S100A7, S100A8, S100A9 and KRT16) as consistently highly upregulated genes across all three diseases. Our hypothesis suggests that these proteins could bind and sequester zinc, potentially leading to localized zinc deficiency and heightened inflammation. We identified high-dose dietary zinc as a promising therapeutic approach and confirmed its effectiveness through validation in an acne mouse model.


Subject(s)
Acne Vulgaris , Gene Expression Profiling , Hidradenitis Suppurativa , Rosacea , Zinc , Acne Vulgaris/drug therapy , Acne Vulgaris/genetics , Zinc/therapeutic use , Zinc/metabolism , Rosacea/drug therapy , Rosacea/genetics , Hidradenitis Suppurativa/drug therapy , Hidradenitis Suppurativa/genetics , Animals , Mice , Humans , S100 Calcium Binding Protein A7/metabolism , S100 Calcium Binding Protein A7/genetics , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Transcriptome , S100 Proteins/genetics , S100 Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Disease Models, Animal , Up-Regulation
9.
Nutrients ; 16(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38999774

ABSTRACT

Dysregulation of zinc and zinc transporters families has been associated with the genesis and progression of prostate cancer. The prostate epithelium utilizes two types of zinc transporters, the ZIP (Zrt-, Irt-related Protein) and the ZnTs (Zinc Transporter), to transport zinc from the blood plasma to the gland lumen. ZIP transporters uptake zinc from extracellular space and organelle lumen, while ZnT transporters release zinc outside the cells or to organelle lumen. In prostate cancer, a commonly observed low zinc concentration in prostate tissue has been correlated with downregulations of certain ZIPs (e.g., ZIP1, ZIP2, ZIP3, ZIP14) and upregulations of specific ZnTs (e.g., ZnT1, ZnT9, ZnT10). These alterations may enable cancer cells to adapt to toxic high zinc levels. While zinc supplementation has been suggested as a potential therapy for this type of cancer, studies have yielded inconsistent results because some trials have indicated that zinc supplementation could exacerbate cancer risk. The reason for this discrepancy remains unclear, but given the high molecular and genetic variability present in prostate tumors, it is plausible that some zinc transporters-comprising 14 ZIP and 10 ZnT members-could be dysregulated in others patterns that promote cancer. From this perspective, this review highlights novel dysregulation, such as ZIP-Up/ZnT-Down, observed in prostate cancer cell lines for ZIP4, ZIP8, ZnT2, ZnT4, ZnT5, etc. Additionally, an in silico analysis of an available microarray from mouse models of prostate cancer (Nkx3.1;Pten) predicts similar dysregulation pattern for ZIP4, ZIP8, and ZnT2, which appear in early stages of prostate cancer progression. Furthermore, similar dysregulation patterns are supported by an in silico analysis of RNA-seq data from human cancer tumors available in cBioPortal. We discuss how these dysregulations of zinc transporters could impact zinc supplementation trials, particularly focusing on how the ZIP-Up/ZnT-Down dysregulation through various mechanisms might promote prostate cancer progression.


Subject(s)
Cation Transport Proteins , Prostatic Neoplasms , Zinc , Humans , Male , Prostatic Neoplasms/metabolism , Zinc/metabolism , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics , Animals , Carrier Proteins/metabolism , Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic , Dietary Supplements , Prostate/metabolism
10.
Sci Rep ; 14(1): 15740, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977862

ABSTRACT

Genome replication is frequently impeded by highly stable DNA secondary structures, including G-quadruplex (G4) DNA, that can hinder the progression of the replication fork. Human WRNIP1 (Werner helicase Interacting Protein 1) associates with various components of the replication machinery and plays a crucial role in genome maintenance processes. However, its detailed function is still not fully understood. Here we show that human WRNIP1 interacts with G4 structures and provide evidence for its contribution to G4 processing. The absence of WRNIP1 results in elevated levels of G4 structures, DNA damage and chromosome aberrations following treatment with PhenDC3, a G4-stabilizing ligand. Additionally, we establish a functional and physical relationship between WRNIP1 and the PIF1 helicase in G4 processing. In summary, our results suggest that WRNIP1 aids genome replication and maintenance by regulating G4 processing and this activity relies on Pif1 DNA helicase.


Subject(s)
DNA Helicases , DNA Replication , G-Quadruplexes , Humans , DNA Helicases/metabolism , DNA Damage , Chromosome Aberrations , Carrier Proteins/metabolism , Carrier Proteins/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics
11.
Cell Death Dis ; 15(7): 504, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39009589

ABSTRACT

Abnormal epigenetic modifications are involved in the regulation of Warburg effect in tumor cells. Protein arginine methyltransferases (PRMTs) mediate arginine methylation and have critical functions in cellular responses. PRMTs are deregulated in a variety of cancers, but their precise roles in Warburg effect in cancer is largely unknown. Experiments from the current study showed that PRMT1 was highly expressed under conditions of glucose sufficiency. PRMT1 induced an increase in the PKM2/PKM1 ratio through upregulation of PTBP1, in turn, promoting aerobic glycolysis in non-small cell lung cancer (NSCLC). The PRMT1 level in p53-deficient and p53-mutated NSCLC remained relatively unchanged while the expression was reduced in p53 wild-type NSCLC under conditions of glucose insufficiency. Notably, p53 activation under glucose-deficient conditions could suppress USP7 and further accelerate the polyubiquitin-dependent degradation of PRMT1. Melatonin, a hormone that inhibits glucose intake, markedly suppressed cell proliferation of p53 wild-type NSCLC, while a combination of melatonin and the USP7 inhibitor P5091 enhanced the anticancer activity in p53-deficient NSCLC. Our collective findings support a role of PRMT1 in the regulation of Warburg effect in NSCLC. Moreover, combination treatment with melatonin and the USP7 inhibitor showed good efficacy, providing a rationale for the development of PRMT1-based therapy to improve p53-deficient NSCLC outcomes.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Membrane Proteins , Protein-Arginine N-Methyltransferases , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Tumor Suppressor Protein p53 , Warburg Effect, Oncologic , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Warburg Effect, Oncologic/drug effects , Tumor Suppressor Protein p53/metabolism , Thyroid Hormones/metabolism , Cell Line, Tumor , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Proliferation/drug effects , Carrier Proteins/metabolism , Carrier Proteins/genetics , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Animals , Glycolysis/drug effects , Mice, Nude , Glucose/metabolism , Mice , Gene Expression Regulation, Neoplastic , A549 Cells , Polypyrimidine Tract-Binding Protein
12.
Cardiovasc Diabetol ; 23(1): 228, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951793

ABSTRACT

BACKGROUND: Glucokinase (GK) plays a key role in glucose metabolism. In the liver, GK is regulated by GK regulatory protein (GKRP) with nuclear sequestration at low plasma glucose level. Some GK activators (GKAs) disrupt GK-GKRP interaction which increases hepatic cytoplasmic GK level. Excess hepatic GK activity may exceed the capacity of glycogen synthesis with excess triglyceride formation. It remains uncertain whether hypertriglyceridemia associated with some GKAs in previous clinical trials was due to direct GK activation or impaired GK-GKRP interaction. METHODS: Using publicly available genome-wide association study summary statistics, we selected independent genetic variants of GCKR and GCK associated with fasting plasma glucose (FPG) as instrumental variables, to mimic the effects of impaired GK-GKRP interaction and direct GK activation, respectively. We applied two-sample Mendelian Randomization (MR) framework to assess their causal associations with lipid-related traits, risks of metabolic dysfunction-associated steatotic liver disease (MASLD) and cardiovascular diseases. We verified these findings in one-sample MR analysis using individual-level statistics from the Hong Kong Diabetes Register (HKDR). RESULTS: Genetically-proxied impaired GK-GKRP interaction increased plasma triglycerides, low-density lipoprotein cholesterol and apolipoprotein B levels with increased odds ratio (OR) of 14.6 (95% CI 4.57-46.4) per 1 mmol/L lower FPG for MASLD and OR of 2.92 (95% CI 1.78-4.81) for coronary artery disease (CAD). Genetically-proxied GK activation was associated with decreased risk of CAD (OR 0.69, 95% CI 0.54-0.88) and not with dyslipidemia. One-sample MR validation in HKDR showed consistent results. CONCLUSIONS: Impaired GK-GKRP interaction, rather than direct GK activation, may worsen lipid profiles and increase risks of MASLD and CAD. Development of future GKAs should avoid interfering with GK-GKRP interaction.


Subject(s)
Adaptor Proteins, Signal Transducing , Blood Glucose , Genetic Predisposition to Disease , Genome-Wide Association Study , Glucokinase , Mendelian Randomization Analysis , Humans , Adaptor Proteins, Signal Transducing/genetics , Risk Factors , Risk Assessment , Blood Glucose/metabolism , Glucokinase/genetics , Glucokinase/metabolism , Biomarkers/blood , Lipids/blood , Phenotype , Carrier Proteins/genetics , Carrier Proteins/metabolism , Polymorphism, Single Nucleotide , Time Factors , Dyslipidemias/genetics , Dyslipidemias/blood , Dyslipidemias/diagnosis , Dyslipidemias/epidemiology , Dyslipidemias/enzymology , Fatty Liver/genetics , Fatty Liver/enzymology , Fatty Liver/blood
13.
Sci Adv ; 10(29): eadl5638, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39018414

ABSTRACT

Viruses can selectively repress the translation of mRNAs involved in the antiviral response. RNA viruses exploit the Grb10-interacting GYF (glycine-tyrosine-phenylalanine) proteins 2 (GIGYF2) and eukaryotic translation initiation factor 4E (eIF4E) homologous protein 4EHP to selectively repress the translation of transcripts such as Ifnb1, which encodes the antiviral cytokine interferon-ß (IFN-ß). Herein, we reveal that GIGYF1, a paralog of GIGYF2, robustly represses cellular mRNA translation through a distinct 4EHP-independent mechanism. Upon recruitment to a target mRNA, GIGYF1 binds to subunits of eukaryotic translation initiation factor 3 (eIF3) at the eIF3-eIF4G1 interaction interface. This interaction disrupts the eIF3 binding to eIF4G1, resulting in transcript-specific translational repression. Depletion of GIGYF1 induces a robust immune response by derepressing IFN-ß production. Our study highlights a unique mechanism of translational regulation by GIGYF1 that involves sequestering eIF3 and abrogating its binding to eIF4G1. This mechanism has profound implications for the host response to viral infections.


Subject(s)
Eukaryotic Initiation Factor-3 , Eukaryotic Initiation Factor-4G , Protein Binding , RNA, Messenger , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-3/genetics , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Interferon-beta/metabolism , Interferon-beta/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Peptide Chain Initiation, Translational , Animals , Protein Biosynthesis , Gene Expression Regulation
14.
Biochemistry ; 63(14): 1824-1836, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38968244

ABSTRACT

Faced with the emergence of multiresistant microorganisms that affect human health, microbial agents have become a serious global threat, affecting human health and plant crops. Antimicrobial peptides have attracted significant attention in research for the development of new microbial control agents. This work's goal was the structural characterization and analysis of antifungal activity of chitin-binding peptides from Capsicum baccatum and Capsicum frutescens seeds on the growth of Candida and Fusarium species. Proteins were initially submitted to extraction in phosphate buffer pH 5.4 and subjected to chitin column chromatography. Posteriorly, two fractions were obtained for each species, Cb-F1 and Cf-F1 and Cb-F2 and Cf-F2, respectively. The Cb-F1 (C. baccatum) and Cf-F1 (C. frutescens) fractions did not bind to the chitin column. The electrophoresis results obtained after chromatography showed two major protein bands between 3.4 and 14.2 kDa for Cb-F2. For Cf-F2, three major bands were identified between 6.5 and 14.2 kDa. One band from each species was subjected to mass spectrometry, and both bands showed similarity to nonspecific lipid transfer protein. Candida albicans and Candida tropicalis had their growth inhibited by Cb-F2. Cf-F2 inhibited the development of C. albicans but did not inhibit the growth of C. tropicalis. Both fractions were unable to inhibit the growth of Fusarium species. The toxicity of the fractions was tested in vivo on Galleria mellonella larvae, and both showed a low toxicity rate at high concentrations. As a result, the fractions have enormous promise for the creation of novel antifungal compounds.


Subject(s)
Antifungal Agents , Candida , Chitin , Fusarium , Molecular Docking Simulation , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Antifungal Agents/metabolism , Chitin/chemistry , Chitin/metabolism , Fusarium/drug effects , Candida/drug effects , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Animals , Capsicum/chemistry , Plant Proteins/chemistry , Plant Proteins/metabolism , Plant Proteins/pharmacology , Microbial Sensitivity Tests , Protein Binding , Protein Conformation
15.
BMC Nephrol ; 25(1): 227, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020292

ABSTRACT

BACKGROUND: End-stage and acquired cystic renal disease (ESRD/ACRD) kidneys are characterized by inflammatory remodelling and multiplex renal cell carcinomas (RCC). Eosinophilic vacuolated tumour (EVT) occurs exclusively in ACRD. The aim of this study was to identify the involvement of thioredoxin-interacting protein (TXNIP) and thioredoxin (TXN) in ESRD/ACRD pathology. METHODS: Expression of TXNIP and TXN was examined in histological slides of 6 ESRD and 6 ACRD kidneys, precursor lesions and associated tumours as well as of RCCs from the general population by immunohistochemistry. RESULTS: Strong TXNIP expression was seen in epithelial cells, myo-fibroblasts and endothelial cells and weak TXN expression in ESRD/ACRD kidneys and tumours. In ACRD specific EVT and its precursors TXN were translocated into nuclei. CONCLUSION: The impaired TXNIP/TXN redox homeostasis might be associated with development of multiplex cancer especially of EVT in ESRD/ACRD kidney.


Subject(s)
Carrier Proteins , Kidney Failure, Chronic , Kidney Neoplasms , Thioredoxins , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Carrier Proteins/metabolism , Thioredoxins/metabolism , Kidney Failure, Chronic/metabolism , Male , Middle Aged , Female , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Aged , Cell Nucleus/metabolism , Adult , Active Transport, Cell Nucleus
16.
PeerJ ; 12: e17625, 2024.
Article in English | MEDLINE | ID: mdl-38948221

ABSTRACT

Plasmodesmata are transmembrane channels embedded within the cell wall that can facilitate the intercellular communication in plants. Plasmodesmata callose-binding (PDCB) protein that associates with the plasmodesmata contributes to cell wall extension. Given that the elongation of cotton fiber cells correlates with the dynamics of the cell wall, this protein can be related to the cotton fiber elongation. This study sought to identify PDCB family members within the Gossypium. hirsutum genome and to elucidate their expression profiles. A total of 45 distinct family members were observed through the identification and screening processes. The analysis of their physicochemical properties revealed the similarity in the amino acid composition and molecular weight across most members. The phylogenetic analysis facilitated the construction of an evolutionary tree, categorizing these members into five groups mainly distributed on 20 chromosomes. The fine mapping results facilitated a tissue-specific examination of group V, revealing that the expression level of GhPDCB9 peaked five days after flowering. The VIGS experiments resulted in a marked decrease in the gene expression level and a significant reduction in the mature fiber length, averaging a shortening of 1.43-4.77 mm. The results indicated that GhPDCB9 played a pivotal role in the cotton fiber development and served as a candidate for enhancing cotton yield.


Subject(s)
Cotton Fiber , Gossypium , Phylogeny , Plant Proteins , Plasmodesmata , Gossypium/genetics , Gossypium/metabolism , Plasmodesmata/metabolism , Cotton Fiber/analysis , Plant Proteins/genetics , Plant Proteins/metabolism , Gene Expression Regulation, Plant , Glucans/metabolism , Multigene Family , Cell Wall/metabolism , Cell Wall/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism
17.
Sci Rep ; 14(1): 15422, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38965264

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is an inherited disorder characterized by left ventricular hypertrophy and diastolic dysfunction, and increases the risk of arrhythmias and heart failure. Some patients with HCM develop a dilated phase of hypertrophic cardiomyopathy (D-HCM) and have poor prognosis; however, its pathogenesis is unclear and few pathological models exist. This study established disease-specific human induced pluripotent stem cells (iPSCs) from a patient with D-HCM harboring a mutation in MYBPC3 (c.1377delC), a common causative gene of HCM, and investigated the associated pathophysiological mechanisms using disease-specific iPSC-derived cardiomyocytes (iPSC-CMs). We confirmed the expression of pluripotent markers and the ability to differentiate into three germ layers in D-HCM patient-derived iPSCs (D-HCM iPSCs). D-HCM iPSC-CMs exhibited disrupted myocardial sarcomere structures and an increased number of damaged mitochondria. Ca2+ imaging showed increased abnormal Ca2+ signaling and prolonged decay time in D-HCM iPSC-CMs. Cell metabolic analysis revealed increased basal respiration, maximal respiration, and spare-respiratory capacity in D-HCM iPSC-CMs. RNA sequencing also showed an increased expression of mitochondrial electron transport system-related genes. D-HCM iPSC-CMs showed abnormal Ca2+ handling and hypermetabolic state, similar to that previously reported for HCM patient-derived iPSC-CMs. Although further studies are required, this is expected to be a useful pathological model for D-HCM.


Subject(s)
Calcium , Cardiomyopathy, Hypertrophic , Carrier Proteins , Frameshift Mutation , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Induced Pluripotent Stem Cells/metabolism , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Calcium/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Calcium Signaling , Cell Differentiation , Male
18.
J Cell Biol ; 223(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39007803

ABSTRACT

Stress triggers the formation of two distinct cytoplasmic biomolecular condensates: stress granules (SGs) and processing bodies (PBs), both of which may contribute to stress-responsive translation regulation. Though PBs can be present constitutively, stress can increase their number and size and lead to their interaction with stress-induced SGs. The mechanism of such interaction, however, is largely unknown. Formation of canonical SGs requires the RNA binding protein Ubiquitin-Associated Protein 2-Like (UBAP2L), which is a central SG node protein in the RNA-protein interaction network of SGs and PBs. UBAP2L binds to the essential SG and PB proteins G3BP and DDX6, respectively. Research on UBAP2L has mostly focused on its role in SGs, but not its connection to PBs. We find that UBAP2L is not solely an SG protein but also localizes to PBs in certain conditions, contributes to PB biogenesis and SG-PB interactions, and can nucleate hybrid granules containing SG and PB components in cells. These findings inform a new model for SG and PB formation in the context of UBAP2L's role.


Subject(s)
RNA Helicases , RNA Recognition Motif Proteins , Stress Granules , Humans , Stress Granules/metabolism , Stress Granules/genetics , RNA Helicases/metabolism , RNA Helicases/genetics , RNA Recognition Motif Proteins/metabolism , RNA Recognition Motif Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Processing Bodies/metabolism , Processing Bodies/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Cytoplasmic Granules/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/genetics , HeLa Cells , DNA Helicases/metabolism , DNA Helicases/genetics , HEK293 Cells , Protein Binding , Carrier Proteins/metabolism , Carrier Proteins/genetics , Proto-Oncogene Proteins
19.
Adv Exp Med Biol ; 1459: 199-215, 2024.
Article in English | MEDLINE | ID: mdl-39017845

ABSTRACT

BCL11A, a zinc finger repressor, is a stage-specific transcription factor that controls the switch from fetal (HbF, α2γ2) to adult (HbA, α2ß2) hemoglobin in erythroid cells. While BCL11A was known as a factor critical for B-lymphoid cell development, its relationship to erythroid cells and HbF arose through genome-wide association studies (GWAS). Subsequent work validated its role as a silencer of γ-globin gene expression in cultured cells and mice. Erythroid-specific loss of BCL11A rescues the phenotype of engineered sickle cell disease (SCD) mice, thereby suggesting that downregulation of BCL11A expression might be beneficial in patients with SCD and ß-thalassemia. Common genetic variation in GWAS resides in an erythroid-specific enhancer within the BCL11A gene that is required for its own expression. CRISPR/Cas9 gene editing of the enhancer revealed a GATA-binding site that confers a large portion of its regulatory function. Disruption of the GATA site leads to robust HbF reactivation. Advancement of a guide RNA targeting the GATA-binding site in clinical trials has recently led to approval of first-in-man use of ex vivo CRISPR editing of hematopoietic stem/progenitor cells (HSPCs) as therapy of SCD and ß-thalassemia. Future challenges include expanding access and infrastructure for delivery of genetic therapy to eligible patients, reducing potential toxicity and costs, exploring prospects for in vivo targeting of hematopoietic stem cells (HSCs), and developing small molecule drugs that impair function of BCL11A protein as an alternative option.


Subject(s)
Erythroid Cells , Repressor Proteins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Humans , Animals , Erythroid Cells/metabolism , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Mice , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , CRISPR-Cas Systems , Gene Editing/methods , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , gamma-Globins/genetics , gamma-Globins/metabolism , Gene Expression Regulation , Genome-Wide Association Study
20.
CNS Neurosci Ther ; 30(6): e14754, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38884369

ABSTRACT

AIMS: Islet cell autoantigen 1 (ICA1) is involved in autoimmune diseases and may affect synaptic plasticity as a neurotransmitter. Databases related to Alzheimer's disease (AD) have shown decreased ICA1 expression in patients with AD. However, the role of ICA1 in AD remains unclear. Here, we report that ICA1 expression is decreased in the brains of patients with AD and an AD mouse model. RESULTS: The ICA1 increased the expression of amyloid precursor protein (APP), disintegrin and metalloprotease 10 (ADAM10), and disintegrin and metalloprotease 17 (ADAM17), but did not affect protein half-life or mRNA levels. Transcriptome sequencing analysis showed that ICA1 regulates the G protein-coupled receptor signaling pathway. The overexpression of ICA1 increased PKCα protein levels and phosphorylation. CONCLUSION: Our results demonstrated that ICA1 shifts APP processing to non-amyloid pathways by regulating the PICK1-PKCα signaling pathway. Thus, this study suggests that ICA1 is a novel target for the treatment of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Protein Kinase C-alpha , Signal Transduction , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Protein Kinase C-alpha/metabolism , Protein Kinase C-alpha/genetics , Signal Transduction/physiology , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Mice , Carrier Proteins/metabolism , Carrier Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Male , Mice, Transgenic , Female , Mice, Inbred C57BL , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Brain/metabolism , Cell Cycle Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...