Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Sci Total Environ ; : 176412, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39322074

ABSTRACT

Although our understanding of the dramatic worldwide loss of biodiversity in recent decades is far from adequate, one of the main factors in areas dominated by agriculture is undoubtedly the widespread use of synthetic pesticides. Unfortunately, the ecological risk assessment (EcoRA) for pesticides is based on a few single-species bioassays which do not allow for the evaluation of risks to whole communities. Here we present the results of an experimental assessment of the risk to the ecosystem service provider (ESP) communities - pest control agents - from exposure to the commonly used pyrethroid insecticide, λ-cyhalothrin. The study was performed in five European countries (Germany, Poland, Portugal, Spain, United Kingdom) representing different pedoclimatic zones. Representatives of the most common species of the ESP communities in each country were exposed in a standardized insecticide-coated glass vials bioassay to five doses of λ-cyhalothrin: 0.8 %, 4 %, 20 %, 100 %, and 200 % of the recommended field dose (RFD) plus an untreated control. Based on the calculated LD50s, species sensitivity distributions (SSDs) were estimated for each country and on combined data. In all five countries, the estimated hazardous concentration for 5 % of the species (HD5) was between 0.23 % and 1.67 % RFD, with HD5 = 0.44 % RFD based on combined data. At the RFD = 7.5 g a.i./ha (active ingredient per hectare), the predicted affected fraction of the ESP communities was between 96.4 % and 99.9 % of the species (98.5 % for combined data). The results indicate an extremely high risk to ESP communities across Europe associated with the use of λ-cyhalothrin at the recommended doses when these species are exposed to insecticide treatment. We recommend that EcoRA should include multi-species approaches, such as SSD, to better protect entire ESP communities from the negative impacts of pesticides.

3.
Environ Sci Technol ; 58(4): 1854-1864, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38251653

ABSTRACT

The mortality of organisms exposed to toxicants has been attributed to either stochastic processes or individual tolerance (IT), leading to the stochastic death (SD) and IT models. While the IT model follows the principles of natural selection, the relevance of the SD model has been debated. To clarify why the idea of stochastic mortality has found its way into ecotoxicology, we investigated the mortality of Poecilus cupreus (Linnaeus, 1758) beetles from pesticide-treated oilseed rape (OSR) fields and unsprayed meadows, subjected to repeated insecticide treatments. We analyzed the mortality with the Kaplan-Meier estimator and general unified threshold model for survival (GUTS), which integrates SD and IT assumptions. The beetles were exposed three times, ca. monthly, to the same dose of Proteus 110 OD insecticide containing thiacloprid and deltamethrin, commonly used in the OSR fields. Kaplan-Meier analysis showed that the mortality of beetles from meadows was much higher after the first treatment than after the next two, indicating the IT model. Beetles from the OSR displayed approximately constant mortality after the first and second treatments, consistent with the SD model. GUTS analysis did not conclusively identify the better model, with the IT being marginally better for beetles from meadows and the SD better for beetles from OSR fields.


Subject(s)
Brassica napus , Coleoptera , Insecticides , Animals , Insecticides/toxicity
4.
Ecotoxicology ; 32(9): 1141-1151, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37755556

ABSTRACT

The intensifications in the agricultural landscape and the application of pesticides can cause adverse effects on the fitness of organisms in that landscape. Here, we investigated whether habitats with different agricultural pressures influenced acetylcholinesterase (AChE) activity - a biomarker for exposure to pesticides, respiration rate, and resistance to starvation in the ground beetle Poecilus cupreus. Two differently structured landscapes were selected for the study, one dominated by small (S) and another by large (L) fields. Within each landscape three habitat types were selected: in the S landscape, these were habitats with medium (M), small (S) and no canola (meadow, 0) coverage (i.e., SM, SS, S0), and in the L landscape habitats with large (L), medium (M) and no canola (meadow, 0) coverage (i.e., LL, LM, L0), representing different levels of agricultural pressure. The activity of AChE was the highest in beetles from canola-free habitats (S0 and L0), being significantly higher than in beetles from the SM and SS habitats. The mean respiration rate corrected for body mass was also the highest in S0 and L0 beetles, with significant differences between populations from L0 vs. SS and from S0 vs. SS. Only beetles from S0, SS, L0, and LM were numerous enough to assess the resistance to starvation. Individuals from the LM habitat showed better survival compared to the canola-free habitat in the same landscape (L0), whereas in S landscape the SS beetles survived worse than those from S0, suggesting that characteristics of L landscape may lead to developing mechanisms of starvation resistance of P. cupreus in response to agricultural pressure.


Subject(s)
Coleoptera , Pesticides , Animals , Acetylcholinesterase , Ecosystem , Agriculture
5.
PLoS One ; 17(4): e0266453, 2022.
Article in English | MEDLINE | ID: mdl-35472211

ABSTRACT

The intensification of agriculture leads to increased pesticide use and significant transformation from small fields towards large-scale monocultures. This may significantly affect populations of non-target arthropods (NTA). We aimed to assess whether the multigenerational exposure to plant protection products has resulted in the evolution of resistance to insecticides in the ground beetle Poecilus cupreus originating from different agricultural landscapes. Two contrasting landscapes were selected for the study, one dominated by small and another by large fields. Within each landscape the beetles were collected at nine sites representing range of canola coverage and a variety of habitat types. Part of the collected beetles, after acclimation to laboratory conditions, were tested for sensitivity to Proteus 110 OD-the most commonly used insecticide in the studied landscapes. The rest were bred in the laboratory for two consecutive generations, and part of the beetles from each generation were also tested for sensitivity to selected insecticide. We showed that the beetles inhabiting areas with medium and large share of canola located in the landscape dominated by large fields were less sensitive to the studied insecticide. The persistence of reduced sensitivity to Proteus 110 OD for two consecutive generations indicates that either the beetles have developed resistance to the insecticide or the chronic exposure to pesticides has led to the selection of more resistant individuals naturally present in the studied populations. No increased resistance was found in the beetles from more heterogeneous landscape dominated by small fields, in which spatio-temporal diversity of crops and abundance of small, linear off-crop landscape elements may provide shelter that allows NTAs to survive without developing any, presumably costly, resistance mechanisms.


Subject(s)
Brassica napus , Coleoptera , Insecticides , Pesticides , Agriculture , Animals , Insecticide Resistance , Insecticides/pharmacology , Plant Breeding
6.
Am J Physiol Heart Circ Physiol ; 320(3): H1185-H1198, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33416452

ABSTRACT

Ischemia/reperfusion (I/R)-induced rapid inflammation involving activation of leukocyte-endothelial adhesive interactions and leukocyte infiltration into tissues is a major contributor to postischemic tissue injury. However, the molecular mediators involved in this pathological process are not fully known. We have previously reported that caveolin-2 (Cav-2), a protein component of plasma membrane caveolae, regulated leukocyte infiltration in mouse lung carcinoma tumors. The goal of the current study was to examine if Cav-2 plays a role in I/R injury and associated acute leukocyte-mediated inflammation. Using a mouse small intestinal I/R model, we demonstrated that I/R downregulates Cav-2 protein levels in the small bowel. Further study using Cav-2-deficient mice revealed aggravated postischemic tissue injury determined by scoring of villi length in H&E-stained tissue sections, which correlated with increased numbers of MPO-positive tissue-infiltrating leukocytes determined by IHC staining. Intravital microscopic analysis of upstream events relative to leukocyte transmigration and tissue infiltration revealed that leukocyte-endothelial cell adhesive interactions in postcapillary venules, namely leukocyte rolling and adhesion were also enhanced in Cav-2-deficient mice. Mechanistically, Cav-2 deficiency increased plasminogen activator inhibitor-1 (PAI-1) protein levels in the intestinal tissue and a pharmacological inhibition of PAI-1 had overall greater inhibitory effect on both aggravated I/R tissue injury and enhanced leukocyte-endothelial interactions in postcapillary venules in Cav-2-deficient mice. In conclusion, our data suggest that Cav-2 protein alleviates tissue injury in response to I/R by dampening PAI-1 protein levels and thereby reducing leukocyte-endothelial adhesive interactions.NEW & NOTEWORTHY The role of caveolin-2 in regulating ischemia/reperfusion (I/R) tissue injury and the mechanisms underlying its effects are unknown. This study uses caveolin-2-deficient mouse and small intestinal I/R injury models to examine the role of caveolin-2 in the leukocyte-dependent reperfusion injury. We demonstrate for the first time that caveolin-2 plays a protective role from the I/R-induced leukocyte-dependent reperfusion injury by reducing PAI-1 protein levels in intestinal tissue and leukocyte-endothelial adhesive interactions in postcapillary venules.


Subject(s)
Caveolin 2/deficiency , Cell Adhesion , Endothelial Cells/metabolism , Jejunal Diseases/metabolism , Jejunum/blood supply , Leukocyte Rolling , Leukocytes/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Reperfusion Injury/metabolism , Transendothelial and Transepithelial Migration , Venules/metabolism , Animals , Caveolin 2/genetics , Disease Models, Animal , Endothelial Cells/pathology , Jejunal Diseases/genetics , Jejunal Diseases/pathology , Leukocytes/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , Venules/pathology
7.
Sci Rep ; 9(1): 18970, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831780

ABSTRACT

Immunosuppression is critical for tumor growth and metastasis as well as obstacle to effective immunotherapy. Here, we demonstrate that host deficiency in caveolin-2, a member of caveolin protein family, increases M1-polarized tumor-associated macrophage (TAM) and CD8 T cell infiltration into subcutaneously implanted murine lung carcinoma tumors. Importantly, increase in M1 TAM-specific markers and cytokines occurs prior to increased numbers of tumor-infiltrating CD8 T cells and tumor regression in caveolin-2 deficient mice, suggesting that an early increase in M1 TAMs is a novel mechanism, via which host deficiency in caveolin-2 inhibits tumor growth. Consistent with the latter, transfer and co-injection of caveolin-2 deficient bone marrow (origin of TAMs) suppresses tumor growth and increases numbers of M1-polarized TAMs in wild type mice. Collectively, our data suggest that lung cancer cells use caveolin-2 expressed in bone marrow-derived cell types including TAMs to promote tumor growth via suppressing the anti-tumor immune response and that caveolin-2 could be a potential target for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Caveolin 2/deficiency , Immunity, Cellular , Lung Neoplasms/immunology , Macrophages/immunology , Neoplasm Proteins/deficiency , Neoplasms, Experimental/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Caveolin 2/immunology , Cell Line, Tumor , Immunotherapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Macrophages/pathology , Mice , Mice, Knockout , Neoplasm Proteins/immunology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy
8.
Bull Environ Contam Toxicol ; 102(2): 191-197, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30666388

ABSTRACT

To test effects of metal contamination on beetles morphology, specimens from species representing herbivores (Strophosoma capitatum), carnivores (Carabus arcensis and C. violaceus) and detritivores (Anoplotrupes stercorosus) were collected from an area polluted with zinc, lead and cadmium and from a control site. Both the length and width of elytra and pronotum were compared. Females of all species from the polluted area were smaller than those from the control site with the average difference of 2.7% (range 0.7%-6%). In contrast, males responded less consistently among species: A. stercorosus showed lower size of elytra and pronotum at the polluted area, while in C. arcensis only the elytra length and pronotum width were smaller. C. violaceus males exhibited smaller elytra length and pronotum length and width at the polluted area. In contrast, no differences between the two sites were found for S. capitatum males. Sex differences may originate from different energy investment strategies in females and males related to the reproduction needs. Even if the observed differences in body size were small, the smaller body size in females of all studied species, irrespectively of the guild, is striking and may indicate on lower fitness of a range of species inhabiting metal polluted areas.


Subject(s)
Cadmium/toxicity , Coleoptera/physiology , Environmental Pollutants/toxicity , Zinc/toxicity , Animals , Body Size/drug effects , Ecosystem , Female , Male , Metals , Sex Characteristics
9.
Am J Physiol Heart Circ Physiol ; 311(2): H415-25, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27317631

ABSTRACT

Caveolin-2 (Cav2) is a major protein component of caveolae in membranes of vascular smooth muscle and endothelium, yet its absence alters the ultrastructure of skeletal muscle fibers. To gain insight into Cav2 function in skeletal muscle, we tested the hypothesis that genetic deletion of Cav2 would alter microvascular reactivity and depress contractile function of skeletal muscle in vivo. In the left gluteus maximus muscle (GM) of anesthetized Cav2(-/-) and wild-type (WT) male mice (age, 6 mo), microvascular responses to physiological agonists and to GM contractions were studied at 34°C. For feed arteries (FA), first- (1A), second- (2A) and third-order (3A) arterioles, respective mean diameters at rest (45, 35, 25, 12 µm) and during maximal dilation (65, 55, 45, 30 µm) were similar between groups. Cumulative dilations to ACh (10(-9) to 10(-5) M) and constrictions to norepinephrine (10(-9) to 10(-5) M) were also similar between groups, as were steady-state dilations during rhythmic twitch contractions (2 and 4 Hz; 30 s). For single tetanic contractions (100 Hz; 100, 250, and 500 ms), rapid onset vasodilation (ROV) increased with contraction duration throughout networks in GM of both groups but was reduced by nearly half in Cav2(-/-) mice compared with WT mice (P < 0.05). Nevertheless, maximal force during tetanic contraction was ∼40% greater in GM of Cav2(-/-) vs. WT mice (152 ± 14 vs. 110 ± 3 mN per square millimeter, respectively; P < 0.05). Thus, while structural and functional properties of resistance networks are well maintained in the GM of Cav2(-/-) mice, diminished ROV with greater force production reveals novel physiological roles for Cav2 in skeletal muscle.


Subject(s)
Arterioles/physiopathology , Caveolin 2/genetics , Muscle Contraction , Muscle, Skeletal/blood supply , Muscle, Smooth, Vascular/physiopathology , Vasodilation/genetics , Acetylcholine/pharmacology , Animals , Arteries/drug effects , Arteries/physiopathology , Arterioles/drug effects , Buttocks , Intravital Microscopy , Male , Mice , Mice, Knockout , Microvessels/drug effects , Microvessels/physiopathology , Muscle, Smooth, Vascular/drug effects , Norepinephrine/pharmacology , Vascular Resistance , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
10.
Cancer Res ; 74(22): 6452-62, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25269481

ABSTRACT

Caveolin-2 (Cav-2), a member of caveolin protein family, is largely different from better known caveolin-1 (Cav-1) and thus might play distinct functions. Here, we provide the first genetic evidence suggesting that host-expressed Cav-2 promotes subcutaneous tumor growth and tumor-induced neovascularization using two independent syngeneic mouse models. Host deficiency in Cav-2 resulted in defective and reduced growth of subcutaneously implanted Lewis lung carcinoma (LLC) and B16-F10 melanoma tumors, respectively. Consistent with the defective growth, LLC and B16-F10 melanoma tumors implanted into Cav-2 KO mice displayed reduced microvascular density (MVD) determined by IHC with anti-CD31 antibodies, suggesting impaired pathologic angiogenesis. Additional studies involving LLC tumors extracted from Cav-2 KO mice just 10 days after implantation determined reduced cell proliferation, massive necrotic cell death, and fibrosis. In contrast with day 10, only MVD but not cell proliferation and survival was reduced in the earliest palpable LLC tumors extracted 6 days after implantation into Cav-2 KO mice, suggesting that impaired angiogenesis is the causative factor. Mechanistically, impaired LLC tumor growth and angiogenesis in Cav-2 KO mice was associated with increased expression levels of antiangiogenic thrombospondin-1 and inhibited S1177 phosphorylation of endothelial nitric oxide synthase. Taken together, our data suggest that host deficiency in Cav-2 impairs tumor-induced angiogenesis, leading to compromised tumor cell survival/proliferation manifested by the defective tumor growth. In conclusion, host-expressed Cav-2 may promote tumor growth via supporting tumor-induced angiogenesis. Thus, Cav-2 expressed in tumor microenvironment may potentially become a novel target for cancer therapy.


Subject(s)
Carcinoma, Lewis Lung/pathology , Caveolin 2/physiology , Neovascularization, Pathologic/prevention & control , Animals , Carcinoma, Lewis Lung/blood supply , Cell Proliferation , Fibrosis , Ki-67 Antigen/analysis , Male , Mice , Mice, Inbred C57BL , Necrosis , Nitric Oxide Synthase Type III/metabolism , Thrombospondin 1/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology
11.
Article in English | MEDLINE | ID: mdl-26005706

ABSTRACT

In addition to cancer cells, primary tumors are composed of a multitude of stromal cell types. Among others, the stromal cell types involved in tumor growth and progression include endothelial cells, fibroblasts, pericytes, stem cells and various cell types of immune origin. While the role of oncogenes or tumor suppressor proteins expressed in cancer cells has been extensively studied, far less is known about potential involvement of proteins expressed in stromal cell types present within the tumor microenvironment. Recent experimental evidence from our laboratory suggests that caveolin-2 (Cav-2) protein expressed in stromal cell types of the tumor microenvironment promotes subcutaneous tumor growth in two independent syngeneic mouse models, i.e., Lewis lung carcinoma (LLC) and B16-F10 melanoma. Mechanistically, the tumor growth promoting role of Cav-2 is associated with enhanced tumor induced neovascularization. At the molecular level, host-expressed Cav-2 appears to prevent excessive expression of anti-angiogenic thrombospondin-1 (TSP-1) and promote phosphorylation of pro-angiogenic endothelial nitric oxide synthase (eNOS) at serine 1177. Taken together, our recent findings suggest that Cav-2 expressed within the tumor microenvironment could be a potential target for anti-cancer therapy.

12.
FEBS Lett ; 586(19): 3317-23, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22819829

ABSTRACT

Here we show that tyrosine phosphorylation of caveolin-2 (Cav-2) negatively regulates the anti-proliferative function of transforming growth factor beta (TGF-beta) in endothelial cells. In contrast to wild-type-Cav-2, retroviral re-expression of Y19/27F-Cav-2 in Cav-2 knockout endothelial cells did not affect anti-proliferative effect of TGF-beta compared to empty vector. Conversely, although less effective than wild-type, re-expression of S23/36A-Cav-2 reduced the effect of TGF-beta compared to empty vector. This differential effect of tyrosine and serine phosphorylation mutants of Cav-2 correlated with TGF-beta-induced Smad3 phosphorylation and transcriptional activation of plasminogen activator inhibitor-1. Thus tyrosine-phosphorylated Cav-2 counteracts anti-proliferative effect of TGF-beta in endothelial cells.


Subject(s)
Caveolin 2/chemistry , Caveolin 2/metabolism , Transforming Growth Factor beta/pharmacology , Amino Acid Substitution , Animals , Caveolin 2/antagonists & inhibitors , Caveolin 2/genetics , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Knockout Techniques , Humans , Mice , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phosphorylation , Plasminogen Activator Inhibitor 1/genetics , Protein Tyrosine Phosphatases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Serpin E2/genetics , Smad3 Protein/metabolism , Transcriptional Activation/drug effects , Tyrosine/chemistry
13.
Front Physiol ; 2: 120, 2012.
Article in English | MEDLINE | ID: mdl-22232608

ABSTRACT

Caveolae are cholesterol and glycosphingolipid-rich flask-shaped invaginations of the plasma membrane which are particularly abundant in vascular endothelium and present in all other cell types of the cardiovascular system, including vascular smooth-muscle cells, macrophages, cardiac myocytes, and fibroblasts. Caveolins and the more recently discovered cavins are the major protein components of caveolae. When caveolae were discovered, their functional role was believed to be limited to transport across the endothelial cell barrier. Since then, however, a large body of evidence has accumulated, suggesting that these microdomains are very important in regulating many other important endothelial cell functions, mostly due to their ability to concentrate and compartmentalize various signaling molecules. Over the course of several years, multiple studies involving knockout mouse and small interfering RNA approaches have considerably enhanced our understanding of the role of caveolae and caveolin-1 in regulating many cardiovascular functions. New findings have been reported implicating other caveolar protein components in endothelial cell signaling and function, such as the understudied caveolin-2 and newly discovered cavin proteins. The aim of this review is to focus primarily on molecular and cellular aspects of the role of caveolae, caveolins, and cavins in endothelial cell signaling and function. In addition, where appropriate, the possible implications for the cardiovascular and pulmonary physiology and pathophysiology will be discussed.

14.
Article in English | MEDLINE | ID: mdl-26605130

ABSTRACT

Caveolae are cholesterol- and glycosphingolipid-rich omega-shaped invaginations of the plasma membrane that are very abundant in vascular endothelial cells and present in most cell types. Caveolins are the major coat protein components of caveolae. Multiple studies using knockout mouse, small interfering RNA, and cell-permeable peptide delivery approaches have significantly enhanced our understanding of the role of endothelial caveolae and caveolin-1 in physiology and disease. Several postnatal pulmonary and cardiovascular pathologies have been reported in caveolin-1 knockout mice, many of which have been recently rescued by selective re-expression of caveolin-1 in endothelium of these mice. A large body of experimental evidence mostly using caveolin-1 knockout mice suggests that, depending on the disease model, endothelial caveolin-1 may play either a protective or a detrimental role. For instance, physiological or higher expression levels of caveolin-1 in endothelium might be beneficial in such diseases as pulmonary hypertension, cardiac hypertrophy, or ischemic injury. On the other hand, endothelial caveolin-1 might contribute to acute lung injury and inflammation, atherosclerosis or pathological angiogenesis associated with inflammatory bowel disease. Moreover, depending on the specific model, endothelial caveolin-1 may either promote or suppress tumor-induced angiogenesis. In addition to overwhelming evidence for the role of endothelial caveolin-1, more recent studies also suggest that endothelial caveolin-2 could possibly play a role in pulmonary disease. The purpose of this review is to focus on how caveolin-1 expressed in endothelial cells regulates endothelial cell signaling and function. The review places particular emphasis on relevance to disease, including but not limited to Pulmonary and cardiovascular disorders as well as cancer. In addition to caveolin-1, possible importance of the less-studied endothelial caveolin-2 in pulmonary diseases will be also discussed.

15.
Pediatr Ther ; 2012(Suppl 2)2012.
Article in English | MEDLINE | ID: mdl-26618071

ABSTRACT

Despite the progress in medical treatment sepsis remains one of the major causes of death in pediatric and elderly patients. Understanding signaling pathways associated with sepsis may be of key significance for designing more efficient therapeutic approaches which could alleviate sepsis outcome. Earlier studies suggested that cholesteroland sphingolipid-rich lipid rafts and their morphologically distinct subset, caveolaecan be utilized by certain bacterial pathogens to enter and invade host cells. Moreover, there is also evidence that the expression levels of the major caveolar coat proteincaveolin-1 can be regulated by the major component of the outer membrane of Gram-negative bacteria,lipopolysaccharide (LPS) in various cell types involved in sepsis. In particular recent studies using caveolin-1 knockout mice and cells have revealed that caveolin-1 is directly involved in regulating numerous signalingpathways and functions in various cell types of the immune system and other cell types involved in sepsis. Moreover, the most recent report implies that in addition to extensively studied caveolin-1, caveolin-2 is also important in regulating LPS-induced sepsis and might possibly play an opposite role to caveolin-1 in regulating certain pro-inflammatory signaling pathways. The purpose of this review is to discuss these new exciting discoveries relatedto the specific role of caveolin-1 and the less studiedcaveolin-2in regulating signaling and outcome associated with sepsis induced by LPS and pathogenic bacteria at molecular, cellular and systemic levels.

16.
Am J Physiol Cell Physiol ; 301(5): C1161-74, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21832243

ABSTRACT

Using a combination of wild-type (WT) and caveolin-2 (Cav-2) knockout along with retroviral reexpression approaches, we provide the evidence for the negative role of Cav-2 in regulating anti-proliferative function and signaling of transforming growth factor ß (TGF-ß) in endothelial cells (ECs). Although, TGF-ß had a modest inhibitory effect on WT ECs, it profoundly inhibited proliferation of Cav-2 knockout ECs. To confirm the specificity of the observed difference in response to TGF-ß, we have stably reexpressed Cav-2 in Cav-2 knockout ECs using a retroviral approach. Similar to WT ECs, the anti-proliferative effect of TGF-ß was dramatically reduced in the Cav-2 reexpressing ECs. The reduced anti-proliferative effect of TGF-ß in Cav-2-positive cells was evidenced by three independent proliferation assays: 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), cell count, and bromodeoxyuridine incorporation and correlated with a loss of TGF-ß-mediated upregulation of cell cycle inhibitor p27 and subsequent reduction of the levels of hyperphosphorylated (inactive) form of the retinoblastoma protein in Cav-2 reexpressing ECs. Mechanistically, Cav-2 inhibits anti-proliferative action of TGF-ß by suppressing Alk5-Smad2/3 pathway manifested by reduced magnitude and length of TGF-ß-induced Smad2/3 phosphorylation as well as activation of activin receptor-like kinase-5 (Alk5)-Smad2/3 target genes plasminogen activator inhibitor-1 and collagen type I in Cav-2-positive ECs. Expression of Cav-2 does not appear to significantly change targeting of TGF-ß receptors I and Smad2/3 to caveolar and lipid raft microdomains as determined by sucrose fractionation gradient. Overall, the negative regulation of TGF-ß signaling and function by Cav-2 is independent of Cav-1 expression levels and is not because of changing targeting of Cav-1 protein to plasma membrane lipid raft/caveolar domains.


Subject(s)
Caveolin 2/metabolism , Cell Proliferation/drug effects , Lung/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/drug effects , Animals , Cells, Cultured , Collagen Type I/metabolism , Endothelial Cells/drug effects , Lung/drug effects , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plasminogen Activator Inhibitor 1/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Retinoblastoma Protein/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism
17.
Biochem Res Int ; 2011: 809259, 2011.
Article in English | MEDLINE | ID: mdl-22229094

ABSTRACT

Caveolin-2 is one of the major protein components of cholesterol- and glycosphingolipid-rich flask-shaped invaginations of plasma membrane caveolae. A new body of evidence suggests that caveolin-2 plays an important, and often more direct, role than caveolin-1 in regulating signaling and function in a cell- and tissue type-specific manner. The purpose of this paper is to primarily focus on discussing how these recent discoveries may help better understand the specific contribution of caveolin-2 to lipid raft- and caveolae-regulated cell/tissue-specific signaling and functions.

18.
Mol Cell Proteomics ; 9(10): 2109-24, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20585024

ABSTRACT

Caveolae are organelles abundant in the plasma membrane of many specialized cells including endothelial cells (ECs), epithelial cells, and adipocytes, and in these cells, caveolin-1 (Cav-1) is the major coat protein essential for the formation of caveolae. To identify proteins that require Cav-1 for stable incorporation into membrane raft domains, a quantitative proteomics analysis using isobaric tagging for relative and absolute quantification was performed on rafts isolated from wild-type and Cav-1-deficient mice. In three independent experiments, 117 proteins were consistently identified in membrane rafts with the largest differences in the levels of Cav-2 and in the caveola regulatory proteins Cavin-1 and Cavin-2. Because the lung is highly enriched in ECs, we validated and characterized the role of the newly described protein Cavin-1 in several cardiovascular tissues and in ECs. Cavin-1 was highly expressed in ECs lining blood vessels and in cultured ECs. Knockdown of Cavin-1 reduced the levels of Cav-1 and -2 and weakly influenced the formation of high molecular weight oligomers containing Cav-1 and -2. Cavin-1 silencing enhanced basal nitric oxide release from ECs but blocked proangiogenic phenotypes such as EC proliferation, migration, and morphogenesis in vitro. Thus, these data support an important role of Cavin-1 as a regulator of caveola function in ECs.


Subject(s)
Caveolin 1/metabolism , DNA Polymerase I/metabolism , Proteomics , Animals , Base Sequence , Blotting, Western , Caveolin 1/genetics , Cell Line , Cell Movement , Cell Proliferation , Chromatography, Ion Exchange , Gene Silencing , Humans , Mass Spectrometry , Mice , Mice, Knockout , Microscopy, Fluorescence , Nitric Oxide/metabolism , RNA, Small Interfering
19.
Am J Physiol Cell Physiol ; 298(3): C693-701, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20007452

ABSTRACT

The goal of this study was to determine whether caveolin-2 (Cav-2) is capable of controlling endothelial cell (EC) proliferation in vitro. To realize this goal, we have directly compared proliferation rates and cell cycle-associated signaling proteins between lung ECs isolated from wild-type (WT) and Cav-2 knockout (KO) mice. Using three independent proliferation assays, we have determined that Cav-2 KO ECs proliferate by ca. 2-fold faster than their WT counterparts. Cell cycle analysis by flow cytometry of propidium iodide-stained cells showed a relatively higher percentage of Cav-2 KO ECs in S and G(2)/M and lower percentage in G(o)/G(1) phases of cell cycle relative to their WT counterparts. Furthermore, an over 2-fold increase in the percentage of S phase-associated Cav-2 KO relative to WT ECs was independently determined with bromodeoxyuridine incorporation assay. Mechanistically, the increase in proliferation/cell cycle progression of Cav-2 KO ECs correlated well with elevated expression levels of predominantly S phase- and G(2)/M phase-associated cyclin A and B1, respectively. Further mechanistic analysis of molecular events controlling cell cycle progression revealed increased level of hyperphosphorylated (inactive) form of G(1) to S phase transition inhibitor, the retinoblastoma protein in hyperproliferating Cav-2 KO ECs. Conversely, the expression level of the two cyclin-dependent kinase inhibitors p16(INK4) and p27(Kip1) was reduced in Cav-2 KO ECs. Finally, increased phosphorylation (activation) of proproliferative extracellular signal-regulated kinase 1/2 was observed in hyperproliferating Cav-2 KO ECs. Overall, our data suggest that Cav-2 negatively regulates lung EC proliferation and cell cycle progression.


Subject(s)
Caveolin 2/deficiency , Cell Cycle , Cell Proliferation , Endothelial Cells/metabolism , Animals , Caveolin 2/genetics , Cells, Cultured , Cyclin A/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Flow Cytometry , Intracellular Signaling Peptides and Proteins/metabolism , Lung/blood supply , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Retinoblastoma Protein/metabolism , Signal Transduction , Time Factors
20.
Biochemistry ; 47(1): 101-11, 2008 Jan 08.
Article in English | MEDLINE | ID: mdl-18081315

ABSTRACT

In the present study, using a combination of reconstituted systems and endothelial cells endogenously expressing caveolins, we show that phosphorylation of caveolin-2 at serines 23 and 36 can be differentially regulated by caveolin-1 mediated subcellular targeting to lipid raft/caveolae and in endothelial cells synchronized in mitosis. Detergent insolubility and sucrose flotation gradient experiments revealed that serine 23 phosphorylation of caveolin-2 preferably occurs in detergent-resistant membranes (DRMs), while serine 36 phosphorylation takes place in non-DRMs. Furthermore, immunofluorescence microscopy studies determined that in the presence of caveolin-1, serine 23-phosphorylated caveolin-2 mostly localizes to plasma membrane, while serine 36-phosphorylated caveolin-2 primarily resides in intracellular compartments. To directly address the role of caveolin-1 in regulating phosphorylation of endogenous caveolin-2, we have used the siRNA approach. The specific knockdown of caveolin-1 in endothelial cells decreases caveolin-2 phosphorylation at serine 23 but not at serine 36. Thus, upregulation of serine 23 phosphorylation of caveolin-2 depends on caveolin-1-driven targeting to plasma membrane lipid rafts and caveolae. Interestingly, although serine 36 phosphorylation does not seem to be regulated in endothelial cells by caveolin-1, it can be selectively upregulated in endothelial cells synchronized in mitosis. The latter data suggests a possible involvement of serine 36-phosphorylated caveolin-2 in modulating mitosis.


Subject(s)
Caveolae/metabolism , Caveolin 2/metabolism , Endothelial Cells/metabolism , Membrane Microdomains/metabolism , Serine/metabolism , Caveolin 1/chemistry , Caveolin 1/genetics , Caveolin 1/metabolism , Caveolin 2/chemistry , Caveolin 2/genetics , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Detergents/chemistry , Dimerization , Endothelial Cells/cytology , Flow Cytometry , Humans , Microscopy, Fluorescence , Mitosis/physiology , Phosphorylation , Protein Transport , RNA, Small Interfering/genetics , Serine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL