1
|
Dan L, Jiamei X, Ning W, Yanxiang G, Mengli T. Predictors and prognostic significance of the volume load trajectory: a longitudinal study in patients on peritoneal dialysis. Ren Fail 2024; 46:2405561. [PMID: 39301865 PMCID: PMC11418055 DOI: 10.1080/0886022x.2024.2405561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/27/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
Volume overload in peritoneal dialysis patients is a common issue that can lead to poor prognosis. We employed a group trajectory model to categorize volume load trajectories and examined the factors associated with each trajectory class to explore the impact of different trajectory groups on clinical prognosis and residual renal function (RRF). This single-center prospective cohort study included 214 patients on maintenance peritoneal dialysis within a tertiary hospital. The ratio of extracellular water to total body water was measured using Bioimpedance analysis. The SAS 9.4 PROC Traj procedure was used to examine the group-based trajectory of the patients. A multivariate logistic regression model was used to calculate the adjusted odds ratios (aOR) of the associated factors to predict the trajectory class of participants. The average age of the included patients was 53.56 (SD: 11.77) years, with a male proportion of 46.7% and a median follow-up time of 6 months. The normal stable group accounted for 35.05% of the total population and maintained a normal and stable level, the moderate stable group accounted for 52.8% of the total population and showed a slightly higher and stable level, and the high fluctuation group accounted for 12.15% of the total population and showed a high and fluctuating level. A multivariate logistic regression analysis revealed that age, diabetes, and albumin levels are significant factors influencing the categorization of volume load trajectories. There were statistically significant differences in both the technical survival rate and the loss of residual renal function among the three trajectory groups.
Collapse
Affiliation(s)
- Liu Dan
- Nephrology Department, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Xu Jiamei
- Nephrology Department, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Weng Ning
- Nephrology Department, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Guo Yanxiang
- Nephrology Department, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Tong Mengli
- Nephrology Department, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Curry FE, Taxt T, Rygh CB, Pavlin T, Bjørnstad R, Døskeland SO, Reed RK. Epac1 -/- mice have elevated baseline permeability and do not respond to histamine as measured with dynamic contrast-enhanced magnetic resonance imaging with contrast agents of different molecular weights. Acta Physiol (Oxf) 2019; 225:e13199. [PMID: 30300965 PMCID: PMC6646910 DOI: 10.1111/apha.13199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 09/29/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022]
Abstract
Aim Epac1−/− mice, but not Epac2−/− mice have elevated baseline permeability to albumin. This study extends the investigations of how Epac‐dependent pathways modulate transvascular exchange in response to the classical inflammatory agent histamine. It also evaluates the limitations of models of blood‐to‐tissue exchange in transgenic mice in DCE‐MRI measurements. Methods We measured DCE‐MRI signal intensity in masseter muscle of wt and Epac1−/− mice with established approaches from capillary physiology to determine how changes in blood flow and vascular permeability contribute to overall changes of microvascular flux. We used two tracers, the high molecular weight tracer (Gadomer‐17, MW 17 kDa, apparent MW 30‐35 kDa) is expected to be primarily limited by diffusion and therefore less dependent on changes in blood flow and the low molecular weight tracer (Dotarem (MW 0.56 kDa) whose transvascular exchange is determined by both blood flow and permeability. Paired experiments in each animal combined with analytical methods provided an internally consistent description of microvascular transport. Results Epac1−/− mice had elevated baseline permeability relative to wt control mice for Dotarem and Gadomer‐17. In contrast to wt mice, Epac1−/− mice failed to increase transvascular permeability in response to histamine. Dotarem underestimated blood flow and vascular volume and Gadomer‐17 has limited sensitivity in extravascular accumulation. Conclusion The study suggests that the normal barrier loosening effect of histamine in venular microvessels do not function when the normal barrier tightening effect of Epac1 is already compromised. The study also demonstrated that the numerical analysis of DCE‐MRI data with tracers of different molecular weight has significant limitations.
Collapse
Affiliation(s)
- Fitz‐Roy E. Curry
- Department of Physiology and Membrane Biology University of California Davis Davis California
| | - Torfinn Taxt
- Department of Biomedicine University of Bergen Bergen Norway
| | - Cecilie Brekke Rygh
- Department of Biomedicine University of Bergen Bergen Norway
- Molecular Imaging Centre Department of Biomedicine University of Bergen Bergen Norway
| | - Tina Pavlin
- Department of Biomedicine University of Bergen Bergen Norway
- Molecular Imaging Centre Department of Biomedicine University of Bergen Bergen Norway
| | - Ronja Bjørnstad
- Department of Biomedicine University of Bergen Bergen Norway
| | | | - Rolf K. Reed
- Department of Biomedicine University of Bergen Bergen Norway
- Centre for Cancer Biomarkers University of Bergen Bergen Norway
| |
Collapse
|
3
|
Fite BZ, Kheirolomoom A, Foiret JL, Seo JW, Mahakian LM, Ingham ES, Tam SM, Borowsky AD, Curry FRE, Ferrara KW. Dynamic contrast enhanced MRI detects changes in vascular transport rate constants following treatment with thermally-sensitive liposomal doxorubicin. J Control Release 2017; 256:203-213. [PMID: 28395970 DOI: 10.1016/j.jconrel.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 03/16/2017] [Accepted: 04/05/2017] [Indexed: 01/03/2023]
Abstract
Temperature-sensitive liposomal formulations of chemotherapeutics, such as doxorubicin, can achieve locally high drug concentrations within a tumor and tumor vasculature while maintaining low systemic toxicity. Further, doxorubicin delivery by temperature-sensitive liposomes can reliably cure local cancer in mouse models. Histological sections of treated tumors have detected red blood cell extravasation within tumors treated with temperature-sensitive doxorubicin and ultrasound hyperthermia. We hypothesize that the local release of drug into the tumor vasculature and resulting high drug concentration can alter vascular transport rate constants along with having direct tumoricidal effects. Dynamic contrast enhanced MRI (DCE-MRI) coupled with a pharmacokinetic model can detect and quantify changes in such vascular transport rate constants. Here, we set out to determine whether changes in rate constants resulting from intravascular drug release were detectable by MRI. We found that the accumulation of gadoteridol was enhanced in tumors treated with temperature-sensitive liposomal doxorubicin and ultrasound hyperthermia. While the initial uptake rate of the small molecule tracer was slower (k1=0.0478±0.011s-1 versus 0.116±0.047s-1) in treated compared to untreated tumors, the tracer was retained after treatment due to a larger reduction in the rate of clearance (k2=0.291±0.030s-1 versus 0.747±0.24s-1). While DCE-MRI assesses a combination of blood flow and permeability, ultrasound imaging of microvascular flow rate is sensitive only to changes in vascular flow rate; based on this technique, blood flow was not significantly altered 30min after treatment. In summary, DCE-MRI provides a means to detect changes that are associated with treatment by thermally-activated particles and such changes can be exploited to enhance local delivery.
Collapse
Affiliation(s)
- Brett Z Fite
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Azadeh Kheirolomoom
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Josquin L Foiret
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Jai W Seo
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| | - Alexander D Borowsky
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA 95616, USA.
| | - Fitz-Roy E Curry
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA.
| | - Katherine W Ferrara
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| |
Collapse
|
4
|
Takaya Y, Yoshihara F, Yokoyama H, Kanzaki H, Kitakaze M, Goto Y, Anzai T, Yasuda S, Ogawa H, Kawano Y, Kangawa K. Impact of decreased serum albumin levels on acute kidney injury in patients with acute decompensated heart failure: a potential association of atrial natriuretic peptide. Heart Vessels 2017; 32:932-943. [PMID: 28176004 DOI: 10.1007/s00380-017-0954-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 01/20/2017] [Indexed: 01/22/2023]
Abstract
Although hypoalbuminemia at admission is a risk for acute kidney injury (AKI) and mortality in patients with acute decompensated heart failure (ADHF), the clinical significance of decreased serum albumin levels (DAL) during ADHF therapy has not been elucidated. This study aimed to evaluate whether DAL was associated with AKI, and whether intravenous atrial natriuretic peptide (ANP) administration, which provides an effective treatment for ADHF but promotes albumin extravasation, was associated with DAL and AKI. A total of 231 consecutive patients with ADHF were enrolled. AKI was defined as ≥0.3 mg/dl absolute or 1.5-fold increase in serum creatinine levels within 48 h. AKI occurred in 73 (32%) of the 231 patients during ADHF therapy. The median value of decreases in serum albumin levels was 0.3 g/dl at 7 days after admission. When DAL was defined as ≥0.3 g/dl decrease in serum albumin levels, DAL occurred in 113 patients, and was independently associated with AKI. Of the 231 patients, 73 (32%) were treated with intravenous ANP. DAL occurred more frequently in patients receiving ANP than in those not receiving ANP (77 vs. 36%, p < 0.001), and ANP was independently associated with DAL. The incidence of AKI was higher in patients receiving ANP than in those not receiving ANP (48 vs. 24%, p < 0.001). ANP was independently associated with AKI. In conclusion, DAL is associated with AKI. Intravenous ANP administration may be one of the promoting factors of DAL, which leads to AKI, indicating a possible novel mechanism of AKI.
Collapse
Affiliation(s)
- Yoichi Takaya
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Fumiki Yoshihara
- Department of Hypertension and Nephrology, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan.
| | - Hiroyuki Yokoyama
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Hideaki Kanzaki
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Masafumi Kitakaze
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yoichi Goto
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Toshihisa Anzai
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Satoshi Yasuda
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Hisao Ogawa
- Department of Cardiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yuhei Kawano
- Department of Hypertension and Nephrology, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka, 565-8565, Japan
| | - Kenji Kangawa
- Research Institute, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
5
|
Kopperud RK, Rygh CB, Karlsen TV, Krakstad C, Kleppe R, Hoivik EA, Bakke M, Tenstad O, Selheim F, Lidén Å, Madsen L, Pavlin T, Taxt T, Kristiansen K, Curry FRE, Reed RK, Døskeland SO. Increased microvascular permeability in mice lacking Epac1 (Rapgef3). Acta Physiol (Oxf) 2017; 219:441-452. [PMID: 27096875 PMCID: PMC5073050 DOI: 10.1111/apha.12697] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/15/2016] [Accepted: 04/14/2016] [Indexed: 12/22/2022]
Abstract
Aim Maintenance of the blood and extracellular volume requires tight control of endothelial macromolecule permeability, which is regulated by cAMP signalling. This study probes the role of the cAMP mediators rap guanine nucleotide exchange factor 3 and 4 (Epac1 and Epac2) for in vivo control of microvascular macromolecule permeability under basal conditions. Methods Epac1−/− and Epac2−/− C57BL/6J mice were produced and compared with wild‐type mice for transvascular flux of radio‐labelled albumin in skin, adipose tissue, intestine, heart and skeletal muscle. The transvascular leakage was also studied by dynamic contrast‐enhanced magnetic resonance imaging (DCE‐MRI) using the MRI contrast agent Gadomer‐17 as probe. Results Epac1−/− mice had constitutively increased transvascular macromolecule transport, indicating Epac1‐dependent restriction of baseline permeability. In addition, Epac1−/− mice showed little or no enhancement of vascular permeability in response to atrial natriuretic peptide (ANP), whether probed with labelled albumin or Gadomer‐17. Epac2−/− and wild‐type mice had similar basal and ANP‐stimulated clearances. Ultrastructure analysis revealed that Epac1−/− microvascular interendothelial junctions had constitutively less junctional complex. Conclusion Epac1 exerts a tonic inhibition of in vivo basal microvascular permeability. The loss of this tonic action increases baseline permeability, presumably by reducing the interendothelial permeability resistance. Part of the action of ANP to increase permeability in wild‐type microvessels may involve inhibition of the basal Epac1‐dependent activity.
Collapse
Affiliation(s)
- R. K. Kopperud
- Department of Biomedicine; University of Bergen; Bergen Norway
- Centre for Cancer Biomarkers (CCBIO); University of Bergen; Bergen Norway
| | - C. Brekke Rygh
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - T. V. Karlsen
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - C. Krakstad
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - R. Kleppe
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - E. A. Hoivik
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - M. Bakke
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - O. Tenstad
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - F. Selheim
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - Å. Lidén
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - L. Madsen
- Department of Biomedicine; University of Bergen; Bergen Norway
- Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - T. Pavlin
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - T. Taxt
- Department of Biomedicine; University of Bergen; Bergen Norway
| | - K. Kristiansen
- Department of Biology; University of Copenhagen; Copenhagen Denmark
| | - F.-R. E. Curry
- Department of Physiology and Membrane Biology; School of Medicine; University of California; Davis CA USA
| | - R. K. Reed
- Department of Biomedicine; University of Bergen; Bergen Norway
- Centre for Cancer Biomarkers (CCBIO); University of Bergen; Bergen Norway
| | - S. O. Døskeland
- Department of Biomedicine; University of Bergen; Bergen Norway
| |
Collapse
|
6
|
Curry FRE, Clark JF, Jiang Y, Kim MH, Adamson RH, Simon SI. The role of atrial natriuretic peptide to attenuate inflammation in a mouse skin wound and individually perfused rat mesenteric microvessels. Physiol Rep 2016; 4:4/18/e12968. [PMID: 27670406 PMCID: PMC5037917 DOI: 10.14814/phy2.12968] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 01/05/2023] Open
Abstract
We tested the hypothesis that the anti‐inflammatory actions of atrial natriuretic peptide (ANP) result from the modulation of leukocyte adhesion to inflamed endothelium and not solely ANP ligation of endothelial receptors to stabilize endothelial barrier function. We measured vascular permeability to albumin and accumulation of fluorescent neutrophils in a full‐thickness skin wound on the flank of LysM‐EGFP mice 24 h after formation. Vascular permeability in individually perfused rat mesenteric microvessels was also measured after leukocytes were washed out of the vessel lumen. Thrombin increased albumin permeability and increased the accumulation of neutrophils. The thrombin‐induced inflammatory responses were attenuated by pretreating the wound with ANP (30 min). During pretreatment ANP did not lower permeability, but transiently increased baseline albumin permeability concomitant with the reduction in neutrophil accumulation. ANP did not attenuate acute increases in permeability to histamine and bradykinin in individually perfused rat microvessels. The hypothesis that anti‐inflammatory actions of ANP depend solely on endothelial responses that stabilize the endothelial barrier is not supported by our results in either individually perfused microvessels in the absence of circulating leukocytes or the more chronic skin wound model. Our results conform to the alternate hypothesis that ANP modulates the interaction of leukocytes with the inflamed microvascular wall of the 24 h wound. Taken together with our previous observations that ANP reduces deformability of neutrophils and their strength of attachment, rolling, and transvascular migration, these observations provide the basis for additional investigations of ANP as an anti‐inflammatory agent to modulate leukocyte–endothelial cell interactions.
Collapse
Affiliation(s)
- Fitz-Roy E Curry
- Department of Physiology & Membrane Biology, University of California, Davis, Davis, California
| | - Joyce F Clark
- Department of Physiology & Membrane Biology, University of California, Davis, Davis, California
| | - Yanyan Jiang
- Department of Physiology & Membrane Biology, University of California, Davis, Davis, California
| | - Min-Ho Kim
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| | - Roger H Adamson
- Department of Physiology & Membrane Biology, University of California, Davis, Davis, California
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, Davis, California
| |
Collapse
|
7
|
Affiliation(s)
- Bengt Rippe
- Department of Nephrology; Faculty of Medicine; Lund University; Clinical Sciences Lund; Lund Sweden
| | - Carl M. Öberg
- Department of Nephrology; Faculty of Medicine; Lund University; Clinical Sciences Lund; Lund Sweden
| |
Collapse
|
8
|
Abstract
cGMP controls many cellular functions ranging from growth, viability, and differentiation to contractility, secretion, and ion transport. The mammalian genome encodes seven transmembrane guanylyl cyclases (GCs), GC-A to GC-G, which mainly modulate submembrane cGMP microdomains. These GCs share a unique topology comprising an extracellular domain, a short transmembrane region, and an intracellular COOH-terminal catalytic (cGMP synthesizing) region. GC-A mediates the endocrine effects of atrial and B-type natriuretic peptides regulating arterial blood pressure/volume and energy balance. GC-B is activated by C-type natriuretic peptide, stimulating endochondral ossification in autocrine way. GC-C mediates the paracrine effects of guanylins on intestinal ion transport and epithelial turnover. GC-E and GC-F are expressed in photoreceptor cells of the retina, and their activation by intracellular Ca(2+)-regulated proteins is essential for vision. Finally, in the rodent system two olfactorial GCs, GC-D and GC-G, are activated by low concentrations of CO2and by peptidergic (guanylins) and nonpeptidergic odorants as well as by coolness, which has implications for social behaviors. In the past years advances in human and mouse genetics as well as the development of sensitive biosensors monitoring the spatiotemporal dynamics of cGMP in living cells have provided novel relevant information about this receptor family. This increased our understanding of the mechanisms of signal transduction, regulation, and (dys)function of the membrane GCs, clarified their relevance for genetic and acquired diseases and, importantly, has revealed novel targets for therapies. The present review aims to illustrate these different features of membrane GCs and the main open questions in this field.
Collapse
Affiliation(s)
- Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
9
|
Curry FRE, Clark JF, Adamson RH. Microperfusion Technique to Investigate Regulation of Microvessel Permeability in Rat Mesentery. J Vis Exp 2015. [PMID: 26436435 DOI: 10.3791/53210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Experiments to measure the permeability properties of individually perfused microvessels provide a bridge between investigation of molecular and cellular mechanisms regulating vascular permeability in cultured endothelial cell monolayers and the functional exchange properties of whole microvascular beds. A method to cannulate and perfuse venular microvessels of rat mesentery and measure the hydraulic conductivity of the microvessel wall is described. The main equipment needed includes an intravital microscope with a large modified stage that supports micromanipulators to position three different microtools: (1) a beveled glass micropipette to cannulate and perfuse the microvessel; (2) a glass micro-occluder to transiently block perfusion and enable measurement of transvascular water flow movement at a measured hydrostatic pressure, and (3) a blunt glass rod to stabilize the mesenteric tissue at the site of cannulation. The modified Landis micro-occlusion technique uses red cells suspended in the artificial perfusate as markers of transvascular fluid movement, and also enables repeated measurements of these flows as experimental conditions are changed and hydrostatic and colloid osmotic pressure difference across the microvessels are carefully controlled. Measurements of hydraulic conductivity first using a control perfusate, then after re-cannulation of the same microvessel with the test perfusates enable paired comparisons of the microvessel response under these well-controlled conditions. Attempts to extend the method to microvessels in the mesentery of mice with genetic modifications expected to modify vascular permeability were severely limited because of the absence of long straight and unbranched microvessels in the mouse mesentery, but the recent availability of the rats with similar genetic modifications using the CRISPR/Cas9 technology is expected to open new areas of investigation where the methods described herein can be applied.
Collapse
Affiliation(s)
- Fitz-Roy E Curry
- Department of Physiology & Membrane Biology, University of California Davis
| | - Joyce F Clark
- Department of Physiology & Membrane Biology, University of California Davis
| | - Roger H Adamson
- Department of Physiology & Membrane Biology, University of California Davis;
| |
Collapse
|
10
|
Arteaga-Marrero N, Rygh CB, Mainou-Gomez JF, Nylund K, Roehrich D, Heggdal J, Matulaniec P, Gilja OH, Reed RK, Svensson L, Lutay N, Olsen DR. Multimodal approach to assess tumour vasculature and potential treatment effect with DCE-US and DCE-MRI quantification in CWR22 prostate tumour xenografts. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 10:428-37. [PMID: 26010530 DOI: 10.1002/cmmi.1645] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/16/2015] [Accepted: 04/04/2015] [Indexed: 01/01/2023]
Abstract
The aim of this study was to compare intratumoural heterogeneity and longitudinal changes assessed by dynamic contrast-enhanced ultrasound (DCE-US) and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in prostate tumour xenografts. In vivo DCE-US and DCE-MRI were obtained 24 h pre- (day 0) and post- (day 2) radiation treatment with a single dose of 7.5 Gy. Characterization of the tumour vasculature was determined by Brix pharmacokinetic analysis of the time-intensity curves. Histogram analysis of voxels showed significant changes (p < 0.001) from day 0 to day 2 in both modalities for kep , the exchange rate constant from the extracellular extravascular space to the plasma, and kel , the elimination rate constant of the contrast. In addition, kep and kel values from DCE-US were significantly higher than those derived from DCE-MRI at day 0 (p < 0.0001) for both groups. At day 2, kel followed the same tendency for both groups, whereas kep showed this tendency only for the treated group in intermediate-enhancement regions. Regarding kep median values, longitudinal changes were not found for any modality. However, at day 2, kep linked to DCE-US was correlated to MVD in high-enhancement areas for the treated group (p = 0.05). In contrast, correlation to necrosis was detected for the control group in intermediate-enhancement areas (p < 0.1). Intratumoural heterogeneity and longitudinal changes in tumour vasculature were assessed for both modalities. Microvascular parameters derived from DCE-US seem to provide reliable biomarkers during radiotherapy as validated by histology. Furthermore, DCE-US could be a stand-alone or a complementary technique.
Collapse
Affiliation(s)
- N Arteaga-Marrero
- Department of Physics and Technology, University of Bergen, Bergen, Norway
| | - C B Rygh
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - J F Mainou-Gomez
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - K Nylund
- National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - D Roehrich
- Department of Physics and Technology, University of Bergen, Bergen, Norway
| | - J Heggdal
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - P Matulaniec
- Department of Physics and Technology, University of Bergen, Bergen, Norway
| | - O H Gilja
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - R K Reed
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Centre for Cancer Biomarkers (CCBIO), University of Bergen, Norway
| | - L Svensson
- Section of Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - N Lutay
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - D R Olsen
- Department of Physics and Technology, University of Bergen, Bergen, Norway
| |
Collapse
|
11
|
Golden MH. Nutritional and other types of oedema, albumin, complex carbohydrates and the interstitium - a response to Malcolm Coulthard's hypothesis: Oedema in kwashiorkor is caused by hypo-albuminaemia. Paediatr Int Child Health 2015; 35:90-109. [PMID: 25844980 DOI: 10.1179/2046905515y.0000000010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The various types of oedema in man are considered in relation to Starling's hypothesis of fluid movement from capillaries, with the main emphasis on nutritional oedema and the nephrotic syndrome in children. It is concluded that each condition has sufficient anomalous findings to render Starling's hypothesis untenable. The finding that the endothelial glycocalyx is key to control of fluid movement from and into the capillaries calls for complete revision of our understanding of oedema formation. The factors so far known to affect the function of the glycocalyx are reviewed. As these depend upon sulphated proteoglycans and other glycosaminoglycans, the argument is advanced that the same abnormalities will extend to the interstitial space and that kwashiorkor is fundamentally related to a defect in sulphur metabolism which can explain all the clinical features of the condition, including the formation of oedema.
Collapse
Key Words
- Albumin,
- Aldosterone,
- Angiotensin,
- Beriberi,
- Edema,
- Epidemic dropsy,
- Famine oedema,
- Glycocalyx,
- Glycosaminoglycans,
- Heart failure,
- Hunger oedema,
- Kwashiorkor,
- Malnutrition,
- Nephrotic syndrome,
- Oedema,
- Potassium deficiency,
- Pre-eclampsia,
- Protein-energy malnutrition,
- Proteoglycans,
- Renin,
- Salt,
- Severe acute malnutrition
- Vitamin E deficiency,
- War oedema,
- Water,
Collapse
|
12
|
González-Esquivel AE, Charles-Niño CL, Pacheco-Moisés FP, Ortiz GG, Jaramillo-Juárez F, Rincón-Sánchez AR. Beneficial effects of quercetin on oxidative stress in liver and kidney induced by titanium dioxide (TiO2) nanoparticles in rats. Toxicol Mech Methods 2015; 25:166-75. [DOI: 10.3109/15376516.2015.1006491] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
13
|
Rygh CB, Løkka G, Heljasvaara R, Taxt T, Pavlin T, Sormunen R, Pihlajaniemi T, Curry FR, Tenstad O, Reed RK. Image-based assessment of microvascular function and structure in collagen XV- and XVIII-deficient mice. J Physiol 2013; 592:325-36. [PMID: 24218547 DOI: 10.1113/jphysiol.2013.263574] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Collagen XV and XVIII are ubiquitous constituents of basement membranes. We aimed to study the physiological roles of these two components of the permeability barrier non-invasively in striated muscle in mice deficient in collagen XV or XVIII by dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Structural information was obtained with transmission electron microscopy (TEM). MR data were analysed by two different analysis methods to quantify tissue perfusion and microcirculatory exchange parameters to rule out data analysis method-dependent results. Control mice (C57BL/6J Ola/Hsd strain) or mice lacking either collagen XV (Col15a1(-/-)) or XVIII (Col18a1(-/-)) were included in the study. MR images were acquired using a preclinical system using gadodiamide (Gd-DTPA-BMA, molecular weight 0.58 kDa) as a tracer. Exchange capacity (permeability (P)-surface area (S) product relative to blood flow (FB)) was increased in test mice compared to controls, but the contributions from P, S, and FB were different in these two phenotypes. FB was significantly increased in Col18a1(-/-), but slightly decreased in Col15a1(-/-). PS was significantly increased only in Col18a1(-/-) even though P was increased in both phenotypes suggesting S might also be reduced in Col15a1(-/-) mice. Immunohistochemistry and electron microscopy demonstrated alterations in capillary density and morphology in both knockout mouse strains in comparison to the control mice. Both collagen XV and XVIII are important for maintaining normal capillary permeability in the striated muscle. DCE-MRI and the perfusion analyses successfully determined microvascular haemodynamic parameters of genetically modified mice and gave results consistent with more invasive methods.
Collapse
Affiliation(s)
- C B Rygh
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Qin S, Fite BZ, Gagnon MKJ, Seo JW, Curry FR, Thorsen F, Ferrara KW. A physiological perspective on the use of imaging to assess the in vivo delivery of therapeutics. Ann Biomed Eng 2013; 42:280-98. [PMID: 24018607 DOI: 10.1007/s10439-013-0895-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 08/14/2013] [Indexed: 12/25/2022]
Abstract
Our goal is to provide a physiological perspective on the use of imaging to optimize and monitor the accumulation of nanotherapeutics within target tissues, with an emphasis on evaluating the pharmacokinetics of organic particles. Positron emission tomography (PET), magnetic resonance imaging (MRI) and ultrasound technologies, as well as methods to label nanotherapeutic constructs, have created tremendous opportunities for preclinical optimization of therapeutics and for personalized treatments in challenging disease states. Within the methodology summarized here, the accumulation of the construct is estimated directly from the image intensity. Particle extravasation is then estimated based on classical physiological measures. Specifically, the transport of nanotherapeutics is described using the concept of apparent permeability, which is defined as the net flux of solute across a blood vessel wall per unit surface area of the blood vessel and per unit solute concentration difference across the blood vessel wall. The apparent permeability to small molecule MRI constructs is accurately shown to be far larger than that estimated for proteins such as albumin or nanoconstructs such as liposomes. Further, the quantitative measurements of vascular permeability are shown to facilitate detection of the transition from a pre-malignant to a malignant cancer and to quantify the delivery enhancement resulting from interventions such as ultrasound. While PET-based estimates facilitate quantitative comparisons of many constructs, high field MRI proves useful in the visualization of model drugs within small lesions and in the evaluation of the release and intracellular trafficking of nanoparticles and cargo.
Collapse
Affiliation(s)
- Shengping Qin
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA,
| | | | | | | | | | | | | |
Collapse
|
15
|
Chen W, Oberwinkler H, Werner F, Gaßner B, Nakagawa H, Feil R, Hofmann F, Schlossmann J, Dietrich A, Gudermann T, Nishida M, Del Galdo S, Wieland T, Kuhn M. Atrial Natriuretic Peptide–Mediated Inhibition of Microcirculatory Endothelial Ca
2+
and Permeability Response to Histamine Involves cGMP-Dependent Protein Kinase I and TRPC6 Channels. Arterioscler Thromb Vasc Biol 2013; 33:2121-9. [DOI: 10.1161/atvbaha.113.001974] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Wen Chen
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Heike Oberwinkler
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Franziska Werner
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Birgit Gaßner
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Hitoshi Nakagawa
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Robert Feil
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Franz Hofmann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Jens Schlossmann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Alexander Dietrich
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Thomas Gudermann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Motohiro Nishida
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Sabrina Del Galdo
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Thomas Wieland
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Michaela Kuhn
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| |
Collapse
|
16
|
Scallan JP, Davis MJ, Huxley VH. Permeability and contractile responses of collecting lymphatic vessels elicited by atrial and brain natriuretic peptides. J Physiol 2013; 591:5071-81. [PMID: 23897233 DOI: 10.1113/jphysiol.2013.260042] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Atrial and brain natriuretic peptides (ANP and BNP, respectively) are cardiac hormones released into the bloodstream in response to hypervolaemia or fluid shifts to the central circulation. The actions of both peptides include natriuresis and diuresis, a decrease in systemic blood pressure, and inhibition of the renin-angiotensin-aldosterone system. Further, ANP and BNP elicit increases in blood microvessel permeability sufficient to cause protein and fluid extravasation into the interstitium to reduce the vascular volume. Given the importance of the lymphatic vasculature in maintaining fluid balance, we tested the hypothesis that ANP or BNP (100 nM) would likewise elevate lymphatic permeability (Ps) to serum albumin. Using a microfluorometric technique adapted to in vivo lymphatic vessels, we determined that rat mesenteric collecting lymphatic Ps to rat serum albumin increased by 2.0 ± 0.4-fold (P = 0.01, n = 7) and 2.7 ± 0.8-fold (P = 0.07, n = 7) with ANP and BNP, respectively. In addition to measuring Ps responses, we observed changes in spontaneous contraction amplitude and frequency from the albumin flux tracings in vivo. Notably, ANP abolished spontaneous contraction amplitude (P = 0.005) and frequency (P = 0.006), while BNP augmented both parameters by ∼2-fold (P < 0.01 each). These effects of ANP and BNP on contractile function were examined further by using an in vitro assay. In aggregate, these data support the theory that an increase in collecting lymphatic permeability opposes the absorptive function of the lymphatic capillaries, and aids in the retention of protein and fluid in the interstitial space to counteract volume expansion.
Collapse
Affiliation(s)
- Joshua P Scallan
- V. H. Huxley: Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, 1 Hospital Dr., MA415, Columbia, MO 65212, USA.
| | | | | |
Collapse
|
17
|
Curry FRE, Adamson RH. Tonic regulation of vascular permeability. Acta Physiol (Oxf) 2013; 207:628-49. [PMID: 23374222 PMCID: PMC4054936 DOI: 10.1111/apha.12076] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 01/10/2013] [Accepted: 01/28/2013] [Indexed: 12/30/2022]
Abstract
Our major theme is that the layered structure of the endothelial barrier requires continuous activation of signalling pathways regulated by sphingosine-1-phosphate (S1P) and intracellular cAMP. These pathways modulate the adherens junction, continuity of tight junction strands, and the balance of synthesis and degradation of glycocalyx components. We evaluate recent evidence that baseline permeability is maintained by constant activity of mechanisms involving the small GTPases Rap1 and Rac1. In the basal state, the barrier is compromised when activities of the small GTPases are reduced by low S1P supply or delivery. With inflammatory stimulus, increased permeability can be understood in part as the action of signalling to reduce Rap1 and Rac1 activation. With the hypothesis that microvessel permeability and selectivity under both normal and inflammatory conditions are regulated by mechanisms that are continuously active, it follows that when S1P or intracellular cAMP are elevated at the time of inflammatory stimulus, they can buffer changes induced by inflammatory agents and maintain normal barrier stability. When endothelium is exposed to inflammatory conditions and subsequently exposed to elevated S1P or intracellular cAMP, the same processes restore the functional barrier by first re-establishing the adherens junction, then modulating tight junctions and glycocalyx. In more extreme inflammatory conditions, loss of the inhibitory actions of Rac1-dependent mechanisms may promote expression of more inflammatory endothelial phenotypes by contributing to the up-regulation of RhoA-dependent contractile mechanisms and the sustained loss of surface glycocalyx allowing access of inflammatory cells to the endothelium.
Collapse
Affiliation(s)
- F-R E Curry
- Department of Physiology & Membrane Biology, School of Medicine, University of California at Davis, Davis, CA 95616, USA.
| | | |
Collapse
|
18
|
Abstract
The cardiac hormone atrial natriuretic peptide (ANP) is critically involved in the maintenance of arterial blood pressure and intravascular volume homeostasis. Its cGMP-producing GC-A receptor is densely expressed in the microvascular endothelium of the lung and systemic circulation, but the functional relevance is controversial. Some studies reported that ANP stimulates endothelial cell permeability, whereas others described that the peptide attenuates endothelial barrier dysfunction provoked by inflammatory agents such as thrombin or histamine. Many studies in vitro addressed the effects of ANP on endothelial proliferation and migration. Again, both pro- and anti-angiogenic properties were described. To unravel the role of the endothelial actions of ANP in vivo, we inactivated the murine GC-A gene selectively in endothelial cells by homologous loxP/Cre-mediated recombination. Our studies in these mice indicate that ANP, via endothelial GC-A, increases endothelial albumin permeability in the microcirculation of the skin and skeletal muscle. This effect is critically involved in the endocrine hypovolaemic, hypotensive actions of the cardiac hormone. On the other hand the homologous GC-A-activating B-type NP (BNP), which is produced by cardiac myocytes and many other cell types in response to stressors such as hypoxia, possibly exerts more paracrine than endocrine actions. For instance, within the ischaemic skeletal muscle BNP released from activated satellite cells can improve the regeneration of neighbouring endothelia. This review will focus on recent advancements in our understanding of endothelial NP/GC-A signalling in the pulmonary versus systemic circulation. It will discuss possible mechanisms accounting for the discrepant observations made for the endothelial actions of this hormone-receptor system and distinguish between (patho)physiological and pharmacological actions. Lastly it will emphasize the potential therapeutical implications derived from the actions of NPs on endothelial permeability and regeneration.
Collapse
Affiliation(s)
- Michaela Kuhn
- Physiologisches Institut der Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
19
|
Muradashvili N, Tyagi R, Lominadze D. A dual-tracer method for differentiating transendothelial transport from paracellular leakage in vivo and in vitro. Front Physiol 2012; 3:166. [PMID: 22754530 PMCID: PMC3385581 DOI: 10.3389/fphys.2012.00166] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 05/08/2012] [Indexed: 12/14/2022] Open
Abstract
Inflammation-induced impaired function of vascular endothelium may cause leakage of plasma proteins that can lead to edema. Proteins may leave the vascular lumen through two main paracellular and transcellular pathways. As the first involves endothelial cell (EC) junction proteins and the second caveolae formation, these two pathways are interconnected. Therefore, it is difficult to differentiate the prevailing role of one or the other pathway during pathology that causes inflammation. Here we present a newly developed dual-tracer probing method that allows differentiation of transcellular from paracellular transport during pathology. This fluorescence-based method can be used in vitro to test changes in EC layer permeability and in vivo in various animal vascular preparations. The method is based on comparison of low molecular weight molecule (LMWM) transport to that of high molecular weight molecule (HMWM) transport through the EC layer or the vascular wall during physiological and pathological conditions. Since the LMWM will leak through mainly the paracellular and HMWM will move through paracellular (when gaps between the ECs are wide enough) and transcellular pathways, the difference in transport rate (during normal conditions and pathology) of these molecules will indicate the prevailing transport pathway involved in overall protein crossing of vascular wall. Thus, the novel approach of assessing the transport kinetics of different size tracers in vivo by intravital microscopy can clarify questions related to identification of target pathways for drug delivery during various pathologies associated with elevated microvascular permeability.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville KY, USA
| | | | | |
Collapse
|
20
|
Watson KD, Lai CY, Qin S, Kruse DE, Lin YC, Seo JW, Cardiff RD, Mahakian LM, Beegle J, Ingham ES, Curry FR, Reed RK, Ferrara KW. Ultrasound increases nanoparticle delivery by reducing intratumoral pressure and increasing transport in epithelial and epithelial-mesenchymal transition tumors. Cancer Res 2012; 72:1485-93. [PMID: 22282664 DOI: 10.1158/0008-5472.can-11-3232] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Acquisition of the epithelial-mesenchymal transition (EMT) tumor phenotype is associated with impaired chemotherapeutic delivery and a poor prognosis. In this study, we investigated the application of therapeutic ultrasound methods available in the clinic to increase nanotherapeutic particle accumulation in epithelial and EMT tumors by labeling particles with a positron emission tomography tracer. Epithelial tumors were highly vascularized with tight cell-cell junctions, compared with EMT tumors where cells displayed an irregular, elongated shape with loosened cell-cell adhesions and a reduction in E-cadherin and cytokeratins 8/18 and 19. Without ultrasound, the accumulation of liposomal nanoparticles administered to tumors in vivo was approximately 1.5 times greater in epithelial tumors than EMT tumors. When ultrasound was applied, both nanoaccumulation and apparent tumor permeability were increased in both settings. Notably, ultrasound effects differed with thermal and mechanical indices, such that increasing the thermal ultrasound dose increased nanoaccumulation in EMT tumors. Taken together, our results illustrate how ultrasound can be used to enhance nanoparticle accumulation in tumors by reducing their intratumoral pressure and increasing their vascular permeability.
Collapse
Affiliation(s)
- Katherine D Watson
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Karlsen TV, McCormack E, Mujic M, Tenstad O, Wiig H. Minimally invasive quantification of lymph flow in mice and rats by imaging depot clearance of near-infrared albumin. Am J Physiol Heart Circ Physiol 2011; 302:H391-401. [PMID: 22101523 DOI: 10.1152/ajpheart.00842.2011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is a lack of available methods to noninvasively quantify lymphatic function in small experimental animals, a necessity for studies on lymphatic system pathophysiology. We present a new method to quantify lymph flow in mice and rats, based on optically monitoring the depot clearance of near-infrared fluorescently labeled albumin and subsequent calculation of removal rate constants (k). BSA was conjugated with Alexa680 NHS ester and remained stable in protein-rich solutions without free dye dissociation. To assess lymph flow, mice or rats were imaged every 30 or 60 min during a 3- to 6-h period following an intradermal injection of 0.5 or 1 μl Alexa680-albumin. Mice were awake between measurements, whereas rats were anesthetized throughout the experiment. The k, a parameter defined as equivalent to lymph flow, was calculated from the slopes of the resultant log-linear washout curves and averaged -0.40 ± 0.03 and -0.30 ± 0.02%/min for control C57BL/6 and C3H mice, respectively. Local administration of the vasoconstrictor endothelin-1 in mice led to a significant reduction in k, whereas overhydration in rats increased k, reflecting the coupling between capillary filtration and lymph flow. Furthermore, k was 50% of wild type in lymphedema Chy mice where dermal lymphatics are absent. We conclude that lymph flow can be determined as its rate constant k by optical imaging of depot clearance of submicroliter amounts of Alexa680-albumin. The method offers a minimally invasive, reproducible, and simple alternative to assess lymphatic function in mice and rats.
Collapse
Affiliation(s)
- Tine V Karlsen
- Department of Biomedicine, Hematology Section, University of Bergen, Jonas Lies Vei 91, Bergen, Norway.
| | | | | | | | | |
Collapse
|
22
|
Lin YC, Adamson RH, Clark JF, Reed RK, Curry FRE. Phosphodiesterase 4 inhibition attenuates plasma volume loss and transvascular exchange in volume-expanded mice. J Physiol 2011; 590:309-22. [PMID: 22083598 DOI: 10.1113/jphysiol.2011.213447] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We tested the hypothesis that inhibition of phosphodiesterase 4 (PDE4) with rolipram to increase vascular endothelial cAMP and stabilize the endothelial barrier would attenuate the action of endogenous atrial natriuretic peptide (ANP) to increase vascular permeability to the plasma protein albumin after an acute plasma volume expansion. After rolipram pretreatment (8 mg (kg body wt)(-1), intraperitoneal, 30 min) more than 95% of the peak increase in plasma volume after volume expansion (4.5% bovine serum albumin, 114 μl (g body wt)(-1) h(-1), 15 min) remained in the vascular space 75 min after the end of infusion, whereas only 67% of the fluid was retained in volume-expanded animals with no rolipram pretreatment. Rolipram significantly decreased 30 min fluorescently labelled albumin clearance (μl (g dry wt)(-1)) relative to untreated volume-expanded controls in skin (e.g. back, 10.4 ± 1.6 vs. 19.5 ± 3.6, P = 0.04), muscle (e.g. hamstring, 15.0 ± 1.9 vs. 20.8 ± 1.4, P = 0.04) and in colon, caecum, and rectum (average reduction close to 50%). The mass of muscle and skin tissue accounted for 70% of volume-expansion-dependent albumin shifts from plasma to interstitium. The results are consistent with observations that the PDE4 inhibitor rolipram attenuates ANP-induced increases in vascular permeability after infusion of exogenous ANP and observations of elevated central venous pressure after a similar volume expansion in mice with selective deletion of the endothelial ANP receptor. These observations may form the basis for new strategies to retain intravenous fluid containing macromolecules.
Collapse
Affiliation(s)
- Yueh-Chen Lin
- Department of Physiology and Membrane Biology, School of Medicine, 1 Shields Avenue, University of California, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
23
|
Do viral infections mimic bacterial sepsis? The role of microvascular permeability: A review of mechanisms and methods. Antiviral Res 2011; 93:2-15. [PMID: 22068147 DOI: 10.1016/j.antiviral.2011.10.019] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 10/12/2011] [Accepted: 10/22/2011] [Indexed: 12/13/2022]
Abstract
A dysregulated immune response and functional immunosuppression have been considered the major mechanisms of the bacterial sepsis syndrome. More recently, the loss of endothelial barrier function and resultant microvascular leak have been found to be a key determinant of the pathogenesis of bacterial sepsis. Whether a similar paradigm applies to systemic viral syndromes is not known. Answering this question has far-reaching implications for the development of future anti-viral therapeutic strategies. In this review, we provide an overview of the structure and function of the endothelium and how its barrier integrity is compromised in bacterial sepsis. The various in vitro and in vivo methodologies available to investigate vascular leak are reviewed. Emphasis is placed on the advantages and limitations of cell culture techniques, which represent the most commonly used methods. Within this context, we appraise recent studies of three viruses - hantavirus, human herpes virus 8 and dengue virus - that suggest microvascular leak may play a role in the pathogenesis of these viral infections. We conclude with a discussion of how endothelial barrier breakdown may occur in other viral infections such as H5N1 avian influenza virus.
Collapse
|
24
|
Chen W, Gassner B, Börner S, Nikolaev VO, Schlegel N, Waschke J, Steinbronn N, Strasser R, Kuhn M. Atrial natriuretic peptide enhances microvascular albumin permeability by the caveolae-mediated transcellular pathway. Cardiovasc Res 2011; 93:141-51. [PMID: 22025581 PMCID: PMC3243041 DOI: 10.1093/cvr/cvr279] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Cardiac atrial natriuretic peptide (ANP) participates in the maintenance of arterial blood pressure and intravascular volume homeostasis. The hypovolaemic effects of ANP result from coordinated actions in the kidney and systemic microcirculation. Hence, ANP, via its guanylyl cyclase-A (GC-A) receptor and intracellular cyclic GMP as second messenger, stimulates endothelial albumin permeability. Ultimately, this leads to a shift of plasma fluid into interstitial pools. Here we studied the role of caveolae-mediated transendothelial albumin transport in the hyperpermeability effects of ANP. METHODS AND RESULTS Intravital microscopy studies of the mouse cremaster microcirculation showed that ANP stimulates the extravasation of fluorescent albumin from post-capillary venules and causes arteriolar vasodilatation. The hyperpermeability effect was prevented in mice with conditional, endothelial deletion of GC-A (EC GC-A KO) or with deleted caveolin-1 (cav-1), the caveolae scaffold protein. In contrast, the vasodilating effect was preserved. Concomitantly, the acute hypovolaemic action of ANP was abolished in EC GC-A KO and Cav-1(-/-) mice. In cultured microvascular rat fat pad and mouse lung endothelial cells, ANP stimulated uptake and transendothelial transport of fluorescent albumin without altering endothelial electrical resistance. The stimulatory effect on albumin uptake was prevented in GC-A- or cav-1-deficient pulmonary endothelia. Finally, preparation of caveolin-enriched lipid rafts from mouse lung and western blotting showed that GC-A and cGMP-dependent protein kinase I partly co-localize with Cav-1 in caveolae microdomains. CONCLUSION ANP enhances transendothelial caveolae-mediated albumin transport via its GC-A receptor. This ANP-mediated cross-talk between the heart and the microcirculation is critically involved in the regulation of intravascular volume.
Collapse
Affiliation(s)
- Wen Chen
- Institute of Physiology, University of Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Bruegger D, Schwartz L, Chappell D, Jacob M, Rehm M, Vogeser M, Christ F, Reichart B, Becker BF. Release of atrial natriuretic peptide precedes shedding of the endothelial glycocalyx equally in patients undergoing on- and off-pump coronary artery bypass surgery. Basic Res Cardiol 2011; 106:1111-21. [DOI: 10.1007/s00395-011-0203-y] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 07/01/2011] [Accepted: 07/06/2011] [Indexed: 10/18/2022]
|
26
|
Rygh CB, Qin S, Seo JW, Mahakian LM, Zhang H, Adamson R, Chen JQ, Borowsky AD, Cardiff RD, Reed RK, Curry FRE, Ferrara KW. Longitudinal investigation of permeability and distribution of macromolecules in mouse malignant transformation using PET. Clin Cancer Res 2011; 17:550-9. [PMID: 21106723 PMCID: PMC3107124 DOI: 10.1158/1078-0432.ccr-10-2049] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE We apply positron emission tomography (PET) to elucidate changes in nanocarrier extravasation during the transition from premalignant to malignant cancer, providing insight into the use of imaging to characterize early cancerous lesions and the utility of nanoparticles in early disease. EXPERIMENTAL DESIGN Albumin and liposomes were labeled with (64)Cu (half-life 12.7 hours), and longitudinal PET and CT imaging studies were conducted in a mouse model of ductal carcinoma in situ. A pharmacokinetic model was applied to estimate the tumor vascular volume and permeability. RESULTS From early time points characterized by disseminated hyperproliferation, the enhanced vascular permeability facilitated lesion detection. During disease progression, the vascular volume fraction increased 1.6-fold and the apparent vascular permeability to albumin and liposomes increased ∼2.5-fold to 6.6 × 10(-8) and 1.3 × 10(-8) cm/s, respectively, with the accumulation of albumin increasing earlier in the disease process. In the malignant tumor, both tracers reached similar mean intratumoral concentrations of ∼6% ID/cc but the distribution of liposomes was more heterogeneous, ranging from 1% to 18% ID/cc compared with 1% to 9% ID/cc for albumin. The tumor-to-muscle ratio was 17.9 ± 8.1 and 7.1 ± 0.5 for liposomes and albumin, respectively, indicating a more specific delivery of liposomes than with albumin. CONCLUSIONS PET imaging of radiolabeled particles, validated by confocal imaging and histology, detected the transition from premalignant to malignant lesions and effectively quantified the associated changes in vascular permeability.
Collapse
Affiliation(s)
- Cecilie B. Rygh
- Department of Physiology & Membrane Biology, University of California. Davis, USA
- Department of Biomedicine, University of Bergen, Norway
- Heart and Circulatory Group, Haukeland University Hospital, Bergen, Norway
| | - Shengping Qin
- Department of Biomedical Engineering, University of California. Davis, USA
| | - Jai W. Seo
- Department of Biomedical Engineering, University of California. Davis, USA
| | - Lisa M. Mahakian
- Department of Biomedical Engineering, University of California. Davis, USA
| | - Hua Zhang
- Department of Biomedical Engineering, University of California. Davis, USA
| | - Roger Adamson
- Department of Physiology & Membrane Biology, University of California. Davis, USA
| | - Jane Q. Chen
- Department of Pathology & Laboratory Medicine, University of California. Davis, USA
| | | | - Robert D. Cardiff
- Department of Pathology & Laboratory Medicine, University of California. Davis, USA
| | - Rolf K. Reed
- Department of Biomedicine, University of Bergen, Norway
| | - Fitz-Roy E. Curry
- Department of Physiology & Membrane Biology, University of California. Davis, USA
- Department of Biomedicine, University of Bergen, Norway
| | | |
Collapse
|
27
|
Axelsson J, Rippe A, Rippe B. Transient and sustained increases in glomerular permeability following ANP infusion in rats. Am J Physiol Renal Physiol 2011; 300:F24-30. [DOI: 10.1152/ajprenal.00347.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The present study was performed to investigate the effects of systemic atrial natriuretic peptide (ANP) infusion on the glomerular permeability to macromolecules in rats. In anesthetized Wistar rats (250–280 g), the left urether was cannulated for urine collection while simultaneously blood access was achieved. Rats were continuously infused intravenously with ANP [30 ng·kg−1·min−1 (Lo-ANP; n = 8) or 800 ng·kg−1·min−1 (Hi-ANP; n = 10)] or 0.9% NaCl (SHAM; n = 16), respectively, and with polydisperse FITC-Ficoll-70/400 (molecular radius 13–90 Å) and 51Cr-EDTA for 2 h. Plasma and urine samples were taken at 5, 15, 30, 60, and 120 min of ANP infusion and analyzed by high-performance size-exclusion chromatography (HPLC) for determination of glomerular sieving coefficients (θ) for Ficoll. GFR was also assessed (51Cr-EDTA). In Hi-ANP, there was a rapid (within 5 min), but bimodal, increase in glomerular permeability. θ to high-molecular-weight Ficoll thus reached a maximum at 15 min, after which θ returned to near control at 30 min, to again increase moderately at 60 and 120 min. In Lo-ANP, there was also a rapid, reversible increase in glomerular θ, returning to near control at 30 min, followed by just a tendency of a sustained increase in permeability, but with a significant increase in “large-pore” radius. In conclusion, in Hi-ANP there was a rapid increase in glomerular permeability, with an early, partly reversible permeability peak, followed by a (moderate) sustained increase in permeability. In Lo-ANP animals, only the initial permeability peak was evident. In both Lo-ANP and Hi-ANP, the glomerular sieving pattern observed was found to mainly reflect an increase in the number and radius of large pores in the glomerular filter.
Collapse
Affiliation(s)
| | - Anna Rippe
- Department of Nephrology, Lund University, Lund, Sweden
| | - Bengt Rippe
- Department of Nephrology, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Lin YC, Samardzic H, Adamson RH, Renkin EM, Clark JF, Reed RK, Curry FRE. Phosphodiesterase 4 inhibition attenuates atrial natriuretic peptide-induced vascular hyperpermeability and loss of plasma volume. J Physiol 2010; 589:341-53. [PMID: 21098005 DOI: 10.1113/jphysiol.2010.199588] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Inhibition of phosphodiesterase 4 (PDE4) to increase endothelial cAMP and stabilize the endothelial barrier attenuates acute inflammatory increases in vascular permeability.We extended this approach to attenuate physiological increases in vascular permeability in response to atrial natriuretic peptide (ANP), which acts with the kidney to regulate plasma volume. We measured blood-to-tissue albumin clearance and changes in plasma volume in isoflurane-anaesthetized mice (C57BL/6J) pre-treated with rolipram (8 mg kg(-1) I.P., 30 min). Rolipram significantly reduced albumin permeability, measured using a dual-label fluorescence method, in skin and skeletal muscle compared with ANP alone (500 ng kg(-1) min(-1)). Skin and muscle tissue accounted for 70% of the reduction in whole body albumin clearance taking into account albumin clearance in gastrointestinal (GI) tissue, heart and kidney. The action of ANP and rolipram to modify albumin clearances in duodenum and jejunum could be accounted for by local increases in vascular perfusion to increase surface area for exchange. ANP increased haematocrit from 40.6% to 46.8%, corresponding to an average loss of 22% plasma fluid volume (227 μl), and this was almost completely reversed with rolipram. Renal water excretion accounted for less than 30% of plasma fluid loss indicating that reduced albumin permeability and reduced filtration into vasodilated GI tissue were the predominant actions of PDE4 inhibition. Similar fluid retention was measured in mice with endothelial-restricted deletion of the guanylyl cyclase-A receptor for ANP. Stabilizing the endothelial barrier to offset ANP-induced increases in vascular permeability may be part of a strategy to maintain plasma volume.
Collapse
Affiliation(s)
- Yueh-Chen Lin
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, CA 95616, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Becker BF, Chappell D, Jacob M. Endothelial glycocalyx and coronary vascular permeability: the fringe benefit. Basic Res Cardiol 2010; 105:687-701. [DOI: 10.1007/s00395-010-0118-z] [Citation(s) in RCA: 191] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 08/25/2010] [Accepted: 08/26/2010] [Indexed: 12/11/2022]
|
30
|
Curry FRE, Adamson RH. Vascular permeability modulation at the cell, microvessel, or whole organ level: towards closing gaps in our knowledge. Cardiovasc Res 2010; 87:218-29. [PMID: 20418473 PMCID: PMC2895542 DOI: 10.1093/cvr/cvq115] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 04/13/2010] [Accepted: 04/19/2010] [Indexed: 01/20/2023] Open
Abstract
Multiple processes modulate net blood-to-tissue exchange in a microvascular unit in normal and pathophysiological conditions. These include mechanisms that control the number and type of microvessels perfused, the balance of adhesion and contractile forces that determine the conductance of the spaces between endothelial cells to water and solutes, the pressure and chemical potential gradients determining the driving forces through these conductive pathways, and the organization of barriers to macromolecules in the endothelial glycocalyx. Powerful methods are available to investigate these mechanisms at the levels of cultured endothelial monolayers, isolated microvessels, and the microvascular units within intact organs. Here we focus on current problems that limit the integration of our knowledge of mechanisms investigated in detail at the cellular level into a more complete understanding of modulation of blood-to-tissue exchange in whole organs when the endothelial barrier is exposed to acute and more long-term inflammatory conditions. First, we review updated methods, applicable in mouse models of vascular permeability regulation, to investigate both acute and long-term changes in permeability. Methods to distinguish tracer accumulation due to change in perfusion from real increases in extravascular accumulation are emphasized. The second part of the review compares normal and increased permeability in individually perfused venular microvessels and endothelial cell monolayers. The heterogeneity of endothelial cell phenotypes in the baseline state and after exposure to injury and inflammatory conditions is emphasized. Lastly, we review new approaches to investigation of the glycocalyx barrier properties in cultured endothelial monolayers and in whole-body investigations.
Collapse
Affiliation(s)
- Fitz-Roy E Curry
- Department of Physiology and Membrane Biology, School of Medicine, University of California, 1 Shields Avenue, Davis, CA 95616, USA.
| | | |
Collapse
|
31
|
Affiliation(s)
- Rupert M Pearse
- Queen Marys University of London, Barts & The London School of Medicine and Dentistry, William Harvey Research Institute, London, UK.
| |
Collapse
|