Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.704
Filter
1.
Artif Cells Nanomed Biotechnol ; 52(1): 384-398, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39101753

ABSTRACT

Nanotechnology-based cancer treatment has received considerable attention, and these treatments generally use drug-loaded nanoparticles (NPs) to target and destroy cancer cells. Nanotechnology combined with photodynamic therapy (PDT) has demonstrated positive outcomes in cancer therapy. Combining nanotechnology and PDT is effective in targeting metastatic cancer cells. Nanotechnology can also increase the effectiveness of PDT by targeting cells at a molecular level. Dendrimer-based nanoconjugates (DBNs) are highly stable and biocompatible, making them suitable for drug delivery applications. Moreover, the hyperbranched structures in DBNs have the capacity to load hydrophobic compounds, such as photosensitizers (PSs) and chemotherapy drugs, and deliver them efficiently to tumour cells. This review primarily focuses on DBNs and their potential applications in cancer treatment. We discuss the chemical design, mechanism of action, and targeting efficiency of DBNs in tumour metastasis, intracellular trafficking in cancer treatment, and DBNs' biocompatibility, biodegradability and clearance properties. Overall, this study will provide the most recent insights into the application of DBNs and PDT in cancer therapy.


DBNs' intracellular journey in cancer-PDT refines targeted therapy, boosting efficacy.DBN in PDT for tumour metastasis: targeting and drug release mechanisms.DBNs' biocompatibility, biodegradability and clearance were explored thoroughly.


Subject(s)
Dendrimers , Nanoconjugates , Neoplasms , Photochemotherapy , Humans , Dendrimers/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Nanoconjugates/chemistry , Nanoconjugates/therapeutic use , Animals , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Biological Transport , Intracellular Space/metabolism , Intracellular Space/drug effects , Drug Carriers/chemistry
2.
Foodborne Pathog Dis ; 21(7): 447-457, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38985570

ABSTRACT

Bacillus cereus causes food poisoning by producing toxins that cause diarrhea and vomiting and, in severe cases, endocarditis, meningitis, and other diseases. It also tends to form biofilms and spores that lead to contamination of the food production environment. Citral is a potent natural antibacterial agent, but its antibacterial activity against B. cereus has not been extensively studied. In this study, we first determined the minimum inhibitory concentrations and minimum bactericidal concentrations, growth curves, killing effect in different media, membrane potential, intracellular adenosine triphosphate (ATP), reactive oxygen species levels, and morphology of vegetative cells, followed by germination rate, morphology, germination state of spores, and finally biofilm clearance effect. The results showed that the minimum inhibitory concentrations and minimum bactericidal concentrations of citral against bacteria ranged from 100 to 800 µg/mL. The lag phase of bacteria was effectively prolonged by citral, and the growth rate of bacteria was slowed down. Bacteria in Luria-Bertani broth were reduced to below the detection limit by citral at 800 µg/mL within 0.5 h. Bacteria in rice were reduced to 3 log CFU/g by citral at 4000 µg/mL within 0.5 h. After treatment with citral, intracellular ATP concentration was reduced, membrane potential was altered, intracellular reactive oxygen species concentration was increased, and normal cell morphology was altered. After treatment with citral at 400 µg/mL, spore germination rate was reduced to 16.71%, spore morphology was affected, and spore germination state was altered. It also had a good effect on biofilm removal. The present study showed that citral had good bacteriostatic activity against B. cereus vegetative cells and its spores and also had a good clearance effect on its biofilm. Citral has the potential to be used as a bacteriostatic substance for the control of B. cereus in food industry production.


Subject(s)
Acyclic Monoterpenes , Bacillus cereus , Biofilms , Acyclic Monoterpenes/pharmacology , Anti-Infective Agents/pharmacology , Bacillus cereus/drug effects , Bacillus cereus/growth & development , Bacillus cereus/ultrastructure , Spores, Bacterial/drug effects , Biofilms/drug effects , Microbial Sensitivity Tests , Oryza/microbiology , Membrane Potentials/drug effects , Intracellular Space/enzymology , Adenosine Triphosphate/metabolism , Reactive Oxygen Species/metabolism , Microscopy, Electron, Scanning , Food Microbiology
3.
Semin Immunopathol ; 46(1-2): 4, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38990375

ABSTRACT

In the galectin family, a group of lectins is united by their evolutionarily conserved carbohydrate recognition domains. These polypeptides play a role in various cellular processes and are implicated in disease mechanisms such as cancer, fibrosis, infection, and inflammation. Following synthesis in the cytosol, manifold interactions of galectins have been described both extracellularly and intracellularly. Extracellular galectins frequently engage with glycoproteins or glycolipids in a carbohydrate-dependent manner. Intracellularly, galectins bind to non-glycosylated proteins situated in distinct cellular compartments, each with multiple cellular functions. This diversity complicates attempts to form a comprehensive understanding of the role of galectin molecules within the cell. This review enumerates intracellular galectin interaction partners and outlines their involvement in cellular processes. The intricate connections between galectin functions and pathomechanisms are illustrated through discussions of intracellular galectin assemblies in immune and cancer cells. This underscores the imperative need to fully comprehend the interplay of galectins with the cellular machinery and to devise therapeutic strategies aimed at counteracting the establishment of galectin-based disease mechanisms.


Subject(s)
Galectins , Neoplasms , Humans , Galectins/metabolism , Animals , Neoplasms/metabolism , Neoplasms/etiology , Neoplasms/pathology , Protein Binding , Disease Susceptibility , Inflammation/metabolism , Intracellular Space/metabolism
4.
Nature ; 630(8018): 943-949, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38898271

ABSTRACT

Spatial transcriptomics measures in situ gene expression at millions of locations within a tissue1, hitherto with some trade-off between transcriptome depth, spatial resolution and sample size2. Although integration of image-based segmentation has enabled impactful work in this context, it is limited by imaging quality and tissue heterogeneity. By contrast, recent array-based technologies offer the ability to measure the entire transcriptome at subcellular resolution across large samples3-6. Presently, there exist no approaches for cell type identification that directly leverage this information to annotate individual cells. Here we propose a multiscale approach to automatically classify cell types at this subcellular level, using both transcriptomic information and spatial context. We showcase this on both targeted and whole-transcriptome spatial platforms, improving cell classification and morphology for human kidney tissue and pinpointing individual sparsely distributed renal mouse immune cells without reliance on image data. By integrating these predictions into a topological pipeline based on multiparameter persistent homology7-9, we identify cell spatial relationships characteristic of a mouse model of lupus nephritis, which we validate experimentally by immunofluorescence. The proposed framework readily generalizes to new platforms, providing a comprehensive pipeline bridging different levels of biological organization from genes through to tissues.


Subject(s)
Cells , Gene Expression Profiling , Intracellular Space , Kidney , Transcriptome , Animals , Female , Humans , Mice , Cells/classification , Cells/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Kidney/cytology , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Lupus Nephritis/pathology , Reproducibility of Results , Intracellular Space/genetics , Intracellular Space/metabolism
5.
Cell Commun Signal ; 22(1): 289, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802925

ABSTRACT

A cell is a dynamic system in which various processes occur simultaneously. In particular, intra- and intercellular signaling pathway crosstalk has a significant impact on a cell's life cycle, differentiation, proliferation, growth, regeneration, and, consequently, on the normal functioning of an entire organ. Hippo signaling and YAP/TAZ nucleocytoplasmic shuttling play a pivotal role in normal development, homeostasis, and tissue regeneration, particularly in lung cells. Intersignaling communication has a significant impact on the core components of the Hippo pathway and on YAP/TAZ localization. This review describes the crosstalk between Hippo signaling and key lung signaling pathways (WNT, SHH, TGFß, Notch, Rho, and mTOR) using lung cells as an example and highlights the remaining unanswered questions.


Subject(s)
Lung , Signal Transduction , Transcription Factors , Humans , Lung/metabolism , Lung/cytology , Animals , Transcription Factors/metabolism , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Hippo Signaling Pathway , Intracellular Space/metabolism
6.
Adv Mater ; 36(25): e2400425, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38574376

ABSTRACT

Active transcytosis-mediated nanomedicine transport presents considerable potential in overcoming diverse delivery barriers, thereby facilitating tumor accumulation and penetration. Nevertheless, the persistent challenge lies in achieving a nuanced equilibrium between intracellular interception for drug release and transcytosis for tumor penetration. In this study, a comprehensive exploration is conducted involving a series of polyglutamine-paclitaxel conjugates featuring distinct hydrophilic/hydrophobic ratios (HHR) and tertiary amine-oxide proportions (TP) (OPGA-PTX). The screening process, meticulously focused on delineating their subcellular distribution, transcytosis capability, and tumor penetration, unveils a particularly promising candidate denoted as OPPX, characterized by an HHR of 10:1 and a TP of 100%. OPPX, distinguished by its rapid cellular internalization through multiple endocytic pathways, selectively engages in trafficking to the Golgi apparatus for transcytosis to facilitate accumulation within and penetration throughout tumor tissues and simultaneously sorted to lysosomes for cathepsin B-activated drug release. This study not only identifies OPPX as an exemplary nanomedicine but also underscores the feasibility of modulating subcellular distribution to optimize the active transport capabilities and intracellular release mechanisms of nanomedicines, providing an alternative approach to designing efficient anticancer nanomedicines.


Subject(s)
Paclitaxel , Transcytosis , Humans , Paclitaxel/pharmacology , Paclitaxel/chemistry , Animals , Drug Liberation , Cell Line, Tumor , Drug Carriers/chemistry , Mice , Intracellular Space/metabolism , Hydrophobic and Hydrophilic Interactions , Lysosomes/metabolism
7.
J Cell Physiol ; 239(6): e31272, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38646844

ABSTRACT

The inhibition of cell surface crystal adhesion and an appropriate increase in crystal endocytosis contribute to the inhibition of kidney stone formation. In this study, we investigated the effects of different degrees of carboxymethylation on these processes. An injury model was established by treating human renal proximal tubular epithelial (HK-2) cells with 98.3 ± 8.1 nm calcium oxalate dihydrate (nanoCOD) crystals. The HK-2 cells were protected with carboxy (-COOH) Desmodium styracifolium polysaccharides at 1.17% (DSP0), 7.45% (CDSP1), 12.2% (CDSP2), and 17.7% (CDSP3). Changes in biochemical indexes and effects on nanoCOD adhesion and endocytosis were detected. The protection of HK-2 cells from nanoCOD-induced oxidative damage by carboxymethylated Desmodium styracifolium polysaccharides (CDSPs) is closely related to the protection of subcellular organelles, such as mitochondria. CDSPs can reduce crystal adhesion on the cell surface and maintain appropriate crystal endocytosis, thereby reducing the risk of kidney stone formation. CDSP2 with moderate -COOH content showed the strongest protective activity among the CDSPs.


Subject(s)
Calcium Oxalate , Endocytosis , Kidney Calculi , Polysaccharides , Humans , Calcium Oxalate/metabolism , Cell Adhesion/drug effects , Cell Line , Crystallization , Endocytosis/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Kidney Calculi/prevention & control , Kidney Calculi/drug therapy , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/metabolism , Oxidative Stress/drug effects , Polysaccharides/pharmacology , Polysaccharides/chemistry , Cell Survival/drug effects , Cell Cycle/drug effects , Calcium/metabolism , Intracellular Space/metabolism , Reactive Oxygen Species/metabolism , Membrane Potential, Mitochondrial/drug effects
8.
Nature ; 628(8008): 664-671, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38600377

ABSTRACT

Bitter taste sensing is mediated by type 2 taste receptors (TAS2Rs (also known as T2Rs)), which represent a distinct class of G-protein-coupled receptors1. Among the 26 members of the TAS2Rs, TAS2R14 is highly expressed in extraoral tissues and mediates the responses to more than 100 structurally diverse tastants2-6, although the molecular mechanisms for recognizing diverse chemicals and initiating cellular signalling are still poorly understood. Here we report two cryo-electron microscopy structures for TAS2R14 complexed with Ggust (also known as gustducin) and Gi1. Both structures have an orthosteric binding pocket occupied by endogenous cholesterol as well as an intracellular allosteric site bound by the bitter tastant cmpd28.1, including a direct interaction with the α5 helix of Ggust and Gi1. Computational and biochemical studies validate both ligand interactions. Our functional analysis identified cholesterol as an orthosteric agonist and the bitter tastant cmpd28.1 as a positive allosteric modulator with direct agonist activity at TAS2R14. Moreover, the orthosteric pocket is connected to the allosteric site via an elongated cavity, which has a hydrophobic core rich in aromatic residues. Our findings provide insights into the ligand recognition of bitter taste receptors and suggest activities of TAS2R14 beyond bitter taste perception via intracellular allosteric tastants.


Subject(s)
Cholesterol , Intracellular Space , Receptors, G-Protein-Coupled , Taste , Humans , Allosteric Regulation/drug effects , Allosteric Site , Cholesterol/chemistry , Cholesterol/metabolism , Cholesterol/pharmacology , Cryoelectron Microscopy , Hydrophobic and Hydrophilic Interactions , Intracellular Space/chemistry , Intracellular Space/metabolism , Ligands , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/ultrastructure , Reproducibility of Results , Taste/drug effects , Taste/physiology , Transducin/chemistry , Transducin/metabolism , Transducin/ultrastructure
9.
Sci Total Environ ; 930: 172695, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38663613

ABSTRACT

General control non-derepressible-2 (GCN2) is widely expressed in eukaryotes and responds to biotic and abiotic stressors. However, the precise function and mechanism of action of GCN2 in response to cadmium (Cd) stress in Nicotiana tabacum L. (tobacco) remains unclear. We investigated the role of NtGCN2 in Cd tolerance and explored the mechanism by which NtGCN2 responds to Cd stress in tobacco by exposing NtGCN2 transgenic tobacco lines to different concentrations of CdCl2. NtGCN2 was activated under 50 µmol·L-1 CdCl2 stress and enhanced the Cd tolerance and photosynthetic capacities of tobacco by increasing chlorophyll content and antioxidant capacity by upregulating NtSOD, NtPOD, and NtCAT expression and corresponding enzyme activities and decreasing malondialdehyde and O2·- contents. NtGCN2 enhanced the osmoregulatory capacity of tobacco by elevating proline (Pro) and soluble sugar contents and maintaining low levels of relative conductivity. Finally, NtGCN2 enhanced Cd tolerance in tobacco by reducing Cd uptake and translocation, promoting Cd efflux, and regulating Cd subcellular distribution. In conclusion, NtGCN2 improves the tolerance of tobacco to Cd through a series of mechanisms, namely, increasing antioxidant, photosynthetic, and osmoregulation capacities and regulating Cd uptake, translocation, efflux, and subcellular distribution. This study provides a scientific basis for further exploration of the role of NtGCN2 in plant responses to Cd stress and enhancement of the Cd stress signaling network in tobacco.


Subject(s)
Cadmium , Drug Resistance , Nicotiana , Plant Proteins , Cadmium/toxicity , Cadmium/metabolism , Nicotiana/physiology , Nicotiana/metabolism , Photosynthesis/drug effects , Photosynthesis/genetics , Plant Proteins/metabolism , Plant Proteins/genetics , Soil Pollutants/metabolism , Soil Pollutants/toxicity , Stress, Physiological/drug effects , Stress, Physiological/genetics , Chlorophyll/metabolism , Plant Leaves/drug effects , Plant Leaves/genetics , Plant Leaves/metabolism , Drug Resistance/genetics , Oxidoreductases/genetics , Oxidoreductases/metabolism , Enzyme Activation/genetics , Osmoregulation/genetics , Intracellular Space/metabolism
10.
New Phytol ; 242(6): 2430-2439, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38586981

ABSTRACT

Calcium ion (Ca2+) serves as a versatile and conserved second messenger in orchestrating immune responses. In plants, plasma membrane-localized Ca2+-permeable channels can be activated to induce Ca2+ influx from extracellular space to cytosol upon pathogen infection. Notably, different immune elicitors can induce dynamic Ca2+ signatures in the cytosol. During pattern-triggered immunity, there is a rapid and transient increase in cytosolic Ca2+, whereas in effector-triggered immunity, the elevation of cytosolic Ca2+ is strong and sustained. Numerous Ca2+ sensors are localized in the cytosol or different intracellular organelles, which are responsible for detecting and converting Ca2+ signals. In fact, Ca2+ signaling coordinated by cytosol and subcellular compartments plays a crucial role in activating plant immune responses. However, the complete Ca2+ signaling network in plant cells is still largely ambiguous. This review offers a comprehensive insight into the collaborative role of intracellular Ca2+ stores in shaping the Ca2+ signaling network during plant immunity, and several intriguing questions for future research are highlighted.


Subject(s)
Calcium Signaling , Calcium , Plant Immunity , Calcium/metabolism , Cytosol/metabolism , Intracellular Space/metabolism , Models, Biological
11.
Adv Mater ; 36(25): e2314184, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38459829

ABSTRACT

Longitudinal analysis of intracellular contents including gene and protein expression is crucial for deciphering the fundamentally dynamic nature of cells. This offers invaluable insights into complex tissue composition and behavior, and drives progress in disease diagnosis, biomarker discovery, and drug development. Traditional longitudinal analysis workflows, involving the destruction of cells at various timepoints, limit insights to singular moments and fail to account for cellular heterogeneity. Current non-destructive approaches, like temporal modeling with single-cell ribonucleic acid sequencing (RNA-seq) and live-cell fluorescence imaging, either rely on biological assumptions or possess the risk of cellular perturbation. Recent advances in nanoscale technologies for non-destructive intracellular content extraction offer a promising solution to these challenges. These novel methods work at the nanoscale to non-destructively access cellular membranes and can be broadly classified into three mechanisms: tip-facilitated aspiration, membrane-based, and probe-based methods. This perspective focuses on these emerging nanotechnologies for repeated intracellular content extraction. Their potential in longitudinal analysis is discussed, the critical requirements for effective repeated sampling are addressed, and the suitability of each technique for various applications is explored. Furthermore, unresolved challenges in repeated sampling are highlighted to encourage further research in this growing field.


Subject(s)
Nanotechnology , Humans , Nanotechnology/methods , Animals , Intracellular Space/metabolism
12.
Small ; 20(30): e2308562, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38441369

ABSTRACT

Diagnosis and treatment of tumor especially drug-resistant tumor remains a huge challenge, which requires intelligent nanomedicines with low toxic side effects and high efficacy. Herein, deformable smart DNA nanomachines are developed for synergistic intracellular cancer-related miRNAs imaging and chemo-gene therapy of drug-resistant tumors. The tetrahedral DNA framework (MA-TDNA) with fluorescence quenched component and five antennas is self-assembled first, and then DOX molecules are loaded on the MA-TDNAs followed by linking MUC1-aptamer and Mcl-1 siRNA to the antennas of MA-TDNA, so that the apt-MA-TDNA@DOX-siRNA (DNA nanomachines) is constructed. The DNA nanomachine can respond to two tumor-related miRNAs in vitro and in vivo, which can undergo intelligent miRNA-triggered opening of the framework, resulting in the "turn on" of the fluorescence for sensitively and specifically sensing intracellular miRNAs. Meanwhile, both miRNA-responded rapid release and pH-responded release of DOX are achieved for chemotherapy of tumor. In addition, the gene therapy of the DNA nanomachines is achieved due to the miRNA-specific capture and the RNase H triggered release of Mcl-1 siRNA. The DNA nanomachines intergrading both tumor imaging and chemo-gene therapy in single nanostructures realized efficient tumor-targeted, image-guided, and microenvironment-responsive tumor diagnosis and treatment, which provides a synergetic antitumor effect on drug-resistant tumor.


Subject(s)
DNA , Doxorubicin , Drug Resistance, Neoplasm , Genetic Therapy , MicroRNAs , MicroRNAs/genetics , Humans , Drug Resistance, Neoplasm/drug effects , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Genetic Therapy/methods , DNA/chemistry , Animals , Neoplasms/therapy , Neoplasms/diagnostic imaging , Neoplasms/genetics , RNA, Small Interfering , Cell Line, Tumor , Intracellular Space/metabolism
13.
J Biol Chem ; 300(4): 107132, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38432636

ABSTRACT

Heme is an iron-containing prosthetic group necessary for the function of several proteins termed "hemoproteins." Erythrocytes contain most of the body's heme in the form of hemoglobin and contain high concentrations of free heme. In nonerythroid cells, where cytosolic heme concentrations are 2 to 3 orders of magnitude lower, heme plays an essential and often overlooked role in a variety of cellular processes. Indeed, hemoproteins are found in almost every subcellular compartment and are integral in cellular operations such as oxidative phosphorylation, amino acid metabolism, xenobiotic metabolism, and transcriptional regulation. Growing evidence reveals the participation of heme in dynamic processes such as circadian rhythms, NO signaling, and the modulation of enzyme activity. This dynamic view of heme biology uncovers exciting possibilities as to how hemoproteins may participate in a range of physiologic systems. Here, we discuss how heme is regulated at the level of its synthesis, availability, redox state, transport, and degradation and highlight the implications for cellular function and whole organism physiology.


Subject(s)
Cell Physiological Phenomena , Heme , Animals , Humans , Circadian Rhythm/physiology , Heme/metabolism , Hemeproteins/metabolism , Oxidation-Reduction , Signal Transduction , Intracellular Space/metabolism , Cell Physiological Phenomena/physiology
14.
J Radiat Res ; 65(3): 263-271, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38461549

ABSTRACT

Ionizing radiation (IR)-induced double-strand breaks (DSBs) are primarily repaired by non-homologous end joining or homologous recombination (HR) in human cells. DSB repair requires adenosine-5'-triphosphate (ATP) for protein kinase activities in the multiple steps of DSB repair, such as DNA ligation, chromatin remodeling, and DNA damage signaling via protein kinase and ATPase activities. To investigate whether low ATP culture conditions affect the recruitment of repair proteins at DSB sites, IR-induced foci were examined in the presence of ATP synthesis inhibitors. We found that p53 binding protein 1 foci formation was modestly reduced under low ATP conditions after IR, although phosphorylated histone H2AX and mediator of DNA damage checkpoint 1 foci formation were not impaired. Next, we examined the foci formation of breast cancer susceptibility gene I (BRCA1), replication protein A (RPA) and radiation 51 (RAD51), which are HR factors, in G2 phase cells following IR. Interestingly, BRCA1 and RPA foci in the G2 phase were significantly reduced under low ATP conditions compared to that under normal culture conditions. Notably, RAD51 foci were drastically impaired under low ATP conditions. These results suggest that HR does not effectively progress under low ATP conditions; in particular, ATP shortages impair downstream steps in HR, such as RAD51 loading. Taken together, these results suggest that the maintenance of cellular ATP levels is critical for DNA damage response and HR progression after IR.


Subject(s)
Adenosine Triphosphate , BRCA1 Protein , Homologous Recombination , Rad51 Recombinase , Radiation, Ionizing , Humans , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/biosynthesis , Homologous Recombination/radiation effects , Rad51 Recombinase/metabolism , BRCA1 Protein/metabolism , DNA Breaks, Double-Stranded/radiation effects , Replication Protein A/metabolism , Cell Line, Tumor , Intracellular Space/metabolism , Intracellular Space/radiation effects , DNA Repair , Histones/metabolism
15.
Annu Rev Biophys ; 53(1): 193-220, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38346244

ABSTRACT

The interior of a living cell is an active, fluctuating, and crowded environment, yet it maintains a high level of coherent organization. This dichotomy is readily apparent in the intracellular transport system of the cell. Membrane-bound compartments called endosomes play a key role in carrying cargo, in conjunction with myriad components including cargo adaptor proteins, membrane sculptors, motor proteins, and the cytoskeleton. These components coordinate to effectively navigate the crowded cell interior and transport cargo to specific intracellular locations, even though the underlying protein interactions and enzymatic reactions exhibit stochastic behavior. A major challenge is to measure, analyze, and understand how, despite the inherent stochasticity of the constituent processes, the collective outcomes show an emergent spatiotemporal order that is precise and robust. This review focuses on this intriguing dichotomy, providing insights into the known mechanisms of noise suppression and noise utilization in intracellular transport processes, and also identifies opportunities for future inquiry.


Subject(s)
Stochastic Processes , Biological Transport , Humans , Models, Biological , Animals , Endosomes/metabolism , Intracellular Space/metabolism
16.
Small ; 20(27): e2310300, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38299477

ABSTRACT

Glutathione (GSH) is the primary antioxidant in cells, and GSH consumption will break the redox balance in cells. Based on this, a method that uses high concentrations of GSH in the tumor microenvironment to trigger the redox reaction of Cu(II) to generate copper nanoprobes with fluorescence and tumor growth inhibition properties is proposed. The nanoprobe mainly exists in the form of Cu(I) and catalyzes the decomposition of hydrogen peroxide into hydroxyl radicals. At the same time, a simple and controllable carbon micro-nano electrode is used to construct a single-cell sensing platform, which enable the detection of glutathione content in single living cells after Cu(II) treatment, providing an excellent example for detecting single-cell biomolecules.


Subject(s)
Copper , Glutathione , Glutathione/metabolism , Copper/chemistry , Humans , Neoplasms/metabolism , Biosensing Techniques/methods , Cell Line, Tumor , Animals , Oxidation-Reduction , Intracellular Space/metabolism
17.
Biophys J ; 123(14): 2024-2037, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38219015

ABSTRACT

Slow deactivation is a critical property of voltage-gated K+ channels encoded by the human Ether-à-go-go-Related Gene 1 (hERG). hERG1 channel deactivation is modulated by interactions between intracellular N-terminal Per-Arnt-Sim (PAS) and C-terminal cyclic nucleotide-binding homology (CNBh) domains. The PAS domain is multipartite, comprising a globular domain (gPAS; residues 26-135) and an N-terminal PAS-cap that is further subdivided into an initial unstructured "tip" (residues 1-12) and an amphipathic α-helical region (residues 13-25). Although the PAS-cap tip has long been considered the effector of slow deactivation, how its position near the gating machinery is controlled has not been elucidated. Here, we show that a triad of hydrophobic interactions among the gPAS, PAS-cap α helix, and the CNBh domains is required to support slow deactivation in hERG1. The primary sequence of this "hydrophobic nexus" is highly conserved among mammalian ERG channels but shows key differences to fast-deactivating Ether-à-go-go 1 (EAG1) channels. Combining sequence analysis, structure-directed mutagenesis, electrophysiology, and molecular dynamics simulations, we demonstrate that polar serine substitutions uncover an intermediate deactivation mode that is also mimicked by deletion of the PAS-cap α helix. Molecular dynamics simulation analyses of the serine-substituted channels show an increase in distance among the residues of the hydrophobic nexus, a rotation of the intracellular gating ring, and a retraction of the PAS-cap tip from its receptor site near the voltage sensor domain and channel gate. These findings provide compelling evidence that the hydrophobic nexus coordinates the respective components of the intracellular gating ring and positions the PAS-cap tip to control hERG1 deactivation gating.


Subject(s)
Hydrophobic and Hydrophilic Interactions , Animals , Humans , Amino Acid Sequence , ERG1 Potassium Channel/metabolism , ERG1 Potassium Channel/chemistry , ERG1 Potassium Channel/genetics , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Ether-A-Go-Go Potassium Channels/genetics , HEK293 Cells , Intracellular Space/metabolism , Ion Channel Gating , Protein Domains
18.
J Biol Chem ; 300(3): 105668, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38272232

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and a critical class of regulators of mammalian physiology. Also known as seven transmembrane receptors (7TMs), GPCRs are ubiquitously expressed and versatile, detecting a diverse set of endogenous stimuli, including odorants, neurotransmitters, hormones, peptides, and lipids. Accordingly, GPCRs have emerged as the largest class of drug targets, accounting for upward of 30% of all prescription drugs. The view that ligand-induced GPCR responses originate exclusively from the cell surface has evolved to reflect accumulating evidence that receptors can elicit additional waves of signaling from intracellular compartments. These events in turn shape unique cellular and physiological outcomes. Here, we discuss our current understanding of the roles and regulation of compartmentalized GPCR signaling.


Subject(s)
Receptors, G-Protein-Coupled , Signal Transduction , Animals , Cell Membrane/metabolism , Receptors, G-Protein-Coupled/metabolism , Humans , Intracellular Space/metabolism , Enzyme Activation
19.
Nucleic Acids Res ; 52(1): e2, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-37953397

ABSTRACT

To gain a better understanding of the complexity of gene expression in normal and diseased tissues it is important to account for the spatial context and identity of cells in situ. State-of-the-art spatial profiling technologies, such as the Nanostring GeoMx Digital Spatial Profiler (DSP), now allow quantitative spatially resolved measurement of the transcriptome in tissues. However, the bioinformatics pipelines currently used to analyse GeoMx data often fail to successfully account for the technical variability within the data and the complexity of experimental designs, thus limiting the accuracy and reliability of the subsequent analysis. Carefully designed quality control workflows, that include in-depth experiment-specific investigations into technical variation and appropriate adjustment for such variation can address this issue. Here, we present standR, an R/Bioconductor package that enables an end-to-end analysis of GeoMx DSP data. With four case studies from previously published experiments, we demonstrate how the standR workflow can enhance the statistical power of GeoMx DSP data analysis and how the application of standR enables scientists to develop in-depth insights into the biology of interest.


Subject(s)
Gene Expression Profiling , Software , Transcriptome , Computational Biology , Reproducibility of Results , Workflow , Intracellular Space/genetics
20.
Small ; 20(17): e2307955, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38148312

ABSTRACT

Unraveling the intricacies between oxygen dynamics and cellular processes in the tumor microenvironment (TME) hinges upon precise monitoring of intracellular and intratumoral oxygen levels, which holds paramount significance. The majority of these reported oxygen nanoprobes suffer compromised lifetime and quantum yield when exposed to the robust ROS activities prevalent in TME, limiting their prolonged in vitro usability. Herein, the ruthenium-embedded oxygen nano polymeric sensor (Ru-ONPS) is proposed for precise oxygen gradient monitoring within the cellular environment and TME. Ru-ONPS (≈64±7 nm) incorporates [Ru(dpp)3]Cl2 dye into F-127 and crosslinks it with urea and paraformaldehyde, ensuring a prolonged lifetime (5.4 µs), high quantum yield (66.65 ± 2.43% in N2 and 49.80 ± 3.14% in O2), superior photostability (>30 min), and excellent stability in diverse environmental conditions. Based on the Stern-Volmer plot, the Ru-ONPS shows complete linearity for a wide dynamic range (0-23 mg L-1), with a detection limit of 10 µg mL-1. Confocal imaging reveals Ru-ONPS cellular uptake and intratumoral distribution. After 72 h, HCT-8 cells show 5.20±1.03% oxygen levels, while NIH3T3 cells have 7.07±1.90%. Co-culture spheroids display declining oxygen levels of 17.90±0.88%, 10.90±0.88%, and 5.10±1.18%, at 48, 120, and 216 h, respectively. Ru-ONPS advances cellular oxygen measurement and facilitates hypoxia-dependent metastatic research and therapeutic target identification.


Subject(s)
Oxygen , Polymers , Oxygen/metabolism , Humans , Polymers/chemistry , Tumor Microenvironment , Cell Line, Tumor , Animals , Ruthenium/chemistry , Mice , Biosensing Techniques/methods , Intracellular Space/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL