Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS One ; 14(8): e0221317, 2019.
Article in English | MEDLINE | ID: mdl-31465445

ABSTRACT

The purpose of this prospective study was to evaluate the effects of single and repeated intra-articular administration of allogeneic, umbilical cord-derived, neonatal mesenchymal stem cells (MSC) in horses with lameness due to osteoarthritis (OA) of a metacarpophalangeal joint (MPJ). Twenty-eight horses were included. Horses were divided into two groups. Horses in group MSC1 received an MSC injection at M0 and a placebo injection at M1 (1 month after M0). Horses in group MSC2 received MSC injections at M0 and at M1. Joint injections were performed with a blinded syringe. Clinical assessment was performed by the treating veterinarian at M1, M2 and M6 (2 and 6 months after M0), including lameness evaluation, palpation and flexion of the joint. Radiographic examination of the treated joints was performed at inclusion and repeated at M6. Radiographs were anonymized and assessed by 2 ECVDI LA associate members. Short term safety assessment was performed by owner survey. A 2-month rehabilitation program was recommended to veterinarians. There was a significant improvement of the total clinical score for horses in both groups. There was no significant difference in the total clinical score between groups MSC1 and MSC2 at any time point in the study. There was no significant difference in the total radiographic OA score, osteophyte score, joint space width score and subchondral bone score between inclusion and M6. Owner-detected adverse effects to MSC injection were recorded in 18% of the horses. Lameness caused by OA improved significantly over the 6-month duration of the study after treatment with allogeneic neonatal umbilical cord-derived MSCs combined with 8 weeks rest and rehabilitation. There is no apparent clinical benefit of repeated intra-articular administration of MSCs at a 1-month interval in horses with MPJ OA when compared to the effect of a single injection.


Subject(s)
Horse Diseases/therapy , Horses , Mesenchymal Stem Cell Transplantation , Metacarpophalangeal Joint , Metatarsophalangeal Joint , Osteoarthritis/therapy , Allografts , Animals , Female , Horse Diseases/pathology , Horse Diseases/physiopathology , Male , Osteoarthritis/pathology , Osteoarthritis/physiopathology
2.
Oncotarget ; 7(26): 39421-39435, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27270646

ABSTRACT

Vascular endothelial growth factor-C (VEGF-C)-induced lymphangiogenesis and increased tissue drainage have been reported to inhibit acute and chronic inflammation, and an activated lymphatic endothelium might mediate peripheral tolerance. Using transgenic mice overexpressing VEGF-C in the skin, we found that under inflammatory conditions, VEGF-C-mediated expansion of the cutaneous lymphatic network establishes an immune-inhibitory microenvironment characterised by increased regulatory T (Treg) cells, immature CD11c+CD11b+ dendritic cells (DCs) and CD8+ cells exhibiting decreased effector function. Strikingly, lymphatic endothelial cell (LEC)-conditioned media (CM) potently suppress DC maturation with reduced expression of MHCII, CD40, and IL-6, and increased IL-10 and CCL2 expression. We identify an imbalance in prostaglandin synthase expression after LEC activation, favoring anti-inflammatory prostacyclin synthesis. Importantly, blockade of LEC prostaglandin synthesis partially restores DC maturity. LECs also produce TGF-ß1, contributing to the immune-inhibitory microenvironment. This study identifies novel mechanisms by which the lymphatic endothelium modulates cellular immune responses to limit inflammation.


Subject(s)
Dendritic Cells/cytology , Endothelial Cells/metabolism , Lymph Nodes/metabolism , Vascular Endothelial Growth Factor C/metabolism , Animals , Antigen Presentation , Bone Marrow Cells/cytology , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cell Movement , Chemokine CCL2/metabolism , Dendritic Cells/metabolism , Flow Cytometry , Humans , Immune Tolerance , Inflammation , Interleukin-10/metabolism , Lymph Nodes/pathology , Lymphangiogenesis/drug effects , Mice , Mice, Transgenic , Phenotype , Vascular Endothelial Growth Factor A/metabolism
3.
Vet Surg ; 44(8): 1012-20, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26455833

ABSTRACT

OBJECTIVE: To describe (1) preoperative findings and surgical technique, (2) intraoperative difficulties, and (3) postoperative complications and long-term outcome of equine cheek tooth extraction using a minimally invasive transbuccal screw extraction (MITSE) technique. STUDY DESIGN: Retrospective case series. ANIMALS: Fifty-four equids; 50 horses, 3 ponies, and 1 mule. METHODS: Fifty-eight MITSE procedures were performed to extract cheek teeth in 54 equids. Peri- and intraoperative difficulties, as well as short- (<1 month) and long-term (>6 months) postoperative complications were recorded. Followup information was obtained through telephone interviews, making specific inquiries about nasal discharge, facial asymmetry, and findings consistent with surgical site infection. RESULTS: Preoperative findings that prompted exodontia included 50 cheek teeth with apical infections, 48 fractures, 4 neoplasia, 2 displacements, and 1 supernumerary tooth. Previous oral extraction was attempted but had failed in 55/58 (95%) animals because of cheek tooth fracture in 28, or insufficient clinical crown for extraction with forceps in 27. MITSE was successful in removing the entire targeted dental structure in 47/58 (81%) procedures. However, MITSE failed to remove the entire targeted dental structure in 11/58 (19%) procedures and was followed by repulsion in 10/11 (91%). Short-term postoperative complications included bleeding (4/58 procedures, 7%) and transient facial nerve paralysis (4/58 procedures, 7%). Owners were satisfied with the functional and cosmetic outcome for 40/41 (98%) animals with followup. CONCLUSION: MITSE offers an alternate for cheek tooth extraction in equids, where conventional oral extraction is not possible or has failed. Overall, there was low morbidity, which compares favorably with invasive buccotomy or repulsion techniques.


Subject(s)
Bone Screws/veterinary , Horses/surgery , Intraoperative Complications/veterinary , Postoperative Complications/veterinary , Preoperative Care/veterinary , Tooth Extraction/veterinary , Animals , Bicuspid/surgery , Equidae/surgery , Female , Male , Molar/surgery , Retrospective Studies , Treatment Outcome
4.
PLoS One ; 9(6): e99297, 2014.
Article in English | MEDLINE | ID: mdl-24911791

ABSTRACT

Contact hypersensitivity (CHS) induced by topical application of haptens is a commonly used model to study dermal inflammatory responses in mice. Several recent studies have indicated that CHS-induced skin inflammation triggers lymphangiogenesis but may negatively impact the immune-function of lymphatic vessels, namely fluid drainage and dendritic cell (DC) migration to draining lymph nodes (dLNs). On the other hand, haptens have been shown to exert immune-stimulatory activity by inducing DC maturation. In this study we investigated how the presence of pre-established CHS-induced skin inflammation affects the induction of adaptive immunity in dLNs. Using a mouse model of oxazolone-induced skin inflammation we observed that lymphatic drainage was reduced and DC migration from skin to dLNs was partially compromised. At the same time, a significantly stronger adaptive immune response towards ovalbumin (OVA) was induced when immunization had occurred in CHS-inflamed skin as compared to uninflamed control skin. In fact, immunization with sterile OVA in CHS-inflamed skin evoked a delayed-type hypersensitivity (DTH) response comparable to the one induced by conventional immunization with OVA and adjuvant in uninflamed skin. Striking phenotypic and functional differences were observed when comparing DCs from LNs draining uninflamed or CHS-inflamed skin. DCs from LNs draining CHS-inflamed skin expressed higher levels of co-stimulatory molecules and MHC molecules, produced higher levels of the interleukin-12/23 p40 subunit (IL-12/23-p40) and more potently induced T cell activation in vitro. Immunization experiments revealed that blockade of IL-12/23-p40 during the priming phase partially reverted the CHS-induced enhancement of the adaptive immune response. Collectively, our findings indicate that CHS-induced skin inflammation generates an overall immune-stimulatory milieu, which outweighs the potentially suppressive effect of reduced lymphatic vessel function.


Subject(s)
Adaptive Immunity , Dermatitis, Contact/immunology , Lymph Nodes/immunology , Lymphangiogenesis/immunology , Adjuvants, Immunologic/adverse effects , Animals , Cell Movement/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis, Contact/genetics , Dermatitis, Contact/pathology , Disease Models, Animal , Interleukin-12/antagonists & inhibitors , Interleukin-12/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Oxazolone/adverse effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
Angiogenesis ; 17(2): 383-93, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24154862

ABSTRACT

Lymphatic vessels have traditionally been regarded as a rather inert drainage system, which just passively transports fluids, leukocytes and antigen. However, it is becoming increasingly clear that the lymphatic vasculature is highly dynamic and plays a much more active role in inflammatory and immune processes. Tissue inflammation induces a rapid, stimulus-specific upregulation of chemokines and adhesion molecules in lymphatic endothelial cells and a proliferative expansion of the lymphatic network in the inflamed tissue and in draining lymph nodes. Moreover, increasing evidence suggests that inflammation-induced changes in the lymphatic vasculature have a profound impact on the course of inflammatory and immune responses, by modulating fluid drainage, leukocyte migration or the removal of inflammatory mediators from tissues. In this review we will summarize and discuss current knowledge of the inflammatory response of lymphatic endothelium and of inflammation-induced lymphangiogenesis and the current perspective on the overall functional significance of these processes.


Subject(s)
Endothelium, Lymphatic/pathology , Inflammation/pathology , Animals , Endothelium, Lymphatic/metabolism , Humans , Inflammation/immunology , Inflammation Mediators/metabolism , Lymphangiogenesis/genetics
6.
Blood ; 122(13): 2271-81, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23963040

ABSTRACT

The cytokine interleukin (IL)-7 exerts essential roles in lymph node (LN) organogenesis and lymphocyte development and homeostasis. Recent studies have identified lymphatic endothelial cells (LECs) as a major source of IL-7 in LNs. Here, we report that LECs not only produce IL-7, but also express the IL-7 receptor chains IL-7Rα and CD132. Stimulation with recombinant IL-7 enhanced LEC in vitro activity and induced lymphangiogenesis in the cornea of wild-type (WT) mice. Whereas in IL-7Rα(-/-) mice, dermal lymphatic vessels (LVs) were abnormally organized and lymphatic drainage was compromised, transgenic overexpression of IL-7 in mice resulted in an expanded dermal LV network with increased drainage function. Moreover, systemic treatment with recombinant IL-7 enhanced lymphatic drainage in the skin of WT mice and of mice devoid of lymphocytes. Experiments in IL-7Rα(-/-) bone marrow chimeras demonstrated that the drainage-enhancing activity of IL-7 was exclusively dependent on IL-7Rα expression in stromal but not in hematopoietic cells. Finally, near-infrared in vivo imaging performed in IL-7Rα(-/-) mice revealed that the pumping activity of collecting vessels was normal but fluid uptake into lymphatic capillaries was defective. Overall, our data point toward an unexpected new role for IL-7 as a potential autocrine mediator of lymphatic drainage.


Subject(s)
Endothelial Cells/metabolism , Interleukin-7/metabolism , Lymphatic Vessels/metabolism , Animals , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout
7.
Cancer Res ; 73(14): 4212-21, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23695550

ABSTRACT

VEGF-C and VEGF-D were identified as lymphangiogenic growth factors and later shown to promote tumor metastasis, but their effects on carcinogenesis are poorly understood. Here, we have studied the effects of VEGF-C and VEGF-D on tumor development in the murine multistep chemical carcinogenesis model of squamous cell carcinoma by using a soluble VEGF-C/VEGF-D inhibitor. After topical treatment with a tumor initiator and repeated tumor promoter applications, transgenic mice expressing a soluble VEGF-C/VEGF-D receptor (sVEGFR-3) in the skin developed significantly fewer squamous cell tumors with a delayed onset when compared with wild-type mice or mice expressing sVEGFR-3 lacking the ligand-binding site. Epidermal proliferation was reduced in the carcinogen-treated transgenic skin, whereas epidermal keratinocyte proliferation in vitro was not affected by VEGF-C or VEGF-D, indicating indirect effects of sVEGFR-3 expression. Importantly, transgenic mouse skin was less sensitive to tumor promoter-induced inflammation, with reduced angiogenesis and blood vessel leakage. Cutaneous leukocytes, especially macrophages, were reduced in transgenic skin without major changes in macrophage polarization or blood monocyte numbers. Several macrophage-associated cytokines were also reduced in transgenic papillomas, although the dermal macrophages themselves did not express VEGFR-3. These findings indicate that VEGF-C/VEGF-D are involved in shaping the inflammatory tumor microenvironment that regulates early tumor progression. Our results support the use of VEGF-C/VEGF-D-blocking agents not only to inhibit metastatic progression, but also during the early stages of tumor growth.


Subject(s)
Carcinogenesis/drug effects , Inflammation/drug therapy , Skin Neoplasms/drug therapy , Skin/drug effects , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Vascular Endothelial Growth Factor D/antagonists & inhibitors , Animals , Carcinogenesis/chemically induced , Carcinogenesis/metabolism , Carcinogens , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation/drug effects , Cytokines/metabolism , Epidermis/drug effects , Epidermis/metabolism , Epidermis/pathology , Female , Inflammation/metabolism , Inflammation/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Leukocytes/drug effects , Leukocytes/metabolism , Leukocytes/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Skin/metabolism , Skin/pathology , Skin Neoplasms/chemically induced , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
8.
J Invest Dermatol ; 133(3): 751-758, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23096716

ABSTRACT

Systemic high-dose IL2 promotes long-term survival in a subset of metastatic melanoma patients, but this treatment is accompanied by severe toxicities. The immunocytokine L19-IL2, in which IL2 is fused to the human L19 antibody capable of selective accumulation on tumor neovasculature, has recently shown encouraging clinical activity in patients with metastatic melanoma. In this study, we have investigated the therapeutic performance of L19-IL2, administered systemically in combination with a murine anti-CTLA-4 antibody or with a second clinical-stage immunocytokine (L19-TNF) in two syngeneic immunocompetent mouse models of cancer. We observed complete tumor eradications when L19-IL2 was used in combination with CTLA-4 blockade. Interestingly, mice cured from F9 tumors developed new lesions when rechallenged with tumor cells after therapy, whereas mice cured from CT26 tumors were resistant to tumor rechallenge. Similarly, L19-IL2 induced complete remissions when administered in a single intratumoral injection in combination with L19-TNF, whereas the two components did not lead to cures when administered as single agents. These findings provide a rationale for combination trials in melanoma, as the individual therapeutic agents have been extensively studied in clinical trials, and the antigen recognized by the L19 antibody has an identical sequence in mouse and man.


Subject(s)
Antibodies, Anti-Idiotypic/therapeutic use , CTLA-4 Antigen/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Recombinant Fusion Proteins/therapeutic use , Teratocarcinoma/drug therapy , Animals , Antibodies, Anti-Idiotypic/administration & dosage , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents , CTLA-4 Antigen/immunology , Cell Line, Tumor , Colonic Neoplasms/pathology , Disease Models, Animal , Drug Therapy, Combination , Female , Humans , Injections, Intralesional , Ipilimumab , Melanoma/drug therapy , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Recombinant Fusion Proteins/administration & dosage , Skin Neoplasms/drug therapy , Teratocarcinoma/pathology
9.
Blood ; 120(11): 2249-58, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22855606

ABSTRACT

Dendritic cell (DC) migration via lymphatic vessels to draining lymph nodes (dLNs) is crucial for the initiation of adaptive immunity. We imaged this process by intravital microscopy (IVM) in the ear skin of transgenic mice bearing red-fluorescent vasculature and yellow-fluorescent DCs. DCs within lymphatic capillaries were rarely transported by flow, but actively migrated within lymphatics and were significantly faster than in the interstitium. Pharmacologic blockade of the Rho-associated protein kinase (ROCK), which mediates nuclear contraction and de-adhesion from integrin ligands, significantly reduced DC migration from skin to dLNs in steady-state. IVM revealed that ROCK blockade strongly reduced the velocity of interstitial DC migration, but only marginally affected intralymphatic DC migration. By contrast, during tissue inflammation, ROCK blockade profoundly decreased both interstitial and intralymphatic DC migration. Inhibition of intralymphatic migration was paralleled by a strong up-regulation of ICAM-1 in lymphatic endothelium, suggesting that during inflammation ROCK mediates de-adhesion of DC-expressed integrins from lymphatic-expressed ICAM-1. Flow chamber assays confirmed an involvement of lymphatic-expressed ICAM-1 and DC-expressed ROCK in DC crawling on lymphatic endothelium. Overall, our findings further define the role of ROCK in DC migration to dLNs and reveal a differential requirement for ROCK in intralymphatic DC crawling during steady-state and inflammation.


Subject(s)
Cell Movement , Dendritic Cells/metabolism , Dermatitis, Contact/metabolism , Endothelium, Lymphatic/immunology , rho-Associated Kinases/metabolism , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Crosses, Genetic , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Dermatitis, Contact/drug therapy , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Endothelium, Lymphatic/drug effects , Endothelium, Lymphatic/metabolism , Endothelium, Lymphatic/pathology , Intercellular Adhesion Molecule-1/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Microscopy, Video , Protein Kinase Inhibitors/pharmacology , Radiation Chimera , Recombinant Fusion Proteins/metabolism , Skin/drug effects , Skin/immunology , Skin/metabolism , Skin/pathology , Up-Regulation/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics
10.
Blood ; 118(1): 205-15, 2011 Jul 07.
Article in English | MEDLINE | ID: mdl-21596851

ABSTRACT

Chemokines and adhesion molecules up-regulated in lymphatic endothelial cells (LECs) during tissue inflammation are thought to enhance dendritic cell (DC) migration to draining lymph nodes, but the in vivo control of this process is not well understood. We performed a transcriptional profiling analysis of LECs isolated from murine skin and found that inflammation induced by a contact hypersensitivity (CHS) response up-regulated the adhesion molecules ICAM-1 and VCAM-1 and inflammatory chemokines. Importantly, the lymphatic markers Prox-1, VEGFR3, and LYVE-1 were significantly down-regulated during CHS. By contrast, skin inflammation induced by complete Freund adjuvant induced a different pattern of chemokine and lymphatic marker gene expression and almost no ICAM-1 up-regulation in LECs. Fluorescein isothiocyanate painting experiments revealed that DC migration to draining lymph nodes was more strongly increased in complete Freund adjuvant-induced than in CHS-induced inflammation. Surprisingly, DC migration did not correlate with the induction of CCL21 and ICAM-1 protein in LECs. Although the requirement for CCR7 signaling became further pronounced during inflammation, CCR7-independent signals had an additional, albeit moderate, impact on enhancing DC migration. Collectively, these findings indicate that DC migration in response to inflammation is stimulus-specific, mainly CCR7-dependent, and overall only moderately enhanced by LEC-induced genes other than CCL21.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Endothelial Cells/immunology , Lymph Nodes/immunology , Animals , Chemokine CCL21/genetics , Chemokine CCL21/immunology , Chemokine CCL21/metabolism , Dendritic Cells/cytology , Ear, External/immunology , Female , Gene Expression/immunology , Gene Expression Profiling , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Receptors, CCR7/metabolism , Up-Regulation/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
11.
PLoS Pathog ; 6(2): e1000755, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20140197

ABSTRACT

Neutrophils are rapidly and massively recruited to sites of microbial infection, where they can influence the recruitment of dendritic cells. Here, we have analyzed the role of neutrophil released chemokines in the early recruitment of dendritic cells (DCs) in an experimental model of Leishmania major infection. We show in vitro, as well as during infection, that the parasite induced the expression of CCL3 selectively in neutrophils from L. major resistant mice. Neutrophil-secreted CCL3 was critical in chemotaxis of immature DCs, an effect lost upon CCL3 neutralisation. Depletion of neutrophils prior to infection, as well as pharmacological or genetic inhibition of CCL3, resulted in a significant decrease in DC recruitment at the site of parasite inoculation. Decreased DC recruitment in CCL3(-/-) mice was corrected by the transfer of wild type neutrophils at the time of infection. The early release of CCL3 by neutrophils was further shown to have a transient impact on the development of a protective immune response. Altogether, we identified a novel role for neutrophil-secreted CCL3 in the first wave of DC recruitment to the site of infection with L. major, suggesting that the selective release of neutrophil-secreted chemokines may regulate the development of immune response to pathogens.


Subject(s)
Chemokine CCL3/immunology , Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Leishmaniasis, Cutaneous/immunology , Neutrophils/immunology , Animals , Chemokine CCL3/metabolism , Dendritic Cells/metabolism , Female , Flow Cytometry , Leishmania major/immunology , Leishmaniasis, Cutaneous/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
Mol Biol Cell ; 20(4): 1132-40, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19109419

ABSTRACT

FXYD3 (Mat-8) proteins are regulators of Na,K-ATPase. In normal tissue, FXYD3 is mainly expressed in stomach and colon, but it is also overexpressed in cancer cells, suggesting a role in tumorogenesis. We show that FXYD3 silencing has no effect on cell proliferation but promotes cell apoptosis and prevents cell differentiation of human colon adenocarcinoma cells (Caco-2), which is reflected by a reduction in alkaline phosphatase and villin expression, a change in several other differentiation markers, and a decrease in transepithelial resistance. Inhibition of cell differentiation in FXYD3-deficient cells is accompanied by an increase in the apparent Na+ and K+ affinities of Na,K-ATPase, reflecting the absence of Na,K-pump regulation by FXYD3. In addition, we observe a decrease in the maximal Na,K-ATPase activity due to a decrease in its turnover number, which correlates with a change in Na,K-ATPase isozyme expression that is characteristic of cancer cells. Overall, our results suggest an important role of FXYD3 in cell differentiation of Caco-2 cells. One possibility is that FXYD3 silencing prevents proper regulation of Na,K-ATPase, which leads to perturbation of cellular Na+ and K+ homeostasis and changes in the expression of Na,K-ATPase isozymes, whose functional properties are incompatible with Caco-2 cell differentiation.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/enzymology , Intestines/cytology , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Apoptosis , Caco-2 Cells , Cell Proliferation , Down-Regulation , Gene Silencing , Humans , Isoenzymes/metabolism , Potassium/metabolism , RNA, Small Interfering/metabolism , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...