Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 970
Filter
1.
Cells ; 13(6)2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38534320

ABSTRACT

The ubiquitous second messenger 3',5'-cyclic adenosine monophosphate (cAMP) regulates cardiac excitation-contraction coupling (ECC) by signaling in discrete subcellular microdomains. Phosphodiesterase subfamilies 4B and 4D are critically involved in the regulation of cAMP signaling in mammalian cardiomyocytes. Alterations of PDE4 activity in human hearts has been shown to result in arrhythmias and heart failure. Here, we sought to systematically investigate specific roles of PDE4B and PDE4D in the regulation of cAMP dynamics in three distinct subcellular microdomains, one of them located at the caveolin-rich plasma membrane which harbors the L-type calcium channels (LTCCs), as well as at two sarco/endoplasmic reticulum (SR) microdomains centered around SR Ca2+-ATPase (SERCA2a) and cardiac ryanodine receptor type 2 (RyR2). Transgenic mice expressing Förster Resonance Energy Transfer (FRET)-based cAMP-specific biosensors targeted to caveolin-rich plasma membrane, SERCA2a and RyR2 microdomains were crossed to PDE4B-KO and PDE4D-KO mice. Direct analysis of the specific effects of both PDE4 subfamilies on local cAMP dynamics was performed using FRET imaging. Our data demonstrate that all three microdomains are differentially regulated by these PDE4 subfamilies. Whereas both are involved in cAMP regulation at the caveolin-rich plasma membrane, there are clearly two distinct cAMP microdomains at the SR formed around RyR2 and SERCA2a, which are preferentially controlled by PDE4B and PDE4D, respectively. This correlates with local cAMP-dependent protein kinase (PKA) substrate phosphorylation and arrhythmia susceptibility. Immunoprecipitation assays confirmed that PDE4B is associated with RyR2 along with PDE4D. Stimulated Emission Depletion (STED) microscopy of immunostained cardiomyocytes suggested possible co-localization of PDE4B with both sarcolemmal and RyR2 microdomains. In conclusion, our functional approach could show that both PDE4B and PDE4D can differentially regulate cardiac cAMP microdomains associated with calcium homeostasis. PDE4B controls cAMP dynamics in both caveolin-rich plasma membrane and RyR2 vicinity. Interestingly, PDE4B is the major regulator of the RyR2 microdomain, as opposed to SERCA2a vicinity, which is predominantly under PDE4D control, suggesting a more complex regulatory pattern than previously thought, with multiple PDEs acting at the same location.


Subject(s)
Calcium , Ryanodine Receptor Calcium Release Channel , Mice , Humans , Animals , Calcium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Cyclic AMP/metabolism , Myocytes, Cardiac/metabolism , Mice, Transgenic , Caveolins/metabolism , Mammals/metabolism
2.
Cells ; 12(23)2023 11 22.
Article in English | MEDLINE | ID: mdl-38067108

ABSTRACT

In the mid-1950s, a groundbreaking discovery revealed the fascinating presence of caveolae, referred to as flask-shaped invaginations of the plasma membrane, sparking renewed excitement in the field of cell biology. Caveolae are small, flask-shaped invaginations in the cell membrane that play crucial roles in diverse cellular processes, including endocytosis, lipid homeostasis, and signal transduction. The structural stability and functionality of these specialized membrane microdomains are attributed to the coordinated activity of scaffolding proteins, including caveolins and cavins. While caveolae and caveolins have been long appreciated for their integral roles in cellular physiology, the accumulating scientific evidence throughout the years reaffirms their association with a broad spectrum of human disorders. This review article aims to offer a thorough account of the historical advancements in caveolae research, spanning from their initial discovery to the recognition of caveolin family proteins and their intricate contributions to cellular functions. Furthermore, it will examine the consequences of a dysfunctional caveolar network in the development of human diseases.


Subject(s)
Caveolae , Caveolins , Humans , Caveolae/metabolism , Caveolins/metabolism , Cell Membrane/metabolism , Membrane Microdomains/metabolism , Signal Transduction
3.
Int J Med Sci ; 20(13): 1662-1670, 2023.
Article in English | MEDLINE | ID: mdl-37928877

ABSTRACT

Lung cancer is one of the most frequently diagnosed cancers worldwide. Due to its late diagnosis, it remains the leading cause of cancer-related deaths. Despite it is mostly associated to tobacco smoking, recent data suggested that genetic factors are of the highest importance. In this context, different processes meaningful for the development and progression of lung cancer such endocytosis, protein secretion and signal transduction, are controlled by membrane rafts. These highly ordered membrane domains contain proteins such as caveolins and flotillins, which were traditionally considered scaffold proteins but have currently been given a preponderant role in lung cancer. Here, we summarize current knowledge regarding the involvement of caveolins and flotillins in lung cancer from a molecular point of view.


Subject(s)
Caveolins , Lung Neoplasms , Humans , Caveolins/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Microdomains
4.
Biophys J ; 122(20): 4082-4090, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37742070

ABSTRACT

Caveolins form complexes of various sizes that deform membranes into polyhedral shapes. However, the recent structure of the 8S complex was disk-like with a flat membrane-binding surface. How can a flat complex deform membranes into nonplanar structures? Molecular dynamics simulations revealed that the 8S complex rapidly takes the form of a suction cup. Simulations on implicit membrane vesicles determined that binding is stronger when E140 gets protonated. In that case, the complex binds much more strongly to 5- and 10-nm-radius vesicles. A concave membrane-binding surface readily explains the membrane-deforming ability of caveolins by direct scaffolding. We propose that the 8S complex sits at the vertices of the caveolar polyhedra, rather than at the center of the polyhedral faces.


Subject(s)
Caveolins , Membrane Proteins , Caveolins/analysis , Caveolins/metabolism , Membrane Proteins/chemistry , Caveolin 1/metabolism , Membranes/metabolism , Cell Membrane/metabolism
5.
J Cell Biol ; 222(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37526691

ABSTRACT

Caveolin-1 (CAV1) and CAV3 are membrane-sculpting proteins driving the formation of the plasma membrane (PM) caveolae. Within the PM mosaic environment, caveola assembly is unique as it requires progressive oligomerization of newly synthesized caveolins while trafficking through the biosynthetic-secretory pathway. Here, we have investigated these early events by combining structural, biochemical, and microscopy studies. We uncover striking trafficking differences between caveolins, with CAV1 rapidly exported to the Golgi and PM while CAV3 is initially retained in the endoplasmic reticulum and laterally moves into lipid droplets. The levels of caveolins in the endoplasmic reticulum are controlled by proteasomal degradation, and only monomeric/low oligomeric caveolins are exported into the cis-Golgi with higher-order oligomers assembling beyond this compartment. When any of those early proteostatic mechanisms are compromised, chemically or genetically, caveolins tend to accumulate along the secretory pathway forming non-functional aggregates, causing organelle damage and triggering cellular stress. Accordingly, we propose a model in which disrupted proteostasis of newly synthesized caveolins contributes to pathogenesis.


Subject(s)
Caveolins , Proteostasis , Caveolins/metabolism , Caveolin 1/metabolism , Membrane Proteins/metabolism , Caveolae/metabolism , Cell Membrane/metabolism , Golgi Apparatus/metabolism
6.
Article in English | MEDLINE | ID: mdl-37277189

ABSTRACT

Caveolae are plasma membrane invaginations with a distinct lipid composition. Membrane lipids cooperate with the structural components of caveolae to generate a metastable surface domain. Recent studies have provided insights into the structure of essential caveolar components and how lipids are crucial for the formation, dynamics, and disassembly of caveolae. They also suggest new models for how caveolins, major structural components of caveolae, insert into membranes and interact with lipids.


Subject(s)
Caveolae , Membrane Lipids , Caveolae/chemistry , Caveolae/metabolism , Membrane Lipids/analysis , Membrane Lipids/metabolism , Caveolins/analysis , Caveolins/metabolism , Endocytosis
7.
FEBS Lett ; 597(11): 1528-1540, 2023 06.
Article in English | MEDLINE | ID: mdl-37051832

ABSTRACT

GPR15 is a G protein-coupled receptor involved in immune disorders such as human immunodeficiency virus-induced enteropathy, multiple sclerosis, and colitis. Yet, the important endocytosis mechanism of GPR15 remained unclear. This study determined the participation of endocytic machinery proteins, including Gα proteins, G protein-coupled receptor kinases (GRKs), protein kinase C, arrestins, clathrin, caveolin, and dynamin in GPR15 internalization. The results demonstrate that GPR15 internalization is moderately dependent on GRKs and clathrin, and highly dependent on caveolin and dynamin. Moreover, a bystander arrestin recruitment assay showed that GPR15 recruits arrestin-3 to the cell membrane upon agonist stimulation, although GPR15 internalizes in an arrestin-independent manner. Overall, our study provides novel insights into ß-arrestin recruitment and receptor internalization mechanisms for the recently deorphanized GPR15.


Subject(s)
Arrestin , Receptors, G-Protein-Coupled , Humans , Arrestin/metabolism , Receptors, G-Protein-Coupled/metabolism , beta-Arrestins/metabolism , Dynamins/metabolism , Endocytosis/physiology , Clathrin/metabolism , Caveolins/metabolism , Receptors, Peptide/metabolism
8.
J Biol Chem ; 299(4): 104574, 2023 04.
Article in English | MEDLINE | ID: mdl-36870682

ABSTRACT

Caveolin-1 (CAV1) is a membrane-sculpting protein that oligomerizes to generate flask-shaped invaginations of the plasma membrane known as caveolae. Mutations in CAV1 have been linked to multiple diseases in humans. Such mutations often interfere with oligomerization and the intracellular trafficking processes required for successful caveolae assembly, but the molecular mechanisms underlying these defects have not been structurally explained. Here, we investigate how a disease-associated mutation in one of the most highly conserved residues in CAV1, P132L, affects CAV1 structure and oligomerization. We show that P132 is positioned at a major site of protomer-protomer interactions within the CAV1 complex, providing a structural explanation for why the mutant protein fails to homo-oligomerize correctly. Using a combination of computational, structural, biochemical, and cell biological approaches, we find that despite its homo-oligomerization defects P132L is capable of forming mixed hetero-oligomeric complexes with WT CAV1 and that these complexes can be incorporated into caveolae. These findings provide insights into the fundamental mechanisms that control the formation of homo- and hetero-oligomers of caveolins that are essential for caveolae biogenesis, as well as how these processes are disrupted in human disease.


Subject(s)
Caveolin 1 , Caveolins , Disease , Humans , Caveolae/metabolism , Caveolin 1/genetics , Caveolin 1/metabolism , Caveolins/metabolism , Cell Membrane/metabolism , Membrane Proteins/metabolism , Mutation , Protein Subunits/metabolism , Disease/genetics
9.
J Biol Chem ; 299(3): 102974, 2023 03.
Article in English | MEDLINE | ID: mdl-36738791

ABSTRACT

In vivo and in vitro assays, particularly reconstitution using artificial membranes, have established the role of synaptic soluble N-Ethylmaleimide-sensitive attachment protein receptors (SNAREs) VAMP2, Syntaxin-1A, and SNAP-25 in membrane fusion. However, using artificial membranes requires challenging protein purifications that could be avoided in a cell-based assay. Here, we developed a synthetic biological approach based on the generation of membrane cisternae by the integral membrane protein Caveolin in Escherichia coli and coexpression of SNAREs. Syntaxin-1A/SNAP-25/VAMP-2 complexes were formed and regulated by SNARE partner protein Munc-18a in the presence of Caveolin. Additionally, Syntaxin-1A/SNAP-25/VAMP-2 synthesis provoked increased length of E. coli only in the presence of Caveolin. We found that cell elongation required SNAP-25 and was inhibited by tetanus neurotoxin. This elongation was not a result of cell division arrest. Furthermore, electron and super-resolution microscopies showed that synaptic SNAREs and Caveolin coexpression led to the partial loss of the cisternae, suggesting their fusion with the plasma membrane. In summary, we propose that this assay reconstitutes membrane fusion in a simple organism with an easy-to-observe phenotype and is amenable to structure-function studies of SNAREs.


Subject(s)
Artificial Cells , Membrane Fusion , SNARE Proteins , Caveolins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Nerve Tissue Proteins/metabolism , Qa-SNARE Proteins/metabolism , SNARE Proteins/genetics , Syntaxin 1/genetics , Vesicle-Associated Membrane Protein 2/metabolism , Vesicular Transport Proteins/metabolism
10.
Nat Commun ; 13(1): 7234, 2022 11 24.
Article in English | MEDLINE | ID: mdl-36433988

ABSTRACT

Caveolae are small coated plasma membrane invaginations with diverse functions. Caveolae undergo curvature changes. Yet, it is unclear which proteins regulate this process. To address this gap, we develop a correlative stimulated emission depletion (STED) fluorescence and platinum replica electron microscopy imaging (CLEM) method to image proteins at single caveolae. Caveolins and cavins are found at all caveolae, independent of curvature. EHD2 is detected at both low and highly curved caveolae. Pacsin2 associates with low curved caveolae and EHBP1 with mostly highly curved caveolae. Dynamin is absent from caveolae. Cells lacking dynamin show no substantial changes to caveolae, suggesting that dynamin is not directly involved in caveolae curvature. We propose a model where caveolins, cavins, and EHD2 assemble as a cohesive structural unit regulated by intermittent associations with pacsin2 and EHBP1. These coats can flatten and curve to enable lipid traffic, signaling, and changes to the surface area of the cell.


Subject(s)
Caveolae , Caveolins , Caveolae/metabolism , Cell Membrane/metabolism , Caveolins/metabolism , Endocytosis , Dynamins/metabolism , Proteins/metabolism
11.
Cells ; 11(20)2022 10 17.
Article in English | MEDLINE | ID: mdl-36291129

ABSTRACT

The plasma membrane (sarcolemma) of skeletal muscle myofibers is susceptible to injury caused by physical and chemical stresses during normal daily movement and/or under disease conditions. These acute plasma membrane disruptions are normally compensated by an intrinsic membrane resealing process involving interactions of multiple intracellular proteins including dysferlin, annexin, caveolin, and Mitsugumin 53 (MG53)/TRIM72. There is new evidence for compromised muscle sarcolemma repair mechanisms in Amyotrophic Lateral Sclerosis (ALS). Mitochondrial dysfunction in proximity to neuromuscular junctions (NMJs) increases oxidative stress, triggering MG53 aggregation and loss of its function. Compromised membrane repair further worsens sarcolemma fragility and amplifies oxidative stress in a vicious cycle. This article is to review existing literature supporting the concept that ALS is a disease of oxidative-stress induced disruption of muscle membrane repair that compromise the integrity of the NMJs and hence augmenting muscle membrane repair mechanisms could represent a viable therapeutic strategy for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Regeneration , Sarcolemma , Humans , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/therapy , Annexins/metabolism , Carrier Proteins/metabolism , Caveolins/metabolism , Dysferlin/metabolism , Membrane Proteins/metabolism , Sarcolemma/metabolism , Sarcolemma/pathology
12.
ACS Appl Mater Interfaces ; 14(37): 41712-41725, 2022 Sep 21.
Article in English | MEDLINE | ID: mdl-36069201

ABSTRACT

The intestinal epithelium is known to be a main hindrance to oral delivery of nanoparticles. Even though surface ligand modification can enhance cellular uptake of nanoparticles, the "easy entry and hard across" was frequently observed for many active targeting nanoparticles. Here, we fabricated polymeric nanoparticles relayed by bile acid transporters with monomethoxy poly(ethylene glycol)-poly(D,l-lactide) and deoxycholic acid-conjugated poly(2-ethyl-2-oxazoline)-poly(D,l-lactide) based on structural characteristics of intestine epithelium and the absorption characteristics of endogenous substances. As anticipated, deoxycholic acid-modified polymeric nanoparticles featuring good stability in simulated gastrointestinal fluid could notably promote the internalization of their payload by Caco-2 cells through mediation of apical sodium-dependent bile acid transporter (ASBT) and transmembrane transport of the nanoparticles across Caco-2 cell monolayers via relay-guide of ASBT, ileal bile acid-binding protein, and the heteromeric organic solute transporter (OSTα-OSTß) along with multidrug resistance-associated protein 3 (MRP3) evidenced by competitive inhibition and fluorescence immunoassay, which was further visually confirmed by the stronger fluorescence from C6-labeled nanoparticles inside enterocytes and the basal side of the intestinal epithelium of mice. The transcellular transport of deoxycholic acid-modified nanoparticles in an intact form was mediated by caveolin/lipid rafts and clathrin with intracellular trafficking trace of endosome-lysosome-ER-Golgi apparatus and bile acid transport route. Furthermore, the increased uptake by HepG2 cells compared with unmodified nanoparticles evidenced the target ability of deoxycholic acid-modified nanoparticles to the liver, which was further supported by ex vivo imaging of excised major organs of mice. Thus, this study provided a feasible and potential strategy to further enhance transepithelial transport efficiency and liver-targeted ability of nanoparticles by means of the specific enterohepatic circulation pathways of bile acid.


Subject(s)
Bile Acids and Salts , Nanoparticles , Animals , Caco-2 Cells , Caveolins/metabolism , Clathrin/metabolism , Deoxycholic Acid/chemistry , Enterohepatic Circulation , Humans , Ligands , Liver/metabolism , Mice , Nanoparticles/chemistry , Polyethylene Glycols/metabolism
13.
PLoS Pathog ; 18(9): e1010759, 2022 09.
Article in English | MEDLINE | ID: mdl-36084159

ABSTRACT

Peste des petits ruminants virus (PPRV) is an important pathogen that seriously influences the productivity of small ruminants worldwide. PPRV is lymphotropic in nature and SLAM was identified as the primary receptor for PPRV and other Morbilliviruses. Many viruses have been demonstrated to engage extracellular vesicles (EVs) to facilitate their replication and pathogenesis. Here, we provide evidence that PPRV infection significantly induced the secretion levels of EVs from goat PBMC, and that PPRV-H protein carried in EVs can enhance SLAM receptor expression in the recipient cells via suppressing miR-218, a negative miRNA directly targeting SLAM gene. Importantly, EVs-mediated increased SLAM expression enhances PPRV infectivity as well as the expression of various cytokines related to SLAM signaling pathway in the recipient cells. Moreover, our data reveal that PPRV associate EVs rapidly entry into the recipient cells mainly through macropinocytosis pathway and cooperated with caveolin- and clathrin-mediated endocytosis. Taken together, our findings identify a new strategy by PPRV to enhance virus infection and escape innate immunity by engaging EVs pathway.


Subject(s)
Extracellular Vesicles , MicroRNAs , Peste-des-Petits-Ruminants , Peste-des-petits-ruminants virus , Virus Diseases , Animals , Caveolins/metabolism , Clathrin/metabolism , Cytokines/metabolism , Extracellular Vesicles/metabolism , Goats/genetics , Leukocytes, Mononuclear , Lymphocyte Activation , MicroRNAs/genetics , MicroRNAs/metabolism , Peste-des-petits-ruminants virus/genetics , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism
14.
Am J Physiol Cell Physiol ; 323(4): C1137-C1148, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-35993515

ABSTRACT

Caveolins, encoded by the Cav gene family, are the main components of caveolae. Caveolin-3 (Cav3) is specifically expressed in muscle cells. Mutations in Cav3 are responsible for a group of muscle diseases called caveolinopathies, and Cav3 deficiency is associated with sarcolemmal membrane alterations, disorganization of T-tubules, and disruption of specific cell-signaling pathways. However, Cav3 overexpression increases the number of sarcolemmal caveolae and muscular dystrophy-like regenerating muscle fibers with central nuclei, suggesting that the alteration of Cav3 expression levels or localization influences muscle cell functions. Here, we used mouse C2C12 myoblasts in which Cav3 expression was suppressed with short hairpin RNA and found that Cav3 suppression impaired myotube differentiation without affecting the expression of MyoD and Myog. We also observed an increase of intracellular Ca2+ levels, total calpain activity, and Ca2+-dependent calmodulin kinase II (CaMKII) levels in Cav3-depleted myoblasts. Importantly, those phenotypes due to Cav3 suppression were caused by the ryanodine receptor activation. Furthermore, pharmacological inhibition of CaMKII rescued the impairment of myoblast differentiation due to Cav3 knockdown. Our results suggest that Cav3 regulates intracellular Ca2+ concentrations by modulating ryanodine receptor activity in muscle cells and that CaMKII suppression in muscle could be a novel therapy for caveolinopathies.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Caveolin 3 , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calpain/genetics , Calpain/metabolism , Caveolin 3/genetics , Caveolin 3/metabolism , Caveolins/metabolism , Mice , Muscle, Skeletal/metabolism , RNA, Small Interfering/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
15.
Cell Signal ; 97: 110399, 2022 09.
Article in English | MEDLINE | ID: mdl-35820545

ABSTRACT

The caveolae-mediated transport across polarized epithelial cell barriers has been largely deciphered in the last decades and is considered the second essential intracellular transfer mechanism, after the clathrin-dependent endocytosis. The basic cell biology knowledge was supplemented recently, with the molecular mechanisms beyond caveolae generation implying the key contribution of the lipid-binding proteins (the structural protein Caveolin and the adapter protein Cavin), along with the bulb coat stabilizing molecules PACSIN-2 and Eps15 homology domain protein-2. The current attention is focused also on caveolae architecture (such as the bulb coat, the neck, the membrane funnel inside the bulb, and the associated receptors), and their specific tasks during the intracellular transport of various cargoes. Here, we resume the present understanding of the assembly, detachment, and internalization of caveolae from the plasma membrane lipid raft domains, and give an updated view on transcytosis and endocytosis, the two itineraries of cargoes transport via caveolae. The review adds novel data on the signalling molecules regulating caveolae intracellular routes and on the transport dysregulation in diseases. The therapeutic possibilities offered by exploitation of Caveolin-1 expression and caveolae trafficking, and the urgent issues to be uncovered conclude the review.


Subject(s)
Caveolae , Caveolins , Carrier Proteins/metabolism , Caveolae/metabolism , Caveolin 1/metabolism , Caveolins/metabolism , Endocytosis/physiology , Membrane Microdomains/metabolism , Protein Transport , Signal Transduction
16.
Biomed Pharmacother ; 153: 113282, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35750009

ABSTRACT

Multiple pathophysiological pathways are activated during the process of myocardial injury. Various cardioprotective strategies protect the myocardium from ischemia, infarction, and ischemia/reperfusion (I/R) injury through different targets, yet the clinical translation remains limited. Caveolae and its structure protein, caveolins, have been suggested as a bridge to transmit damage-preventing signals and mediate the protection of ultrastructure in cardiomyocytes under pathological conditions. In this review, we first briefly introduce caveolae and caveolins. Then we review the cardioprotective strategies mediated by caveolins through various pathophysiological pathways. Finally, some possible research directions are proposed to provide future experiments and clinical translation perspectives targeting caveolin based on the investigative evidence.


Subject(s)
Caveolins , Myocardial Reperfusion Injury , Caveolae/metabolism , Caveolae/pathology , Caveolae/ultrastructure , Caveolin 1/metabolism , Caveolins/metabolism , Humans , Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardium/pathology
17.
J Virol ; 96(7): e0020222, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35297667

ABSTRACT

In the United States, most new cases of human immunodeficiency virus (HIV) belong to the at-risk group of gay and bisexual men. Developing therapies to reverse viral latency and prevent spread is paramount for the HIV cure agenda. In gay and bisexual men, a major, yet poorly characterized, route of HIV entry is via transport across the colonic epithelial barrier. While colonic tears and paracellular transport contribute to infection, we hypothesize that HIV entry through the colonic mucosa proceeds via a process known as transcytosis, involving (i) virion binding to the apical surface of the colonic epithelium, (ii) viral endocytosis, (iii) transport of virions across the cell, and (iv) HIV release from the basolateral membrane. Using Caco-2 colonic epithelial cells plated as a polarized monolayer in transwells, we characterized the mechanism of HIV transport. After exposing the monolayer to HIV apically, reverse transcription quantitative PCR (RT-qPCR) of the viral genome present in the basolateral chamber revealed that transport is dose dependent, cooperative, and inefficient, with released virus first detectable at 12 h. Inefficiency may be associated with >50% decline in detectable intracellular virus that correlates temporally with increased association of the virion with lysosomal-associated membrane protein 1 (LAMP-1+) endosomes. Microscopy revealed green fluorescent protein (GFP)-labeled HIV within the confines of the epithelial monolayer, with no virus detectable between cells, suggesting that viral transport is transcellular. Treatment of the monolayer with endocytosis inhibitors, cholesterol reducing agents, and small interfering RNA (siRNA) to caveolin showed that viral endocytosis is mediated by caveolin-coated endosomes contained in lipid rafts. These results indicate that HIV transport across the intestinal epithelial barrier via transcytosis is a viable mechanism for viral spread and a potential therapeutic target. IMPORTANCE Despite the success of combination antiretroviral therapy in suppressing HIV replication and the emergence and effectiveness of PrEP-based prevention strategies, in 2018, 37,968 people in the United States received a new HIV diagnosis, accompanied by 15,820 deaths. While the annual number of new diagnoses decreased 7% from 2014 to 2018, 14% of people with HIV did not know they were infected. Gay and bisexual men accounted for 69% of all HIV diagnoses and 83% of diagnoses among males. Due to the scope of the HIV epidemic, determining and understanding precise routes of infection and the mechanisms of viral spread are paramount to ending the epidemic. Since transcellular transport of HIV across an intact colonic epithelial barrier is poorly understood, our overall goal is to characterize the molecular events involved in HIV transcytosis across the intestinal epithelial cell.


Subject(s)
Colon , Endocytosis , HIV Infections , HIV , Intestinal Mucosa , Caco-2 Cells , Caveolins/metabolism , Colon/immunology , Colon/virology , Endosomes/metabolism , HIV/metabolism , HIV Infections/metabolism , HIV Infections/prevention & control , HIV Infections/transmission , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/virology , Male
18.
Gene ; 809: 146013, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34655718

ABSTRACT

Plant pathogenesis-related (PR) proteins are a large group of proteins, classified in 17 families, that are induced by pathological conditions. Here, we characterized the soybean PR-1 (GmPR-1) gene repertoire at the sequence, structural and expression levels. We found 24 GmPR-1 genes, clustered in two phylogenetic groups. GmPR-1 genes are under strong purifying selection, particularly those that emerged by tandem duplications. GmPR-1 promoter regions are abundant in cis-regulatory elements associated with major stress-related transcription factor families, namely WRKY, ERF, HD-Zip, C2H2, NAC, and GATA. We observed that 23 GmPR-1 genes are induced by stress conditions or exclusively expressed upon stress. We explored 1972 transcriptome samples, including 26 stress conditions, revealing that most GmPR-1 genes are differentially expressed in a plethora of biotic and abiotic stresses. Our findings highlight stress-responsive GmPR-1 genes with potential biotechnological applications, such as the development of transgenic lines with increased resistance to biotic and abiotic stresses.


Subject(s)
Gene Expression Regulation, Plant , Glycine max/genetics , Phylogeny , Plant Proteins/genetics , Stress, Physiological/genetics , Binding Sites , Caveolins/metabolism , Evolution, Molecular , Gene Duplication , Genome, Plant , Genome-Wide Association Study , Multigene Family , Plant Proteins/chemistry , Plant Proteins/metabolism , Regulatory Sequences, Nucleic Acid , Selection, Genetic , Glycine max/physiology , Transcription Factors/genetics
19.
Cell Biochem Biophys ; 80(2): 311-320, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34796419

ABSTRACT

The endocrine system is closely related to the development of the breast cancer. Many studies have shown that FGF1 (Fibroblast growth factor-1) is involved the occurrence and development of the breast cancer. But up to now, the cellular behavior and characteristics of FGF1 in breast cancer have not been fully revealed. In the current study, breast cancer cell was used as an in vitro cell model to investigate FGF's cell property. The results showed that FGF1 internalized into cells in a time-dependent manner. Further study indicated that both clathrin-mediated and caveolin-mediated endocytic pathway are involved in the internalization of FGF/FGFR (Fibroblast growth factor receptor), and both clathrin-mediated endocytosis and caveolin-mediated endocytosis are involved in the process of FGF1's nuclear localization. Further study showed that Rab5 also plays an important role in the process of nuclear localization of FGF-1. In addition, we found that FGF1 and FGFR transported to the cell nuclei of breast cancer. Further experimental results indicated that the nuclear-localized FGF1 and/or FGFR is closely associated to cell proliferation of breast cancer cell. Taken together, the current work lays the foundation for exploring the relationship between nuclear-localized FGF1/FGFR and the occurrence and development of breast cancer.


Subject(s)
Breast Neoplasms , Fibroblast Growth Factor 1 , Receptors, Fibroblast Growth Factor , Active Transport, Cell Nucleus , Breast Neoplasms/metabolism , Caveolins/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Clathrin/metabolism , Female , Fibroblast Growth Factor 1/metabolism , Humans , Receptors, Fibroblast Growth Factor/metabolism
20.
Nucleic Acids Res ; 49(21): 12306-12319, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34865123

ABSTRACT

DNA damage and apoptosis lead to the release of free nucleosomes-the basic structural repeating units of chromatin-into the blood circulation system. We recently reported that free nucleosomes that enter the cytoplasm of mammalian cells trigger immune responses by activating cGMP-AMP synthase (cGAS). In the present study, we designed experiments to reveal the mechanism of nucleosome uptake by human cells. We showed that nucleosomes are first absorbed on the cell membrane through nonspecific electrostatic interactions between positively charged histone N-terminal tails and ligands on the cell surface, followed by internalization via clathrin- or caveolae-dependent endocytosis. After cellular internalization, endosomal escape occurs rapidly, and nucleosomes are released into the cytosol, maintaining structural integrity for an extended period. The efficient endocytosis of extracellular nucleosomes suggests that circulating nucleosomes may lead to cellular disorders as well as immunostimulation, and thus, the biological effects exerted by endocytic nucleosomes should be addressed in the future.


Subject(s)
Caveolins/metabolism , Cell Membrane/metabolism , Clathrin/metabolism , Endocytosis , Nucleosomes/metabolism , Animals , Cell Line , Cholera Toxin/metabolism , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Golgi Apparatus/metabolism , HeLa Cells , Hep G2 Cells , Humans , Lysosomes/metabolism , Mice , Microscopy, Confocal , Nucleosomes/genetics , THP-1 Cells , Transferrin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...