Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Expert Rev Mol Med ; 25: e16, 2023 05 03.
Article in English | MEDLINE | ID: mdl-37132370

ABSTRACT

This review discusses current research on acute paediatric leukaemia, the leukaemic bone marrow (BM) microenvironment and recently discovered therapeutic opportunities to target leukaemia-niche interactions. The tumour microenvironment plays an integral role in conferring treatment resistance to leukaemia cells, this poses as a key clinical challenge that hinders management of this disease. Here we focus on the role of the cell adhesion molecule N-cadherin (CDH2) within the malignant BM microenvironment and associated signalling pathways that may bear promise as therapeutic targets. Additionally, we discuss microenvironment-driven treatment resistance and relapse, and elaborate the role of CDH2-mediated cancer cell protection from chemotherapy. Finally, we review emerging therapeutic approaches that directly target CDH2-mediated adhesive interactions between the BM cells and leukaemia cells.


Subject(s)
Bone Marrow , Leukemia, Myeloid, Acute , Child , Humans , Bone Marrow/metabolism , Bone Marrow/pathology , Cadherins/genetics , Cadherins/metabolism , Cadherins/therapeutic use , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/drug therapy , Cell Adhesion , Tumor Microenvironment , Antigens, CD/metabolism , Antigens, CD/therapeutic use
2.
Front Cell Dev Biol ; 10: 866200, 2022.
Article in English | MEDLINE | ID: mdl-35309924

ABSTRACT

This review focuses on the cell adhesion molecule (CAM), known as neural (N)-cadherin (CDH2). The molecular basis of N-cadherin-mediated intercellular adhesion is discussed, as well as the intracellular signaling pathways regulated by this CAM. N-cadherin antagonists and agonists are then described, and several potential therapeutic applications of these intercellular adhesion modulators are considered. The usefulness of N-cadherin antagonists in treating fibrotic diseases and cancer, as well as manipulating vascular function are emphasized. Biomaterials incorporating N-cadherin modulators for tissue regeneration are also presented. N-cadherin antagonists and agonists have potential for broad utility in the treatment of numerous maladies.

3.
Biochem Biophys Res Commun ; 529(2): 162-168, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32703405

ABSTRACT

Glioblastoma multiforme (GBM) is a deadly type of brain cancer. There is a need to identify novel therapies for GBM as current treatments only marginally increase survival. Modelling the complexity of cancerous tissues using 3D bioprinted constructs serves as a novel approach for preclinical testing of anticancer drugs. A novel small molecule antagonist of the cell adhesion molecule, N-cadherin (NCAD), (S)-1-(3,4-Dichlorophenoxy)-3-(4-((S)-2-hydroxy-3-(4-methoxyphenoxy)propylamino)piperidin-1-yl)propan-2-ol has shown promise as an anticancer agent. This study investigated the influence of this antagonist on GBM cells bioprinted with astrocytes into 3D constructs. The NCAD antagonist prevented spheroid formation and induced cell death in the 3D model. This is the first demonstration that an NCAD antagonist can cause GBM cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Bioprinting/instrumentation , Brain Neoplasms/drug therapy , Cadherins/antagonists & inhibitors , Coculture Techniques/instrumentation , Glioblastoma/drug therapy , Antigens, CD , Cell Death/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor/instrumentation , Equipment Design , Humans
4.
FASEB Bioadv ; 2(6): 339-353, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32617520

ABSTRACT

N-cadherin is a homophilic cell-cell adhesion molecule that plays a critical role in maintaining vascular stability and modulating endothelial barrier permeability. Pre-clinical studies have shown that the N-cadherin antagonist peptide, ADH-1, increases the permeability of tumor-associated vasculature thereby increasing anti-cancer drug delivery to tumors and enhancing tumor response. Small molecule library screens have identified a novel compound, LCRF-0006, that is a mimetic of the classical cadherin His-Ala-Val sequence-containing region of ADH-1. Here, we evaluated the vascular permeability-enhancing and anti-cancer properties of LCRF-0006 using in vitro vascular disruption and cell apoptosis assays, and a well-established pre-clinical model (C57BL/KaLwRij/5TGM1) of the hematological cancer multiple myeloma (MM). We found that LCRF-0006 disrupted endothelial cell junctions in a rapid, transient and reversible manner, and increased vascular permeability in vitro and at sites of MM tumor in vivo. Notably, LCRF-0006 synergistically increased the in vivo anti-MM tumor response to low-dose bortezomib, a frontline anti-MM agent, leading to regression of disease in 100% of mice. Moreover, LCRF-0006 and bortezomib synergistically induced 5TGM1 MM tumor cell apoptosis in vitro. Our findings demonstrate the potential clinical utility of LCRF-0006 to significantly increase bortezomib effectiveness and enhance the depth of tumor response in patients with MM.

5.
Br J Haematol ; 171(3): 387-99, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26194766

ABSTRACT

Elevated expression of the cell adhesion molecule N-cadherin (cadherin 2, type 1, N-cadherin (neuronal); CDH2) is associated with poor prognosis in newly-diagnosed multiple myeloma (MM) patients. In this study, we investigated whether targeting of N-cadherin represents a potential treatment for the ~50% of MM patients with elevated N-cadherin. Initially, we stably knocked-down N-cadherin in the mouse MM plasma cell (PC) line 5TGM1 to assess the functional role of N-cadherin in MM pathogenesis. When compared with 5TGM1-scramble-shRNA cells, 5TGM1-Cdh2-shRNA cells had significantly reduced adhesion to bone marrow endothelial cells. However, N-cadherin knock-down did not affect 5TGM1 cell proliferation or adhesion to bone marrow stromal cells. In the C57BL/KaLwRij murine MM model, mice intravenously inoculated with 5TGM1-Cdh2-shRNA cells showed significantly decreased tumour burden after 4 weeks, compared with animals bearing 5TGM1-scramble-shRNA cells. Finally, the N-cadherin antagonist ADH-1 had no effect on tumour burden in the established disease setting, whereas up-front ADH-1 treatment resulted in significantly reduced tumour burden after 4 weeks. Our findings demonstrate that N-cadherin may play a key role in the extravasation of circulating MM PCs promoting bone marrow homing. Moreover, these studies suggest that N-cadherin may represent a viable therapeutic target to prevent the dissemination of MM PCs and delay MM disease progression.


Subject(s)
Cadherins/antagonists & inhibitors , Cell Proliferation/drug effects , Multiple Myeloma/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Oligopeptides/pharmacology , Peptides, Cyclic/pharmacology , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Knockdown Techniques , Humans , Mice , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism
6.
Philos Trans R Soc Lond B Biol Sci ; 370(1661): 20140039, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25533096

ABSTRACT

The cell adhesion molecule (CAM), N-cadherin, has emerged as an important oncology therapeutic target. N-cadherin is a transmembrane glycoprotein mediating the formation and structural integrity of blood vessels. Its expression has also been documented in numerous types of poorly differentiated tumours. This CAM is involved in regulating the proliferation, survival, invasiveness and metastasis of cancer cells. Disruption of N-cadherin homophilic intercellular interactions using peptide or small molecule antagonists is a promising novel strategy for anti-cancer therapies. This review discusses: the discovery of N-cadherin, the mechanism by which N-cadherin promotes cell adhesion, the role of N-cadherin in blood vessel formation and maintenance, participation of N-cadherin in cancer progression, the different types of N-cadherin antagonists and the use of N-cadherin antagonists as anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Cadherins/antagonists & inhibitors , Animals , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/genetics , Neoplasms/metabolism
7.
Prog Histochem Cytochem ; 48(3): 103-40, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24100070

ABSTRACT

Islets form in the pancreas after the first endocrine cells have arisen as either single cells or small cell clusters in the epithelial cords. These cords constitute the developing pancreas in one of its earliest recognizable stages. Islet formation begins at the time the cords transform into a branching ductal system, continues while the ductal system expands, and finally stops before the exocrine tissue of ducts and acini reaches its final expansion. Thus, islets continuously arise from founder cells located in the branching and ramifying ducts. Islets arising from proximal duct cells locate between the exocrine lobules, develop strong autonomic and sensory innervations, and pass their blood to efferent veins (insulo-venous efferent system). Islets arising from cells of more distal ducts locate within the exocrine lobules, respond to nerve impulses ending at neighbouring blood vessels, and pass their blood to the surrounding acini (insulo-acinar portal system). Consequently, the section of the ductal system from which an islet arises determines to a large extent its future neighbouring tissue, architecture, properties, and functions. We note that islets interlobular in position are frequently found in rodents (rats and mice), whereas intralobularly-located, peripheral duct islets prevail in humans and cattle. Also, we expound on bovine foetal Laguesse islets as a prominent foetal type of type 1 interlobular neuro-insular complexes, similar to neuro-insular associations frequently found in rodents. Finally, we consider the probable physiological and pathophysiological implications of the different islet positions within and between species.


Subject(s)
Islets of Langerhans/embryology , Islets of Langerhans/growth & development , Morphogenesis , Pancreatic Ducts/embryology , Pancreatic Ducts/growth & development , Animals , Cattle , Humans , Islets of Langerhans/cytology , Mice , Models, Biological , Pancreatic Ducts/cytology , Rats , Species Specificity
8.
Cell Tissue Res ; 348(2): 309-13, 2012 May.
Article in English | MEDLINE | ID: mdl-22281688

ABSTRACT

This article describes over 20 years of research on antagonists of the cell adhesion molecule, N-cadherin. Four types of antagonists are discussed: synthetic linear peptides, synthetic cyclic peptides, non-peptidyl peptidomimetics of the disulfide linked cyclic peptide N-Ac-CHAVC-NH(2) and monoclonal antibodies directed against the N-cadherin ectodomain. The biological activities of these antagonists are also discussed. In particular, the ability of N-cadherin antagonists to act as anti-cancer drugs is considered.


Subject(s)
Cadherins/antagonists & inhibitors , Peptides/pharmacology , Amino Acid Sequence , Humans , Peptides/chemistry
9.
J Vasc Surg ; 52(5): 1301-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20630685

ABSTRACT

OBJECTIVES: Inhibition of vascular smooth muscle cell (VSMC) migration is a potential strategy for reducing intimal thickening during in-stent restenosis and vein graft failure. In this study, we examined the effect of disrupting the function of the VSMC adhesion molecule, N-cadherin, using antagonists, neutralizing antibodies, and a dominant negative, on VSMC migration and intimal thickening. Migration was assessed by the scratch-wound assay of human saphenous vein VSMCs and in a human saphenous vein ex vivo organ culture model of intimal thickening. RESULTS: Inhibition of cadherin function using a pan-cadherin antagonist, significantly reduced migration by 53%±8% compared with the control peptide (n=3; P<.05). Furthermore, inhibition of N-cadherin function with an N-cadherin antagonist, neutralizing antibodies, and adenoviral expression of dominant negative N-cadherin (RAd dn-N-cadherin), significantly reduced migration by 31%±2%, 23%±1% and 32%±7% compared with controls, respectively (n=3; P<.05). Inhibition of cadherin function significantly increased apoptosis by between 1.5- and 3.3-fold at the wound edge. In an ex vivo model of intimal thickening, inhibition of N-cadherin function by infection of human saphenous vein segments with RAd dn-N-cadherin significantly reduced VSMC migration by 55% and increased VSMC apoptosis by 2.7-fold. As a result, intimal thickening was significantly suppressed by 54%±14%. Importantly, there was no detrimental effect of dn-N-cadherin on endothelial coverage; in fact, it was significantly increased, as was survival of cultured human saphenous vein endothelial cells. CONCLUSIONS: Under the condition of this study, cell-cell adhesion mediated by N-cadherin regulates VSMC migration via modulation of viability. Interestingly, inhibition of N-cadherin function significantly retards intimal thickening via inhibition of VSMC migration and promotion of endothelial cell survival. We suggest that disruption of N-cadherin-mediated cell-cell contacts is a potential strategy for reducing VSMC migration and intimal thickening.


Subject(s)
Antibodies/pharmacology , Apoptosis/drug effects , Cadherins/antagonists & inhibitors , Cell Movement/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Peptides, Cyclic/pharmacology , Tunica Intima/drug effects , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Survival/drug effects , Cells, Cultured , Humans , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Organ Culture Techniques , Saphenous Vein/drug effects , Saphenous Vein/metabolism , Saphenous Vein/pathology , Time Factors , Transfection , Tunica Intima/metabolism , Tunica Intima/pathology
10.
Eur J Pharmacol ; 625(1-3): 195-8, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19836380

ABSTRACT

The cell adhesion molecules N-, VE- and OB-cadherin have been implicated as regulators of tumor growth and metastasis. We discuss evidence that N- and VE-cadherin play a key role in promoting blood vessel formation and stability, processes which are essential for tumor growth. Secondly, we describe the potential involvement of N- and OB-cadherin in the metastatic process. Finally, studies concerning the effects of the N-cadherin antagonist designated ADH-1 on tumor growth are presented. Collectively, these observations suggest that antagonists of N-, VE- and OB-cadherin would be useful as anti-cancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Cadherins/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Cadherins/metabolism , Cell Adhesion/drug effects , Clinical Trials as Topic , Drug Delivery Systems , Humans , Neoplasm Metastasis/pathology , Neoplasms/physiopathology , Peptides, Cyclic/pharmacology
11.
Peptides ; 30(8): 1539-47, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19465078

ABSTRACT

E- and N-cadherin are related calcium-dependent cell adhesion molecules that exert an influence over multiple biological and disease processes. Antagonists of these cadherins can therefore be envisaged as therapeutically useful drugs. We have used phage display technology to discover such antagonists. A peptide phage library was screened against a chimeric protein composed of the human E-cadherin ectodomain fused to the Fc fragment of human immunoglobulin G1 (E-cad/Fc). All of the phage clones that were isolated also bound a chimeric protein composed of the human N-cadherin ectodomain fused to the Fc fragment of human immunoglobulin G1 (N-cad/Fc). A peptide displayed by several of the isolated phage clones was synthesized (H-SWELYYPLRANL-NH2) and found to bind both E- and N-cad/Fc chimeric proteins with affinities (K(D)) of 9.4 microM and 323 nM, respectively, as judged by surface plasmon resonance spectroscopy. This peptide was also capable of blocking the aggregation of E- and N-cad/Fc chimeric protein-coated beads, as well as the aggregation of MCF-7 and MDA-MB435 human breast cancer cells (these cells express E- and N-cadherin, respectively). Finally, we showed that the peptide disrupted MCF-7 and MDA-MB435 cell monolayers. The peptide, H-SWELYYPLRANL-NH(2) thus proved to be a biologically active, dual E- and N-cadherin antagonist. Such an antagonist has application in a wide variety of biological contexts.


Subject(s)
Cadherins/antagonists & inhibitors , Peptides/metabolism , Amino Acid Sequence , Cadherins/metabolism , Cell Aggregation/drug effects , Cell Line, Tumor , Humans , Molecular Sequence Data , Peptide Library , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Protein Binding
12.
Peptides ; 29(11): 1853-61, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18655820

ABSTRACT

The cell adhesion molecule, N-cadherin plays a pivotal role in many biological and disease processes. Drugs that modulate N-cadherin function should therefore be useful therapeutic agents. We have used phage display technology to identify amino acid sequences capable of binding to N-cadherin. All of these sequences harbor a Trp residue in the second position from the N-terminus. A synthetic linear peptide containing one of these sequences, H-SWTLYTPSGQSK-NH(2) was found to bind a chimeric protein composed of the N-cadherin ectodomain fused to the immunoglobulin G1 Fc fragment with an affinity (K(D)) of 10.7microM, as determined by surface plasmon resonance. It also blocked the aggregation of beads coated with this chimeric protein. Furthermore, this peptide disrupted adhesion and tube formation by N-cadherin-expressing human umbilical vein endothelial cells in vitro. These observations suggest that N-cadherin antagonists have the potential of serving as anti-angiogenic agents. The peptide, H-SWTLYTPSGQSK-NH(2) should prove useful for studies designed to evaluate N-cadherin function in various biological processes.


Subject(s)
Cadherins/antagonists & inhibitors , Oligopeptides/isolation & purification , Oligopeptides/pharmacology , Peptides/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Humans , Immunoglobulin Fc Fragments , Peptide Library , Surface Plasmon Resonance
13.
Arterioscler Thromb Vasc Biol ; 25(5): 982-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15774907

ABSTRACT

OBJECTIVE: Vascular smooth muscle cell (VSMC) apoptosis is thought to contribute to atherosclerotic plaque instability. Cadherin mediates calcium-dependent homophilic cell-cell contact. We studied the role of N-cadherin in VSMC apoptosis. METHODS AND RESULTS: Human saphenous vein VSMCs were grown in agarose-coated wells to allow cadherin-mediated aggregate formation. Cell death and apoptosis were determined after disruption of cadherins using several approaches (n> or =3 per approach). Calcium removal from culture medium or addition of nonspecific cadherin antagonist peptides significantly decreased aggregate formation and increased cell death by apoptosis (34+/-6% versus 75+/-1% and 19+/-1% versus 40+/-5%, respectively; P<0.05). Specific inhibition of N-cadherin using antagonists and neutralizing antibodies similarly increased apoptosis. Supporting this, overexpression of full-length N-cadherin significantly reduced VSMC apoptosis from 44+/-10% to 20+/-3% (P<0.05), whereas abolishing N-cadherin expression by overexpression of a dominant-negative N-cadherin significantly, even in the presence of cell-matrix contacts, increased apoptosis from 9+/-2% to 50+/-1% (P<0.05). Interestingly, cell-cell contacts provided a similar degree of protection from apoptosis to cell-matrix contacts. Finally, N-cadherin-mediated cell-cell contacts initiated anti-apoptotic signaling by increasing Akt and Bad phosphorylation. CONCLUSIONS: Our results indicate that VSMC survival is dependent on N-cadherin-mediated cell-cell contacts, which could be important in the context of plaque instability.


Subject(s)
Atherosclerosis/pathology , Cadherins/metabolism , Cell Communication/physiology , Muscle, Smooth, Vascular/cytology , Saphenous Vein/cytology , Antibodies/pharmacology , Apoptosis/physiology , Atherosclerosis/metabolism , Cadherins/genetics , Cadherins/immunology , Calcium/metabolism , Calcium/pharmacology , Cell Aggregation/physiology , Cell Communication/drug effects , Cell Survival/physiology , Cells, Cultured , Extracellular Matrix/physiology , Gene Expression , Humans , Muscle, Smooth, Vascular/metabolism , Signal Transduction/physiology
14.
Exp Cell Res ; 294(2): 366-78, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15023527

ABSTRACT

Cadherins are a family of transmembrane glycoproteins mediating calcium-dependent, homophilic cell-cell adhesion. In addition, these molecules are involved in signaling events, regulating such processes as cell motility, proliferation, and apoptosis. Members of the cadherin subfamily, called either classical or type I cadherins, contain a highly conserved sequence at their homophilic binding site consisting of the three amino acids--histidine-alanine-valine (HAV). Previous studies have shown that peptides containing the HAV motif inhibit cadherin-dependent events such as cell aggregation, compaction, and neurite outgrowth. We report here that a cyclic peptide, N-Ac-CHAVC-NH2 can perturb cadherin-mediated endothelial cell interactions, resulting in a progressive apoptotic cell death. This effect depends on cell density, as it is only observed when dense cultures are treated with the peptide. Adherens junction (AJ)-associated cadherin and catenins are differentially affected by the N-Ac-CHAVC-NH2 treatment, as judged by double immunofluorescence labeling followed by immunofluorescence-ratio imaging. However, cell-cell adhesions are largely retained during the first few hours after addition of the peptide. It was also observed that following treatment, actin filaments partially lose their plasma membrane anchorage at AJs and translocate towards the cell center. Interestingly, addition of basic fibroblast growth factor to confluent, peptide-treated, endothelial cell cultures, completely blocks apoptosis and the inhibitory peptide reduce the phosphorylation of the FGF receptor target protein FRS2, suggesting that the peptide exerts its effect by inhibiting cadherin-mediated activation of fibroblast growth factor receptor signaling. We propose that cadherin-mediated signaling is essential for maintaining viability of confluent endothelial cells, and that its perturbation by N-Ac-CHAVC-NH2 drives these cells to apoptosis.


Subject(s)
Apoptosis/physiology , Cadherins/metabolism , Cell Adhesion/physiology , Cell Communication/physiology , Endothelial Cells/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Amino Acid Motifs/physiology , Amino Acid Sequence/physiology , Animals , Apoptosis/drug effects , Cadherins/drug effects , Cattle , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/pharmacology , Fluorescent Antibody Technique , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mice , Peptides/pharmacology , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Phosphorylation/drug effects , Receptors, Fibroblast Growth Factor/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
15.
Inflammation ; 26(4): 193-8, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12184633

ABSTRACT

The molecular mechanisms by which the tight junction integral membrane protein, occludin promotes cell adhesion and establishes an endothelial monolayer permeability barrier have not been elucidated. In particular, the amino acid sequences of the occludin cell adhesion recognition (CAR) sites have not been determined. Here we demonstrate that a cyclic peptide containing the sequence LYHY, which is found in the second extracellular domain of occludins in all mammalian species, inhibits the establishment of endothelial cell barriers in vitro and in vivo. This cyclic peptide also prevents the aggregation of fibroblasts stably transfected with cDNA encoding occludin. The data suggest that the LYHY motif is an occludin CAR sequence.


Subject(s)
Endothelium, Vascular/physiology , Membrane Proteins/physiology , Amino Acid Sequence/genetics , Animals , Capillary Permeability/physiology , Cell Adhesion/physiology , Cell Aggregation , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fluorescent Antibody Technique , Humans , Membrane Proteins/chemistry , Microcirculation/drug effects , Occludin , Peptide Fragments/pharmacology , Protein Structure, Tertiary/genetics , Rats , Rats, Sprague-Dawley
16.
Mol Membr Biol ; 19(2): 75-80, 2002.
Article in English | MEDLINE | ID: mdl-12126233

ABSTRACT

This review focuses on the three known plasma membrane components of adherens junctions: E-cadherin, nectin-2 and vezatin. The structures of these three components are discussed, with particular emphasis on the molecular mechanisms by which E-cadherin and nectin-2 promote cell adhesion.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Humans , Nectins
17.
Mol Reprod Dev ; 61(2): 142-54, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11803548

ABSTRACT

Classical and atypical cadherins mediate calcium-dependent cell adhesion and play an important role in morphogenetic processes. We have shown, previously, N- and E-cadherin expression in the rat ovary. This expression, however, was not associated with specific follicle-restructuring events such as antrum formation and segregation of mural from cumulus granulosa cells suggesting that other cadherins may serve this function. In this study, RT-PCR and immunostaining techniques showed that three other cadherins are expressed throughout prepubertal ovarian development in the rat: one classical (P-) cadherin, and two atypical (K- and OB-) cadherins. RT-PCR analysis of isolated ovarian tissue compartments (granulosa cells and the residual ovarian tissue) agreed with the immunostaining results. Immunostaining showed P- and K-cadherin expression by granulosa, as well as thecal/interstitial cells, and also in oocytes of primordial follicles. P-cadherin expression was absent in oocytes of follicles in later stages of development compared to K-cadherin, which was found in oocytes at all stages of folliculogenesis. P-, K-, and OB-cadherin were expressed by the ovarian surface epithelial cells of neonatal animals but only P- and OB-cadherin expression were maintained in these cells in 25 day-old animals. Cellular OB-cadherin staining was absent in follicles at all stages of development and its expression was restricted to the ovarian hilar region and portions of the stroma. In summary, cadherin expression and distribution profiles changed during ovarian growth and folliculogenesis suggesting a role for cadherins in organizational and morphogenetic processes within the developing rat ovary.


Subject(s)
Cadherins/metabolism , Ovary/growth & development , Ovary/metabolism , Animals , Cadherins/genetics , Female , Immunoblotting , Immunohistochemistry , Ovary/cytology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL