Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Publication year range
1.
J Proteome Res ; 23(4): 1433-1442, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38488493

ABSTRACT

MDR3 (multidrug resistance 3) deficiency in humans (MDR2 in mice) causes progressive familial intrahepatic cholestasis type 3 (PFIC3). PFIC3 is a lethal disease characterized by an early onset of intrahepatic cholestasis progressing to liver cirrhosis, a preneoplastic condition, putting individuals at risk of hepatocellular carcinoma (HCC). Hepatocyte-like organoids from MDR2-deficient mice (MDR2KO) were used in this work to study the molecular alterations caused by the deficiency of this transporter. Proteomic analysis by mass spectrometry allowed characterization of 279 proteins that were differentially expressed in MDR2KO compared with wild-type organoids. Functional enrichment analysis indicated alterations in three main cellular functions: (1) interaction with the extracellular matrix, (2) remodeling intermediary metabolism, and (3) cell proliferation and differentiation. The affected cellular processes were validated by orthogonal molecular biology techniques. Our results point to molecular mechanisms associated with PFIC3 that may drive the progression to liver cirrhosis and HCC and suggest proteins and cellular processes that could be targeted for the development of early detection strategies for these severe liver diseases.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B , Carcinoma, Hepatocellular , Cholestasis, Intrahepatic , Cholestasis , Liver Neoplasms , Animals , Humans , Mice , ATP Binding Cassette Transporter, Subfamily B/deficiency , Carcinoma, Hepatocellular/pathology , Cholestasis/genetics , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice, Knockout , Proteomics
2.
Arterioscler Thromb Vasc Biol ; 43(11): 2213-2222, 2023 11.
Article in English | MEDLINE | ID: mdl-37732482

ABSTRACT

BACKGROUND: Systemic inflammatory diseases, such as sepsis and severe COVID-19, provoke acute respiratory distress syndrome in which the pathological hyperpermeability of the microvasculature, induced by uncontrolled inflammatory stimulation, causes pulmonary edema. Identifying the inflammatory mediators that induce human lung microvascular endothelial cell barrier dysfunction is essential to find the best anti-inflammatory treatments for critically ill acute respiratory distress syndrome patients. METHODS: We have compared the responses of primary human lung microvascular endothelial cells to the main inflammatory mediators involved in cytokine storms induced by sepsis and SARS-CoV2 pulmonary infection and to sera from healthy donors and severely ill patients with sepsis. Endothelial barrier function was measured by electric cell-substrate impedance sensing, quantitative confocal microscopy, and Western blot. RESULTS: The human lung microvascular endothelial cell barrier was completely disrupted by IL (interleukin)-6 conjugated with soluble IL-6R (IL-6 receptor) and by IL-1ß (interleukin-1beta), moderately affected by TNF (tumor necrosis factor)-α and IFN (interferon)-γ and unaffected by other cytokines and chemokines, such as IL-6, IL-8, MCP (monocyte chemoattractant protein)-1 and MCP-3. The inhibition of IL-1 and IL-6R simultaneously, but not separately, significantly reduced endothelial hyperpermeability on exposing human lung microvascular endothelial cells to a cytokine storm consisting of 8 inflammatory mediators or to sera from patients with sepsis. Simultaneous inhibition of IL-1 and JAK (Janus kinase)-STAT (signal transducer and activator of transcription protein), a signaling node downstream IL-6 and IFN-γ, also prevented septic serum-induced endothelial barrier disruption. CONCLUSIONS: These findings strongly suggest a major role for both IL-6 trans-signaling and IL-1ß signaling in the pathological increase in permeability of the human lung microvasculature and reveal combinatorial strategies that enable the gradual control of pulmonary endothelial barrier function in response to a cytokine storm.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Sepsis , Humans , Interleukin-6/metabolism , Cytokine Release Syndrome , Endothelial Cells/metabolism , RNA, Viral/metabolism , Lung/metabolism , Interferon-gamma/metabolism , Tumor Necrosis Factor-alpha/metabolism , COVID-19/metabolism , Sepsis/metabolism , Interleukin-1/metabolism
3.
Cell Mol Life Sci ; 79(1): 61, 2022 Jan 09.
Article in English | MEDLINE | ID: mdl-34999972

ABSTRACT

Apical localization of Intercellular Adhesion Receptor (ICAM)-1 regulates the adhesion and guidance of leukocytes across polarized epithelial barriers. Here, we investigate the molecular mechanisms that determine ICAM-1 localization into apical membrane domains of polarized hepatic epithelial cells, and their effect on lymphocyte-hepatic epithelial cell interaction. We had previously shown that segregation of ICAM-1 into apical membrane domains, which form bile canaliculi and bile ducts in hepatic epithelial cells, requires basolateral-to-apical transcytosis. Searching for protein machinery potentially involved in ICAM-1 polarization we found that the SNARE-associated protein plasmolipin (PLLP) is expressed in the subapical compartment of hepatic epithelial cells in vitro and in vivo. BioID analysis of ICAM-1 revealed proximal interaction between this adhesion receptor and PLLP. ICAM-1 colocalized and interacted with PLLP during the transcytosis of the receptor. PLLP gene editing and silencing increased the basolateral localization and reduced the apical confinement of ICAM-1 without affecting apicobasal polarity of hepatic epithelial cells, indicating that ICAM-1 transcytosis is specifically impaired in the absence of PLLP. Importantly, PLLP depletion was sufficient to increase T-cell adhesion to hepatic epithelial cells. Such an increase depended on the epithelial cell polarity and ICAM-1 expression, showing that the epithelial transcytotic machinery regulates the adhesion of lymphocytes to polarized epithelial cells. Our findings strongly suggest that the polarized intracellular transport of adhesion receptors constitutes a new regulatory layer of the epithelial inflammatory response.


Subject(s)
Cell Adhesion/physiology , Epithelial Cells/metabolism , Hepatocytes/metabolism , Intercellular Adhesion Molecule-1/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , T-Lymphocytes/metabolism , Cell Line, Tumor , Hep G2 Cells , Humans , Liver/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins/genetics , Transcytosis/physiology
4.
Cell Mol Life Sci ; 79(10): 535, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36180740

ABSTRACT

Preservation of blood vessel integrity, which is critical for normal physiology and organ function, is controlled at multiple levels, including endothelial junctions. However, the mechanism that controls the adequate assembly of endothelial cell junctions is not fully defined. Here, we uncover TAp73 transcription factor as a vascular architect that orchestrates transcriptional programs involved in cell junction establishment and developmental blood vessel morphogenesis and identify Angiomotin (AMOT) as a TAp73 direct transcriptional target. Knockdown of p73 in endothelial cells not only results in decreased Angiomotin expression and localization at intercellular junctions, but also affects its downstream function regarding Yes-associated protein (YAP) cytoplasmic sequestration upon cell-cell contact. Analysis of adherens junctional morphology after p73-knockdown in human endothelial cells revealed striking alterations, particularly a sharp increase in serrated junctions and actin bundles appearing as stress fibers, both features associated with enhanced barrier permeability. In turn, stabilization of Angiomotin levels rescued those junctional defects, confirming that TAp73 controls endothelial junction dynamics, at least in part, through the regulation of Angiomotin. The observed defects in monolayer integrity were linked to hyperpermeability and reduced transendothelial electric resistance. Moreover, p73-knockout retinas showed a defective sprout morphology coupled with hemorrhages, highlighting the physiological relevance of p73 regulation in the maintenance of vessel integrity in vivo. We propose a new model in which TAp73 acts as a vascular architect integrating transcriptional programs that will impinge with Angiomotin/YAP signaling to maintain junctional dynamics and integrity, while balancing endothelial cell rearrangements in angiogenic vessels.


Subject(s)
Angiomotins , Endothelial Cells , Actins/metabolism , Cadherins/metabolism , Endothelial Cells/metabolism , Humans , Intercellular Junctions/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins
5.
Cell Mol Life Sci ; 77(11): 2125-2140, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31396656

ABSTRACT

VE-cadherin plays a central role in controlling endothelial barrier function, which is transiently disrupted by proinflammatory cytokines such as tumor necrosis factor (TNFα). Here we show that human endothelial cells compensate VE-cadherin degradation in response to TNFα by inducing VE-cadherin de novo synthesis. This compensation increases adherens junction turnover but maintains surface VE-cadherin levels constant. NF-κB inhibition strongly reduced VE-cadherin expression and provoked endothelial barrier collapse. Bacterial lipopolysaccharide and TNFα upregulated the transcription factor ETS1, in vivo and in vitro, in an NF-κB dependent manner. ETS1 gene silencing specifically reduced VE-cadherin protein expression in response to TNFα and exacerbated TNFα-induced barrier disruption. We propose that TNFα induces not only the expression of genes involved in increasing permeability to small molecules and immune cells, but also a homeostatic transcriptional program in which NF-κB- and ETS1-regulated VE-cadherin expression prevents the irreversible damage of endothelial barriers.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adherens Junctions/genetics , Adherens Junctions/metabolism , Animals , Antigens, CD/genetics , Cadherins/genetics , Capillary Permeability , Endothelial Cells/cytology , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Mice , Proteolysis , Proto-Oncogene Protein c-ets-1/genetics , Up-Regulation
6.
Int J Mol Sci ; 22(17)2021 Sep 02.
Article in English | MEDLINE | ID: mdl-34502456

ABSTRACT

Emerging evidence has suggested that dysbiosis of the gut microbiota may influence the drug efficacy of colorectal cancer (CRC) patients during cancer treatment by modulating drug metabolism and the host immune response. Moreover, gut microbiota can produce metabolites that may influence tumor proliferation and therapy responsiveness. In this study we have investigated the potential contribution of the gut microbiota and microbial-derived metabolites such as short chain fatty acids and polyamines to neoadjuvant radiochemotherapy (RCT) outcome in CRC patients. First, we established a profile for healthy gut microbiota by comparing the microbial diversity and composition between CRC patients and healthy controls. Second, our metagenomic analysis revealed that the gut microbiota composition of CRC patients was relatively stable over treatment time with neoadjuvant RCT. Nevertheless, treated patients who achieved clinical benefits from RTC (responders, R) had significantly higher microbial diversity and richness compared to non-responder patients (NR). Importantly, the fecal microbiota of the R was enriched in butyrate-producing bacteria and had significantly higher levels of acetic, butyric, isobutyric, and hexanoic acids than NR. In addition, NR patients exhibited higher serum levels of spermine and acetyl polyamines (oncometabolites related to CRC) as well as zonulin (gut permeability marker), and their gut microbiota was abundant in pro-inflammatory species. Finally, we identified a baseline consortium of five bacterial species that could potentially predict CRC treatment outcome. Overall, our results suggest that the gut microbiota may have an important role in the response to cancer therapies in CRC patients.


Subject(s)
Colorectal Neoplasms/therapy , Fatty Acids, Volatile , Gastrointestinal Microbiome , Neoadjuvant Therapy , Polyamines/blood , Aged , Case-Control Studies , Colorectal Neoplasms/microbiology , Feces/chemistry , Feces/microbiology , Female , Humans , Intestinal Mucosa/metabolism , Male , Middle Aged , Permeability , Treatment Outcome
7.
Cell Mol Life Sci ; 76(7): 1299-1317, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30552441

ABSTRACT

The vasculature is the paradigm of a compartment generated by parallel cellular barriers that aims to transport oxygen, nutrients and immune cells in complex organisms. Vascular barrier dysfunction leads to fatal acute and chronic inflammatory diseases. The endothelial barrier lines the inner side of vessels and is the main regulator of vascular permeability. Cadherins comprise a superfamily of 114 calcium-dependent adhesion proteins that contain conserved cadherin motifs and form cell-cell junctions in metazoans. In mature human endothelial cells, only VE (vascular endothelial)-cadherin and N (neural)-cadherin have been investigated in detail. Although both cadherins are essential for regulating endothelial permeability, no comprehensive expression studies to identify which other family members could play a relevant role in endothelial cells has so far been performed. Here, we have reviewed gene and protein expression databases to analyze cadherin expression in mature human endothelium and found that at least 24 cadherin superfamily members are significantly expressed. Based on data obtained from other cell types, organisms and experimental models, we discuss their potential functions, many of them unrelated to the formation of endothelial cell-cell junctions. The expression of this new set of endothelial cadherins highlights the important but still poorly defined roles of planar cell polarity, the Hippo pathway and mitochondria metabolism in human vascular homeostasis.


Subject(s)
Cadherins/metabolism , Endothelial Cells/metabolism , Calcium-Binding Proteins/metabolism , Endothelium, Vascular/metabolism , Humans , Protein Isoforms/metabolism , Protein Precursors/metabolism , Proto-Oncogene Proteins c-ret/metabolism
8.
Proc Natl Acad Sci U S A ; 114(45): 11838-11843, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078386

ABSTRACT

Controlling interactions between proteins and nanoparticles in electrolyte solutions is crucial for advancing biological sciences and biotechnology. The assembly of charged nanoparticles (NPs) and proteins in aqueous solutions can be directed by modifying the salt concentration. High concentrations of monovalent salt can induce the solubilization or crystallization of NPs and proteins. By using a multiscale coarse-grained molecular dynamics approach, we show that, due to ionic correlations in the electrolyte, NPs pairs at high monovalent salt concentrations interact via remarkably strong long-range attractions or repulsions, which can be split into three regimes depending on the surface charge densities of the NPs. NPs with zero-to-low surface charge densities interact via a long-range attraction that is stronger and has a similar range to the depletion attraction induced by polymers with radius of gyrations comparable to the NP diameter. On the other hand, moderately charged NPs with smooth surfaces as well as DNA-functionalized NPs with no possibility of hybridization between them interact via a strong repulsion of range and strength larger than the repulsion predicted by models that neglect ionic correlations, including the Derjaguin-Landau-Vervey-Overbeek (DLVO) model. Interactions between strongly charged NPs (>2 e/nm2), both types smooth and DNA-functionalized NPs, show an attractive potential well at intermediate-to-high salt concentrations, which demonstrates that electrolytes can induce aggregation of strongly charged NPs. Our work provides an improved understanding of the role of ionic correlations in NP assembly and design rules to utilize the salting-out process to crystallize NPs.

9.
Int J Mol Sci ; 21(18)2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32947866

ABSTRACT

Obesity is considered an important factor that increases the risk of colorectal cancer (CRC). So far, the association of gut microbiota with both obesity and cancer has been described independently. Nevertheless, a specific obesity-related microbial profile linked to CRC development has not been identified. The aim of this study was to determine the gut microbiota composition in fecal samples from CRC patients with (OB-CRC) and without obesity (L-CRC) compared to the microbiota profile present in non-obese healthy controls (L-HC), in order to unravel the possible relationship between gut microbiota and microbial-derived metabolite trimethylamine N-oxide (TMAO), the inflammatory status, and the intestinal permeability in the context of obesity-associated CRC. The presence of obesity does not induce significant changes in the diversity and richness of intestinal bacteria of CRC patients. Nevertheless, OB-CRC patients display a specific gut microbiota profile characterized by a reduction in butyrate-producing bacteria and an overabundance of opportunistic pathogens, which in turn could be responsible, at least in part, for the higher levels of proinflammatory cytokine IL-1ß, the deleterious bacterial metabolite TMAO, and gut permeability found in these patients. These results suggest a possible role of obesity-related gut microbiota in the development of CRC, which could give new clues for the design of new diagnostic tools for CRC prevention.


Subject(s)
Bacteria/isolation & purification , Colorectal Neoplasms/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Inflammation/microbiology , Obesity/microbiology , Aged , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Biomarkers , Body Mass Index , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Dysbiosis/complications , Dysbiosis/pathology , Dysbiosis/physiopathology , Feces/microbiology , Female , Haptoglobins , Humans , Inflammation/blood , Inflammation Mediators/blood , Interleukins/blood , Male , Metagenome , Methylamines/adverse effects , Methylamines/blood , Middle Aged , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Permeability , Protein Precursors/blood
10.
Proc Natl Acad Sci U S A ; 113(38): 10485-90, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27601636

ABSTRACT

In this work, we present a joint experimental and molecular dynamics simulations effort to understand and map the crystallization behavior of polyhedral nanoparticles assembled via the interaction of DNA surface ligands. In these systems, we systematically investigated the interplay between the effects of particle core (via the particle symmetry and particle size) and ligands (via the ligand length) on crystallization behavior. This investigation revealed rich phase diagrams, previously unobserved phase transitions in polyhedral crystallization behavior, and an unexpected symmetry breaking in the ligand distribution on a particle surface. To understand these results, we introduce the concept of a zone of anisotropy, or the portion of the phase space where the anisotropy of the particle is preserved in the crystallization behavior. Through comparison of the zone of anisotropy for each particle we develop a foundational roadmap to guide future investigations.


Subject(s)
Anisotropy , Crystallization , DNA/chemistry , Nanoparticles/chemistry , DNA/drug effects , Ligands , Molecular Dynamics Simulation , Particle Size , Surface Properties , X-Ray Diffraction
11.
Nano Lett ; 17(8): 5126-5132, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28731353

ABSTRACT

Colloidal crystal engineering with DNA can be used to realize precise control over nanoparticle (NP) arrangement. Here, we investigate a case of DNA-based assembly where the properties of DNA as a polyelectrolyte brush are employed to alter a hybridization-driven NP crystallization pathway. Using the coassembly of DNA-conjugated proteins and spherical gold nanoparticles (AuNPs) as a model system, we explore how steric repulsion between noncomplementary, neighboring NPs due to overlapping DNA shells can influence their ligand-directed behavior. Specifically, our experimental data coupled with coarse-grained molecular dynamics (MD) simulations reveal that, by changing factors related to NP repulsion, two structurally distinct outcomes can be achieved. When steric repulsion between DNA-AuNPs is significantly greater than that between DNA-proteins, a lower packing density crystal lattice is favored over the structure that is predicted by design rules based on DNA hybridization considerations alone. This is enabled by the large difference in DNA density on AuNPs versus proteins and can be tuned by modulating the flexibility, and thus conformational entropy, of the DNA on the constituent particles. At intermediate ligand flexibility, the crystallization pathways are energetically similar, and the structural outcome can be adjusted using the density of DNA duplexes on DNA-AuNPs and by screening the Coulomb potential between them. Such lattices are shown to undergo dynamic reorganization upon changing the salt concentration. These data help elucidate the structural considerations necessary for understanding repulsive forces in DNA-mediated assembly and lay the groundwork for using them to increase architectural diversity in engineering colloidal crystals.


Subject(s)
DNA/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , Animals , Catalase/chemistry , Cattle , Corynebacterium glutamicum/enzymology , Crystallization , Molecular Dynamics Simulation , Nucleic Acid Conformation , Nucleic Acid Hybridization , Protein Binding , Protein Multimerization , Thermodynamics
12.
J Mol Cell Cardiol ; 105: 12-23, 2017 04.
Article in English | MEDLINE | ID: mdl-28223221

ABSTRACT

Endothelial progenitor cells (EPCs) constitute a promising alternative in cardiovascular regenerative medicine due to their assigned role in angiogenesis and vascular repair. In response to injury, EPCs promote vascular remodeling by replacement of damaged endothelial cells and/or by secreting angiogenic factors over the damaged tissue. Nevertheless, such mechanisms need to be further characterized. In the current approach we have evaluated the initial response of early EPCs (eEPCs) from healthy individuals after direct contact with the factors released by carotid arteries complicated with atherosclerotic plaques (AP), in order to understand the mechanisms underlying the neovascularization and remodeling properties assigned to these cells. Herein, we found that the AP secretome stimulated eEPCs proliferation and mobilization ex vivo, and such increase was accompanied by augmented permeability, cell contraction and also an increase of cell-cell adhesion in association with raised vinculin levels. Furthermore, a comparative mass spectrometry analysis of control versus stimulated eEPCs revealed a differential expression of proteins in the AP treated cells, mostly involved in cell migration, proliferation and vascular remodeling. Some of these protein changes were also detected in the eEPCs isolated from atherosclerotic patients compared to eEPCs from healthy donors. We have shown, for the first time, that the AP released factors activate eEPCs ex vivo by inducing their mobilization together with the expression of vasculogenic related markers. The present approach could be taken as a ex vivo model to study the initial activation of vascular cells in atherosclerosis and also to evaluate strategies looking to potentiate the mobilization of EPCs prior to clinical applications.


Subject(s)
Endothelial Progenitor Cells/metabolism , Plaque, Atherosclerotic/metabolism , Proteome , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Permeability , Plaque, Atherosclerotic/pathology , Proteomics/methods
13.
J Am Chem Soc ; 139(5): 1754-1757, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28121437

ABSTRACT

Herein, we investigate the use of proteins with tunable DNA modification distributions to modulate nanoparticle superlattice structure. Using beta-galactosidase (ßgal) as a model system, we have employed the orthogonal chemical reactivities of surface amines and thiols to synthesize protein-DNA conjugates with 36 evenly distributed or 8 specifically positioned oligonucleotides. When these are assembled into crystalline superlattices with gold nanoparticles, we find that the distribution of DNA modifications modulates the favored structure: ßgal with uniformly distributed DNA bonding elements results in body-centered cubic crystals, whereas DNA functionalization of cysteines results in AB2 packing. We probe the role of protein oligonucleotide number and conjugate size on this observation, which revealed the importance of oligonucleotide distribution in this observed assembly behavior. These results indicate that proteins with defined DNA modification patterns are powerful tools for controlling nanoparticle superlattices architecture, and establish the importance of oligonucleotide distribution in the assembly behavior of protein-DNA conjugates.

14.
J Surg Oncol ; 115(3): 301-306, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28335081

ABSTRACT

BACKGROUND AND OBJECTIVE: We examined the prognostic value of obesity and nuclear ß-catenin in patients with locally advanced rectal cancer. METHODS: We prospectively recruited a total of 98 eligible patients with locally advanced cancer for preoperative radiochemotherapy followed by total mesorectal excision. Patients' height and weight were reaorded before radiochemotherapy, and the immunohistochemical expression of nuclear ß-catenin was analyzed. Disease-free survival (DFS) was analyzed using the Kaplan-Meier method and a Cox regression model was employed for the multivariate analysis. RESULTS: Obese patients were associated with a lower number of recurrences (3.6% vs. 34.3%, P = 0.001), and a higher DFS (95% vs. 53%; HR, 0.09; 95%CI, 0.01-0.64; P = 0.005) than non-obese patients. In the multivariate analysis, body mass index, nuclear ß-catenin expression, and the absence of lymph node metastases showed a significant increase in DFS. CONCLUSIONS: Obesity and nuclear ß-catenin are independent favorable prognostic factors for DFS in locally advanced cancer treated with preoperative radiochemotherapy. J. Surg. Oncol. 2017;115:301-306. © 2017 Wiley Periodicals, Inc.


Subject(s)
Obesity/pathology , Rectal Neoplasms/pathology , Rectal Neoplasms/therapy , beta Catenin/biosynthesis , Body Mass Index , Cell Nucleus/metabolism , Chemoradiotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Obesity/metabolism , Preoperative Care/methods , Prospective Studies , Rectal Neoplasms/metabolism , Rectal Neoplasms/surgery
15.
Exp Eye Res ; 153: 8-13, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27697549

ABSTRACT

Correct corneal endothelial barrier function is essential for maintaining corneal transparency. However, research on cell signaling pathways mediating corneal endothelial barrier dysfunction has progressed more slowly than that involving other cellular barriers because of the lack of human corneal endothelial cell models. Here we have optimized the culture of the human corneal endothelial cell (HCEC) line B4G12 as a model for studying paracellular permeability. We show that B4G12-HCECs form confluent monolayers with stable cell-cell junctions when cultured on plastic, but not glass, surfaces precoated with various extracellular matrix components. Cell morphometry and measuring intercellular spaces and transendothelial electric resistance indicate that B4G12-HCECs form optimal monolayers on collagen and fibronectin. Based on the use of specific inhibitors, it has been proposed that the Rho-regulated kinases, ROCK-I and ROCK-II, mediate actomyosin-induced contraction in corneal endothelial cell barriers. ROCKs are effectors of RhoA, RhoB and RhoC. We show that the GTPase RhoA and its effector ROCK-II are predominantly expressed in B4G12-HCECs and primary human corneal endothelial cells. The activation of Rho GTPases during acute barrier disruption has not been investigated in corneal endothelial cells. RhoA, but not other related GTPases that are highly expressed in B4G12-HCECs, such as Rac1 and Cdc42, is transiently activated during barrier disruption in response to the inflammatory mediator thrombin. Pharmacological inhibition of RhoA and ROCK reduces B4G12-HCEC acute contraction. We propose that exploiting B4G12-HCECs is a useful experimental strategy for gaining further insight into the signaling pathways involved in human corneal endothelial barrier function.


Subject(s)
Capillary Permeability/physiology , Endothelium, Corneal/metabolism , Intercellular Junctions/metabolism , Cell Membrane Permeability , Cells, Cultured , Endothelium, Corneal/cytology , Humans , Signal Transduction
16.
Mediators Inflamm ; 2016: 7650260, 2016.
Article in English | MEDLINE | ID: mdl-26941485

ABSTRACT

During the inflammatory response, immune cells egress from the circulation and follow a chemotactic and haptotactic gradient within the tissue, interacting with matrix components in the stroma and with parenchymal cells, which guide them towards the sites of inflammation. Polarized epithelial cells compartmentalize tissue cavities and are often exposed to inflammatory challenges such as toxics or infections in non-lymphoid tissues. Apicobasal polarity is critical to the specialized functions of these epithelia. Indeed, a common feature of epithelial dysfunction is the loss of polarity. Here we review evidence showing that apicobasal polarity regulates the inflammatory response: various polarized epithelia asymmetrically secrete chemotactic mediators and polarize adhesion receptors that dictate the route of leukocyte migration within the parenchyma. We also discuss recent findings showing that the loss of apicobasal polarity increases leukocyte adhesion to epithelial cells and the consequences that this could have for the inflammatory response towards damaged, infected or transformed epithelial cells.


Subject(s)
Epithelium/metabolism , Inflammation/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Animals , Cell Adhesion/physiology , Cell Polarity/physiology , Humans
17.
J Cell Sci ; 126(Pt 20): 4589-601, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23943871

ABSTRACT

The microtubule (MT) cytoskeleton is essential for many cellular processes, including cell polarity and migration. Cortical platforms, formed by a subset of MT plus-end-tracking proteins, such as CLASP2, and non-MT binding proteins such as LL5ß, attach distal ends of MTs to the cell cortex. However, the mechanisms involved in organizing these platforms have not yet been described in detail. Here we show that 4.1R, a FERM-domain-containing protein, interacts and colocalizes with cortical CLASP2 and is required for the correct number and dynamics of CLASP2 cortical platforms. Protein 4.1R also controls binding of CLASP2 to MTs at the cell edge by locally altering GSK3 activity. Furthermore, in 4.1R-knockdown cells MT plus-ends were maintained for longer in the vicinity of cell edges, but instead of being tethered to the cell cortex, MTs continued to grow, bending at cell margins and losing their radial distribution. Our results suggest a previously unidentified role for the scaffolding protein 4.1R in locally controlling CLASP2 behavior, CLASP2 cortical platform turnover and GSK3 activity, enabling correct MT organization and dynamics essential for cell polarity.


Subject(s)
Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Cell Movement/physiology , Cell Polarity/physiology , Gene Knockdown Techniques , HEK293 Cells , Humans , Microtubule-Associated Proteins/genetics , Protein Binding , Protein Structure, Tertiary
18.
Soft Matter ; 11(7): 1386-96, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25579173

ABSTRACT

Recent reports of the synthesis and assembly of faceted nanoplates with a wide range of shapes and composition motivates the possibility of a new class of two-dimensional materials with specific patterns targeted for a host of exciting properties. Yet, studies of how nanoplate shape controls their assembly - knowledge necessary for their inverse design from target structures - has been performed for only a handful of systems. By constructing a general framework in which many known faceted nanoplates may be described in terms of four anisotropy dimensions, we discover design rules to guide future synthesis and assembly. We study via Monte Carlo simulations attractive polygons whose shape is altered systematically under the following four transformations: faceting, pinching, elongation and truncation. We report that (i) faceting leads to regular porous structures (ii) pinching stabilizes complex structures such as dodecagonal quasicrystals (iii) elongation leads to asymmetric phase behavior, where low and high aspect ratio nanoplates self-assemble completely different structures and (iv) low and high degrees of truncation transform a complex self-assembler into a disk-like assembler, providing design ideas that could lead to switchable structures. We provide important insight into how the shape and attractive interactions of a nanoplate can be exploited or designed to target specific classes of structures, including space-filling, porous, and complex tilings.

19.
BMC Neurosci ; 15: 110, 2014 Sep 22.
Article in English | MEDLINE | ID: mdl-25242463

ABSTRACT

BACKGROUND: Cell-derived microparticles are secreted in response to cell damage or dysfunction. Endothelial and platelet dysfunction are thought to contribute to the development of multiple sclerosis (MS). Our aim here is, first, to compare the presence of microparticles of endothelial and platelet origin in plasma from patients with different clinical forms of MS and with clinically isolated syndrome. Second, to investigate the effect of microparticles on endothelial barrier function. RESULTS: Platelet-poor plasma from 95 patients (12 with clinically isolated syndrome, 51 relapsing-remitting, 23 secondary progressive, 9 primary progressive) and 49 healthy controls were analyzed for the presence of platelet-derived and endothelium-derived microparticles by flow cytometry. The plasma concentration of platelet-derived and endothelium-derived microparticles increased in all clinical forms of MS and in clinically isolated syndrome versus controls. The response of endothelial barriers to purified microparticles was measured by electric cell-substrate impedance sensing. Microparticles from relapsing-remitting MS patients induced, at equivalent concentrations, a stronger disruption of endothelial barriers than those from healthy donors or from patients with clinically isolated syndrome. MS microparticles acted synergistically with the inflammatory mediator thrombin to disrupt the endothelial barrier function. CONCLUSIONS: Plasma microparticles should be considered not only as markers of early stages of MS, but also as pathological factors with the potential to increase endothelial permeability and leukocyte infiltration.


Subject(s)
Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Demyelinating Diseases/physiopathology , Endothelium, Vascular/metabolism , Multiple Sclerosis, Chronic Progressive/physiopathology , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Adolescent , Adult , Aged , Capillary Permeability , Child , Electric Impedance , Female , Flow Cytometry , Humans , Male , Middle Aged , Thrombin/metabolism , Young Adult
20.
BMC Cancer ; 14: 511, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-25015560

ABSTRACT

BACKGROUND: New biomarkers are needed for the prognosis of advanced colorectal cancer, which remains incurable by conventional treatments. O6-methylguanine DNA methyltransferase (MGMT) methylation and protein expression have been related to colorectal cancer treatment failure and tumor progression. Moreover, the presence in these tumors of cancer stem cells, which are characterized by CD133 expression, has been associated with chemoresistance, radioresistance, metastasis, and local recurrence. The objective of this study was to determine the prognostic value of CD133 and MGMT and their possible interaction in colorectal cancer patients. METHODS: MGMT and CD133 expression was analyzed by immunohistochemistry in 123 paraffin-embedded colorectal adenocarcinoma samples, obtaining the percentage staining and intensity. MGMT promoter methylation status was obtained by using bisulfite modification and methylation-specific PCR (MSP). These values were correlated with clinical data, including overall survival (OS), disease-free survival (DFS), tumor stage, and differentiation grade. RESULTS: Low MGMT expression intensity was significantly correlated with shorter OS and was a prognostic factor independently of treatment and histopathological variables. High percentage of CD133 expression was significantly correlated with shorter DFS but was not an independent factor. Patients with low-intensity MGMT expression and ≥50% CD133 expression had the poorest DFS and OS outcomes. CONCLUSIONS: Our results support the hypothesis that MGMT expression may be an OS biomarker as useful as tumor stage or differentiation grade and that CD133 expression may be a predictive biomarker of DFS. Thus, MGMT and CD133 may both be useful for determining the prognosis of colorectal cancer patients and to identify those requiring more aggressive adjuvant therapies. Future studies will be necessary to determine its clinical utility.


Subject(s)
Adenocarcinoma/pathology , Antigens, CD/metabolism , Colorectal Neoplasms/pathology , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Glycoproteins/metabolism , Peptides/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , AC133 Antigen , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cross-Sectional Studies , DNA Methylation , Disease-Free Survival , Female , Humans , Male , Middle Aged , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL